1
|
Costa B, Gouveia MJ, Vale N. Oxidative Stress Induced by Antivirals: Implications for Adverse Outcomes During Pregnancy and in Newborns. Antioxidants (Basel) 2024; 13:1518. [PMID: 39765846 PMCID: PMC11727424 DOI: 10.3390/antiox13121518] [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: 09/27/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/15/2025] Open
Abstract
Oxidative stress plays a critical role in various physiological and pathological processes, particularly during pregnancy, where it can significantly affect maternal and fetal health. In the context of viral infections, such as those caused by Human Immunodeficiency Virus (HIV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), oxidative stress may exacerbate complications by disrupting cellular function and immune responses. Antiviral drugs, while essential in managing these infections, can also contribute to oxidative stress, potentially impacting both the mother and the developing fetus. Understanding the mechanisms by which antivirals can contribute to oxidative stress and examination of pharmacokinetic changes during pregnancy that influence drug metabolism is essential. Some research indicates that antiretroviral drugs can induce oxidative stress and mitochondrial dysfunction during pregnancy, while other studies suggest that their use is generally safe. Therefore, concerns about long-term health effects persist. This review delves into the complex interplay between oxidative stress, antioxidant defenses, and antiviral therapies, focusing on strategies to mitigate potential oxidative damage. By addressing gaps in our understanding, we highlight the importance of balancing antiviral efficacy with the risks of oxidative stress. Moreover, we advocate for further research to develop safer, more effective therapeutic approaches during pregnancy. Understanding these dynamics is essential for optimizing health outcomes for both mother and fetus in the context of viral infections during pregnancy.
Collapse
Affiliation(s)
- Bárbara Costa
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Centre for Parasite Biology and Immunology, Department of Infectious Diseases, National Health Institute Dr. Ricardo Jorge, 4000-055 Porto, Portugal;
| | - Maria João Gouveia
- Centre for Parasite Biology and Immunology, Department of Infectious Diseases, National Health Institute Dr. Ricardo Jorge, 4000-055 Porto, Portugal;
- Center for the Study in Animal Science (CECA/ICETA), University of Porto, 4051-401 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
2
|
Xu Q, Zhang C, Lu J, Qian H, Wang X, Guo W, Cheng H. Azithromycin induces liver injury in mice by targeting the AMPK/Nrf2 pathway. Immunopharmacol Immunotoxicol 2024; 46:850-860. [PMID: 39406691 DOI: 10.1080/08923973.2024.2415115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 10/05/2024] [Indexed: 11/19/2024]
Abstract
BACKGROUND Azithromycin is an antibacterial and anti-inflammatory drug widely used for the treatment of various diseases, including those caused by atypical pathogens, bacterial or viral infections, chronic sinusitis, and bronchial asthma, particularly in pediatric patients. However, concerns have emerged regarding its hepatotoxicity and its precise mechanism of action remains unclear. OBJECTIVE To investigate the molecular mechanisms responsible for azithromycin-induced acute liver injury to advance our understanding of the progression and pathogenesis of antibiotic-induced liver damage, and to improve prevention and treatment strategies. MATERIALS AND METHODS C57BL/6 mice, Nrf2-/- mice, and primary hepatocytes were used. Primary hepatocytes from mice were isolated using a two-step perfusion method and cultured in vitro via the 'sandwich' culture model. RESULTS The exposure to azithromycin resulted in increased apoptosis and reactive oxygen species (ROS) levels. In mouse models, intraperitoneal administration of azithromycin at varying concentrations and time points substantially induced hepatic disarray, swelling, and dysfunction. Azithromycin markedly upregulated the mRNA and protein levels of phosphorylated adenosine-activated protein kinase (AMPK) while downregulating nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase 1 (HO-1), and NADPH: quinone oxidoreductase 1 (NQO-1). Moreover, HO-1 and NQO-1 protein levels remained largely unaffected in primary hepatocytes co-cultured with azithromycin in Nrf2-/- mice. CONCLUSIONS Our findings suggest that azithromycin-induced acute liver injury is mediated by suppression of Nrf2 activation and ROS production. This sheds light on the potential mechanisms involved in azithromycin-induced liver damage, underscoring the importance of exploring targeted interventions to mitigate the hepatotoxic effects.
Collapse
Affiliation(s)
- Qixiang Xu
- School of Pharmacology, Wannan Medical College, Wuhu, China
| | - Cuifeng Zhang
- Anesthesia Laboratory and Training Center, School of Anesthesiology, Wannan Medical College, Wuhu, China
- Perioperative Monitoring and Prognostic Technology Research and Development Center of Wuhu, Wuhu, China
| | - Jingwen Lu
- Anesthesia Laboratory and Training Center, School of Anesthesiology, Wannan Medical College, Wuhu, China
- Perioperative Monitoring and Prognostic Technology Research and Development Center of Wuhu, Wuhu, China
| | - Haiyi Qian
- School of Pharmacology, Wannan Medical College, Wuhu, China
| | - Xiaodong Wang
- Anesthesia Laboratory and Training Center, School of Anesthesiology, Wannan Medical College, Wuhu, China
- Perioperative Monitoring and Prognostic Technology Research and Development Center of Wuhu, Wuhu, China
| | - Wenjun Guo
- Perioperative Monitoring and Prognostic Technology Research and Development Center of Wuhu, Wuhu, China
- Department of Anesthesiology, Yijishan Hospital, The First Affiliated of Wannan Medical College, Wuhu, China
| | - Huixian Cheng
- Perioperative Monitoring and Prognostic Technology Research and Development Center of Wuhu, Wuhu, China
- Department of Anesthesiology, Yijishan Hospital, The First Affiliated of Wannan Medical College, Wuhu, China
| |
Collapse
|
3
|
Zhao Y, Park JY, Yang D, Zhang M. A computational framework to in silico screen for drug-induced hepatocellular toxicity. Toxicol Sci 2024; 201:14-25. [PMID: 38902949 PMCID: PMC11347774 DOI: 10.1093/toxsci/kfae078] [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: 06/22/2024] Open
Abstract
Drug-induced liver injury (DILI) is the most common trigger for acute liver failure and the leading cause of attrition in drug development. In this study, we developed an in silico framework to screen drug-induced hepatocellular toxicity (INSIGHT) by integrating the post-treatment transcriptomic data from both rodent models and primary human hepatocytes. We first built an early prediction model using logistic regression with elastic net regularization for 123 compounds and established the INSIGHT framework that can screen for drug-induced hepatotoxicity. The 235 signature genes identified by INSIGHT were involved in metabolism, bile acid synthesis, and stress response pathways. Applying the INSIGHT to an independent transcriptomic dataset treated by 185 compounds predicted that 27 compounds show a high DILI risk, including zoxazolamine and emetine. Further integration with cell image data revealed that predicted compounds with high DILI risk can induce abnormal morphological changes in the endoplasmic reticulum and mitochondrion. Clustering analysis of the treatment-induced transcriptomic changes delineated distinct DILI mechanisms induced by these compounds. Our study presents a computational framework for a mechanistic understanding of long-term liver injury and the prospective prediction of DILI risk.
Collapse
Affiliation(s)
- Yueshan Zhao
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Ji Youn Park
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Da Yang
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, United States
- UPMC Hillman Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15261, United States
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15261, United States
| | - Min Zhang
- Department of Pharmaceutical Sciences, Center for Pharmacogenetics, University of Pittsburgh, Pittsburgh, PA 15261, United States
| |
Collapse
|
4
|
Garcia-Fossa F, Moraes-Lacerda T, Rodrigues-da-Silva M, Diaz-Rohrer B, Singh S, Carpenter AE, Cimini BA, de Jesus MB. Live Cell Painting: image-based profiling in live cells using Acridine Orange. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.28.610144. [PMID: 39257795 PMCID: PMC11383656 DOI: 10.1101/2024.08.28.610144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Image-based profiling has been used to analyze cell health, drug mechanism of action, CRISPR-edited cells, and overall cytotoxicity. Cell Painting is a broadly used image-based assay that uses morphological features to capture how cells respond to treatments. However, this method requires cell fixation for staining, which prevents examining live cells. To address this limitation, here we present Live Cell Painting (LCP), a high-content method based on Acridine orange, a metachromatic dye that labels different organelles and cellular structures. We began by showing that LCP can be applied to follow acidic vesicle redistribution of cells exposed to acidic vesicles inhibitors. Next, we show that LCP can identify subtle changes in cells exposed to silver nanoparticles that are not detected by techniques such as MTT assay. In drug treatments, LCP was helpful in assessing the dose-response relationship and creating profiles that allow clustering of drugs that cause liver injury. Here, we present an affordable and easy-to-use image-based assay capable of assessing overall cell health and showing promise for use in various applications such as assessing drugs and nanoparticles. We envisage the use of Live Cell Painting as an initial screening of overall cell health while providing insights into new biological questions.
Collapse
|
5
|
Liu J, Du H, Huang L, Xie W, Liu K, Zhang X, Chen S, Zhang Y, Li D, Pan H. AI-Powered Microfluidics: Shaping the Future of Phenotypic Drug Discovery. ACS APPLIED MATERIALS & INTERFACES 2024; 16:38832-38851. [PMID: 39016521 DOI: 10.1021/acsami.4c07665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/18/2024]
Abstract
Phenotypic drug discovery (PDD), which involves harnessing biological systems directly to uncover effective drugs, has undergone a resurgence in recent years. The rapid advancement of artificial intelligence (AI) over the past few years presents numerous opportunities for augmenting phenotypic drug screening on microfluidic platforms, leveraging its predictive capabilities, data analysis, efficient data processing, etc. Microfluidics coupled with AI is poised to revolutionize the landscape of phenotypic drug discovery. By integrating advanced microfluidic platforms with AI algorithms, researchers can rapidly screen large libraries of compounds, identify novel drug candidates, and elucidate complex biological pathways with unprecedented speed and efficiency. This review provides an overview of recent advances and challenges in AI-based microfluidics and their applications in drug discovery. We discuss the synergistic combination of microfluidic systems for high-throughput screening and AI-driven analysis for phenotype characterization, drug-target interactions, and predictive modeling. In addition, we highlight the potential of AI-powered microfluidics to achieve an automated drug screening system. Overall, AI-powered microfluidics represents a promising approach to shaping the future of phenotypic drug discovery by enabling rapid, cost-effective, and accurate identification of therapeutically relevant compounds.
Collapse
Affiliation(s)
- Junchi Liu
- Department of Anesthesiology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130012, China
| | - Hanze Du
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Lei Huang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Wangni Xie
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Kexuan Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Xue Zhang
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Shi Chen
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| | - Yuan Zhang
- Department of Anesthesiology, The First Hospital of Jilin University, 71 Xinmin Street, Changchun 130012, China
| | - Daowei Li
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, 1500 Qinghua Road, Changchun 130012, China
| | - Hui Pan
- Department of Endocrinology, Key Laboratory of Endocrinology of National Health Commission, Translation Medicine Centre, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
6
|
Wu W, Han Y, Niu B, Yang B, Liu R, Fang X, Chen H, Xiao S, Farag MA, Zheng S, Xiao J, Chen H, Gao H. Recent advances in Zizania latifolia: A comprehensive review on phytochemical, health benefits and applications that maximize its value. Crit Rev Food Sci Nutr 2024; 64:7535-7549. [PMID: 36908217 DOI: 10.1080/10408398.2023.2186125] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Zizania latifolia is an aquatic and medicinal plant with a long history of development in China and the East Asian region. The smut fungus "Ustilago esculenta" parasitizes Z. latifolia and induces culm expansion to form a vegetable named Jiaobai, which has a unique taste and nutritional attributes. However, the postharvest quality of water bamboo shoots is still a big challenge for farmers and merchants. This paper traced the origin, development process, and morphological characteristics of Z. latifolia. Subsequently, the compilation of the primary nutrients and bioactive substances are presented in context to their effects on ecology a postharvest storage and preservation methods. Furthermore, the industrial, environmental, and material science applications of Z. latifolia in the fields of industry were discussed. Finally, the primary objective of the review proposes future directions for research to support the development of Z. latifolia industry and aid in maximizing its value. To sum up, Z. latifolia, aside from its potential as material it can be utilized to make different productions and improve the existing applications. This paper provides an emerging strategy for researchers undertaking Z. latifolia.
Collapse
Affiliation(s)
- Weijie Wu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key laboratory of postharvest handling of fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Key laboratory of fruits and vegetables postharvest and processing technology research of Zhejiang province, Key laboratory of postharvest preservation and processing of fruits and vegetables, China National Light Industry, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Yanchao Han
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key laboratory of postharvest handling of fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Key laboratory of fruits and vegetables postharvest and processing technology research of Zhejiang province, Key laboratory of postharvest preservation and processing of fruits and vegetables, China National Light Industry, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Ben Niu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key laboratory of postharvest handling of fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Key laboratory of fruits and vegetables postharvest and processing technology research of Zhejiang province, Key laboratory of postharvest preservation and processing of fruits and vegetables, China National Light Industry, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Baiqi Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key laboratory of postharvest handling of fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Key laboratory of fruits and vegetables postharvest and processing technology research of Zhejiang province, Key laboratory of postharvest preservation and processing of fruits and vegetables, China National Light Industry, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Ruiling Liu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key laboratory of postharvest handling of fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Key laboratory of fruits and vegetables postharvest and processing technology research of Zhejiang province, Key laboratory of postharvest preservation and processing of fruits and vegetables, China National Light Industry, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Xiangjun Fang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key laboratory of postharvest handling of fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Key laboratory of fruits and vegetables postharvest and processing technology research of Zhejiang province, Key laboratory of postharvest preservation and processing of fruits and vegetables, China National Light Industry, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Huizhi Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key laboratory of postharvest handling of fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Key laboratory of fruits and vegetables postharvest and processing technology research of Zhejiang province, Key laboratory of postharvest preservation and processing of fruits and vegetables, China National Light Industry, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Shangyue Xiao
- Department of Analytical Chemistry and Food Science, University of Vigo, Vigo, Spain
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Cairo, Egypt
| | - Shiqi Zheng
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key laboratory of postharvest handling of fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Key laboratory of fruits and vegetables postharvest and processing technology research of Zhejiang province, Key laboratory of postharvest preservation and processing of fruits and vegetables, China National Light Industry, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Jianbo Xiao
- Department of Analytical Chemistry and Food Science, University of Vigo, Vigo, Spain
| | - Hangjun Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key laboratory of postharvest handling of fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Key laboratory of fruits and vegetables postharvest and processing technology research of Zhejiang province, Key laboratory of postharvest preservation and processing of fruits and vegetables, China National Light Industry, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Haiyan Gao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Key laboratory of postharvest handling of fruits, Ministry of Agriculture and Rural Affairs, Key Laboratory of Postharvest Preservation and Processing of Vegetables (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, Key laboratory of fruits and vegetables postharvest and processing technology research of Zhejiang province, Key laboratory of postharvest preservation and processing of fruits and vegetables, China National Light Industry, Food Science Institute, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| |
Collapse
|
7
|
Wojtunik-Kulesza K, Rudkowska M, Klimek K, Agacka-Mołdoch M, Mołdoch J, Michalak A. Expanding Knowledge about the Influence of Citral on Cognitive Functions-In Vitro, In Vivo and Ex Vivo Studies. Int J Mol Sci 2024; 25:6866. [PMID: 38999975 PMCID: PMC11241199 DOI: 10.3390/ijms25136866] [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: 05/13/2024] [Revised: 06/14/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Citral, a common monoterpene found in numerous plants, is an interesting compound that has been shown to have various biological activities. Although it is widely distributed in nature and there are many studies presenting its biological activities, its anti-neurodegenerative activity, especially under in vivo conditions, is very poorly understood. Thus, this paper aimed to deepen knowledge about citral activity towards factors and symptoms of neurodegeneration. To accomplish this, several comprehensive tests were conducted, including the estimation of butyrylcholinesterase inhibition, the evaluation of hepatotoxicity and the detection of oxidative stress and lipid peroxidation in vitro, as well as an in vivo behavioral assessment using mice models. Additionally, ex vivo determination of level of the compound in the brain and blood of a tested animal was undertaken. The results obtained revealed that citral is able to inhibit butyrylcholinesterase activity and protect hepatic cells against oxidative stress and lipid peroxidation in vitro. Moreover, behavioral tests in vivo indicated that citral (50 mg/kg) improves memory processes associated with acquisition (passive avoidance test), both in acute and subchronic administration. Additionally, we found that the administration of citral at 25 mg/kg and 50 mg/kg did not significantly affect the locomotor activity. Beyond the aforementioned, gas chromatography-mass spectrometry analysis revealed the presence of the compound in the blood and brain after subchronic administration of citral. Taken together, the results obtained in vitro, in vivo and ex vivo clearly indicate that citral is a promising monoterpene that can potentially be used towards cognition improvement.
Collapse
Affiliation(s)
| | - Monika Rudkowska
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, 4A Chodźki, 20-093 Lublin, Poland
| | - Katarzyna Klimek
- Department of Biochemistry and Biotechnology, Medical University of Lublin, 1 Chodźki Street, 20-093 Lublin, Poland
| | - Monika Agacka-Mołdoch
- Department of Plant Breeding and Biotechnology, Institute of Soil Science and Plant Cultivation, State Research Institute, Czartoryskich 8, 24-100 Puławy, Poland
| | - Jarosław Mołdoch
- Department of Biochemistry and Crop Quality, Institute of Soil Science and Plant Cultivation, State Research Institute, Czartoryskich 8, 24-100 Puławy, Poland
| | - Agnieszka Michalak
- Independent Laboratory of Behavioral Studies, Medical University of Lublin, 4A Chodźki, 20-093 Lublin, Poland
| |
Collapse
|
8
|
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
|
9
|
Alatawi FS, Omran AME, Alatawi MS, Rashad E, Yasin NAE, Soliman AF. Network Pharmacology Prediction and Experimental Validation of Ferulic Acid’s Protective Effects against Diclofenac‐Induced Liver Injury. J Food Biochem 2024; 2024. [DOI: 10.1155/2024/5592390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 07/04/2024] [Indexed: 07/31/2024]
Abstract
Despite being one of the most consumed analgesics worldwide, liver injury is an adverse effect of diclofenac (DF). In pursuit of reliable hepatoprotective natural remedies, this study aimed to investigate the potential protective effect of ferulic acid (FA) and its mechanism against DF‐induced liver injury. Various network databases and datasets were used to collect targets corresponding to FA and DF‐induced liver injury. Enrichment analyses of common targets were performed, a protein‐protein interaction (PPI) network was constructed, the hub genes were identified, and the upstream miRNA interacting with the top hub gene was later predicted. A DF‐induced liver injury rat model was established to verify FA’s protective effects, and the selected hub gene expression level with its upstream regulatory miRNA and a downstream set of targets was examined to elucidate the underlying mechanism. A total of 18 genes were identified as potential targets of FA to protect against DF‐induced liver injury. Data from the enrichment and PPI analyses and the prediction of the upstream miRNAs indicated that the most worthwhile pair to study was miR‐296‐5p/Jun. In vivo findings showed that coadministration of FA significantly reduced the DF‐induced alterations in the liver function indices, oxidative stress, and liver histology. Mechanistically, FA downregulated the expression of Jun, Bim, Bax, Casp3, IL‐1β, IL‐6, and TNF‐α, whereas it upregulated the expression of rno‐miR‐296‐5p and Bcl2. In conclusion, combining network pharmacology and an in vivo study revealed that miR‐296‐5p/Jun axis could mediate the mitigative effect of FA against DF‐induced liver injury.
Collapse
|
10
|
Sansoucy M, Naud JF. Using Proteins As Markers for Anabolic Steroid Abuse: A New Perspective in Doping Control? Chem Res Toxicol 2023; 36:1168-1173. [PMID: 37561919 DOI: 10.1021/acs.chemrestox.3c00166] [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: 08/12/2023]
Abstract
Drug toxicity is a major concern and has motivated numerous studies to elucidate specific adverse mechanisms, with acetaminophen being the favorite candidate in toxicology studies. Conversely, androgenic anabolic steroids (AASs) also represent a severe public health issue in sports for elite and non-elite athletes. Supraphysiological dosages of AASs are associated with various adverse effects, from cardiovascular to neurological repercussions including liver dysfunction. Yet, few studies have addressed the toxicity of anabolic steroids, and a significant amount of work will be needed to elucidate and understand steroid toxicity properly. This Perspective suggests ideas on how proteomics and liquid chromatography coupled with high-resolution tandem mass spectrometry (LC-HRMS/MS) can contribute to (1) pinpoint serum proteins affected by substantial doses of anabolic steroids that would represent interesting novel candidates for routine testing and (2) provide additional knowledge on androgenic anabolic steroid toxicity to help raise awareness on the harmful effects.
Collapse
Affiliation(s)
- Maxime Sansoucy
- Laboratoire de contrôle du dopage, Institut National de la Recherche Scientifique Centre Armand-Frappier Santé Biotechnologie, 531 Boulevard des Prairies, Laval, QC H7V 1B7, Canada
| | - Jean-François Naud
- Laboratoire de contrôle du dopage, Institut National de la Recherche Scientifique Centre Armand-Frappier Santé Biotechnologie, 531 Boulevard des Prairies, Laval, QC H7V 1B7, Canada
| |
Collapse
|
11
|
Sumner G, Keller S, Huleatt J, Staack RF, Wagner L, Azadeh M, Bandukwala A, Cao L, Du X, Salinas GF, Garofolo F, Harris S, Hopper S, Irwin C, Ji Q, Joseph J, King L, Kinhikar A, Lu Y, Luo R, Mabrouk O, Malvaux L, Marshall JC, McGuire K, Mikol V, Neely R, Qiu X, Saito Y, Salaun B, Scully I, Smeraglia J, Solstad T, Stoop J, Tang H, Teixeira P, Wang Y, Wright M, Mendez L, Beaver C, Eacret J, Au-Yeung A, Decman V, Dessy F, Eck S, Goihberg P, Alcaide EG, Gonneau C, Grugan K, Hedrick MN, Kar S, Sehra S, Stevens E, Stevens C, Sun Y, McCush F, Williams L, Fischer S, Wu B, Jordan G, Burns C, Cludts I, Coble K, Grimaldi C, Henderson N, Joyce A, Lotz G, Lu Y, Luo L, Neff F, Sperinde G, Stubenrauch KG, Wang Y, Ware M, Xu W. 2022 White Paper on Recent Issues in Bioanalysis: Enzyme Assay Validation, BAV for Primary End Points, Vaccine Functional Assays, Cytometry in Tissue, LBA in Rare Matrices, Complex NAb Assays, Spectral Cytometry, Endogenous Analytes, Extracellular Vesicles Part 2 - Recommendations on Biomarkers/CDx, Flow Cytometry, Ligand-Binding Assays Development & Validation; Emerging Technologies; Critical Reagents Deep Characterization. Bioanalysis 2023; 15:861-903. [PMID: 37584363 DOI: 10.4155/bio-2023-0151] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2023] Open
Abstract
The 16th Workshop on Recent Issues in Bioanalysis (16th WRIB) took place in Atlanta, GA, USA on September 26-30, 2022. Over 1000 professionals representing pharma/biotech companies, CROs, and multiple regulatory agencies convened to actively discuss the most current topics of interest in bioanalysis. The 16th WRIB included 3 Main Workshops and 7 Specialized Workshops that together spanned 1 week in order to allow exhaustive and thorough coverage of all major issues in bioanalysis, biomarkers, immunogenicity, gene therapy, cell therapy and vaccines. Moreover, in-depth workshops on ICH M10 BMV final guideline (focused on this guideline training, interpretation, adoption and transition); mass spectrometry innovation (focused on novel technologies, novel modalities, and novel challenges); and flow cytometry bioanalysis (rising of the 3rd most common/important technology in bioanalytical labs) were the special features of the 16th edition. As in previous years, WRIB continued to gather a wide diversity of international, industry opinion leaders and regulatory authority experts working on both small and large molecules as well as gene, cell therapies and vaccines to facilitate sharing and discussions focused on improving quality, increasing regulatory compliance, and achieving scientific excellence on bioanalytical issues. This 2022 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop and is aimed to provide the bioanalytical community with key information and practical solutions on topics and issues addressed, in an effort to enable advances in scientific excellence, improved quality and better regulatory compliance. Due to its length, the 2022 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 2) covers the recommendations on LBA, Biomarkers/CDx and Cytometry. Part 1 (Mass Spectrometry and ICH M10) and Part 3 (Gene Therapy, Cell therapy, Vaccines and Biotherapeutics Immunogenicity) are published in volume 15 of Bioanalysis, issues 16 and 14 (2023), respectively.
Collapse
Affiliation(s)
| | | | | | - Roland F Staack
- Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | - Qin Ji
- AbbVie, North Chicago, IL, USA
| | | | | | | | - Yang Lu
- US FDA, Silver Spring, MD, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Priscila Teixeira
- Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | - Yixin Wang
- Bristol-Myers Squibb, Lawrenceville, NJ, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Gregor Jordan
- Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | | | | | | | | | - Neil Henderson
- Integrated Bioanalysis, Clinical Pharmacology & Safety Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | | | - Gregor Lotz
- Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | | | | | - Florian Neff
- Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | | | | | | | | | | |
Collapse
|
12
|
Shah R, Agarwal A, Kavoussi P, Rambhatla A, Saleh R, Cannarella R, Harraz AM, Boitrelle F, Kuroda S, Hamoda TAAAM, Zini A, Ko E, Calik G, Toprak T, Kandil H, Gül M, Bakırcıoğlu ME, Parekh N, Russo GI, Tadros N, Kadioglu A, Arafa M, Chung E, Rajmil O, Dimitriadis F, Malhotra V, Salvio G, Henkel R, Le TV, Sogutdelen E, Vij S, Alarbid A, Gudeloglu A, Tsujimura A, Calogero AE, El Meliegy A, Crafa A, Kalkanli A, Baser A, Hazir B, Giulioni C, Cho CL, Ho CCK, Salzano C, Zylbersztejn DS, Tien DMB, Pescatori E, Borges E, Serefoglu EC, Saïs-Hamza E, Huyghe E, Ceyhan E, Caroppo E, Castiglioni F, Bahar F, Gokalp F, Lombardo F, Gadda F, Duarsa GWK, Pinggera GM, Busetto GM, Balercia G, Cito G, Blecher G, Franco G, Liguori G, Elbardisi H, Keskin H, Lin H, Taniguchi H, Park HJ, Ziouziou I, de la Rosette J, Hotaling J, Ramsay J, Molina JMC, Lo KL, Bocu K, Khalafalla K, Bowa K, Okada K, Nagao K, Chiba K, Hakim L, Makarounis K, Hehemann M, Rodriguez Peña M, Falcone M, Bendayan M, Martinez M, Timpano M, Altan M, Fode M, Al-Marhoon MS, Sadighi Gilani MA, Soebadi MA, Gherabi N, Sofikitis N, Kahraman O, Birowo P, et alShah R, Agarwal A, Kavoussi P, Rambhatla A, Saleh R, Cannarella R, Harraz AM, Boitrelle F, Kuroda S, Hamoda TAAAM, Zini A, Ko E, Calik G, Toprak T, Kandil H, Gül M, Bakırcıoğlu ME, Parekh N, Russo GI, Tadros N, Kadioglu A, Arafa M, Chung E, Rajmil O, Dimitriadis F, Malhotra V, Salvio G, Henkel R, Le TV, Sogutdelen E, Vij S, Alarbid A, Gudeloglu A, Tsujimura A, Calogero AE, El Meliegy A, Crafa A, Kalkanli A, Baser A, Hazir B, Giulioni C, Cho CL, Ho CCK, Salzano C, Zylbersztejn DS, Tien DMB, Pescatori E, Borges E, Serefoglu EC, Saïs-Hamza E, Huyghe E, Ceyhan E, Caroppo E, Castiglioni F, Bahar F, Gokalp F, Lombardo F, Gadda F, Duarsa GWK, Pinggera GM, Busetto GM, Balercia G, Cito G, Blecher G, Franco G, Liguori G, Elbardisi H, Keskin H, Lin H, Taniguchi H, Park HJ, Ziouziou I, de la Rosette J, Hotaling J, Ramsay J, Molina JMC, Lo KL, Bocu K, Khalafalla K, Bowa K, Okada K, Nagao K, Chiba K, Hakim L, Makarounis K, Hehemann M, Rodriguez Peña M, Falcone M, Bendayan M, Martinez M, Timpano M, Altan M, Fode M, Al-Marhoon MS, Sadighi Gilani MA, Soebadi MA, Gherabi N, Sofikitis N, Kahraman O, Birowo P, Kothari P, Sindhwani P, Javed Q, Ambar RF, Kosgi R, Ghayda RA, Adriansjah R, Condorelli RA, La Vignera S, Micic S, Kim SHK, Fukuhara S, Ahn ST, Mostafa T, Ong TA, Takeshima T, Amano T, Barrett T, Arslan U, Karthikeyan VS, Atmoko W, Yumura Y, Yuan Y, Kato Y, Jezek D, Cheng BKC, Hatzichristodoulou G, Dy J, Castañé ER, El-Sakka AI, Nguyen Q, Sarikaya S, Boeri L, Tan R, Moussa MA, El-Assmy A, Alali H, Alhathal N, Osman Y, Perovic D, Sajadi H, Akhavizadegan H, Vučinić M, Kattan S, Kattan MS, Mogharabian N, Phuoc NHV, Ngoo KS, Alkandari MH, Alsuhaibani S, Sokolakis I, Babaei M, King MS, Diemer T, Gava MM, Henrique R, Silva RSE, Paul GM, Mierzwa TC, Glina S, Siddiqi K, Wu H, Wurzacher J, Farkouh A, Son H, Minhas S, Lee J, Magsanoc N, Capogrosso P, Albano GJ, Lewis SEM, Jayasena CN, Alvarez JG, Teo C, Smith RP, Chua JBM, Jensen CFS, Parekattil S, Finelli R, Durairajanayagam D, Karna KK, Ahmed A, Evenson D, Umemoto Y, Puigvert A, Çeker G, Colpi GM. Consensus and Diversity in the Management of Varicocele for Male Infertility: Results of a Global Practice Survey and Comparison with Guidelines and Recommendations. World J Mens Health 2023; 41:164-197. [PMID: 35791302 PMCID: PMC9826919 DOI: 10.5534/wjmh.220048] [Show More Authors] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/15/2022] [Accepted: 03/20/2022] [Indexed: 02/06/2023] Open
Abstract
PURPOSE Varicocele is a common problem among infertile men. Varicocele repair (VR) is frequently performed to improve semen parameters and the chances of pregnancy. However, there is a lack of consensus about the diagnosis, indications for VR and its outcomes. The aim of this study was to explore global practice patterns on the management of varicocele in the context of male infertility. MATERIALS AND METHODS Sixty practicing urologists/andrologists from 23 countries contributed 382 multiple-choice-questions pertaining to varicocele management. These were condensed into an online questionnaire that was forwarded to clinicians involved in male infertility management through direct invitation. The results were analyzed for disagreement and agreement in practice patterns and, compared with the latest guidelines of international professional societies (American Urological Association [AUA], American Society for Reproductive Medicine [ASRM], and European Association of Urology [EAU]), and with evidence emerging from recent systematic reviews and meta-analyses. Additionally, an expert opinion on each topic was provided based on the consensus of 16 experts in the field. RESULTS The questionnaire was answered by 574 clinicians from 59 countries. The majority of respondents were urologists/uro-andrologists. A wide diversity of opinion was seen in every aspect of varicocele diagnosis, indications for repair, choice of technique, management of sub-clinical varicocele and the role of VR in azoospermia. A significant proportion of the responses were at odds with the recommendations of AUA, ASRM, and EAU. A large number of clinical situations were identified where no guidelines are available. CONCLUSIONS This study is the largest global survey performed to date on the clinical management of varicocele for male infertility. It demonstrates: 1) a wide disagreement in the approach to varicocele management, 2) large gaps in the clinical practice guidelines from professional societies, and 3) the need for further studies on several aspects of varicocele management in infertile men.
Collapse
Affiliation(s)
- Rupin Shah
- Division of Andrology, Department of Urology, Lilavati Hospital and Research Centre, Mumbai, India
| | - Ashok Agarwal
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA.
| | - Parviz Kavoussi
- Austin Fertility & Reproductive Medicine/Westlake IVF, TX, USA
| | - Amarnath Rambhatla
- Department of Urology, Vattikuti Urology Institute, Henry Ford Health System, Detroit, MI, USA
| | - Ramadan Saleh
- Department of Dermatology, Venereology and Andrology, Faculty of Medicine, Sohag University, Sohag, Egypt
| | - Rossella Cannarella
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Ahmed M Harraz
- Mansoura University Urology and Nephrology Center, Mansoura, Egypt
- Department of Surgery, Urology Unit, Farwaniya Hospital, Farwaniya, Kuwait
- Department of Urology, Sabah Al Ahmad Urology Center, Kuwait City, Kuwait
| | - Florence Boitrelle
- Department of Reproductive Biology, Fertility Preservation, Andrology, CECOS, Poissy Hospital, Poissy, France
- Department of Biology, Reproduction, Epigenetics, Environment and Development, Paris Saclay University, UVSQ, INRAE, BREED, Jouy-en-Josas, France
| | - Shinnosuke Kuroda
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Taha Abo-Almagd Abdel-Meguid Hamoda
- Department of Urology, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Urology, Faculty of Medicine, Minia University, Minia, Egypt
| | - Armand Zini
- Division of Urology, Department of Surgery, McGill University, Montreal, QC, Canada
| | - Edmund Ko
- Department of Urology, Loma Linda University Health, Loma Linda, CA, USA
| | - Gokhan Calik
- Department of Urology, Faculty of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Tuncay Toprak
- Department of Urology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | | | - Murat Gül
- Department of Urology, Selcuk University School of Medicine, Konya, Turkey
| | | | - Neel Parekh
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Nicholas Tadros
- Division of Urology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Ates Kadioglu
- Section of Andrology, Department of Urology, Istanbul University, Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Mohamed Arafa
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
- Department of Urology, Hamad Medical Corporation, Doha, Qatar
- Department of Urology, Weill Cornell Medical-Qatar, Doha, Qatar
| | - Eric Chung
- Department of Urology, Princess Alexandra Hospital, University of Queensland, Brisbane, Australia
| | - Osvaldo Rajmil
- Department of Andrology, Fundacio Puigvert, Barcelona, Spain
| | - Fotios Dimitriadis
- Department of Urology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | | | - Gianmaria Salvio
- Department of Endocrinology, Polytechnic University of Marche, Ancona, Italy
| | - Ralf Henkel
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Medical Bioscience, University of the Western Cape, Bellville, South Africa
| | - Tan V Le
- Department of Andrology, Binh Dan Hospital, Ho Chi Minh City, Vietnam
- Department of Urology and Andrology, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, Vietnam
| | | | - Sarah Vij
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Abdullah Alarbid
- Department of Surgery, Urology Unit, Farwaniya Hospital, Farwaniya, Kuwait
| | - Ahmet Gudeloglu
- Department of Urology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Akira Tsujimura
- Department of Urology, Juntendo University Urayasu Hospital, Chiba, Japan
| | - Aldo E Calogero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Amr El Meliegy
- Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Andrea Crafa
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Arif Kalkanli
- Department of Urology, Taksim Education and Research Hospital, Istanbul, Turkey
| | - Aykut Baser
- Department of Urology, Faculty of Medicine, Bandırma Onyedi Eylül University, Balıkesir, Turkey
| | - Berk Hazir
- Reproductive medicine, Clinical Research Centre, Lund University, Malmö, Sweden
| | - Carlo Giulioni
- Department of Urology, Polytechnic University of Marche Region, Ancona, Italy
| | - Chak-Lam Cho
- S. H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Christopher C K Ho
- Department of Urology, Universiti Teknologi Mara (UiTM) Specialist Centre, Selangor, Malaysia
| | - Ciro Salzano
- PO San Giovanni Bosco, ASL Napoli 1 Centro, Napoli, Italy
| | | | - Dung Mai Ba Tien
- Department of Andrology, Binh Dan Hospital, Ho Chi Minh City, Vietnam
| | - Edoardo Pescatori
- Andrology and Reproductive Medicine Unit, Gynepro Medical, Bologna, Italy
| | | | - Ege Can Serefoglu
- Department of Urology, Biruni University School of Medicine, Istanbul, Turkey
| | - Emine Saïs-Hamza
- Department of Reproductive Biology, Fertility Preservation, Andrology, CECOS, Poissy Hospital, Poissy, France
- Department of Biology, Reproduction, Epigenetics, Environment and Development, Paris Saclay University, UVSQ, INRAE, BREED, Jouy-en-Josas, France
| | - Eric Huyghe
- Department of Urology and Andrology, University Hospital of Toulouse, France
| | - Erman Ceyhan
- Department of Urology, School of Medicine, Baskent University, Konya, Turkey
| | - Ettore Caroppo
- Asl Bari, PTA "F Jaia", Reproductive and IVF unit, Andrology Outpatients Clinic, Asl Bari, Conversano (Ba), Italy
| | | | - Fahmi Bahar
- Andrology Section, Siloam Sriwijaya Hospital, Palembang, Indonesia
| | - Fatih Gokalp
- Department of Urology, Hatay Mustafa Kemal University, Antakya, Turkey
| | - Francesco Lombardo
- Laboratory of Seminology-Sperm Bank "Loredana Gandini", Department of Experimental Medicine, University of Rome "Sapienza", Rome, Italy
| | - Franco Gadda
- Department of Urology, IRCCS Fondazione Ca Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | | | - Gian Maria Busetto
- Department of Urology and Organ Transplantation, University of Foggia, Ospedali Riuniti of Foggia, Foggia, Italy
| | - Giancarlo Balercia
- Department of Endocrinology, Polytechnic University of Marche, Ancona, Italy
| | - Gianmartin Cito
- Department of Urology, University of Florence, Florence, Italy
| | - Gideon Blecher
- Department of Surgery, School of Clinical Sciences, Monash University, Melbourne, Australia
- Department of Urology, The Alfred Hospital, Melbourne, Australia
| | - Giorgio Franco
- Department of Urology, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | | | - Haitham Elbardisi
- Department of Urology, Hamad Medical Corporation, Doha, Qatar
- Department of Urology, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Hakan Keskin
- Department of Urology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Haocheng Lin
- Department of Urology, Peking University Third Hospital, Peking University, Beijing, China
| | - Hisanori Taniguchi
- Department of Urology and Andrology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Hyun Jun Park
- Department of Urology, Pusan National University School of Medicine, Busan, Korea
- Medical Research Institute of Pusan National University Hospital, Busan, Korea
| | - Imad Ziouziou
- Department of Urology, College of medicine and pharmacy, Ibn Zohr University, Agadir, Morocco
| | - Jean de la Rosette
- Department of urology, Faculty of Medicine, Istanbul Medipol Mega University Hospital, Istanbul, Turkey
| | - Jim Hotaling
- Department of Surgery (Urology), University of Utah, Salt Lake City, UT, USA
| | - Jonathan Ramsay
- Department of Andrology, Hammersmith Hospital, Imperial, London, UK
| | | | - Ka Lun Lo
- Division of Urology, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Kadir Bocu
- Department of Urology, Silopi State Hospital, Sirnak, Turkey
| | - Kareim Khalafalla
- Department of Urology, Hamad Medical Corporation, Doha, Qatar
- Department of Urology, University of Illinois, Chicago, IL, USA
| | - Kasonde Bowa
- Department of Urology, School of Medicine and Health Sciences, University of Lusaka, Lusaka, Zambia
| | - Keisuke Okada
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Koichi Nagao
- Department of Urology, Toho University Faculty of Medicine, Tokyo, Japan
| | - Koji Chiba
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Lukman Hakim
- Department of Urology, Universitas Airlangga/Rumah Sakit Universitas Airlangga Teaching Hospital, Surabaya, Indonesia
| | | | - Marah Hehemann
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Marcelo Rodriguez Peña
- Instituto de Ginecología y Fertilidad (IFER), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Marco Falcone
- Department of Urology, Molinette Hospital, A.O.U. Città della Salute e della Scienza, University of Turin, Torino, Italy
| | - Marion Bendayan
- Department of Reproductive Biology, Fertility Preservation, Andrology, CECOS, Poissy Hospital, Poissy, France
- Department of Biology, Reproduction, Epigenetics, Environment and Development, Paris Saclay University, UVSQ, INRAE, BREED, Jouy-en-Josas, France
| | - Marlon Martinez
- Section of Urology, Department of Surgery, University of Santo Tomas Hospital, Manila, Philippines
| | - Massimiliano Timpano
- Department of Urology, Molinette Hospital, A.O.U. Città della Salute e della Scienza, University of Turin, Torino, Italy
| | - Mesut Altan
- Department of Urology, Haceppete University, Ankara, Turkey
| | - Mikkel Fode
- Department of Urology, Herlev and Gentofte University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Mohammad Ali Sadighi Gilani
- Department of Andrology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | | | - Nazim Gherabi
- Andrology Committee of the Algerian Association of Urology, Algiers, Algeria
| | - Nikolaos Sofikitis
- Department of Urology, Ioannina University School of Medicine, Ioannina, Greece
| | - Oğuzhan Kahraman
- Department of Urology, School of Medicine, Baskent University, Konya, Turkey
| | - Ponco Birowo
- Department of Urology, Cipto Mangunkusumo General Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | | | - Puneet Sindhwani
- Department of Urology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Qaisar Javed
- Department of Urology, Al - Ahlia Hospital Abu Dhabi UAE, Abu Dhabi, UAE
| | - Rafael F Ambar
- Department of Urology, Centro Universitario em Saude do ABC, Santo André, Brazil
- Andrology Group at Ideia Fertil Institute of Human Reproduction, Santo André, Brazil
| | - Raghavender Kosgi
- Department of Urology and Andrology, AIG Hospitals, Hyderabad, Telangana, India
| | - Ramy Abou Ghayda
- Urology Institute, University Hospitals, Case Western Reserve University, Cleveland, OH, USA
| | - Ricky Adriansjah
- Department of Urology, Faculty of Medicine Universitas Padjadjaran, Hasan Sadikin General Hospital, Banding, Indonesia
| | | | - Sandro La Vignera
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Sava Micic
- Department of Andrology, Uromedica Polyclinic, Belgrade, Serbia
| | - Shannon Hee Kyung Kim
- IVF Australia, Sydney, New South Wales, Australia
- Macquarie School of Medicine, Macquaire University, Sydney, New South Wales, Australia
| | - Shinichiro Fukuhara
- Department of Urology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Sun Tae Ahn
- Department of Urology, Korea University College of Medicine, Seoul, Korea
| | - Taymour Mostafa
- Department of Andrology, Sexology and STIs, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Teng Aik Ong
- Department of Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Teppei Takeshima
- Department of Urology, Reproduction Center, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Toshiyasu Amano
- Department of Urology, Nagano Red Cross Hospital, Nagano, Japan
| | | | - Umut Arslan
- Department of Urology, Fatih Sultan Mehmet Training and Research Hospital, University of Health Sciences, Istanbul, Turkey
| | | | - Widi Atmoko
- Department of Urology and Andrology, AIG Hospitals, Hyderabad, Telangana, India
| | - Yasushi Yumura
- Department of Urology, Reproduction Center, Yokohama City University Medical Center, Yokohama, Kanagawa, Japan
| | - Yiming Yuan
- Andrology Center & Urology Department, Peking University First Hospital, Peking University, Beijing, China
| | - Yuki Kato
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Davor Jezek
- Department for Transfusion Medicine and Transplantation Biology, Reproductive Tissue Bank, University of Zagreb, School of Medicine, University Hospital Zagreb, Zagreb, Croatia
| | | | | | - Jun Dy
- Stone and Prostate Treatment Services/Pelvic Floor Center and Anorectal Diseases, St. Luke's Medical Center, Quezon City, Metro Manila, Philippines
| | - Eduard Ruiz Castañé
- Department of Andrology, Fundació Puigvert, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ahmed I El-Sakka
- Department of Urology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Quang Nguyen
- Center for Andrology and Sexual Medicine, Viet Duc University Hospital, Hanoi, Vietnam
- Department of Urology, Andrology and Sexual Medicine, University of Medicine and Pharmacy, Vietnam National University, Hanoi, Vietnam
| | - Selcuk Sarikaya
- Department of Urology, Gülhane Research and Training Hospital, University of Health Sciences, Ankara, Turkey
| | - Luca Boeri
- Department of Urology, IRCCS Fondazione Ca Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Ronny Tan
- Advanced Urology Associates, Singapore
- Mount Elizabeth Hospital, Singapore
| | - Mohamad A Moussa
- Department of Urology, Lebanese University, Beirut, Lebanon
- Department of Urology, Al Zahraa Hospital, UMC, Lebanon
| | - Ahmed El-Assmy
- Mansoura University Urology and Nephrology Center, Mansoura, Egypt
| | - Hamed Alali
- King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Naif Alhathal
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Yasser Osman
- Mansoura University Urology and Nephrology Center, Mansoura, Egypt
| | - Dragoljub Perovic
- Urology and Andrology Center, CODRA Hospital, University of Montenegro, Podgorica, Montenegro
| | | | - Hamed Akhavizadegan
- Department of Urology, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Said Kattan
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mohamed S Kattan
- King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Nasser Mogharabian
- Sexual Health and Fertility Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
| | | | - Kay Seong Ngoo
- Urology Unit, Department of Surgery, Hospital Angkatan Tentera Tuanku Mizan, Kuala Lumpur, Malaysia
| | - Mohammad H Alkandari
- Department of Urology, Mubarak Al-Kabeer Teaching Hospital, Kuwait University, Jabriya, Kuwait
| | - Shaheed Alsuhaibani
- Department of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Ioannis Sokolakis
- Department of Urology, Martha-Maria Hospital Nuremberg, Nuremberg, Germany
| | - Mehdi Babaei
- Department of Andrology, Shariati Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mak Siu King
- Division of Urology, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Thorsten Diemer
- Department of Urology, Pediatric Urology and Andrology, Justus-Liebig-University Giessen (GER), University Hospital Giessen and Marburg GmbH, Campus Giessen, Giessen, Germany
| | - Marcelo M Gava
- Department of Urology, Centro Universitario em Saude do ABC, Santo André, Brazil
- Andrology Group at Ideia Fertil Institute of Human Reproduction, Santo André, Brazil
| | | | - Rodrigo Spinola E Silva
- Department of Urology, Centro Universitario em Saude do ABC, Santo André, Brazil
- Andrology Group at Ideia Fertil Institute of Human Reproduction, Santo André, Brazil
| | - Gustavo Marquesine Paul
- Department of Urology, Hospital de Clínicas of the Federal University of Paraná, Curitiba, Brazil
| | | | - Sidney Glina
- Department of Urology, Centro Universitario em Saude do ABC, Santo André, Brazil
| | | | - Han Wu
- Department of Andrology, PKU 3rd Hospital Reproductive Medicine Center, Beijing, China
| | - Jana Wurzacher
- Department of Urology, University Hospital Innsbruck, Innsbruck, Austria
| | - Ala'a Farkouh
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Hwancheol Son
- Department of Urology, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Suks Minhas
- Division of Surgery, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Joe Lee
- Department of Urology, National University Hospital, Singapore
| | - Nikko Magsanoc
- Department of Surgery, University of the Philippines College of Medicine, Manila, Philippines
| | | | - German Jose Albano
- Section of Urology, Department of Surgery, University of Santo Tomas Hospital, Manila, Philippines
| | | | - Channa N Jayasena
- Department of Reproductive Endocrinology and Andrology, Imperial College London, London, UK
- Department of Andrology, Hammersmith & St. Mary's Hospitals, London, UK
| | - Juan G Alvarez
- Department of Andrology, Centro ANDROGEN, La Coruña, Spain
| | - Colin Teo
- Department of Urology, Gleneagles Hospital, Singapore
| | - Ryan P Smith
- Department of Urology, University of Virginia School of Medicine, Virginia, USA
| | - Jo Ben M Chua
- Department of Urology, East Avenue Medical Center, Quezon City, Philippines
| | | | - Sijo Parekattil
- Avant Concierge Urology & University of Central Florida, Winter Garden, FL, USA
| | - Renata Finelli
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Damayanthi Durairajanayagam
- Department of Physiology, Faculty of Medicine, Universiti Teknologi MARA, Sungai Buloh Campus, Selangor, Malaysia
| | - Keshab Kumar Karna
- American Center for Reproductive Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Abdelkareem Ahmed
- Department of Urology, Sabah Al Ahmad Urology Center, Kuwait City, Kuwait
| | | | - Yukihiro Umemoto
- Department of Nephro-Urology, Nagoya City West Medical Center, Nagoya, Japan
| | - Ana Puigvert
- Urological and Human Reproduction Unit, Quiron Hospital, Barcelona, Spain
| | - Gökhan Çeker
- Department of Urology, Başakşehir Çam and Sakura City Hospital, Istanbul, Turkey
- Department of Embryology and Histology, Zonguldak Bülent Ecevit University Institute of Health Sciences, Zonguldak, Turkey
| | | |
Collapse
|
13
|
Zhang C, Fan S, Zhao JQ, Jiang Y, Sun JX, Li HJ. Transcriptomics and metabolomics reveal the role of CYP1A2 in psoralen/isopsoralen-induced metabolic activation and hepatotoxicity. Phytother Res 2023; 37:163-180. [PMID: 36056681 DOI: 10.1002/ptr.7604] [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: 06/14/2022] [Revised: 07/23/2022] [Accepted: 08/12/2022] [Indexed: 01/19/2023]
Abstract
Psoralen and isopsoralen are the pharmacologically important but hepatotoxic components in Psoraleae Fructus. The purpose of this study was to reveal the underlying mechanism of psoralen/isopsoralen-induced hepatotoxicity. Initially, we applied integrated analyses of transcriptomic and metabolomic profiles in mice treated with psoralen and isopsoralen, highlighting the xenobiotic metabolism by cytochromes P450 as a potential pathway. Then, with verifications of expression levels by qRT-PCR and western blot, affinities by molecular docking, and metabolic contributions by recombinant human CYP450 and mouse liver microsomes, CYP1A2 was screened out as the key metabolic enzyme. Afterwards, CYP1A2 induction and inhibition models in HepG2 cells and mice were established to verify the role of CYP1A2, demonstrating that induction of CYP1A2 aggravated the hepatotoxicity, and conversely inhibition alleviated the hepatotoxic effects. Additionally, we detected glutathione adducts with reactive intermediates of psoralen and isopsoralen generated by CYP1A2 metabolism in biosystems of recombinant human CYP1A2 and mouse liver microsomes, CYP1A2-overexpressed HepG2 cells, mice livers and the chemical reaction system using UPLC-Q-TOF-MS/MS. Ultimately, the high-content screening presented the cellular oxidative stress and relevant hepatotoxicity due to glutathione depletion by reactive intermediates. In brief, our findings illustrated that CYP1A2-mediated metabolic activation is responsible for the psoralen/isopsoralen-induced hepatotoxicity.
Collapse
Affiliation(s)
- Cai Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Song Fan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jin-Quan Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yan Jiang
- College of Chemical Engineering, Nanjing Forestry University, Nanjing, China
| | - Jia-Xing Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Hui-Jun Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
14
|
Effects of torularhodin against acetaminophen induced liver injury base on antioxidation, anti-inflammation and anti-apoptosis. FOOD BIOSCI 2023. [DOI: 10.1016/j.fbio.2023.102388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
15
|
Varışlı B, Caglayan C, Kandemir FM, Gür C, Ayna A, Genç A, Taysı S. Chrysin mitigates diclofenac-induced hepatotoxicity by modulating oxidative stress, apoptosis, autophagy and endoplasmic reticulum stress in rats. Mol Biol Rep 2023; 50:433-442. [PMID: 36344803 DOI: 10.1007/s11033-022-07928-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/06/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Diclofenac (DF) is a non-steroidal anti-inflammatory drug (NSAID) generally prescribed for the treatment of pain. In spite of the widespread use of DF, hepatotoxicity has been reported after its administration. The current study discloses new evidence as regards of the curative effects of chrysin (CHR) on DF-induced hepatotoxicity by regulating oxidative stress, apoptosis, autophagy, and endoplasmic reticulum (ER) stress. METHODS The animals were separated into five different groups. Group-I was in control. Group-II received CHR-only (50 mg/kg bw, p.o.) on all 5 days. Group-III received DF-only (50 mg/kg bw, i.p.) on 4th and 5th day. Group-IV received DF (50 mg/kg bw) + CHR (25 mg/kg, bw) and group-V received DF (50 mg/kg, bw) + CHR (50 mg/kg, bw) for 5 days. RESULTS DF injection was associated with increased MDA while reduced GSH level, activities of superoxide dismutase, glutathione peroxidase, and catalase and mRNA levels of HO-1 and Nrf2 in the liver. DF injection caused apoptosis and autophagy in the liver by up-regulating caspase-3, Bax, LC3A, and LC3B levels and down-regulating Bcl-2. DF also caused ER stress by increasing mRNA transcript levels of ATF-6, IRE1, PERK, and GRP78. Additionally, it was observed that DF administration up-regulated MMP2 and MMP9. However, treatment with CHR at a dose of 25 and 50 mg/kg considerably ameliorated oxidative stress, apoptosis, autophagy, and ER stress in liver tissue. CONCLUSION Overall, the data of this study indicate that liver damage associated with DF toxicity could be ameliorated by CHR administration.
Collapse
Affiliation(s)
- Behçet Varışlı
- Vocational School of Health Sevices, Final International University, Cyprus, Turkey
| | - Cuneyt Caglayan
- Department of Medical Biochemistry, Faculty of Medicine, Bilecik Seyh Edebali University, Bilecik, Turkey.
| | - Fatih Mehmet Kandemir
- Department of Medical Biochemistry, Faculty of Medicine, Aksaray University, Aksaray, Turkey.
| | - Cihan Gür
- Department of Biochemistry, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - Adnan Ayna
- Department of Chemistry, Faculty of Science and Literature, Bingol University, Bingol, Turkey
| | - Aydın Genç
- Department of Biochemistry, Faculty of Veterinary Medicine, Bingol University, Bingol, Turkey
| | - Seyithan Taysı
- Department of Medical Biochemistry, Medical School, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
16
|
Role of FOXO3a Transcription Factor in the Regulation of Liver Oxidative Injury. Antioxidants (Basel) 2022; 11:antiox11122478. [PMID: 36552685 PMCID: PMC9774119 DOI: 10.3390/antiox11122478] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Oxidative stress has been identified as a key mechanism in liver damage caused by various chemicals. The transcription factor FOXO3a has emerged as a critical regulator of redox imbalance. Multiple post-translational changes and epigenetic processes closely regulate the activity of FOXO3a, resulting in synergistic or competing impacts on its subcellular localization, stability, protein-protein interactions, DNA binding affinity, and transcriptional programs. Depending on the chemical nature and subcellular context, the oxidative-stress-mediated activation of FOXO3a can induce multiple transcriptional programs that play crucial roles in oxidative injury to the liver by chemicals. Here, we mainly review the role of FOXO3a in coordinating programs of genes that are essential for cellular homeostasis, with an emphasis on exploring the regulatory mechanisms and potential application of FOXO3a as a therapeutic target to prevent and treat liver oxidative injury.
Collapse
|
17
|
In Vitro Models for Studying Chronic Drug-Induced Liver Injury. Int J Mol Sci 2022; 23:ijms231911428. [PMID: 36232728 PMCID: PMC9569683 DOI: 10.3390/ijms231911428] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/08/2022] [Accepted: 09/22/2022] [Indexed: 11/17/2022] Open
Abstract
Drug-induced liver injury (DILI) is a major clinical problem in terms of patient morbidity and mortality, cost to healthcare systems and failure of the development of new drugs. The need for consistent safety strategies capable of identifying a potential toxicity risk early in the drug discovery pipeline is key. Human DILI is poorly predicted in animals, probably due to the well-known interspecies differences in drug metabolism, pharmacokinetics, and toxicity targets. For this reason, distinct cellular models from primary human hepatocytes or hepatoma cell lines cultured as 2D monolayers to emerging 3D culture systems or the use of multi-cellular systems have been proposed for hepatotoxicity studies. In order to mimic long-term hepatotoxicity in vitro, cell models, which maintain hepatic phenotype for a suitably long period, should be used. On the other hand, repeated-dose administration is a more relevant scenario for therapeutics, providing information not only about toxicity, but also about cumulative effects and/or delayed responses. In this review, we evaluate the existing cell models for DILI prediction focusing on chronic hepatotoxicity, highlighting how better characterization and mechanistic studies could lead to advance DILI prediction.
Collapse
|
18
|
Xu M, Cui Q, Su W, Zhang D, Pan J, Liu X, Pang Z, Zhu Q. High-content screening of active components of Traditional Chinese Medicine inhibiting TGF-β-induced cell EMT. Heliyon 2022; 8:e10238. [PMID: 36042745 PMCID: PMC9420491 DOI: 10.1016/j.heliyon.2022.e10238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 05/13/2022] [Accepted: 08/05/2022] [Indexed: 11/30/2022] Open
Abstract
The epithelial mesenchymal transition (EMT) has roles in metastasis and invasion during fibrotic diseases and cancer progression. Some Traditional Chinese Medicines (TCMs) have shown inhibitory effects with respect to the EMT. The current study attempted to establish a multiparametric high-content method to screen for active monomeric compounds in TCM with the ability to target cellular EMT by assessing phenotypic changes. A total of 306 monomeric compounds from the MedChemExpress (MCE) compound library were screened by the high-content screening (HCS) system and 5 compounds with anti-EMT activity, including camptothecin (CPT), dimethyl curcumin (DMC), artesunate (ART), sinapine (SNP) and berberine (BER) were identified. To confirm anti-EMT activity, expression of EMT markers was assessed by qRT-PCR and Western blotting, and cell adhesion and migration measured by cell function assays. The results revealed that CPT, DMC, ART, SNP and BER inhibited transforming growth factor-β1 (TGF-β1)-induced expression of vimentin and α-SMA, upregulated expression of E-cadherin, increased cell adhesion and reduced cell migration. In summary, by quantifying the cell morphological changes during TGF-β1-induced EMT through multi-parametric analysis, TCM compounds with anti-EMT activity were successfully screened using the HCS system, a faster and more economical approach than conventional methods.
Collapse
Affiliation(s)
- Mengzhen Xu
- College of Pharmaceutical Science, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qinghua Cui
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Wen Su
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Dan Zhang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jiaxu Pan
- College of Pharmaceutical Science, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Xiangqi Liu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zheng Pang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qingjun Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
19
|
Yousuf S, Shabir S, Singh MP. Protection Against Drug-Induced Liver Injuries Through Nutraceuticals via Amelioration of Nrf-2 Signaling. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2022; 42:495-515. [PMID: 35771985 DOI: 10.1080/27697061.2022.2089403] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hepatotoxicity caused by the overdose of various medications is a leading cause of drug-induced liver injury. Overdose of drugs causes hepatocellular necrosis. Nutraceuticals are reported to prevent drug-induced liver failure. The present article aims to review the protection provided by various medicinal plants against hepatotoxic drugs. Ayurveda is considered a conventional restorative arrangement in India. It is consistently used for ages and is still used today to cure drug-induced hepatotoxicity by focusing on antioxidant stress response pathways such as the nuclear factor erythroid-2 (Nrf-2) antioxidant response element signaling pathway. Nrf-2 is a key transcription factor that entangles Kelch-like ECH-associating protein 1, a protein found in the cell cytoplasm. Some antioxidant enzymes, such as gamma glycine cysteine ligase (γ-GCL) and heme oxygenase-1 (HO-1), are expressed in Nrf-2 targeted genes. Their expression, in turn, decreases the stimulation of hepatic macrophages and induces the messenger RNA (mRNA) articulation of proinflammatory factors including tumor necrosis factor α. This review will cover various medicinal plants from a mechanistic view and how they stimulate and interact with Nrf-2, the master regulator of the antioxidant response to counterbalance oxidative stress. Interestingly, therapeutic plants have become popular in the medical sector due to safer yet effective supplementation for the prevention and treatment of new human diseases. The contemporary study is expected to collect information on a variety of therapeutic traditional herbs that have been studied in the context of drug-induced liver toxicity, as nutraceuticals are the most effective treatments for oxidative stress-induced hepatotoxicity. They are less genotoxic, have a lower cost, and are readily available. Together, nutraceuticals exert protective effects against drug-induced hepatotoxicity through the inhibition of oxidative stress, inflammation, and apoptosis. Its mechanism(s) are considered to be associated with the γ-GCL/HO-1 and Nrf-2 signaling pathways. KEY TEACHING POINTSThe liver is the most significant vital organ that carries out metabolic activities of the body such as the synthesis of glycogen, the formation of triglycerides and cholesterol, as well as the formation of bile.Acute liver failure is caused by the consumption of certain drugs; drug-induced liver injury is the major condition.The chemopreventive activity of nutraceuticals may be related to oxidative stress reduction and attenuation of biosynthetic processes involved in hepatic injury via amelioration of the nuclear factor erythroid-2 (Nrf-2) signaling pathway.Nrf-2 is a key transcription factor that is found in the cell cytoplasm resulting in the expression of various genes such as gamma glycine cysteine ligase and heme oxygenase-1.Nutraceutical-rich phytochemicals possess high antioxidant activity, which helps in the prevention of hepatic injury.
Collapse
Affiliation(s)
- Sumaira Yousuf
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Shabnam Shabir
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| | - Mahendra P Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
20
|
Mirahmad M, Sabourian R, Mahdavi M, Larijani B, Safavi M. In vitro cell-based models of drug-induced hepatotoxicity screening: progress and limitation. Drug Metab Rev 2022; 54:161-193. [PMID: 35403528 DOI: 10.1080/03602532.2022.2064487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Drug-induced liver injury (DILI) is one of the major causes of post-approval withdrawal of therapeutics. As a result, there is an increasing need for accurate predictive in vitro assays that reliably detect hepatotoxic drug candidates while reducing drug discovery time, costs, and the number of animal experiments. In vitro hepatocyte-based research has led to an improved comprehension of the underlying mechanisms of chemical toxicity and can assist the prioritization of therapeutic choices with low hepatotoxicity risk. Therefore, several in vitro systems have been generated over the last few decades. This review aims to comprehensively present the development and validation of 2D (two-dimensional) and 3D (three-dimensional) culture approaches on hepatotoxicity screening of compounds and highlight the main factors affecting predictive power of experiments. To this end, we first summarize some of the recognized hepatotoxicity mechanisms and related assays used to appraise DILI mechanisms and then discuss the challenges and limitations of in vitro models.
Collapse
Affiliation(s)
- Maryam Mirahmad
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Reyhaneh Sabourian
- Department of Drug and Food Control, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Mahdavi
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Maliheh Safavi
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran
| |
Collapse
|
21
|
Oxidative-stress and long-term hepatotoxicity: comparative study in Upcyte human hepatocytes and hepaRG cells. Arch Toxicol 2022; 96:1021-1037. [PMID: 35156134 DOI: 10.1007/s00204-022-03236-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/24/2022] [Indexed: 12/14/2022]
Abstract
Drug-induced liver injury (DILI) is one of the most common and serious adverse drug reactions and a major cause of drug development failure and withdrawal. Although different molecular mechanisms are implicated in DILI, enhanced ROS levels have been described as a major mechanism. Human-derived cell models are increasingly used in preclinical safety assessment because they provide quick and relatively inexpensive information in early stages of drug development. We have analyzed and compared the phenotype and functionality of two liver cell models (Upcyte human hepatocytes and HepaRG cells) to demonstrate their suitability for long-term hepatotoxicity assessments and mechanistic studies. The transcriptomic and functional analysis revealed the maintenance of phase I and phase II enzymes, and antioxidant enzymes along time in culture, although the differences found between both test systems underlie the differential sensitivity to hepatotoxins. The evaluation of several mechanisms of cell toxicity, including oxidative stress, by high-content screening, demonstrated that, by combining the stable phenotype of liver cells and repeated-dose exposure regimes to 12 test compounds at clinically relevant concentrations, both Upcyte hepatocytes and HepaRG offer suitable properties to be used in routine screening assays for toxicological assessments during drug preclinical testing.
Collapse
|
22
|
CHCHD2 Regulates Mitochondrial Function and Apoptosis of Ectopic Endometrial Stromal Cells in the Pathogenesis of Endometriosis. Reprod Sci 2022; 29:2152-2164. [DOI: 10.1007/s43032-021-00831-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/12/2021] [Indexed: 10/19/2022]
|
23
|
Rampa KM, Van De Venter M, Koekemoer TC, Swanepoel B, Venables L, Hattingh AC, Viljoen AM, Kamatou GP. Exploring four South African Croton species for potential anti-inflammatory properties: in vitro activity and toxicity risk assessment. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114596. [PMID: 34492319 DOI: 10.1016/j.jep.2021.114596] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/24/2021] [Accepted: 09/01/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The African Continent harbours approximately 26 Croton species. Many Croton species are used in traditional medicine in southern Africa to treat a variety of ailments including malaria, tuberculosis, microbial infection and inflammation. Considering the high diversity of the genus Croton, the ethnopharmacological information available on southern African species is rather limited. Furthermore, the potential for novel anti-inflammatory drug scaffolds has not previously been investigated. AIM OF THE STUDY The aim of the study was to evaluate the potential of four South African Croton species extracts (Croton gratissimus, Croton pseudopulchellus, Croton sylvaticus, and Croton steenkampianus) for anti-inflammatory activity targeting the TLR4 signalling pathway and to assess the potential risk for hepatotoxicity and genotoxicity using an in vitro cellomics approach. MATERIAL AND METHODS Leaf extracts of C. gratissimus, C. pseudopulchellus, C. sylvaticus and C. steenkampianus were prepared using methanol and chloroform (1:1, v/v). The anti-inflammatory activity was determined using LPS induced nitric oxide production in RAW 264.7 macrophages, while the hepatotoxicity and genotoxicity was evaluated using multi-parameter end point analysis in C3A and Vero cells, respectively. Mitochondrial membrane potential, mitochondrial mass, oxidative stress, lysosomal content and lipid accumulation were used as markers to assess the risk for hepatotoxicity. RESULTS All four species attenuated nitric oxide production with negligible cytotoxicity. However, C. gratissimus yielded the most favorable profile. Cell density was significantly reduced in both C3A and Vero cells with the C. gratissimus extract providing a suitable toxicity profile amenable to further high content analysis. While there was no meaningful effect on mitochondrial dynamics, a strong dose dependent increase in lipid content, paralleled by an expansion of the lysosomal compartment, identifies a potential risk for steatosis. Risk for genotoxicity was investigated using the micronucleus assay which revealed a dose dependent increase in micronuclei formation. Changes in nuclear morphology and cell ploidy further strengthens the associated risk for genotoxicity and suggests the extract from C. gratissimus may function as an aneugen. Collectively, the data demonstrates that although the selected species possess anti-inflammatory components, the risk for possible hepatotoxic and genotoxic side effects may negate their prospect towards further drug development.
Collapse
Affiliation(s)
- Khumo M Rampa
- Department of Pharmaceutical Sciences, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
| | - Maryna Van De Venter
- Department of Biochemistry and Microbiology, PO Box 77000, Nelson Mandela University, 6031, South Africa
| | - Trevor C Koekemoer
- Department of Biochemistry and Microbiology, PO Box 77000, Nelson Mandela University, 6031, South Africa
| | - Bresler Swanepoel
- Department of Biochemistry and Microbiology, PO Box 77000, Nelson Mandela University, 6031, South Africa
| | - Luanne Venables
- Department of Biochemistry and Microbiology, PO Box 77000, Nelson Mandela University, 6031, South Africa
| | - Anna C Hattingh
- Department of Biochemistry and Microbiology, PO Box 77000, Nelson Mandela University, 6031, South Africa
| | - Alvaro M Viljoen
- Department of Pharmaceutical Sciences, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa; SAMRC Herbal Drugs Research Unit, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
| | - Guy P Kamatou
- Department of Pharmaceutical Sciences, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa; SAMRC Herbal Drugs Research Unit, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa.
| |
Collapse
|
24
|
Guo Z, Li P, Wang C, Kang Q, Tu C, Jiang B, Zhang J, Wang W, Wang T. Five Constituents Contributed to the Psoraleae Fructus-Induced Hepatotoxicity via Mitochondrial Dysfunction and Apoptosis. Front Pharmacol 2021; 12:682823. [PMID: 34950022 PMCID: PMC8688997 DOI: 10.3389/fphar.2021.682823] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 11/08/2021] [Indexed: 01/22/2023] Open
Abstract
Backgrounds: Psoraleae Fructus (PF)-induced hepatotoxicity has been reported in clinical and animal experiments. However, the hepatotoxic constituents and mechanisms underlying PF-induced toxicity have remained unclear. Therefore, this study explored the potentially toxic PF components and revealed their relative mechanisms. Methods: The hepatotoxicity of PF water (PFW) and ethanol (PFE) extracts was compared using Kunming mice. The different compositions between PFW and PFE, which were considered toxic compositions, were identified using the UHPLC-Q-Exactive MS method. Then, L02 and HepG2 cell lines were used to evaluate the toxicity of these compositions. Cell viability and apoptosis were determined through the Cell Counting Kit-8 (CCK-8) assay and flow cytometry, respectively. An automatic biochemical analyzer detected the aspartate aminotransferase (AST), alanine aminotransferase (ALT), and alkaline phosphatase (ALP). Lastly, we used high-content screening (HCS) to determine the levels of reactive oxygen species (ROS), lipid, and mitochondrial membrane potential (MMP). Results: The ethanol extraction process aggravated the hepatotoxicity of PF, causing more severe injuries. The content of psoralen, isopsoralen, bavachin, psoralidin, bavachinin, neobavaisoflavone, and bakuchiol was higher in the PFE than PFW. Bavachin, psoralidin, bavachinin, neobavaisoflavone, and bakuchiol induced cell apoptosis and the AST, ALT, and ALP leakages. Furthermore, these five constituents increased intracellular lipid accumulation and ROS levels but decreased the MMP level. Conclusion: The ethanol extraction process could induce severe PF hepatotoxicity. Bavachin, psoralidin, bavachinin, neobavaisoflavone, and bakuchiol are the main hepatotoxic ingredients. This mechanism could be associated with oxidative stress and mitochondrial damage-mediated apoptosis. Taken together, this study provides a basis for the clinical application of PF that formulates and improves its herbal standards.
Collapse
Affiliation(s)
- Zhaojuan Guo
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Pin Li
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Chunguo Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Qianjun Kang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Can Tu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Bingqian Jiang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Jingxuan Zhang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Weiling Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Ting Wang
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China.,NMPA Key Laboratory for Research and Evaluation of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
25
|
Segovia-Zafra A, Di Zeo-Sánchez DE, López-Gómez C, Pérez-Valdés Z, García-Fuentes E, Andrade RJ, Lucena MI, Villanueva-Paz M. Preclinical models of idiosyncratic drug-induced liver injury (iDILI): Moving towards prediction. Acta Pharm Sin B 2021; 11:3685-3726. [PMID: 35024301 PMCID: PMC8727925 DOI: 10.1016/j.apsb.2021.11.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Idiosyncratic drug-induced liver injury (iDILI) encompasses the unexpected harms that prescription and non-prescription drugs, herbal and dietary supplements can cause to the liver. iDILI remains a major public health problem and a major cause of drug attrition. Given the lack of biomarkers for iDILI prediction, diagnosis and prognosis, searching new models to predict and study mechanisms of iDILI is necessary. One of the major limitations of iDILI preclinical assessment has been the lack of correlation between the markers of hepatotoxicity in animal toxicological studies and clinically significant iDILI. Thus, major advances in the understanding of iDILI susceptibility and pathogenesis have come from the study of well-phenotyped iDILI patients. However, there are many gaps for explaining all the complexity of iDILI susceptibility and mechanisms. Therefore, there is a need to optimize preclinical human in vitro models to reduce the risk of iDILI during drug development. Here, the current experimental models and the future directions in iDILI modelling are thoroughly discussed, focusing on the human cellular models available to study the pathophysiological mechanisms of the disease and the most used in vivo animal iDILI models. We also comment about in silico approaches and the increasing relevance of patient-derived cellular models.
Collapse
Affiliation(s)
- Antonio Segovia-Zafra
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
| | - Daniel E. Di Zeo-Sánchez
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| | - Carlos López-Gómez
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga 29010, Spain
| | - Zeus Pérez-Valdés
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| | - Eduardo García-Fuentes
- Unidad de Gestión Clínica de Aparato Digestivo, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Universitario Virgen de la Victoria, Málaga 29010, Spain
| | - Raúl J. Andrade
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
| | - M. Isabel Lucena
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
- Centro de Investigación Biomédica en Red en el Área Temática de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid 28029, Spain
- Platform ISCIII de Ensayos Clínicos, UICEC-IBIMA, Málaga 29071, Spain
| | - Marina Villanueva-Paz
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga 29071, Spain
| |
Collapse
|
26
|
Rocco R, Cambindo Botto AE, Muñoz MJ, Reingruber H, Wainstok R, Cochón A, Gazzaniga S. Early redox homeostasis disruption contributes to the differential cytotoxicity of imiquimod on transformed and normal endothelial cells. Exp Dermatol 2021; 31:608-614. [PMID: 34758172 DOI: 10.1111/exd.14499] [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: 03/26/2021] [Revised: 11/01/2021] [Accepted: 11/06/2021] [Indexed: 11/28/2022]
Abstract
The off-label use of imiquimod (IQ) for hemangioma treatment has shown clinical benefits. We have previously reported a selective direct IQ-cytotoxic effect on transformed (H5V) vs. normal (1G11) endothelial cells (EC). In the present study, we investigated the mechanism underlying this selective cytotoxicity in terms of TLR7/8 receptor expression, NF-κB signalling and time-dependent modifications of oxidative stress parameters (ROS: reactive oxygen species, catalase and superoxide dismutase activities, GSH/GSSG and lipid peroxidation). TLR7/8 level was extremely low in both cell lines, and IQ did not upregulate TLR7/8 expression or activate NF-κB signalling. IQ significantly induced ROS in H5V after 2 h and strongly affected antioxidant defenses. After 12 h, enzyme activities were restored to baseline levels but a robust drop in GSH/GSSG persisted together with increased lipid peroxidation levels and a marked mitochondrial dysfunction. Although in normal IQ-treated EC some oxidative stress parameters were affected after 4 h, mitochondrial health and GSH/GSSG ratio remained notably unaffected after 12 h. Therefore, the early alterations (0-2 h) in transformed EC breached redox homeostasis as strongly as to enhance their susceptibility to IQ. This interesting facet of IQ as redox disruptor could broaden its therapeutic potential for other skin malignancies, alone or in adjuvant schemes.
Collapse
Affiliation(s)
- Rodrigo Rocco
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Adrián E Cambindo Botto
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina
| | - Manuel J Muñoz
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Instituto de Fisiología, Biología Molecular y Neurociencias (IFIBYNE-UBA-CONICET), Universidad de Buenos Aires, Ciudad Universitaria, Buenos Aires, Argentina.,Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy.,Departamento de Biodiversidad y Biología Experimental, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Hernán Reingruber
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Rosa Wainstok
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires, Argentina.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Adriana Cochón
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Silvina Gazzaniga
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
27
|
Xu Q. Human Three-Dimensional Hepatic Models: Cell Type Variety and Corresponding Applications. Front Bioeng Biotechnol 2021; 9:730008. [PMID: 34631680 PMCID: PMC8497968 DOI: 10.3389/fbioe.2021.730008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/30/2021] [Indexed: 12/23/2022] Open
Abstract
Owing to retained hepatic phenotypes and functions, human three-dimensional (3D) hepatic models established with diverse hepatic cell types are thought to recoup the gaps in drug development and disease modeling limited by a conventional two-dimensional (2D) cell culture system and species-specific variability in drug metabolizing enzymes and transporters. Primary human hepatocytes, human hepatic cancer cell lines, and human stem cell-derived hepatocyte-like cells are three main hepatic cell types used in current models and exhibit divergent hepatic phenotypes. Primary human hepatocytes derived from healthy hepatic parenchyma resemble in vivo-like genetic and metabolic profiling. Human hepatic cancer cell lines are unlimitedly reproducible and tumorigenic. Stem cell-derived hepatocyte-like cells derived from patients are promising to retain the donor's genetic background. It has been suggested in some studies that unique properties of cell types endue them with benefits in different research fields of in vitro 3D modeling paradigm. For instance, the primary human hepatocyte was thought to be the gold standard for hepatotoxicity study, and stem cell-derived hepatocyte-like cells have taken a main role in personalized medicine and regenerative medicine. However, the comprehensive review focuses on the hepatic cell type variety, and corresponding applications in 3D models are sparse. Therefore, this review summarizes the characteristics of different cell types and discusses opportunities of different cell types in drug development, liver disease modeling, and liver transplantation.
Collapse
Affiliation(s)
- Qianqian Xu
- School of Chinese Medicine, and Centre for Cancer and Inflammation Research, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
28
|
Hofer T. Oxidative Stress in Human Toxicology. Antioxidants (Basel) 2021; 10:antiox10081159. [PMID: 34439406 PMCID: PMC8388969 DOI: 10.3390/antiox10081159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 11/25/2022] Open
Affiliation(s)
- Tim Hofer
- Department of Environmental Health, Norwegian Institute of Public Health, P.O. Box 222 Skøyen, N-0213 Oslo, Norway
| |
Collapse
|
29
|
Hedenmalm K, Pacurariu A, Slattery J, Kurz X, Candore G, Flynn R. Is There an Increased Risk of Hepatotoxicity with Metamizole? A Comparative Cohort Study in Incident Users. Drug Saf 2021; 44:973-985. [PMID: 34273099 DOI: 10.1007/s40264-021-01087-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2021] [Indexed: 11/27/2022]
Abstract
INTRODUCTION The analgesic metamizole, which has been withdrawn from the market in several countries due to the risk of agranulocytosis but is still available on the market in Germany and some other countries, has been associated with liver injury in published case reports; however, epidemiological studies on the risk of liver injury are limited. OBJECTIVE The aim of this study was to compare the risk of liver injury up to 270 days after the first start of treatment with metamizole with the corresponding risk in patients starting treatment with paracetamol, using a retrospective cohort incident user design. METHODS The first prescription for either metamizole or paracetamol in the Intercontinental Medical Statistics (IMS)® Disease Analyzer Germany database during the study period (2009-2018) was identified in patients with at least 365 days of observation and no prior diagnosis of liver events, cancer or HIV, or treatment within the last 6 months with hepatotoxic drugs typically administered for chronic conditions. Each patient was followed for specific liver events for 90 days after the prescription. In case of a new prescription within 90 days, a new 90-day observation period started, up to a maximum of 270 days. Cox regression was used to compare the risk of liver injury in the two groups. RESULTS Metamizole was associated with a higher risk of liver injury compared with paracetamol (adjusted hazard ratio 1.69, 95% confidence interval 1.46-1.97). Sensitivity analyses were performed to evaluate the robustness of these findings. In all the sensitivity analyses, metamizole was still associated with a higher risk of liver injury, including an analysis where naproxen was used as a comparator instead of paracetamol. CONCLUSIONS Results from this study support previous studies suggesting that metamizole is associated with a significant risk of liver injury. Nevertheless, a possible impact of residual confounding cannot be excluded.
Collapse
Affiliation(s)
- Karin Hedenmalm
- Data Analytics and Methods Task force, European Medicines Agency, Domenico Scarlattilaan 6, 1083 HS, Amsterdam, The Netherlands. .,Department of Laboratory Medicine, Karolinska Institutet, Solna, Stockholm, Sweden.
| | - Alexandra Pacurariu
- Pharmacovigilance and Epidemiology Department, European Medicines Agency, Amsterdam, The Netherlands
| | - Jim Slattery
- Data Analytics and Methods Task force, European Medicines Agency, Domenico Scarlattilaan 6, 1083 HS, Amsterdam, The Netherlands
| | - Xavier Kurz
- Data Analytics and Methods Task force, European Medicines Agency, Domenico Scarlattilaan 6, 1083 HS, Amsterdam, The Netherlands
| | - Gianmario Candore
- Data Analytics and Methods Task force, European Medicines Agency, Domenico Scarlattilaan 6, 1083 HS, Amsterdam, The Netherlands
| | - Rob Flynn
- Data Analytics and Methods Task force, European Medicines Agency, Domenico Scarlattilaan 6, 1083 HS, Amsterdam, The Netherlands
| |
Collapse
|
30
|
Enzyme-Treated Zizania latifolia Extract Protects against Alcohol-Induced Liver Injury by Regulating the NRF2 Pathway. Antioxidants (Basel) 2021; 10:antiox10060960. [PMID: 34203789 PMCID: PMC8232714 DOI: 10.3390/antiox10060960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/12/2022] Open
Abstract
Binge drinking patterns easily produce a state of oxidative stress that disturbs liver function. Eventually, this leads to alcoholic liver disease. A safe and effective therapy for alcoholic liver disease remains elusive. Enzyme-treated Z. latifolia extract (ETZL) was studied as a potential agent for treating alcohol-induced liver disease. In addition, its underlying mechanisms were elucidated. In the binge model, ETZL was pretreated with alcohol (5 g/kg) three times at 12-h intervals. Our results showed that ETZL pretreatment decreased the serum levels of ALT, AST, ALP, and TG. ETZL treatment appeared to prevent an increase in hepatic TG and MDA levels, and there was a decrease in total GSH following alcohol treatment. Histopathological examination showed that lipid droplets were significantly reduced in the ETZL group compared to the control group. ETZL also exhibited radical scavenging activity. It significantly reduced t-BHP-induced cytotoxicity and the production of reactive oxygen species (ROS) in HepG2 cells. ETZL also enhanced NRF2 nuclear translocation and increased expression of the downstream target genes HO-1, NQO1, and GCLC as an antioxidant defense. Finally, ETZL treatment significantly reduced cell death. Our study suggests that ETZL ameliorates binge ethanol-induced liver injury by upregulating the antioxidant defense mechanism.
Collapse
|
31
|
Villanueva-Paz M, Morán L, López-Alcántara N, Freixo C, Andrade RJ, Lucena MI, Cubero FJ. Oxidative Stress in Drug-Induced Liver Injury (DILI): From Mechanisms to Biomarkers for Use in Clinical Practice. Antioxidants (Basel) 2021; 10:390. [PMID: 33807700 PMCID: PMC8000729 DOI: 10.3390/antiox10030390] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 03/02/2021] [Indexed: 12/11/2022] Open
Abstract
Idiosyncratic drug-induced liver injury (DILI) is a type of hepatic injury caused by an uncommon drug adverse reaction that can develop to conditions spanning from asymptomatic liver laboratory abnormalities to acute liver failure (ALF) and death. The cellular and molecular mechanisms involved in DILI are poorly understood. Hepatocyte damage can be caused by the metabolic activation of chemically active intermediate metabolites that covalently bind to macromolecules (e.g., proteins, DNA), forming protein adducts-neoantigens-that lead to the generation of oxidative stress, mitochondrial dysfunction, and endoplasmic reticulum (ER) stress, which can eventually lead to cell death. In parallel, damage-associated molecular patterns (DAMPs) stimulate the immune response, whereby inflammasomes play a pivotal role, and neoantigen presentation on specific human leukocyte antigen (HLA) molecules trigger the adaptive immune response. A wide array of antioxidant mechanisms exists to counterbalance the effect of oxidants, including glutathione (GSH), superoxide dismutase (SOD), catalase, and glutathione peroxidase (GPX), which are pivotal in detoxification. These get compromised during DILI, triggering an imbalance between oxidants and antioxidants defense systems, generating oxidative stress. As a result of exacerbated oxidative stress, several danger signals, including mitochondrial damage, cell death, and inflammatory markers, and microRNAs (miRNAs) related to extracellular vesicles (EVs) have already been reported as mechanistic biomarkers. Here, the status quo and the future directions in DILI are thoroughly discussed, with a special focus on the role of oxidative stress and the development of new biomarkers.
Collapse
Affiliation(s)
- Marina Villanueva-Paz
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, CIBERehd, 29071 Málaga, Spain; (M.V.-P.); (M.I.L.)
| | - Laura Morán
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 28040 Madrid, Spain; (L.M.); (N.L.-A.)
- Health Research Institute Gregorio Marañón (IiSGM), 28009 Madrid, Spain
| | - Nuria López-Alcántara
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 28040 Madrid, Spain; (L.M.); (N.L.-A.)
| | - Cristiana Freixo
- CINTESIS, Center for Health Technology and Services Research, do Porto University School of Medicine, 4200-319 Porto, Portugal;
| | - Raúl J. Andrade
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, CIBERehd, 29071 Málaga, Spain; (M.V.-P.); (M.I.L.)
| | - M Isabel Lucena
- Unidad de Gestión Clínica de Gastroenterología, Servicio de Farmacología Clínica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, CIBERehd, 29071 Málaga, Spain; (M.V.-P.); (M.I.L.)
| | - Francisco Javier Cubero
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, 28040 Madrid, Spain; (L.M.); (N.L.-A.)
- 12 de Octubre Health Research Institute (imas12), 28041 Madrid, Spain
| |
Collapse
|