1
|
Hsueh WY, Wu YL, Weng MT, Liu SY, Santavanond JP, Liu YC, Lin CI, Lai CN, Lu YR, Hsu JY, Gao HY, Lee JC, Wei SC, Lyu PC, Poon IKH, Hsieh HP, Chiu YH. Novel Naphthyridones Targeting Pannexin 1 for Colitis Management. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2411538. [PMID: 39739600 DOI: 10.1002/advs.202411538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/29/2024] [Indexed: 01/02/2025]
Abstract
Pannexin 1 (PANX1) forms cell-surface channels capable of releasing signaling metabolites for diverse patho-physiological processes. While inhibiting dysregulated PANX1 has been proposed as a therapeutic strategy for many pathological conditions, including inflammatory bowel disease (IBD), low efficacy, or poor specificity of classical PANX1 inhibitors introduces uncertainty for their applications in basic and translational research. Here, hit-to-lead optimization is performed and a naphthyridone, compound 12, is identified as a new PANX1 inhibitor with an IC50 of 0.73 µm that does not affect pannexin-homologous LRRC8/SWELL1 channels. Using structure-activity relationship analysis, mutagenesis, cell thermal shift assays, and molecular docking, it is revealed that compound 12 directly engages PANX1 Trp74 residue. Using a dextran sodium sulfate mouse model of IBD, it is found that compound 12 markedly reduced colitis severity, highlighting new PANX1 inhibitors as a proof-of-concept treatment for IBD. These data describe the mechanism of action for a new PANX1 inhibitor, uncover the binding site for future drug design, and present a targeted strategy for treating IBD.
Collapse
Affiliation(s)
- Wen-Yun Hsueh
- Department of Chemistry, National Tsing Hua University, Hsinchu, 300044, Taiwan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County, 350401, Taiwan
| | - Yi-Ling Wu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Meng-Tzu Weng
- Department of Internal Medicine, National Taiwan University Hospital, Taipei City, 100229, Taiwan
- Department of Medical Research, National Taiwan University Hospital, Hsin-Chu Branch, Hsinchu, 302058, Taiwan
| | - Shin-Yun Liu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei City, 100229, Taiwan
| | - Jascinta P Santavanond
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, VIC, 3086, Australia
| | - Yi-Chung Liu
- Institute of Population Health Sciences, National Health Research Institutes, Miaoli County, 350401, Taiwan
| | - Ching-I Lin
- Department of Internal Medicine, National Taiwan University Hospital, Taipei City, 100229, Taiwan
| | - Cheng-Nong Lai
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Yi-Ru Lu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Jing Yin Hsu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Hong-Yu Gao
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Jinq-Chyi Lee
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County, 350401, Taiwan
| | - Shu-Chen Wei
- Department of Internal Medicine, National Taiwan University Hospital, Taipei City, 100229, Taiwan
| | - Ping-Chiang Lyu
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - Ivan K H Poon
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
- Research Centre for Extracellular Vesicles, La Trobe University, VIC, 3086, Australia
| | - Hsing-Pang Hsieh
- Department of Chemistry, National Tsing Hua University, Hsinchu, 300044, Taiwan
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County, 350401, Taiwan
| | - Yu-Hsin Chiu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, 300044, Taiwan
- Departments of Medical Science, Life Science, and Medicine, National Tsing Hua University, Hsinchu, 300044, Taiwan
| |
Collapse
|
2
|
Liu W, Zeng X, Wang X, Hu Y, Chen L, Luo N, Ouyang D, Rao T. 2,3,5,4'- tetrahydroxystilbene-2-O-β-D- glucopyranoside (TSG)-Driven immune response in the hepatotoxicity of Polygonum multiflorum. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117865. [PMID: 38369066 DOI: 10.1016/j.jep.2024.117865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 01/31/2024] [Accepted: 02/03/2024] [Indexed: 02/20/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE 2,3,5,4'-tetrahydroxystilbene-2-O-β-D-glucopyranoside (TSG) as the primary constituent of Polygonum multiflorum Thumb. (PM) possesses anti-oxidative, antihypercholesterolemic, anti-tumor and many more biological activities. The root of PM has been used as a tonic medicine for thousands of years. However, cases of PM-induced liver injury are occasionally reported, and considered to be related to the host immune status. AIM OF THE STUDY The primary toxic elements and specific mechanisms PM causing liver damage are still not thoroughly clear. Our study aimed to investigate the influences of TSG on the immune response in idiosyncratic hepatotoxicity of PM. MATERIALS AND METHODS The male C57BL/6 mice were treated with different doses of TSG and the alterations in liver histology, serum liver enzyme levels, proportions of T cells and cytokines secretion were evaluated by hematoxylin and eosin (HE), RNA sequencing, quantitative real time polymerase chain reaction (qRT-PCR), Flow cytometry (FCM), and enzyme-linked immunosorbent assay (ELISA), respectively. Then, primary spleen cells from drug-naive mice were isolated and cultured with TSG in vitro. T cell subsets proliferation and cytokines secretion after treated with TSG were assessed by CCK8, FCM and ELISA. In addition, mice were pre-treated with anti-CD25 for depleting regulatory T cells (Tregs), and then administered with TSG. Liver functions and immunological alterations were analyzed to evaluate liver injury. RESULTS Data showed that TSG induced liver damage, and immune cells infiltration in the liver tissues. FCM results showed that TSG could activate CD4+T and CD8+T in the liver. Results further confirmed that TSG notably up-regulated the levels of inflammatory cytokines including TNF-α, IFN-γ, IL-18, perforin and granzyme B in the liver tissues. Furthermore, based on transcriptomics profiles, some immune system-related pathways including leukocyte activation involved in inflammatory response, leukocyte cell-cell adhesion, regulation of interleukin-1 beta production, mononuclear cell migration, antigen processing and presentation were altered in TSG treated mice. CD8+T/CD4+T cells were also stimulated by TSG in vitro. Interestingly, increased proportion of Tregs was observed after TSG treatment in vitro and in vivo. Foxp3 and TGF-β1 mRNA expressions were up-regulated in the liver tissues. Depletion of Tregs moderately enhanced TSG induced the secretion of inflammatory cytokines in serum. CONCLUSIONS Our findings showed that TSG could trigger CD4+T and CD8+T cells proliferation, promote cytokines secretion, which revealed that adaptive immune response associated with the mild liver injury cause by TSG administration. Regulatory T cells (Tregs) mainly sustain immunological tolerance, and in this study, the progression of TSG induced liver injury was limited by Tregs. The results of our investigations allow us to preliminarily understand the mechanisms of PM related idiosyncratic hepatotoxicity.
Collapse
Affiliation(s)
- Wenhui Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan Province, 410078, China; Department of Clinical Laboratory, The Second Affiliated Hospital of Guilin Medical University, Guilin, Guangxi Province, 541001, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan Province, 410078, China
| | - Xiangchang Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan Province, 410078, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan Province, 410078, China; Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, Hunan Province, 410221, China
| | - Xinfeng Wang
- Department of Human Anatomy, College of Basic Medicine, Guilin Medical University, Guilin, Guangxi Province, 541199, China
| | - Yuwei Hu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan Province, 410078, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan Province, 410078, China
| | - Lulu Chen
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, Hunan Province, 410221, China
| | - Naixiang Luo
- Department of Immunology, College of Basic Medicine, Guilin Medical University, Guilin, Guangxi Province, 541199, China.
| | - Dongsheng Ouyang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan Province, 410078, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan Province, 410078, China; Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha Duxact Biotech Co., Ltd., Changsha, Hunan Province, 410221, China.
| | - Tai Rao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan Province, 410078, China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan Province, 410078, China.
| |
Collapse
|
3
|
Chen S, Li X, Li Y, He X, Bryant M, Qin X, Li F, Seo JE, Guo X, Mei N, Guo L. The involvement of hepatic cytochrome P450s in the cytotoxicity of lapatinib. Toxicol Sci 2023; 197:69-78. [PMID: 37788138 PMCID: PMC10734604 DOI: 10.1093/toxsci/kfad099] [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: 10/05/2023] Open
Abstract
Lapatinib, an oral tyrosine kinase inhibitor used as a first-line treatment for HER2-positive breast cancer, has been reported to be associated with hepatotoxicity; however, the underlying mechanisms remain unclear. In this study, we report that lapatinib causes cytotoxicity in multiple types of hepatic cells, including primary human hepatocytes, HepaRG cells, and HepG2 cells. A 24-h treatment with lapatinib induced cell cycle disturbances, apoptosis, and DNA damage, and decreased the protein levels of topoisomerase in HepG2 cells. We investigated the role of cytochrome P450 (CYP)-mediated metabolism in lapatinib-induced cytotoxicity using our previously established HepG2 cell lines, which express each of 14 CYPs (1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 3A4, 3A5, and 3A7). We demonstrate that lapatinib is metabolized by CYP1A1, 3A4, 3A5, and 3A7. Among these, lapatinib-induced cytotoxicity and DNA damage were attenuated in cells overexpressing CYP3A5 or 3A7. Additionally, we measured the production of three primary metabolites of lapatinib (O-dealkylated lapatinib, N-dealkylated lapatinib, and N-hydroxy lapatinib) in CYP1A1-, 3A4-, 3A5-, and 3A7-overexpressing HepG2 cells. We compared the cytotoxicity of lapatinib and its 3 metabolites in primary human hepatocytes, HepaRG cells, and HepG2 cells and demonstrated that N-dealkylated lapatinib is more toxic than the parent drug and the other metabolites. Taken together, our results indicate that lapatinib-induced cytotoxicity involves multiple mechanisms, such as apoptosis and DNA damage; that N-dealkylated lapatinib is a toxic metabolite contributing to the toxic effect of lapatinib; and that CYP3A5- and 3A7-mediated metabolism plays a role in attenuating the cytotoxicity of lapatinib.
Collapse
Affiliation(s)
- Si Chen
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079, USA
| | - Xilin Li
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079, USA
| | - Yuxi Li
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079, USA
| | - Xiaobo He
- Office of Scientific Coordination, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079, USA
| | - Matthew Bryant
- Office of Scientific Coordination, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079, USA
| | - Xuan Qin
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Feng Li
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Ji-Eun Seo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079, USA
| | - Xiaoqing Guo
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079, USA
| | - Nan Mei
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079, USA
| | - Lei Guo
- Division of Biochemical Toxicology, National Center for Toxicological Research (NCTR), U.S. Food and Drug Administration (FDA), Jefferson, Arkansas 72079, USA
| |
Collapse
|
4
|
Mino T, Ureshino H, Ueshima T, Kashimoto N, Yamaguchi T, Naka K, Inaba T, Ichinohe T. A novel anticancer quinolone, (R)-WAC-224, has anti-leukemia activities against acute myeloid leukemia. Invest New Drugs 2023; 41:751-760. [PMID: 37702844 DOI: 10.1007/s10637-023-01393-0] [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: 08/03/2023] [Accepted: 09/08/2023] [Indexed: 09/14/2023]
Abstract
Approximately 60%-80% of patients who achieve complete remission eventually relapse after conventional chemotherapy and have poor prognoses despite the recent advances of novel anticancer agents. Continuing development of more effective novel treatments for acute myeloid leukemia (AML) is necessary. We developed (R)-WAC-224 (R-WAC), which is an anticancer quinolone, targeting topoisomerase II. This study evaluated the anti-leukemia potential of R-WAC or racemic WAC-224 (WAC) in vitro and in vivo. R-WAC significantly inhibited the human AML cell line proliferation (MV4-11, HL60, and KG1a), which was comparable to daunorubicin and cytarabine, not affected by P-glycoprotein overexpression. WAC did neither increase serum troponin-T nor decrease the crypt numbers in the small intestine, indicating WAC was less toxic than doxorubicin. R-WAC monotherapy demonstrated prolonged survival in the AML mice model and inhibited tumor growth in the MV4-11 xenograft mice model. Moreover, the combination of R-WAC and cytarabine demonstrated more active anti-leukemia effects than daunorubicin and cytarabine. Finally, R-WAC inhibited the colony-forming abilities using primary AML cells. These results indicate that R-WAC is a promising therapeutic agent for AML.
Collapse
Affiliation(s)
- Tatsuji Mino
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Hiroshi Ureshino
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.
| | - Taichi Ueshima
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co., Ltd, Hiroshima, Japan
| | - Naoki Kashimoto
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co., Ltd, Hiroshima, Japan
| | - Tomonori Yamaguchi
- Drug Discovery Laboratory, Wakunaga Pharmaceutical Co., Ltd, Hiroshima, Japan
| | - Kazuhito Naka
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Toshiya Inaba
- Department of Molecular Oncology and Leukemia Program Project, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
5
|
Dong Y, Qi Y, Jiang H, Mi T, Zhang Y, Peng C, Li W, Zhang Y, Zhou Y, Zang Y, Li J. The development and benefits of metformin in various diseases. Front Med 2023; 17:388-431. [PMID: 37402952 DOI: 10.1007/s11684-023-0998-6] [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: 12/23/2022] [Accepted: 04/01/2023] [Indexed: 07/06/2023]
Abstract
Metformin has been used for the treatment of type II diabetes mellitus for decades due to its safety, low cost, and outstanding hypoglycemic effect clinically. The mechanisms underlying these benefits are complex and still not fully understood. Inhibition of mitochondrial respiratory-chain complex I is the most described downstream mechanism of metformin, leading to reduced ATP production and activation of AMP-activated protein kinase (AMPK). Meanwhile, many novel targets of metformin have been gradually discovered. In recent years, multiple pre-clinical and clinical studies are committed to extend the indications of metformin in addition to diabetes. Herein, we summarized the benefits of metformin in four types of diseases, including metabolic associated diseases, cancer, aging and age-related diseases, neurological disorders. We comprehensively discussed the pharmacokinetic properties and the mechanisms of action, treatment strategies, the clinical application, the potential risk of metformin in various diseases. This review provides a brief summary of the benefits and concerns of metformin, aiming to interest scientists to consider and explore the common and specific mechanisms and guiding for the further research. Although there have been countless studies of metformin, longitudinal research in each field is still much warranted.
Collapse
Affiliation(s)
- Ying Dong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yingbei Qi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Haowen Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tian Mi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yunkai Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chang Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wanchen Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongmei Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Yubo Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Yi Zang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Lingang Laboratory, Shanghai, 201203, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Open Studio for Druggability Research of Marine Natural Products, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao, 266237, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China.
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, 264117, China.
| |
Collapse
|
6
|
Liu XC, Zhou PK. Tissue Reactions and Mechanism in Cardiovascular Diseases Induced by Radiation. Int J Mol Sci 2022; 23:ijms232314786. [PMID: 36499111 PMCID: PMC9738833 DOI: 10.3390/ijms232314786] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
The long-term survival rate of cancer patients has been increasing as a result of advances in treatments and precise medical management. The evidence has accumulated that the incidence and mortality of non-cancer diseases have increased along with the increase in survival time and long-term survival rate of cancer patients after radiotherapy. The risk of cardiovascular disease as a radiation late effect of tissue damage reactions is becoming a critical challenge and attracts great concern. Epidemiological research and clinical trials have clearly shown the close association between the development of cardiovascular disease in long-term cancer survivors and radiation exposure. Experimental biological data also strongly supports the above statement. Cardiovascular diseases can occur decades post-irradiation, and from initiation and development to illness, there is a complicated process, including direct and indirect damage of endothelial cells by radiation, acute vasculitis with neutrophil invasion, endothelial dysfunction, altered permeability, tissue reactions, capillary-like network loss, and activation of coagulator mechanisms, fibrosis, and atherosclerosis. We summarize the most recent literature on the tissue reactions and mechanisms that contribute to the development of radiation-induced cardiovascular diseases (RICVD) and provide biological knowledge for building preventative strategies.
Collapse
|
7
|
Tu C, Gao Y, Song D, Niu M, Ma RR, Zhou MX, He X, Xiao XH, Wang JB. Screening for Susceptibility-Related Biomarkers of Diclofenac-Induced Liver Injury in Rats Using Metabolomics. Front Pharmacol 2021; 12:693928. [PMID: 34630079 PMCID: PMC8494976 DOI: 10.3389/fphar.2021.693928] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 08/03/2021] [Indexed: 11/22/2022] Open
Abstract
Early identification of individuals susceptible to idiosyncratic drug-induced liver injury (IDILI) is a challenging unmet demand. Diclofenac, one of the most widely available over-the-counter drugs for pain management worldwide, may induce liver dysfunction, acute liver failure, and death. Herein, we report that diclofenac-related hepatobiliary adverse reactions occurred more frequently in cases with immune activation. Furthermore, experiments with rats demonstrated divergent hepatotoxicity responses in individuals exposed to diclofenac, and modest inflammation potentiated diclofenac-induced liver injury. Susceptible rats had unique plasma metabolomic characteristics, and as such, the metabolomic approach could be used to distinguish susceptible individuals. The 23 identified susceptibility-related metabolites were enriched by several metabolic pathways related to acute-phase reactions of immunocytes and inflammatory responses, including sphingolipid, tyrosine, phenylalanine, tryptophan, and lipid metabolism pathways. This finding implies a mechanistic role of metabolic and immune disturbances affects susceptibility to diclofenac-IDILI. Further nine metabolite biomarkers with potent diagnostic capabilities were identified using receiver operating characteristic curves. These findings elucidated the potential utility of metabolomic biomarkers to identify individuals susceptible to drug hepatotoxicity and the underlying mechanism of metabolic and immune disturbances occurring in IDILI.
Collapse
Affiliation(s)
- Can Tu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Di Song
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ming Niu
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Run-Ran Ma
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Ming-Xi Zhou
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xian He
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xiao-He Xiao
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jia-Bo Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.,China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
8
|
Lin Y, Xiao R, Xia BH, Zhang ZM, Li C, Wu P, Liao DF, Lin LM. Investigation of the idiosyncratic hepatotoxicity of Polygonum multiflorum Thunb. through metabolomics using GC-MS. BMC Complement Med Ther 2021; 21:120. [PMID: 33845816 PMCID: PMC8043067 DOI: 10.1186/s12906-021-03276-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 03/15/2021] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND The idiosyncratic hepatotoxicity of Polygonum multiflorum (PM) has attracted considerable interest, but the idiosyncratically hepatotoxic components and endogenous metabolite changes resulting from idiosyncratic hepatotoxicity of PM are not well understood. The aim of this study was to identify the idiosyncratically hepatotoxic components and potential endogenous metabolic biomarkers for PM-induced liver injury. METHODS Serum biochemical indicators and hematoxylin and eosin (H&E) staining were evaluated to identify pathological changes. Gas chromatography/mass spectrometry (GC-MS) was performed to identify changes in metabolic biomarkers. Orthogonal projection to latent structures discriminant analysis (OPLS-DA) was applied to determine group clustering trends and differential metabolites. RESULTS The results for the liver index, the liver function index and liver pathology showed that Polygonum multiflorum ethanol extract (PME), 50% ethanol elution fractions and tetrahydroxystilbene glucoside (TSG) from PME can induce idiosyncratic hepatotoxicity. TSG was the main idiosyncratically hepatotoxic component. Forty endogenous metabolites were identified in the rat liver. Six biomarkers, including lower levels of L-valine and higher levels of 3-hydroxybutyric acid, hexadecanoic acid, ribose, phosphoric acid and oxalic acid, were related to PM-induced liver injury. These differential biomarkers led to disruptions in amino acid, fatty acid, oxalate, energy and glucose metabolism. A total of 32 types of endogenous metabolites were identified in rat serum. Ten biomarkers were related to the liver injury induced by TSG, including lower levels of L-valine and L-proline and higher levels of urea, caproic acid, DL-malic acid, D-mannose, 3-hydroxybutyric acid, D-galactose, octadecane and hexadecanoic acid. These differential biomarkers led to disruptions in amino acid, glucose and fat metabolism. The mechanism of idiosyncratic hepatotoxicity in PM involves TSG-induced disruptions in amino acid metabolism, lipid metabolism, energy metabolism and glucose metabolism. CONCLUSIONS These findings reflect the material basis and metabolic mechanism of idiosyncratic PM hepatotoxicity.
Collapse
Affiliation(s)
- Yan Lin
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Rong Xiao
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Bo-Hou Xia
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Zhi-Min Zhang
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chun Li
- China Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Ping Wu
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Duan-Fang Liao
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Li-Mei Lin
- College of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
9
|
Tu C, Niu M, Wei AW, Tang JF, Zhang L, Jing J, Xiao XH, Wang JB. Susceptibility-Related Cytokine Panel for Prediction of Polygonum multiflorum-Induced Hepatotoxicity in Humans. J Inflamm Res 2021; 14:645-655. [PMID: 33692634 PMCID: PMC7939510 DOI: 10.2147/jir.s299892] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/13/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Drug-induced liver injury is a common adverse effect in clinical practice, with severe cases resulting in liver failure and even death. Identification and prediction of individuals susceptible to idiosyncratic DILI continues to remain a challenge. METHODS In this study, we report that cytokines in human serum can be used to identify and predict individuals susceptible to Polygonum multiflorum-induced DILI (PM-DILI) in retrospective and prospective cohort studies. FINDINGS In the retrospective pilot study, we compared serum cytokine expression profiles of the PM-DILI group (n=10) and the PM-Tolerant group (n=12) and found 10 cytokines with significant differences. In the replication cohort study, differences in the 10 cytokines between PM-DILI (n =11) and PM-Tolerant (n=13) groups were verified. Among them, 6 cytokines showed no significant differences at two time points, including liver injury and recovery stage of PM-DILI, suggesting that these 6 cytokines have no correlation with PM-DILI, however, they may be related to susceptibility. Furthermore, all the retrospective cohorts were combined, and a PM-DILI susceptibility prediction model was built by screening the 6 cytokines. The combination of (TNF-α and CCL-2) or VEGF showed the highest sensitivity and specificity. Finally, the efficacy of the above 3 cytokine combination models in predicting PM-DILI-susceptible individuals was verified before PM exposure in another independent prospective cohort (n=24), with sensitivity and specificity of 66.7% and 83.3%, respectively. CONCLUSION This proof-of-concept study demonstrates that the serum cytokine combination reflecting dysimmunity could be used as a new method to predict PM-DILI, thus providing a new perspective for improving the clinical management of IDILI.
Collapse
Affiliation(s)
- Can Tu
- Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, People’s Republic of China
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People’s Republic of China
| | - Ming Niu
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People’s Republic of China
| | - Ai-Wu Wei
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| | - Jin-Fa Tang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, 450000, People’s Republic of China
| | - Le Zhang
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People’s Republic of China
| | - Jing Jing
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People’s Republic of China
| | - Xiao-He Xiao
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People’s Republic of China
| | - Jia-Bo Wang
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, People’s Republic of China
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, People’s Republic of China
| |
Collapse
|
10
|
Gao Y, Xu G, Ma L, Shi W, Wang Z, Zhan X, Qin N, He T, Guo Y, Niu M, Wang J, Bai Z, Xiao X. Icariside I specifically facilitates ATP or nigericin-induced NLRP3 inflammasome activation and causes idiosyncratic hepatotoxicity. Cell Commun Signal 2021; 19:13. [PMID: 33573688 PMCID: PMC7879676 DOI: 10.1186/s12964-020-00647-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 08/12/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Epimedii Folium (EF) is commonly used for treating bone fractures and joint diseases, but the potential hepatotoxicity of EF limits its clinical application. Our previous study confirms that EF could lead to idiosyncratic drug-induced liver injury (IDILI) and hepatocyte apoptosis, but the mechanism remains unknown. Studies have shown that NLRP3 inflammasome plays an important role in the development of various inflammatory diseases such as IDILI. Specific stimulus-induced NLRP3 inflammasome activation may has been a key strategy for lead to liver injury. Therefore, main compounds derived from EF were chosen to test whether the ingredients in EF could activate the NLRP3 inflammasome and to induce IDILI. METHODS Bone-marrow-derived macrophages (BMDMs) were treated with Icariside I, and then stimulated with inflammasome stimuli and assayed for the production of caspase-1 and interleukin 1β (IL-1β) and the release of lactate dehydrogenase (LDH). Determination of intracellular potassium, ASC oligomerization as well as reactive oxygen species (ROS) production were used to evaluate the stimulative mechanism of Icariside I on inflammasome activation. Mouse models of NLRP3 diseases were used to test whether Icariside I has hepatocyte apoptosis effects and promoted NLRP3 inflammasome activation in vivo. RESULTS Icariside I specifically enhances NLRP3 inflammasome activation triggered by ATP or nigericin but not SiO2, poly(I:C) or cytosolic LPS. Additionally, Icariside I does not alter the activation of NLRC4 and AIM2 inflammasomes. Mechanically, Icariside I alone does not induce mitochondrial reactive oxygen species (mtROS), which is one of the critical upstream events of NLRP3 inflammasome activation; however, Icariside I increases mtROS production induced by ATP or nigericin but not SiO2. Importantly, Icariside I leads to liver injury and NLRP3 inflammasome activation in an LPS-mediated susceptibility mouse model of IDILI, but the effect of Icariside I is absent in the LPS-mediated mouse model pretreated with MCC950, which is used to mimic knockdown of NLRP3 inflammasome activation. CONCLUSIONS Our study reveals that Icariside I specifically facilitates ATP or nigericin-induced NLRP3 inflammasome activation and causes idiosyncratic hepatotoxicity. The findings suggest that Icariside I or EF should be avoided in patients with diseases related to ATP or nigericin-induced NLRP3 inflammasome activation, which may be risk factors for IDILI. Video abstract.
Collapse
Affiliation(s)
- Yuan Gao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, No. 100 Xisihuan, Beijing, 100039, China
| | - Guang Xu
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, No. 100 Xisihuan, Beijing, 100039, China
| | - Li Ma
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Wei Shi
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, No. 100 Xisihuan, Beijing, 100039, China
| | - Zhilei Wang
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, No. 100 Xisihuan, Beijing, 100039, China
| | - Xiaoyan Zhan
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, No. 100 Xisihuan, Beijing, 100039, China
| | - Nan Qin
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, No. 100 Xisihuan, Beijing, 100039, China
| | - Tingting He
- Integrative Medical Center, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China
| | - Yuming Guo
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, No. 100 Xisihuan, Beijing, 100039, China
| | - Ming Niu
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, No. 100 Xisihuan, Beijing, 100039, China
| | - Jiabo Wang
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, No. 100 Xisihuan, Beijing, 100039, China
| | - Zhaofang Bai
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, No. 100 Xisihuan, Beijing, 100039, China.
| | - Xiaohe Xiao
- China Military Institute of Chinese Materia, the Fifth Medical Centre, Chinese PLA General Hospital, No. 100 Xisihuan, Beijing, 100039, China.
- Integrative Medical Center, the Fifth Medical Centre, Chinese PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
11
|
Hastings KL, Green MD, Gao B, Ganey PE, Roth RA, Burleson GR. Beyond Metabolism: Role of the Immune System in Hepatic Toxicity. Int J Toxicol 2021; 39:151-164. [PMID: 32174281 DOI: 10.1177/1091581819898399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The liver is primarily thought of as a metabolic organ; however, the liver is also an important mediator of immunological functions. Key perspectives on this emerging topic were presented in a symposium at the 2018 annual meeting of the American College of Toxicology entitled "Beyond metabolism: Role of the immune system in hepatic toxicity." Viral hepatitis is an important disease of the liver for which insufficient preventive vaccines exist. Host immune responses inadequately clear these viruses and often potentiate immunological inflammation that damages the liver. In addition, the liver is a key innate immune organ against bacterial infection. Hepatocytes and immune cells cooperatively control systemic and local bacterial infections. Conversely, bacterial infection can activate multiple types of immune cells and pathways to cause hepatocyte damage and liver injury. Finally, the immune system and specifically cytokines and drugs can interact in idiosyncratic drug-induced liver injury. This rare disease can result in a disease spectrum that ranges from mild to acute liver failure. The immune system plays a role in this disease spectrum.
Collapse
Affiliation(s)
| | | | - Bin Gao
- Laboratory of Liver Diseases, NIH, Bethesda, MD, USA
| | - Patricia E Ganey
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Robert A Roth
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Gary R Burleson
- BRT-Burleson Research Technologies, Inc, Morrisville, NC, USA
| |
Collapse
|
12
|
Angel SO, Vanagas L, Ruiz DM, Cristaldi C, Saldarriaga Cartagena AM, Sullivan WJ. Emerging Therapeutic Targets Against Toxoplasma gondii: Update on DNA Repair Response Inhibitors and Genotoxic Drugs. Front Cell Infect Microbiol 2020; 10:289. [PMID: 32656097 PMCID: PMC7325978 DOI: 10.3389/fcimb.2020.00289] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/15/2020] [Indexed: 12/11/2022] Open
Abstract
Toxoplasma gondii is the causative agent of toxoplasmosis in animals and humans. This infection is transmitted to humans through oocysts released in the feces of the felines into the environment or by ingestion of undercooked meat. This implies that toxoplasmosis is a zoonotic disease and T. gondii is a foodborne pathogen. In addition, chronic toxoplasmosis in goats and sheep is the cause of recurrent abortions with economic losses in the sector. It is also a health problem in pets such as cats and dogs. Although there are therapies against this infection in its acute stage, they are not able to permanently eliminate the parasite and sometimes they are not well tolerated. To develop better, safer drugs, we need to elucidate key aspects of the biology of T. gondii. In this review, we will discuss the importance of the homologous recombination repair (HRR) pathway in the parasite's lytic cycle and how components of these processes can be potential molecular targets for new drug development programs. In that sense, the effect of different DNA damage agents or HHR inhibitors on the growth and replication of T. gondii will be described. Multitarget drugs that were either associated with other targets or were part of general screenings are included in the list, providing a thorough revision of the drugs that can be tested in other scenarios.
Collapse
Affiliation(s)
- Sergio O Angel
- Laboratorio de Parasitología Molecular, Instituto Tecnológico Chascomús (INTECH), Consejo Nacional de Investigaciones Científicas (CONICET)-Universidad Nacional General San Martin (UNSAM), Chascomús, Argentina
| | - Laura Vanagas
- Laboratorio de Parasitología Molecular, Instituto Tecnológico Chascomús (INTECH), Consejo Nacional de Investigaciones Científicas (CONICET)-Universidad Nacional General San Martin (UNSAM), Chascomús, Argentina
| | - Diego M Ruiz
- Laboratorio de Parasitología Molecular, Instituto Tecnológico Chascomús (INTECH), Consejo Nacional de Investigaciones Científicas (CONICET)-Universidad Nacional General San Martin (UNSAM), Chascomús, Argentina
| | - Constanza Cristaldi
- Laboratorio de Parasitología Molecular, Instituto Tecnológico Chascomús (INTECH), Consejo Nacional de Investigaciones Científicas (CONICET)-Universidad Nacional General San Martin (UNSAM), Chascomús, Argentina
| | - Ana M Saldarriaga Cartagena
- Laboratorio de Parasitología Molecular, Instituto Tecnológico Chascomús (INTECH), Consejo Nacional de Investigaciones Científicas (CONICET)-Universidad Nacional General San Martin (UNSAM), Chascomús, Argentina
| | - William J Sullivan
- Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States.,Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
13
|
Li CY, Niu M, Liu YL, Tang JF, Chen W, Qian G, Zhang MY, Shi YF, Lin JZ, Li XJ, Li RS, Xiao XH, Li GH, Wang JB. Screening for Susceptibility-Related Factors and Biomarkers of Xianling Gubao Capsule-Induced Liver Injury. Front Pharmacol 2020; 11:810. [PMID: 32547402 PMCID: PMC7274038 DOI: 10.3389/fphar.2020.00810] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/18/2020] [Indexed: 12/14/2022] Open
Abstract
Although increasing reports from the literature on herbal-related hepatotoxicity, the identification of susceptibility-related factors and biomarkers remains challenging due to idiosyncratic drug-induced liver injury (IDILI). As a well-known Chinese medicine prescription, Xianling Gubao Capsule (XLGB) has attracted great attention due to reports of potential liver toxicity. But the mechanism behind it is difficult to determine. In this paper, we found that XLGB-induced liver injury belongs to IDILI through the analysis of clinical liver injury cases. In toxicological experiment assessment, co-exposure to XLGB and non-toxic dose of lipopolysaccharide (LPS) could cause evident liver injury as manifested by significantly increased plasma alanine aminotransferase activity and obvious liver histological damage. However, it failed to induce observable liver injury in normal rats, suggesting that mild immune stress may be a susceptibility factor for XLGB-induced idiosyncratic liver injury. Furthermore, plasma cytokines were determined and 15 cytokines (such as IL-1β, IFN-γ, and MIP-2α etc) were acquired by receiver operating characteristic (ROC) curves analysis. The expression of these 15 cytokines in LPS group was significantly up-regulated in contrast to the normal group. Meanwhile, the metabolomics profile showed that mild immune stress caused metabolic reprogramming, including sphingolipid metabolism, phenylalanine metabolism, and glycerophospholipid metabolism. 8 potential biomarkers (such as sphinganine, glycerophosphoethanolamine, and phenylalanine etc.) were identified by correlation analysis. Therefore, these results suggested that intracellular metabolism and immune changes induced by mild immune stress may be important susceptibility mechanisms for XLGB IDILI.
Collapse
Affiliation(s)
- Chun-Yu Li
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming Niu
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ya-Lei Liu
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jin-Fa Tang
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Wei Chen
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Geng Qian
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Yu Zhang
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ya-Fei Shi
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jun-Zhi Lin
- Central Laboratory, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xing-Jie Li
- Research Center for Clinical and Translational Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Rui-Sheng Li
- Research Center for Clinical and Translational Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiao-He Xiao
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Guo-Hui Li
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia-Bo Wang
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
14
|
Chen S, Wu Q, Li X, Li D, Fan M, Ren Z, Bryant M, Mei N, Ning B, Guo L. The role of hepatic cytochrome P450s in the cytotoxicity of sertraline. Arch Toxicol 2020; 94:2401-2411. [PMID: 32372212 DOI: 10.1007/s00204-020-02753-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/16/2020] [Indexed: 12/19/2022]
Abstract
Sertraline, an antidepressant, is commonly used to manage mental health symptoms related to depression, anxiety disorders, and obsessive-compulsive disorder. The use of sertraline has been associated with rare but severe hepatotoxicity. Previous research demonstrated that mitochondrial dysfunction, apoptosis, and endoplasmic reticulum stress were involved in sertraline-associated cytotoxicity. In this study, we reported that after a 24-h treatment in HepG2 cells, sertraline caused cytotoxicity, suppressed topoisomerase I and IIα, and damaged DNA in a concentration-dependent manner. We also investigated the role of cytochrome P450 (CYP)-mediated metabolism in sertraline-induced toxicity using our previously established HepG2 cell lines individually expressing 14 CYPs (1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 3A4, 3A5, and 3A7). We demonstrated that CYP2D6, 2C19, 2B6, and 2C9 metabolize sertraline, and sertraline-induced cytotoxicity was significantly decreased in the cells expressing these CYPs. Western blot analysis demonstrated that the induction of ɣH2A.X (a hallmark of DNA damage) and topoisomerase inhibition were partially reversed in CYP2D6-, 2C19-, 2B6-, and 2C9-overexpressing HepG2 cells. These data indicate that DNA damage and topoisomerase inhibition are involved in sertraline-induced cytotoxicity and that CYPs-mediated metabolism plays a role in decreasing the toxicity of sertraline.
Collapse
Affiliation(s)
- Si Chen
- Divisions of Biochemical Toxicology, National Center for Toxicological Research (NCTR)/U.S. Food and Drug Administration (FDA), Jefferson, AR, 72079, USA.
| | - Qiangen Wu
- Divisions of Biochemical Toxicology, National Center for Toxicological Research (NCTR)/U.S. Food and Drug Administration (FDA), Jefferson, AR, 72079, USA
| | - Xilin Li
- Genetic and Molecular Toxicology, National Center for Toxicological Research (NCTR)/U.S. Food and Drug Administration (FDA), Jefferson, AR, 72079, USA
| | - Dongying Li
- Bioinformatics and Biostatistics, HFT-110, National Center for Toxicological Research (NCTR)/U.S. Food and Drug Administration (FDA), 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Michelle Fan
- Life Health and Medical Sciences, Brown University, Providence, RI, 02912, USA
| | - Zhen Ren
- Divisions of Biochemical Toxicology, National Center for Toxicological Research (NCTR)/U.S. Food and Drug Administration (FDA), Jefferson, AR, 72079, USA
| | - Matthew Bryant
- Divisions of Biochemical Toxicology, National Center for Toxicological Research (NCTR)/U.S. Food and Drug Administration (FDA), Jefferson, AR, 72079, USA
| | - Nan Mei
- Genetic and Molecular Toxicology, National Center for Toxicological Research (NCTR)/U.S. Food and Drug Administration (FDA), Jefferson, AR, 72079, USA
| | - Baitang Ning
- Bioinformatics and Biostatistics, HFT-110, National Center for Toxicological Research (NCTR)/U.S. Food and Drug Administration (FDA), 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Lei Guo
- Divisions of Biochemical Toxicology, National Center for Toxicological Research (NCTR)/U.S. Food and Drug Administration (FDA), Jefferson, AR, 72079, USA.
| |
Collapse
|
15
|
Giustarini G, Huppelschoten S, Barra M, Oppelt A, Wagenaar L, Weaver RJ, Bol-Schoenmakers M, Smit JJ, van de Water B, Klingmüller U, Pieters RHH. The hepatotoxic fluoroquinolone trovafloxacin disturbs TNF- and LPS-induced p65 nuclear translocation in vivo and in vitro. Toxicol Appl Pharmacol 2020; 391:114915. [PMID: 32035082 DOI: 10.1016/j.taap.2020.114915] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Revised: 01/10/2020] [Accepted: 02/05/2020] [Indexed: 01/26/2023]
Abstract
Idiosyncratic drug-induced liver injury (IDILI) is a severe disease that cannot be detected during drug development. It has been shown that hepatotoxicity of some compounds associated with IDILI becomes apparent when these are combined in vivo and in vitro with LPS or TNF. Among these compounds trovafloxacin (TVX) induced apoptosis in the liver and increased pro-inflammatory cytokines in mice exposed to LPS/TNF. The hepatocyte survival and the cytokine release after TNF/LPS stimulation relies on a pulsatile activation of NF-κB. We set out to evaluate the dynamic activation of NF-κB in response to TVX + TNF or LPS models, both in mouse and human cells. Remarkably, TVX prolonged the first translocation of NF-κB induced by TNF both in vivo and in vitro. The prolonged p65 translocation caused by TVX was associated with an increased phosphorylation of IKK and MAPKs and accumulation of inhibitors of NF-κB such as IκBα and A20 in HepG2. Coherently, TVX suppressed further TNF-induced NF-κB translocations in HepG2 leading to decreased transcription of ICAM-1 and inhibitors of apoptosis. TVX prolonged LPS-induced NF-κB translocation in RAW264.7 macrophages increasing the secretion of TNF. In summary, this study presents new, relevant insights into the mechanism of TVX-induced liver injury underlining the resemblance between mouse and human models. In this study we convincingly show that regularly used toxicity models provide a coherent view of relevant pathways for IDILI. We propose that assessment of the kinetics of activation of NF-κB and MAPKs is an appropriate tool for the identification of hepatotoxic compounds during drug development.
Collapse
Affiliation(s)
- Giulio Giustarini
- Immunotoxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| | - Suzanna Huppelschoten
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Marco Barra
- Immunotoxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands; University of Pisa, Department of Pharmacy, Italy
| | - Angela Oppelt
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Laura Wagenaar
- Immunotoxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Richard J Weaver
- Biopharmacy, Institut de Recherches Internationales Servier (I.R.I.S.), Suresnes 92284, France
| | - Marianne Bol-Schoenmakers
- Immunotoxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Joost J Smit
- Immunotoxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands
| | - Bob van de Water
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, the Netherlands
| | - Ursula Klingmüller
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Raymond H H Pieters
- Immunotoxicology, Institute for Risk Assessment Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
16
|
Ali SE, Waddington JC, Park BK, Meng X. Definition of the Chemical and Immunological Signals Involved in Drug-Induced Liver Injury. Chem Res Toxicol 2019; 33:61-76. [PMID: 31682113 DOI: 10.1021/acs.chemrestox.9b00275] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Idiosyncratic drug-induced liver injury (iDILI), which is rare and often recognized only late in drug development, poses a major public health concern and impediment to drug development due to its high rate of morbidity and mortality. The mechanisms of DILI are not completely understood; both non-immune- and immune-mediated mechanisms have been proposed. Non-immune-mediated mechanisms including direct damage to hepatocytes, mitochondrial toxicity, interference with transporters, and alteration of bile ducts are well-known to be associated with drugs such as acetaminophen and diclofenac; whereas immune-mediated mechanisms involving activation of both adaptive and innate immune cells and the interactions of these cells with parenchymal cells have been proposed. The chemical signals involved in activation of both innate and adaptive immune responses are discussed with respect to recent scientific advances. In addition, the immunological signals including cytokine and chemokines that are involved in promoting liver injury are also reviewed. Finally, we discuss how liver tolerance and regeneration can have profound impact on the pathogenesis of iDILI. Continuous research in developing in vitro systems incorporating immune cells with liver cells and animal models with impaired liver tolerance will provide an opportunity for improved prediction and prevention of immune-mediated iDILI.
Collapse
Affiliation(s)
- Serat-E Ali
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology , University of Liverpool , Liverpool L69 3GE , United Kingdom
| | - James C Waddington
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology , University of Liverpool , Liverpool L69 3GE , United Kingdom
| | - B Kevin Park
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology , University of Liverpool , Liverpool L69 3GE , United Kingdom
| | - Xiaoli Meng
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology , University of Liverpool , Liverpool L69 3GE , United Kingdom
| |
Collapse
|
17
|
Mir TM, Ma G, Ali Z, Khan IA, Ashfaq MK. Effect of Raspberry Ketone on Normal, Obese and Health-Compromised Obese Mice: A Preliminary Study. J Diet Suppl 2019; 18:1-16. [PMID: 31603036 DOI: 10.1080/19390211.2019.1674996] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Raspberry ketone (RK)-an aromatic compound found mostly in red raspberries (Rubus idaeus) is widely used as an over the counter product for weight loss. The present study was conducted to determine adverse effects associated with RK in obese and health-compromised obese mice. Two sets of experiments were conducted on normal obese and health-compromised obese mice treated with RK for a duration of 10 days. Obese conditions were induced by feeding mice a high fat diet for 10 weeks, while the health compromised obese mouse model was developed by a single intraperitoneal injection of a nontoxic dose of lipopolysaccharide (LPS) (6 mg/kg) to obese mice. Results showed that RK (165, 330, and 500 mg/kg) under obese as well as health-compromised condition retarded the gain in body weights as compared to the control groups. RK at doses 330 and 500 mg/kg resulted in 67.6 and 50% mortality, respectively in normal obese mice and 70% mortality was observed in health-compromised obese mice treated with RK at 500 mg/kg. At higher doses deaths were observed earlier than those given lower doses of RK. Significant elevations in blood alanine transaminase (ALT) were also observed with RK treatment in obese mice. Blood glucose levels were significantly elevated in all groups of mice treated with RK. This study suggests that higher doses of RK may cause adverse effects in health compromised conditions. Under these conditions, prolonged use of RK, especially in high doses, may pose a health hazard.
Collapse
Affiliation(s)
- Tahir Maqbool Mir
- National Center for Natural Product Research, School of Pharmacy, University of Mississippi, University, MS, USA
| | - Guoyi Ma
- Drug Discovery Division, Southern Research, Birmingham, AL, UK
| | - Zulfiqar Ali
- National Center for Natural Product Research, School of Pharmacy, University of Mississippi, University, MS, USA
| | - Ikhlas A Khan
- National Center for Natural Product Research, School of Pharmacy, University of Mississippi, University, MS, USA
| | - Mohammad K Ashfaq
- National Center for Natural Product Research, School of Pharmacy, University of Mississippi, University, MS, USA
| |
Collapse
|
18
|
Hepatotoxicity Induced by Isoniazid-Lipopolysaccharide through Endoplasmic Reticulum Stress, Autophagy, and Apoptosis Pathways in Zebrafish. Antimicrob Agents Chemother 2019; 63:AAC.01639-18. [PMID: 30858204 DOI: 10.1128/aac.01639-18] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 03/02/2019] [Indexed: 02/06/2023] Open
Abstract
Isoniazid (INH) is a first-line antituberculosis drug. The incidence of adverse reactions accompanied by inflammation in the liver during drug administration to tuberculosis patients is high and severely affects clinical treatment. To better understand the mechanism of hepatotoxicity induced by INH under the inflammatory state, we compared the differences in levels of hepatotoxicity from INH between normal zebrafish and zebrafish in an inflammatory state to elucidate the hepatotoxic mechanism using different endpoints such as mortality, malformation, inflammatory effects, liver morphology, histological changes, transaminase analysis, and expression levels of certain genes. The results showed that the toxic effect of INH in zebrafish in an inflammatory state was more obvious than that in normal zebrafish, that liver size was significantly decreased as measured by liver fatty acid binding protein (LFABP) reporter fluorescence and intensity, and that alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels were significantly increased. Hematoxylin and eosin (HE) staining and electron microscopy showed that hepatocyte injury was more obvious in the inflammatory state. In the inflammatory state, INH significantly increased the expression levels of endoplasmic reticulum stress (ERS)-related factors (GRP78, ATF6, PERK, IRE1, XBP1s, GRP94, and CHOP), autophagy-related factors (beclin 1, LC3, Atg3, and Atg12), and apoptosis-related factors (caspase-3, caspase-8, caspase-9, Bax, p53, and Cyt) in larvae. Correlational analyses indicated that the transcription levels of the inflammatory factors interleukin-1b (IL-1b), tumor necrosis factor beta (TNF-β), cyclooxygenase 2 (COX-2), and TNF-ɑ were strongly positively correlated with ALT and AST. Furthermore, the ERS inhibitor sodium 4-phenylbutyrate (4-PBA) could ameliorate the hepatotoxicity of INH-lipopolysaccharide (LPS) in zebrafish larvae. These results indicated that INH hepatotoxicity was enhanced in the inflammatory state. ERS and its mediated autophagy and apoptosis pathways might be involved in INH-induced liver injury promoted by inflammation.
Collapse
|
19
|
The role of hepatic cytochrome P450s in the cytotoxicity of dronedarone. Arch Toxicol 2018; 92:1969-1981. [PMID: 29616291 DOI: 10.1007/s00204-018-2196-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/21/2018] [Indexed: 02/06/2023]
Abstract
Dronedarone is used to treat patients with cardiac arrhythmias and has been reported to be associated with liver injury. Our previous mechanistic work demonstrated that DNA damage-induced apoptosis contributes to the cytotoxicity of dronedarone. In this study, we examined further the underlying mechanisms and found that after a 24-h treatment of HepG2 cells, dronedarone caused cytotoxicity, G1-phase cell cycle arrest, suppression of topoisomerase II, and DNA damage in a concentration-dependent manner. We also investigated the role of cytochrome P450s (CYPs)-mediated metabolism in the dronedarone-induced toxicity using our previously established HepG2 cell lines expressing individually 14 human CYPs (1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C18, 2C19, 2D6, 2E1, 3A4, 3A5, and 3A7). We demonstrated that CYP3A4, 3A5, and 2D6 were the major enzymes that metabolize dronedarone, and that CYP3A7, 2E1, 2C19, 2C18, 1A1, and 2B6 also metabolize dronedarone, but to a lesser extent. Our data showed that the cytotoxicity of dronedarone was decreased in CYP3A4-, 3A5-, or 2D6-overexpressing cells compared to the control HepG2 cells, indicating that the parent dronedarone has higher potency than the metabolites to induce cytotoxicity in these cells. In contrast, cytotoxicity was increased in CYP1A1-overexpressing cells, demonstrating that CYP1A1 exerts an opposite effect in dronedarone's toxicity, comparing to CYP3A4, 3A5, or 2D6. We also studied the involvement of topoisomerase II in dronedarone-induced toxicity, and demonstrated that the overexpression of topoisomerase II caused an increase in cell viability and a decrease in γ-H2A.X induction, suggesting that suppression of topoisomerase II may be one of the mechanisms involved in dronedarone-induced liver toxicity.
Collapse
|
20
|
Ren Z, Chen S, Ning B, Guo L. Use of Liver-Derived Cell Lines for the Study of Drug-Induced Liver Injury. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2018. [DOI: 10.1007/978-1-4939-7677-5_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
21
|
Zhang QL, Zhu QH, Xie ZQ, Xu B, Wang XQ, Chen JY. Genome-wide gene expression analysis of amphioxus (Branchiostoma belcheri) following lipopolysaccharide challenge using strand-specific RNA-seq. RNA Biol 2017; 14:1799-1809. [PMID: 28837390 PMCID: PMC5731807 DOI: 10.1080/15476286.2017.1367890] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Amphioxus is the closest living proxy for exploring the evolutionary origin of the immune system in vertebrates. To understand the immune responses of amphioxus to lipopolysaccharide (LPS), 5 ribosomal RNA (rRNA)-depleted libraries of amphioxus were constructed, including one control (0 h) library and 4 treatment libraries at 6, 12, 24, and 48 h post-injection (hpi) with LPS. The transcriptome of Branchiostoma belcheri was analyzed using strand-specific RNA sequencing technology (RNA-seq). A total of 6161, 6665, 7969, and 6447 differentially expressed genes (DEGs) were detected at 6, 12, 24, and 48 hpi, respectively, compared with expression levels at 0 h. We identified amphioxus genes active during the acute-phase response to LPS at different time points after stimulation. Moreover, to better visualize the resolution phase of the immune process during immune response, we identified 6057 and 5235 DEGs at 48 hpi by comparing with 6 and 24 hpi, respectively. Through real-time quantitative PCR (qRT-PCR) analysis of 12 selected DEGs, we demonstrated the accuracy of the RNA-seq data in this study. Functional enrichment analysis of DEGs demonstrated that most terms were related to defense and immune responses, disease and infection, cell apoptosis, and metabolism and catalysis. Subsequently, we identified 1330, 485, 670, 911, and 1624 time-specific genes (TSGs) at 0, 6, 12, 24, and 48 hpi. Time-specific terms at each of 5 time points were primarily involved in development, immune signaling, signal transduction, DNA repair and stability, and metabolism and catalysis, respectively. As this is the first study to report the transcriptome of an organism with primitive immunity following LPS challenge at multiple time points, it provides gene expression information for further research into the evolution of immunity in vertebrates.
Collapse
Affiliation(s)
- Qi-Lin Zhang
- a LPS , Nanjing Institute of Geology and Paleontology, Chinese Academy of Science , Nanjing , China ; State Key Laboratory of Pharmaceutical Biotechnology , School of Life Science, Nanjing University , Nanjing , China
| | | | - Zheng-Qing Xie
- a LPS , Nanjing Institute of Geology and Paleontology, Chinese Academy of Science , Nanjing , China ; State Key Laboratory of Pharmaceutical Biotechnology , School of Life Science, Nanjing University , Nanjing , China
| | - Bin Xu
- a LPS , Nanjing Institute of Geology and Paleontology, Chinese Academy of Science , Nanjing , China ; State Key Laboratory of Pharmaceutical Biotechnology , School of Life Science, Nanjing University , Nanjing , China
| | - Xiu-Qiang Wang
- a LPS , Nanjing Institute of Geology and Paleontology, Chinese Academy of Science , Nanjing , China ; State Key Laboratory of Pharmaceutical Biotechnology , School of Life Science, Nanjing University , Nanjing , China
| | - Jun-Yuan Chen
- a LPS , Nanjing Institute of Geology and Paleontology, Chinese Academy of Science , Nanjing , China ; State Key Laboratory of Pharmaceutical Biotechnology , School of Life Science, Nanjing University , Nanjing , China
| |
Collapse
|
22
|
Screening for main components associated with the idiosyncratic hepatotoxicity of a tonic herb, Polygonum multiflorum. Front Med 2017; 11:253-265. [PMID: 28315126 DOI: 10.1007/s11684-017-0508-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 11/24/2016] [Indexed: 01/30/2023]
Abstract
The main constituents of a typical medicinal herb, Polygonum multiflorum (Heshouwu in Chinese), that induces idiosyncratic liver injury remain unclear. Our previous work has shown that cotreatment with a nontoxic dose of lipopolysaccharide (LPS) and therapeutic dose of Heshouwu can induce liver injury in rats, whereas the solo treatment cannot induce observable injury. In the present work, using the constituent "knock-out" and "knock-in" strategy, we found that the ethyl acetate (EA) extract of Heshouwu displayed comparable idiosyncratic hepatotoxicity to the whole extract in LPS-treated rats. Results indicated a significant elevation of plasma alanine aminotransferase, aspartate aminotransferase, and liver histologic changes, whereas other separated fractions failed to induce liver injury. The mixture of EA extract with other separated fractions induced comparable idiosyncratic hepatotoxicity to the whole extract in LPS-treated rats. Chemical analysis further revealed that 2,3,5,4'-tetrahydroxy trans-stilbene-2-O-β-glucoside (trans-SG) and its cis-isomer were the two major compounds in EA extract. Furthermore, the isolated cis-, and not its trans-isomer, displayed comparable idiosyncratic hepatotoxicity to EA extract in LPS-treated rats. Higher contents of cis-SG were detected in Heshouwu liquor or preparations from actual liver intoxication patients associated with Heshouwu compared with general collected samples. In addition, plasma metabolomics analysis showed that cis-SG-disturbing enriched pathways remarkably differed from trans-SG ones in LPS-treated rats. All these results suggested that cis-SG was closely associated with the idiosyncratic hepatotoxicity of Heshouwu. Considering that the cis-trans isomerization of trans-SG was mediated by ultraviolet light or sunlight, our findings serve as reference for controlling photoisomerization in drug discovery and for the clinical use of Heshouwu and stilbene-related medications.
Collapse
|
23
|
Li CY, Tu C, Gao D, Wang RL, Zhang HZ, Niu M, Li RY, Zhang CE, Li RS, Xiao XH, Yang MH, Wang JB. Metabolomic Study on Idiosyncratic Liver Injury Induced by Different Extracts of Polygonum multiflorum in Rats Integrated with Pattern Recognition and Enriched Pathways Analysis. Front Pharmacol 2016; 7:483. [PMID: 28018221 PMCID: PMC5156827 DOI: 10.3389/fphar.2016.00483] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/28/2016] [Indexed: 12/26/2022] Open
Abstract
Currently, numerous liver injury cases related to a famous Chinese herb- Polygonum Multiflorum (Heshouwu in Chinese) have attracted great attention in many countries. Our previous work showed that Heshouwu-induced hepatotoxicity belonged to idiosyncratic drug-induced liver injury (IDILI). Unfortunately, the components and mechanisms attributed to IDILI of Heshouwu are difficult to determine and thus remain unknown. Attempts to explore puzzles, we prepared the chloroform (CH)-, ethyl acetate (EA)-, and residue (RE) extracts of Heshouwu to investigate IDILI constituents and underlying mechanisms, using biochemistry, histopathology, and metabolomics examinations. The results showed that co-treatment with non-toxic dose of lipopolysaccharide (LPS) and EA extract could result in evident liver injury, indicated by the significant elevation of plasma alanine aminotransferase and aspartate aminotransferase activities, as well as obvious liver histologic damage; whereas other two separated fractions, CH and RE extracts, failed to induce observable liver injury. Furthermore, 21 potential metabolomic biomarkers that differentially expressed in LPS/EA group compared with other groups without liver injury were identified by untargeted metabolomics, mainly involved two pathways: tricarboxylic acid cycle and sphingolipid metabolism. This work illustrated EA extract had close association with the idiosyncratic hepatotoxicity of Heshouwu and provided a metabolomic insight into IDILI of different extracts from Heshouwu.
Collapse
Affiliation(s)
- Chun-Yu Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China; China Military Institute of Chinese Medicine, 302 Military HospitalBeijing, China
| | - Can Tu
- China Military Institute of Chinese Medicine, 302 Military HospitalBeijing, China; School of Pharmacy, Chengdu University of Traditional Chinese MedicineChengdu, China
| | - Dan Gao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing, China; China Military Institute of Chinese Medicine, 302 Military HospitalBeijing, China
| | - Rui-Lin Wang
- Integrative Medical Center, 302 Military Hospital Beijing, China
| | - Hai-Zhu Zhang
- China Military Institute of Chinese Medicine, 302 Military HospitalBeijing, China; School of Pharmacy, Chengdu University of Traditional Chinese MedicineChengdu, China
| | - Ming Niu
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Rui-Yu Li
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Cong-En Zhang
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Rui-Sheng Li
- Research Center for Clinical and Translational Medicine, 302 Hospital of People's Liberation Army Beijing, China
| | - Xiao-He Xiao
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Mei-Hua Yang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing, China
| | - Jia-Bo Wang
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| |
Collapse
|
24
|
Roth RA, Maiuri AR, Ganey PE. Idiosyncratic Drug-Induced Liver Injury: Is Drug-Cytokine Interaction the Linchpin? J Pharmacol Exp Ther 2016; 360:461-470. [PMID: 28104833 DOI: 10.1124/jpet.116.237578] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 11/11/2016] [Indexed: 12/11/2022] Open
Abstract
Idiosyncratic drug-induced liver injury continues to be a human health problem in part because drugs that cause these reactions are not identified in current preclinical testing and because progress in prevention is hampered by incomplete knowledge of mechanisms that underlie these adverse responses. Several hypotheses involving adaptive immune responses, inflammatory stress, inability to adapt to stress, and multiple, concurrent factors have been proposed. Yet much remains unknown about how drugs interact with the liver to effect death of hepatocytes. Evidence supporting hypotheses implicating adaptive or innate immune responses in afflicted patients has begun to emerge and is bolstered by results obtained in experimental animal models and in vitro systems. A commonality in adaptive and innate immunity is the production of cytokines, including interferon-γ (IFNγ). IFNγ initiates cell signaling pathways that culminate in cell death or inhibition of proliferative repair. Tumor necrosis factor-α, another cytokine prominent in immune responses, can also promote cell death. Furthermore, tumor necrosis factor-α interacts with IFNγ, leading to enhanced cellular responses to each cytokine. In this short review, we propose that the interaction of drugs with these cytokines contributes to idiosyncratic drug-induced liver injury, and mechanisms by which this could occur are discussed.
Collapse
Affiliation(s)
- Robert A Roth
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Ashley R Maiuri
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Patricia E Ganey
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
25
|
Lee S, Mattingly A, Lin A, Sacramento J, Mannent L, Castel MN, Canolle B, Delbary-Gossart S, Ferzaz B, Morganti JM, Rosi S, Ferguson AR, Manley GT, Bresnahan JC, Beattie MS. A novel antagonist of p75NTR reduces peripheral expansion and CNS trafficking of pro-inflammatory monocytes and spares function after traumatic brain injury. J Neuroinflammation 2016; 13:88. [PMID: 27102880 PMCID: PMC4840857 DOI: 10.1186/s12974-016-0544-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 04/10/2016] [Indexed: 12/17/2022] Open
Abstract
Background Traumatic brain injury (TBI) results in long-term neurological deficits, which may be mediated in part by pro-inflammatory responses in both the injured brain and the circulation. Inflammation may be involved in the subsequent development of neurodegenerative diseases and post-injury seizures. The p75 neurotrophin receptor (p75NTR) has multiple biological functions, affecting cell survival, apoptotic cell death, axonal growth, and degeneration in pathological conditions. We recently found that EVT901, a novel piperazine derivative that inhibits p75NTR oligomerization, is neuroprotective, reduces microglial activation, and improves outcomes in two models of TBI in rats. Since TBI elicits both CNS and peripheral inflammation, we used a mouse model of TBI to examine whether EVT901 would affect peripheral immune responses and trafficking to the injured brain. Methods Cortical contusion injury (CCI)-TBI of the sensory/motor cortex was induced in C57Bl/6 wild-type mice and CCR2+/RFP heterozygote transgenic mice, followed by treatment with EVT901, a selective antagonist of p75NTR, or vehicle by i.p. injection at 4 h after injury and then daily for 7 days. Brain and blood were collected at 1 and 6 weeks after injury. Flow cytometry and histological analysis were used to determine peripheral immune responses and trafficking of peripheral immune cells into the lesion site at 1 and 6 weeks after TBI. A battery of behavioral tests administered over 6 weeks was used to evaluate neurological outcome, and stereological estimation of brain tissue volume at 6 weeks was used to assess tissue damage. Finally, multivariate principal components analysis (PCA) was used to evaluate the relationships between inflammatory events, EVT901 treatment, and neurological outcomes. Results EVT901 is neuroprotective in mouse CCI-TBI and dramatically reduced the early trafficking of CCR2+ and pro-inflammatory monocytes into the lesion site. EVT901 reduced the number of CD45highCD11b+ and CD45highF4/80+ cells in the injured brain at 6 weeks. TBI produced a significant increase in peripheral pro-inflammatory monocytes (Ly6Cint-high pro-inflammatory monocytes), and this peripheral effect was also blocked by EVT901 treatment. Further, we found that blocking p75NTR with EVT901 reduces the expansion of pro-inflammatory monocytes, and their response to LPS in vitro, supporting the idea that there is a peripheral EVT901 effect that blunts inflammation. Further, 1 week of EVT901 blocks the expansion of pro-inflammatory monocytes in the circulation after TBI, reduces the number of multiple subsets of pro-inflammatory monocytes that enter the injury site at 1 and 6 weeks post-injury, and is neuroprotective, as it was in the rat. Conclusions Together, these findings suggest that p75NTR signaling participates in the production of the peripheral pro-inflammatory response to CNS injury and implicates p75NTR as a part of the pro-inflammatory cascade. Thus, the neuroprotective effects of p75NTR antagonists might be due to a combination of central and peripheral effects, and p75NTR may play a role in the production of peripheral inflammation in addition to its many other biological roles. Thus, p75NTR may be a therapeutic target in human TBI. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0544-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sangmi Lee
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA
| | - Aaron Mattingly
- Biomedical Science Graduate Program, University of California at San Francisco, San Francisco, CA, 94143-0899, USA
| | - Amity Lin
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA
| | - Jeffrey Sacramento
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA
| | - Leda Mannent
- Early to Candidate, Sanofi Research, 195 route d'Espagne, Chilly-Mazarin, France
| | - Marie-Noelle Castel
- Early to Candidate, Sanofi Research, 195 route d'Espagne, Chilly-Mazarin, France
| | - Benoit Canolle
- Early to Candidate, Sanofi Research, 195 route d'Espagne, Chilly-Mazarin, France
| | | | - Badia Ferzaz
- Early to Candidate, Sanofi Research, 195 route d'Espagne, Chilly-Mazarin, France
| | - Josh M Morganti
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA
| | - Susanna Rosi
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA.,Biomedical Science Graduate Program, University of California at San Francisco, San Francisco, CA, 94143-0899, USA
| | - Adam R Ferguson
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA.,Biomedical Science Graduate Program, University of California at San Francisco, San Francisco, CA, 94143-0899, USA
| | - Geoffrey T Manley
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA
| | - Jacqueline C Bresnahan
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA
| | - Michael S Beattie
- Department of Neurological Surgery, Brain and Spinal Injury Center, University of California at San Francisco, Box 0899, 1001 Potrero Ave, Bldg 1, Rm 101, San Francisco, CA, 94143-0899, USA. .,Biomedical Science Graduate Program, University of California at San Francisco, San Francisco, CA, 94143-0899, USA.
| |
Collapse
|
26
|
Beggs KM, Maiuri AR, Fullerton AM, Poulsen KL, Breier AB, Ganey PE, Roth RA. Trovafloxacin-induced replication stress sensitizes HepG2 cells to tumor necrosis factor-alpha-induced cytotoxicity mediated by extracellular signal-regulated kinase and ataxia telangiectasia and Rad3-related. Toxicology 2015; 331:35-46. [PMID: 25748550 DOI: 10.1016/j.tox.2015.03.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 02/11/2015] [Accepted: 03/04/2015] [Indexed: 01/23/2023]
Abstract
Use of the fluoroquinolone antibiotic trovafloxacin (TVX) was restricted due to idiosyncratic, drug-induced liver injury (IDILI). Previous studies demonstrated that tumor necrosis factor-alpha (TNF) and TVX interact to cause death of hepatocytes in vitro that was associated with prolonged activation of c-Jun N-terminal kinase (JNK), activation of caspases 9 and 3, and DNA damage. The purpose of this study was to explore further the mechanism by which TVX interacts with TNF to cause cytotoxicity. Treatment with TVX caused cell cycle arrest, enhanced expression of p21 and impaired proliferation, but cell death only occurred after cotreatment with TVX and TNF. Cell death involved activation of extracellular signal-related kinase (ERK), which in turn activated caspase 3 and ataxia telangiectasia and Rad3-related (ATR), both of which contributed to cytotoxicity. Cotreatment of HepG2 cells with TVX and TNF caused double-strand breaks in DNA, and ERK contributed to this effect. Inhibition of caspase activity abolished the DNA strand breaks. The data suggest a complex interaction of TVX and TNF in which TVX causes replication stress, and the downstream effects are exacerbated by TNF, leading to hepatocellular death. These results raise the possibility that IDILI from TVX results from MAPK and ATR activation in hepatocytes initiated by interaction of cytokine signaling with drug-induced replication stress.
Collapse
Affiliation(s)
- Kevin M Beggs
- Michigan State University, Department of Pharmacology & Toxicology, Center for Integrative Toxicology, 1129 Farm Lane, East Lansing, MI 48824, United States
| | - Ashley R Maiuri
- Michigan State University, Department of Pharmacology & Toxicology, Center for Integrative Toxicology, 1129 Farm Lane, East Lansing, MI 48824, United States
| | - Aaron M Fullerton
- Michigan State University, Department of Pharmacology & Toxicology, Center for Integrative Toxicology, 1129 Farm Lane, East Lansing, MI 48824, United States
| | - Kyle L Poulsen
- Michigan State University, Department of Pharmacology & Toxicology, Center for Integrative Toxicology, 1129 Farm Lane, East Lansing, MI 48824, United States
| | - Anna B Breier
- Michigan State University, Department of Pharmacology & Toxicology, Center for Integrative Toxicology, 1129 Farm Lane, East Lansing, MI 48824, United States
| | - Patricia E Ganey
- Michigan State University, Department of Pharmacology & Toxicology, Center for Integrative Toxicology, 1129 Farm Lane, East Lansing, MI 48824, United States
| | - Robert A Roth
- Michigan State University, Department of Pharmacology & Toxicology, Center for Integrative Toxicology, 1129 Farm Lane, East Lansing, MI 48824, United States.
| |
Collapse
|