1
|
Li K, Cui Y, Zheng X, Min C, Zhang J, Yan Z, Ji Y, Ge F, Ji H, Zhu F. Jian Gan powder ameliorates immunological liver injury in mice by modulating the gut microbiota and metabolic profiles. Eur J Med Res 2024; 29:240. [PMID: 38641655 PMCID: PMC11031866 DOI: 10.1186/s40001-024-01827-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/03/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Immunological liver injury (ILI) is a common liver disease associated with the microbiota-gut-liver axis. Jian Gan powder (JGP) exhibits both protective and therapeutic effects on hepatitis virus-induced ILI in the clinic. However, the underlying mechanisms remain elusive. The aim of this study is to investigate the hepatoprotective effects and associated mechanisms of JGP in the context of gut microbiota, utilizing a mouse model of ILI. METHODS The mouse model was established employing Bacillus Calmette-Guérin (BCG) plus lipopolysaccharide (LPS). Following treatment with JGP (7.5, 15, or 30 g/kg), serum, liver, and fresh fecal samples were analyzed. 16S rRNA gene sequencing and untargeted metabolomics profiling were performed to assess the role of JGP on the gut microbiota and its metabolites. RESULTS JGP treatment markedly reduced serum IFN-γ, IL-6, IL-22, and hepatic p-STAT3 (phosphorylated transducer and activator of transcription-3) expression. In contrast, JGP increased the percentage of proliferating cell nuclear antigen-positive liver cells in treated mice. Fecal 16S rRNA gene sequencing revealed that JGP treatment restored the levels of Alloprevotella, Burkholderia-Caballeronia-Paraburkholderia, Muribaculum, Streptococcus, and Stenotrophomonas. Additionally, metabolomics analysis of fecal samples showed that JGP restored the levels of allylestrenol, eplerenone, phosphatidylethanolamine (PE) (P-20:0/0:0), sphingomyelin (SM) d27:1, soyasapogenol C, chrysin, and soyasaponin I. CONCLUSIONS JGP intervention improves ILI by restoring gut microbiota and modifying its metabolic profiles. These results provide a novel insight into the mechanism of JGP in treating ILI and the scientific basis to support its clinical application.
Collapse
Affiliation(s)
- Kun Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Building 9, Nanjing, 210046, Jiangsu, People's Republic of China
- Department of Gastroenterology, Hai'an Hospital of Traditional Chinese Medicine Affiliated to Medical College of Yangzhou University, Nantong, People's Republic of China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, People's Republic of China
| | - Yadong Cui
- College of Pharmaceutical Science, Soochow University, Suzhou, People's Republic of China
| | - Xue Zheng
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Building 9, Nanjing, 210046, Jiangsu, People's Republic of China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, People's Republic of China
| | - Chunyan Min
- Suzhou Institute for Drug Control, Suzhou, People's Republic of China
| | - Jian Zhang
- College of Pharmaceutical Science, Soochow University, Suzhou, People's Republic of China
| | - Zhanpeng Yan
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Building 9, Nanjing, 210046, Jiangsu, People's Republic of China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, People's Republic of China
| | - Yu Ji
- Department of Gastroenterology, Hai'an Hospital of Traditional Chinese Medicine Affiliated to Medical College of Yangzhou University, Nantong, People's Republic of China
| | - Fei Ge
- Department of Gastroenterology, Hai'an Hospital of Traditional Chinese Medicine Affiliated to Medical College of Yangzhou University, Nantong, People's Republic of China
| | - Hualiang Ji
- Department of Gastroenterology, Affiliated Haian People's Hospital of Nantong University, 17 Zhong Ba Zhong Road, Hai'an, 226600, Jiangsu, People's Republic of China.
| | - Fangshi Zhu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, 100 Hongshan Road, Building 9, Nanjing, 210046, Jiangsu, People's Republic of China.
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, People's Republic of China.
| |
Collapse
|
2
|
Zhou B, Liu P, Yao X, Cao H, Zhu H, Wang Q, Liu Y, Fang M, Wu Y, Gong Z. Hepatoprotective effects of peach gum polysaccharides against alcoholic liver injury: moderation of oxidative stress and promotion of lipid metabolism. Front Nutr 2024; 10:1325450. [PMID: 38283909 PMCID: PMC10811791 DOI: 10.3389/fnut.2023.1325450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 12/15/2023] [Indexed: 01/30/2024] Open
Abstract
Natural polysaccharides extracted from plants have received increasing attention due to their rich bioactivity. In our study, peach gum polysaccharides (PGPs) were extracted by water extraction-alcohol precipitation method. PGPs are typical pyranose polysaccharides with a mean molecular weight of 3.68 × 106 g/mol. The antioxidant activity and hepatoprotective capacity of PGPs were studied. In vitro, assays showed that PGPs scavenged DPPH, OH, and O2- in a dose-dependent manner. PGPs exhibited antioxidative properties against alcohol-induced HL7702 cells, as evidenced by the normalization of MDA, SOD, ROS, and GSH levels. To further elucidate the hepatoprotective mechanism of PGPs, we carried out in vivo experiments in male mice. PGPs exerted hepatoprotective effects in alcohol liver disease (ALD) mice by exerting antioxidant effects, decreasing the inflammatory response and modulating lipid metabolism. In addition, metabolomic analysis indicated that PGPs mainly regulate D-glutamine and D-glutamate metabolism, alanine, aspartate and glutamate metabolism, and arginine biosynthesis to promote hepatic metabolism and maintain body functions. Overall, this study revealed that the hepatoprotective mechanism of PGPs against ALD might be associated with the regulation of oxidative stress and lipid metabolism.
Collapse
Affiliation(s)
- Bingjie Zhou
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Key Laboratory for Deep Processing of Major Grain and Oil (The Chinese Ministry of Education), Food Safety Research Center for Key Research Institute of Humanities and Social Sciences of Hubei Province, Wuhan Polytechnic University, Wuhan, China
| | - Pinpin Liu
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Key Laboratory for Deep Processing of Major Grain and Oil (The Chinese Ministry of Education), Food Safety Research Center for Key Research Institute of Humanities and Social Sciences of Hubei Province, Wuhan Polytechnic University, Wuhan, China
| | - Xiangao Yao
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Key Laboratory for Deep Processing of Major Grain and Oil (The Chinese Ministry of Education), Food Safety Research Center for Key Research Institute of Humanities and Social Sciences of Hubei Province, Wuhan Polytechnic University, Wuhan, China
| | - Huijie Cao
- Suizhou Center for Disease Control and Prevention, Hubei Province, China
| | - Hang Zhu
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Key Laboratory for Deep Processing of Major Grain and Oil (The Chinese Ministry of Education), Food Safety Research Center for Key Research Institute of Humanities and Social Sciences of Hubei Province, Wuhan Polytechnic University, Wuhan, China
| | - Qiao Wang
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Key Laboratory for Deep Processing of Major Grain and Oil (The Chinese Ministry of Education), Food Safety Research Center for Key Research Institute of Humanities and Social Sciences of Hubei Province, Wuhan Polytechnic University, Wuhan, China
| | - Yan Liu
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Key Laboratory for Deep Processing of Major Grain and Oil (The Chinese Ministry of Education), Food Safety Research Center for Key Research Institute of Humanities and Social Sciences of Hubei Province, Wuhan Polytechnic University, Wuhan, China
| | - Min Fang
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Key Laboratory for Deep Processing of Major Grain and Oil (The Chinese Ministry of Education), Food Safety Research Center for Key Research Institute of Humanities and Social Sciences of Hubei Province, Wuhan Polytechnic University, Wuhan, China
| | - Yongning Wu
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Key Laboratory for Deep Processing of Major Grain and Oil (The Chinese Ministry of Education), Food Safety Research Center for Key Research Institute of Humanities and Social Sciences of Hubei Province, Wuhan Polytechnic University, Wuhan, China
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Food Safety Research Unit (2019RU014) of Chinese Academy of Medical Sciences, Beijing, China
| | - Zhiyong Gong
- Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Key Laboratory for Deep Processing of Major Grain and Oil (The Chinese Ministry of Education), Food Safety Research Center for Key Research Institute of Humanities and Social Sciences of Hubei Province, Wuhan Polytechnic University, Wuhan, China
| |
Collapse
|
3
|
Zhang X, Han S, Jiang X, Duan S, Gao Y, Ding J, Li X, Sun B, Hu X, Zhang X, Zhang W. Comparative analysis of bile metabolic profile in patients with biliary obstruction complicated by Clonorchis sinensis infection. Front Cell Infect Microbiol 2023; 13:1254016. [PMID: 37868349 PMCID: PMC10585366 DOI: 10.3389/fcimb.2023.1254016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/21/2023] [Indexed: 10/24/2023] Open
Abstract
Background Clonorchiasis is an important foodborne parasitic disease. However, eggs of Clonorchis sinensis (C. sinensis) cannot be detected in feces during biliary obstruction. Moreover, many diseases can cause biliary obstruction, such as gallstones, adenocarcinoma, cholangiocarcinoma and Ascaris lumbricoides infection. Therefore, it is of great significance to distinguish between patients of biliary obstruction and biliary obstruction with C. sinensis infection. Methods A total of 48 biliary obstruction patients were enrolled, including 23 infected with C. sinensis (C. sinensis) (OB+C.s) and 25 non-infected subjects (OB). The bile samples were collected by endoscopic retrograde cholangiopancreatography and analyzed using ultra-high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UHPLC-QTOF MS). Additionally, multivariate statistical analysis methods were employed to identify differential metabolites. Next, bile amino acid levels were determined by targeted metabolomics analysis. Result A total of 146 and 132 significant metabolites were identified in electrospray ionization (ESI)+ and ESI- modes, respectively. The levels of amino acids (asparagine, glutamate, ornithine) and polyamines (spermidine and spermine) were significantly changed. Targeted analysis showed that the levels of amino acids (such as L-arginine, L-glutamine, L-lysine, L-propionic, and L-tyrosine) were lower in OB+C.s patients compared to those in OB patients. Marked metabolic pathways were involved in "Glutathione metabolism", "Caffeine metabolism", "Alanine, aspartate and glutamate metabolism", "Arginine and proline metabolism", "Purine metabolism", "Beta-Alanine metabolism", and "D-glutamine and D-glutamate metabolism". Conclusion These results show that there were significant differences between OB+C.s and OB patients, especially in amino acids. The metabolic signature and perturbations in metabolic pathways may help to better distinguish OB+C.s and OB patients.
Collapse
Affiliation(s)
- Xueli Zhang
- Department of Parasitology, Harbin Medical University, Harbin, China
| | - Su Han
- Department of Parasitology, Harbin Medical University, Harbin, China
- Department of Public Health and Preventive Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xu Jiang
- Department of Parasitology, Harbin Medical University, Harbin, China
| | - Shanshan Duan
- Beijing Obstetrics and Gynecology Hospital Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, China
| | - Yannan Gao
- Department of Orthopaedic Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jian Ding
- Department of Parasitology, Harbin Medical University, Harbin, China
| | - Xiang Li
- Department of Parasitology, Harbin Medical University, Harbin, China
| | - Beibei Sun
- Clinical Laboratory, Zhuhai Maternal and Child Health Hospital, Zhuhai, China
| | - Xinyi Hu
- Department of Stomatology, Laixi People's Hospital, Shandong, China
| | - Xiaoli Zhang
- Department of Parasitology, Harbin Medical University, Harbin, China
| | - Weizhe Zhang
- Department of Parasitology, Harbin Medical University, Harbin, China
| |
Collapse
|
4
|
Men L, Wang Z, Gou M, Li Z, Li W, Li C, Li K, Gong X. Metabolomics and targeted amino acid analysis reveal the liver protective effect of arginyl-fructosyl-glucose from red ginseng on acute liver injury in mice. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023] Open
|
5
|
Jiang HY, Gao HY, Li J, Zhou TY, Wang ST, Yang JB, Hao RR, Pang F, Wei F, Liu ZG, Kuang L, Ma SC, He JM, Jin HT. Integrated spatially resolved metabolomics and network toxicology to investigate the hepatotoxicity mechanisms of component D of Polygonum multiflorum Thunb. JOURNAL OF ETHNOPHARMACOLOGY 2022; 298:115630. [PMID: 35987407 DOI: 10.1016/j.jep.2022.115630] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 07/25/2022] [Accepted: 08/06/2022] [Indexed: 06/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The liver toxicity of Reynoutria multiflora (Thunb.) Moldenke. (Polygonaceae) (Polygonum multiflorum Thunb, PM) has always attracted much attention, but the related toxicity materials and mechanisms have not been elucidated due to multi-component and multi-target characteristics. In previous hepatotoxicity screening, different components of PM were first evaluated and the hepatotoxicity of component D [95% ethanol (EtOH) elution] in a 70% EtOH extract of PM (PM-D) showed the highest hepatotoxicity. Furthermore, the main components of PM-D were identified and their hepatotoxicity was evaluated based on a zebrafish embryo model. However, the hepatotoxicity mechanism of PM-D is unknown. AIM OF THE STUDY This work is to explore the hepatotoxicity mechanisms of PM-D by integrating network toxicology and spatially resolved metabolomics strategy. MATERIALS AND METHODS A hepatotoxicity interaction network of PM-D was constructed based on toxicity target prediction for eight key toxic ingredients and a hepatotoxicity target collection. Then the key signaling pathways were enriched, and molecular docking verification was implemented to evaluate the ability of toxic ingredients to bind to the core targets. The pathological changes of liver tissues and serum biochemical assays of mice were used to evaluate the liver injury effect of mice with oral administration of PM-D. Furthermore, spatially resolved metabolomics was used to visualize significant differences in metabolic profiles in mice after drug administration, to screen hepatotoxicity-related biomarkers and analyze metabolic pathways. RESULTS The contents of four key toxic compounds in PM-D were detected. Network toxicology identified 30 potential targets of liver toxicity of PM-D. GO and KEGG enrichment analyses indicated that the hepatotoxicity of PM-D involved multiple biological activities, including cellular response to endogenous stimulus, organonitrogen compound metabolic process, regulation of the apoptotic process, regulation of kinase, regulation of reactive oxygen species metabolic process and signaling pathways including PI3K-Akt, AMPK, MAPK, mTOR, Ras and HIF-1. The molecular docking confirmed the high binding activity of 8 key toxic ingredients with 10 core targets, including mTOR, PIK3CA, AKT1, and EGFR. The high distribution of metabolites of PM-D in the liver of administrated mice was recognized by mass spectrometry imaging. Spatially resolved metabolomics results revealed significant changes in metabolic profiles after PM-D administration, and metabolites such as taurine, taurocholic acid, adenosine, and acyl-carnitines were associated with PM-D-induced liver injury. Enrichment analyses of metabolic pathways revealed tht linolenic acid and linoleic acid metabolism, carnitine synthesis, oxidation of branched-chain fatty acids, and six other metabolic pathways were significantly changed. Comprehensive analysis revealed that the hepatotoxicity caused by PM-D was closely related to cholestasis, mitochondrial damage, oxidative stress and energy metabolism, and lipid metabolism disorders. CONCLUSIONS In this study, the hepatotoxicity mechanisms of PM-D were comprehensively identified through an integrated spatially resolved metabolomics and network toxicology strategy, providing a theoretical foundation for the toxicity mechanisms of PM and its safe clinical application.
Collapse
Affiliation(s)
- Hai-Yan Jiang
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Hui-Yu Gao
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, China
| | - Jie Li
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tian-Yu Zhou
- College of Pharmacy, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| | - Shu-Ting Wang
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jian-Bo Yang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, China
| | - Rui-Rui Hao
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fei Pang
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Feng Wei
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, China
| | - Zhi-Gang Liu
- School of Biological Science and Engineering, South China University of Technology, Guangzhou, China
| | - Lian Kuang
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuang-Cheng Ma
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, China.
| | - Jiu-Ming He
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; NMPA Key Laboratory for Safety Research and Evaluation of Innovative Drug, Beijing, China.
| | - Hong-Tao Jin
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China; Beijing Union-Genius Pharmaceutical Technology Development Co., Ltd., Beijing, China; NMPA Key Laboratory for Safety Research and Evaluation of Innovative Drug, Beijing, China.
| |
Collapse
|
6
|
Arnold CG, Dylla L, Monte AA, Heard K, Heard S, D'Alessandro A, Reynolds K, Dart R, Rumack B, Sonn B. Metabolomic Evaluation of N-Acetyl-p-Benzoquinone Imine Protein Adduct Formation with Therapeutic Acetaminophen Administration: Sex-based Physiologic Differences. J Med Toxicol 2022; 18:297-310. [PMID: 35751009 PMCID: PMC9492831 DOI: 10.1007/s13181-022-00903-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/24/2022] [Accepted: 05/30/2022] [Indexed: 10/17/2022] Open
Abstract
BACKGROUND Acetaminophen (APAP)-associated transaminase elevation, induced by N-acetyl-p-benzoquinone imine (NAPQI) protein adduction, remains an area of research interest. Distinct from known genetic, physiologic, and dosage associations dictating severity of hepatic injury, no known factors predict an absence of protein adduct formation at therapeutic APAP dosing. HYPOTHESIS Sex-based physiology is predictive of APAP-induced protein adduct formation and differential metabolite expression at therapeutic doses. METHODS This retrospective study interrogated serum samples collected for a prior study investigating fluctuations of alanine aminotransferase (ALT) over time with 4G daily APAP dosing for ≥ 16 days in subjects from Denver, Colorado. Subjects were grouped by adduct formation (n = 184) vs no adducts (n = 20). Samples were run on ultra-high-performance liquid chromatography mass spectrometry from study days 0, 7, 16, and 31. Significant metabolite expressions were identified using t-tests with false discovery rate correction (FDR), partial least squares discriminant, and ANOVA simultaneous comparison analyses. Demographic and clinical data were explored using t-tests with FDR (age, weight, BMI, ALT) and Chi-square (sex, ethnicity, race) analyses. RESULTS In pre-treatment samples, relative quantitation caprylic acid was expressed ninefold higher and 6-carboxyhexanoate was expressed threefold lower in subjects who did not develop adducts. Lactate had greater expression in the no adducts group (p = 0.001). Using absolute quantitation, glutathione was expressed 2.6-fold greater among no adduct subjects. Odds of males developing NAPQI protein adducts at therapeutic APAP dosing were 5.91 times lower than females (95% CI = 2.3-14.9; p = 0.0001). CONCLUSION Multiple metabolites were differentially expressed based on adduct group and sex. Metabolites were identified unique to adduct development independent of sex. At therapeutic APAP dosing, males were less likely to develop APAP protein adducts. Further research into lipid biosynthesis and metabolism may provide further insight into physiology associated with adduct production.
Collapse
Affiliation(s)
- Cosby G Arnold
- Department of Emergency Medicine, University of Colorado School of Medicine, University of Colorado Denver-Anschutz Medical Center, Mail Stop B-215, 12401 East 17thAvenue, Aurora, CO, 80045, USA.
| | - Layne Dylla
- Department of Emergency Medicine, University of Colorado School of Medicine, University of Colorado Denver-Anschutz Medical Center, Mail Stop B-215, 12401 East 17thAvenue, Aurora, CO, 80045, USA
| | - Andrew A Monte
- Department of Emergency Medicine, University of Colorado School of Medicine, University of Colorado Denver-Anschutz Medical Center, Mail Stop B-215, 12401 East 17thAvenue, Aurora, CO, 80045, USA
- Rocky Mountain Poison & Drug Center, Denver Health and Hospital Authority, 1391 Speer Blvd Unit 600, Denver, CO, 80204, USA
| | - Kennon Heard
- Department of Emergency Medicine, University of Colorado School of Medicine, University of Colorado Denver-Anschutz Medical Center, Mail Stop B-215, 12401 East 17thAvenue, Aurora, CO, 80045, USA
- Rocky Mountain Poison & Drug Center, Denver Health and Hospital Authority, 1391 Speer Blvd Unit 600, Denver, CO, 80204, USA
| | - Susan Heard
- Rocky Mountain Poison & Drug Center, Denver Health and Hospital Authority, 1391 Speer Blvd Unit 600, Denver, CO, 80204, USA
| | - Angelo D'Alessandro
- Division of Bioinformatics & Personalized Medicine, University of Colorado School of Medicine, 13001 E 17th Place, Fitzimons Building, 5th floor, #563, Aurora, CO, 80045, USA
| | - Kate Reynolds
- Rocky Mountain Poison & Drug Center, Denver Health and Hospital Authority, 1391 Speer Blvd Unit 600, Denver, CO, 80204, USA
| | - Richard Dart
- Rocky Mountain Poison & Drug Center, Denver Health and Hospital Authority, 1391 Speer Blvd Unit 600, Denver, CO, 80204, USA
| | - Barry Rumack
- Rocky Mountain Poison & Drug Center, Denver Health and Hospital Authority, 1391 Speer Blvd Unit 600, Denver, CO, 80204, USA
- Department of Pediatrics, University of Colorado School of Medicine, 13123 E 16th Ave, Aurora, CO, 80045, USA
| | - Brandon Sonn
- Department of Emergency Medicine, University of Colorado School of Medicine, University of Colorado Denver-Anschutz Medical Center, Mail Stop B-215, 12401 East 17thAvenue, Aurora, CO, 80045, USA
| |
Collapse
|
7
|
Hong W, Peng X, Zhou X, Li P, Ye Z, Liang W. FXR/ASS1 axis attenuates the TAA-induced liver injury through arginine metabolism. Biochem Biophys Res Commun 2022; 611:31-37. [PMID: 35477090 DOI: 10.1016/j.bbrc.2022.04.073] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/15/2022] [Indexed: 11/19/2022]
Abstract
Previous studies demonstrated that arginine biosynthesis was frequently impaired in acute liver injury. However, the underlying mechanisms remain elusive. In this study, we found that Argininosuccinate synthetase 1 (ASS1), a rate-limiting enzyme in arginine metabolism, was downregulated in the TAA-induced liver injury model. Single-cell RNA-seq data found that ASS1 was highly enriched in the hepatocytes. The reduction of ASS1 was attributed to the decreased expression of Farnesoid X receptor (FXR), which is a bile acid-activated nuclear hormone receptor with high expression in the liver. Subsequent studies demonstrated that activation of FXR by its agonist obeticholic acid (OCA) directly promoted ASS1 transcription and enhanced arginine synthesis, leading to the alleviation of TAA-mediated liver injury. Further experiments found that OCA, ASS1, and arginine supplement can rescue TAA-mediated hepatocytes apoptosis by decreasing the protein levels of Cyto C, PARP, and Caspase 3. Taken together, our study illustrated a protective role of the FXR/ASS1 axis in TAA-induced liver injury by targeting arginine metabolism, which might shed light on the development of novel therapeutic approaches for acute liver injury.
Collapse
Affiliation(s)
- Weilong Hong
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Xuyun Peng
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China; Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Xue Zhou
- Department of Ultrasonic Medicine, Guangzhou Women and Children's Medical Center, Guangzhou, PR China
| | - Panlong Li
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China; Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China
| | - Zhiqiang Ye
- Department of Emergency, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China.
| | - Weicheng Liang
- Biotherapy Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China; Cell-gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China; Vaccine Research Institute, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, PR China.
| |
Collapse
|
8
|
Miura A, Hosono T, Seki T. Macrophage potentiates the recovery of liver zonation and metabolic function after acute liver injury. Sci Rep 2021; 11:9730. [PMID: 33958644 PMCID: PMC8102573 DOI: 10.1038/s41598-021-88989-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
The liver is an exclusive organ with tremendous regenerative capacity. Liver metabolic functions exhibit spatial heterogeneity, reflecting liver zonation. The mechanisms controlling the proliferation of hepatocytes and the accompanying matrix reconstruction during regeneration have been well explored, but the recovery potential of differentiated metabolic functions and zonation after liver injury remains unclear. We employed a mouse model of carbon tetrachloride (CCl4) induced-acute liver injury with clodronate-induced macrophage depletion to clarify the impact of liver injury on liver metabolism and recovery dynamics of metabolic function and liver zonation during regeneration. Depleting macrophages suppressed tissue remodelling and partially delayed cell proliferation during regeneration after liver injury. In addition, recovery of metabolic functions was delayed by suppressing the tissue remodelling caused by the depleted macrophages. The model revealed that drug metabolic function was resilient against the dysfunction caused by liver injury, but glutamine synthesis was not. Metabolomic analysis revealed that liver branched-chain amino acid (BCAA) and carbohydrate metabolism were suppressed by injury. The plasma BCAA concentration reflected recovery of hepatic function during regeneration. Our study reveals one aspect of the regenerative machinery for hepatic metabolism following acute liver injury.
Collapse
Affiliation(s)
- Atsushi Miura
- General Research Institute, Nihon University Collage of Bioresource Sciences, Fujisawa, Kanagawa 252-0880 Japan
| | - Takashi Hosono
- Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa 252-0880 Japan ,Department of Chemistry and Life Science, Nihon University Collage of Bioresource Sciences, Fujisawa, Kanagawa 252-0880 Japan
| | - Taiichiro Seki
- General Research Institute, Nihon University Collage of Bioresource Sciences, Fujisawa, Kanagawa 252-0880 Japan ,Department of Applied Life Sciences, Nihon University Graduate School of Bioresource Sciences, Fujisawa, Kanagawa 252-0880 Japan ,Department of Chemistry and Life Science, Nihon University Collage of Bioresource Sciences, Fujisawa, Kanagawa 252-0880 Japan
| |
Collapse
|
9
|
Gao H, Yang T, Chen X, Song Y. Changes of Lipopolysaccharide-Induced Acute Kidney and Liver Injuries in Rats Based on Metabolomics Analysis. J Inflamm Res 2021; 14:1807-1825. [PMID: 33986608 PMCID: PMC8110281 DOI: 10.2147/jir.s306789] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/07/2021] [Indexed: 12/28/2022] Open
Abstract
Background The bacterial endotoxin lipopolysaccharide (LPS) was the classic inducer to establish many inflammatory disease models, especially multiple organ injury. Evidences indicated that the mechanism that causes inflammation response is not just related to cytokine release. The main aim of this study was to better elucidate the possible links between metabolic changes and the pathogenesis of LPS-induced acute liver and kidney in order to understand the mechanisms and screening therapeutic targets for developing early diagnostic strategies and treatments. Methods An experimental rat model was established by intraperitoneal injection of 10 mg/kg LPS. An untargeted metabolomics analysis of the serum in the LPS and control groups was carried out using ultra-performance liquid chromatography/quadrupole time-of-flight mass spectrometry (UPLC/QTOF-MS). LPS-induced pathological damage in the lungs, liver, kidneys, and colon was observed, along with changes in biochemical indexes, indicating that there was a severe inflammatory response in many organs after administration of LPS for 8 h. Principal component analysis (PCA) and partial least squares-discriminant analysis (PLS-DA) showed distinct separation in the serum metabolite profiles between the LPS and control groups, indicating significant changes in endogenous metabolites. Results The untargeted metabolomics analysis showed that there were 127 significantly different serum metabolites and 53 altered pathways after LPS administration, including pathways related to the metabolism of D-glutamine and D-glutamate, taurine and hypotaurine, beta-alanine, glutathione, and butanoate, which are involved in the inflammatory response, oxidative stress, and amino acid metabolism. Conclusion The study suggested that LPS-induced acute liver and kidney injury mainly involves inflammatory response, oxidative stress, and protein synthesis, finally causing multi-organ damage. Correcting the disturbances to the metabolites and metabolic pathways may help to prevent and/or treat LPS-induced acute liver and kidney damage.
Collapse
Affiliation(s)
- Huan Gao
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Tao Yang
- Houde Food Co., Ltd, Liaoyuan, 136200, People's Republic of China
| | - Xuan Chen
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Yanqing Song
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| |
Collapse
|
10
|
Sun Y, Wang X, Zhou Y, Zhang J, Cui W, Wang E, Du J, Wei B, Xu X. Protective effect of metformin on BPA-induced liver toxicity in rats through upregulation of cystathionine β synthase and cystathionine γ lyase expression. THE SCIENCE OF THE TOTAL ENVIRONMENT 2021; 750:141685. [PMID: 32862004 DOI: 10.1016/j.scitotenv.2020.141685] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 06/11/2023]
Abstract
Human exposure to bisphenol A (BPA) is unavoidable in daily life. Recently, research has showen that BPA could induce oxidative imbalance, thereby causing reproductive toxicity and liver dysfunction. Accumulated evidence has demonstrated that metformin possesses strong anti-oxidative properties. This study aimed to study the mechanism underlying the hepatic-protective effect of metformin on liver injury induced by BPA in rats via the UPLC-MS/MS metabolomics approach. Forty-two male rats were randomly divided into six groups (n = 7), namely the saline group (control), the corn oil group (vehicle), the metformin group (Met), the bisphenol A group (BPA), the bisphenol A and metformin group (BPA + Met), and the bisphenol A and diammonium glycyrrhizinate (positive control) group (BPA + DG). Serum was collected for biochemical analysis and metabolomics, and liver tissue was collected for histopathology and metabolomics in each group. We found that metformin could significantly reduce the levels of liver function enzymes (ALT, AST and GGT) and ameliorate inflammatory cell infiltration and hepatocyte necrosis induced by BPA. On the other hand, metformin could significantly enhance the total antioxidant capacity in BPA rats. Notably, metabolomics data indicated that the principal altered metabolic pathways based on the 26 differential metabolites in liver tissue, and 21 in serum among vehicle, BPA and BPA + Met groups, respectively, including cysteine and methionine metabolism, glutathione metabolism, and arginine biosynthesis and purine metabolism. Additionally, metformin significantly increased cystathionine β synthase (CBS) and cystathionine γ lyase (CSE), thus reducing serum levels of homocysteine and increasing hepatic levels of cysteine and glutathione in BPA-treated rats. Overall, this study's results provided new insights into the role and mechanism of metformin in BPA-induced liver injury in rats.
Collapse
Affiliation(s)
- Yaxin Sun
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Co-innovation Center of Henan Province for New drug R & D and preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Xinying Wang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Co-innovation Center of Henan Province for New drug R & D and preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Yuanyuan Zhou
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Co-innovation Center of Henan Province for New drug R & D and preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Junhong Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Co-innovation Center of Henan Province for New drug R & D and preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Weiqi Cui
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Co-innovation Center of Henan Province for New drug R & D and preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China
| | - Enyin Wang
- Department of Reproductive Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, PR China
| | - Juan Du
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Co-innovation Center of Henan Province for New drug R & D and preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Bo Wei
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Co-innovation Center of Henan Province for New drug R & D and preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| | - Xia Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Co-innovation Center of Henan Province for New drug R & D and preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, PR China.
| |
Collapse
|
11
|
Liu F, Sun Z, Hu P, Tian Q, Xu Z, Li Z, Tian X, Chen M, Huang C. Determining the protective effects of Yin-Chen-Hao Tang against acute liver injury induced by carbon tetrachloride using 16S rRNA gene sequencing and LC/MS-based metabolomics. J Pharm Biomed Anal 2019; 174:567-577. [PMID: 31261038 DOI: 10.1016/j.jpba.2019.06.028] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/20/2019] [Accepted: 06/21/2019] [Indexed: 12/12/2022]
Abstract
Yin-Chen-Hao Tang (YCHT), consisting of Artemisia annua L., Gardenia jasminoides Ellis, and Rheum Palmatum L., has been used to relieve liver diseases in China for thousands of years. Several protective mechanisms of YCHT on liver injury have been investigated based on metabolomics, but the effects of YCHT on the alterations in the gut microbiota are still unclear. In this study, an integrated approach based on 16S rRNA gene sequencing combined with high-performance liquid chromatography-mass spectrometry (HPLC-MS) metabolic profiling was performed to assess the effects of YCHT on liver injury induced by carbon tetrachloride (CCl4) through the regulation of the relative abundances of gut microbiota and their relationships with biomarker candidates. A total of twelve significantly altered bacterial genera and nine metabolites were identified, which returned to normal levels after YCHT treatment. The relative abundances of the identified microbiota, including significantly elevated amounts of p_Firmicutes, c_Clostridia, o_Clostridiales, f_Ruminococcaceae, g_[Eubacterium]_coprostanoligenes_group, s_uncultured_bacterium_f_Lachnospiraceae and remarkedly increased amounts of p_Bacteroidetes, c_Bacteroidia, o_Bacteroidales, f_Bacteroidaceae, g_Bacteroides and s_uncultured_bacterium_g_Bacteroides, were found in model rats compared with controls. Potential biomarkers, including lower levels of LysoPC (16:1(9Z)/0:0), LysoPC (20:3(5Z,8Z,11Z)), LysoPC (17:0), LysoPC (20:1(11Z)) and 3-hydroxybutyric acid and higher amounts of ornithine, L-kynurenine, hippuric acid and taurocholic acid are involved in several custom metabolic pathways, such as arginine and proline metabolism, tryptophan metabolism, glycerophospholipid metabolism and primary bile acid biosynthesis. Interestingly, there was a strong correlation between the perturbed gut microbiota in genera c_Clostridia and o_Clostridiales and the altered plasma metabolite 3-hydroxybutyric acid. This finding means that the hepatoprotective effects of YCHT may be due to the regulation of the production of the functional metabolite 3-hydroxybutyric acid through changes in the proportions of c_Clostridia and o_Clostridiales. These results showed that the hepatoprotective effects of YCHT not only focused on custom metabolic pathways but also depended on the changes in the gut microbiota in liver injury. These findings suggest that the 16S rRNA gene sequencing and LC-MS based metabolomics approach can be applied to comprehensively evaluate the effects of traditional Chinese medicines (TCMs).
Collapse
Affiliation(s)
- Fang Liu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Material Medica, Chinese Academy of Science, Shanghai, 201203, PR China
| | - Zhaolin Sun
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Material Medica, Chinese Academy of Science, Shanghai, 201203, PR China
| | - Pei Hu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Material Medica, Chinese Academy of Science, Shanghai, 201203, PR China
| | - Qiang Tian
- School of Life Science and Engineering, Southwest University of Science and Technology, Mianyang, Sichuan, 621010, PR China
| | - Zhou Xu
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Material Medica, Chinese Academy of Science, Shanghai, 201203, PR China
| | - Zhixiong Li
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Material Medica, Chinese Academy of Science, Shanghai, 201203, PR China
| | - Xiaoting Tian
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Material Medica, Chinese Academy of Science, Shanghai, 201203, PR China
| | - Mingcang Chen
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Material Medica, Chinese Academy of Science, Shanghai, 201203, PR China.
| | - Chenggang Huang
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, Shanghai Institute of Material Medica, Chinese Academy of Science, Shanghai, 201203, PR China.
| |
Collapse
|
12
|
Tu C, He Q, Li CY, Niu M, Han ZX, Ge FL, Zhou YY, Zhang L, Wang XH, Zhu JX, Li RS, Song HB, Xiao XH, Wang JB. Susceptibility-Related Factor and Biomarkers of Dietary Supplement Polygonum multiflorum-Induced Liver Injury in Rats. Front Pharmacol 2019; 10:335. [PMID: 31024306 PMCID: PMC6459954 DOI: 10.3389/fphar.2019.00335] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 03/19/2019] [Indexed: 12/21/2022] Open
Abstract
Polygonum multiflorum [PM, synonym Reynoutria multiflora (Thunb.) Moldenke.], a well-known and commonly used Traditional Chinese Medicine and herbal dietary supplement for nourishing the kidney and liver, etc., has aroused wide concern for its reported potential hepatotoxicity. Previous clinical cases and experimental studies have suggested that mild immune stress (MIS) may be one of the susceptibility-related factors of idiosyncratic drug-induced liver injury (IDILI) caused by PM. In this paper, we found that the same dose of PM caused abnormal liver biochemical indicators and liver tissue damage in MIS model rats, while it did not result in liver injury in normal rats, further confirming that MIS is a susceptibility factor for PM-IDILI. Plasma chemokine/cytokine profiling indicated that the MIS model group was significantly different from the other groups, showing a significant upregulation of plasma chemokines, while the MIS/PM group showed upregulated expression of chemokines or pro-inflammatory cytokines. Liver histopathological examination indicated a small amount of inflammatory cytokine infiltration in the MIS group, but no hepatocyte injury, consistent with the plasma profiles of increased chemokines and unchanged inflammatory cytokines. Notably, metabolomics characterization showed that MIS caused reprogramming of these metabolic pathways (such as phenylalanine and glutamate pathways), which was associated with acute phase reactions and inflammatory responses. These results suggested that MIS may promote an immune response to the initial cellular injury induced by PM in the liver, and MIS-induced upregulation of chemokines and metabolic reprogramming may an important mechanism that mediates the susceptibility to PM-IDILI. Furthermore, via receiver operating characteristic (ROC) curves analysis, we identified 12 plasma cytokines (e.g., IP-10, MCP-1 and MIP-1α) and nine metabolomics biomarkers (e.g., L-Phenylalanine, Creatinine, and L-glutamine) with differential capabilities (all ROC AUC > 0.9) of identifying susceptibility model animals from normal ones, which might be of referable value for the clinical recognition of PM-IDILI susceptible individuals.
Collapse
Affiliation(s)
- Can Tu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- China Military Institute of Chinese Medicine, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Qin He
- China Military Institute of Chinese Medicine, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Chun-Yu Li
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ming Niu
- China Military Institute of Chinese Medicine, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Zi-Xin Han
- China Military Institute of Chinese Medicine, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Fei-Lin Ge
- China Military Institute of Chinese Medicine, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Yuan-Yuan Zhou
- China Military Institute of Chinese Medicine, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Le Zhang
- China Military Institute of Chinese Medicine, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Xiao-Hui Wang
- China Military Institute of Chinese Medicine, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jing-Xiao Zhu
- China Military Institute of Chinese Medicine, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Rui-Sheng Li
- China Military Institute of Chinese Medicine, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Hai-Bo Song
- Center for Drug Reevaluation, China National Medical Product Administration, Beijing, China
| | - Xiao-He Xiao
- China Military Institute of Chinese Medicine, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| | - Jia-Bo Wang
- China Military Institute of Chinese Medicine, The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
13
|
Liposomal Curcumin is Better than Curcumin to Alleviate Complications in Experimental Diabetic Mellitus. Molecules 2019; 24:molecules24050846. [PMID: 30818888 PMCID: PMC6429477 DOI: 10.3390/molecules24050846] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/19/2019] [Accepted: 02/26/2019] [Indexed: 01/14/2023] Open
Abstract
Curcumin (CC) is known to have anti-inflammatory and anti-oxidative properties and has already been tested for its efficiency in different diseases including diabetes mellitus (DM). New formulations and route administration were designed to obtain products with higher bioavailability. Our study aimed to test the effect of intraperitoneal (i.p.) administration of liposomal curcumin (lCC) as pre-treatment in streptozotocin(STZ)-induced DM in rats on oxidative stress, liver, and pancreatic functional parameters. Forty-two Wistar-Bratislava rats were randomly divided into six groups (seven animals/group): control (no diabetes), control-STZ (STZ-induced DM —60 mg/100g body weight a single dose intraperitoneal administration, and no CC pre-treatment), two groups with DM and CC pre-treatment (1mg/100g bw—STZ + CC1, 2 mg/100g bw—STZ + CC2), and two groups with DM and lCC pre-treatment (1 mg/100g bw—STZ + lCC1, 2 mg/100g bw—STZ + lCC1). Intraperitoneal administration of Curcumin in diabetic rats showed a significant reduction of nitric oxide, malondialdehyde, total oxidative stress, and catalase for both evaluated formulations (CC and lCC) compared to control group (p < 0.005), with higher efficacy of lCC formulation compared to CC solution (p < 0.002, excepting catalase for STZ + CC2vs. STZ + lCC1when p = 0.0845). The CC and lCC showed hepatoprotective and hypoglycemic effects, a decrease in oxidative stress and improvement in anti-oxidative capacity status against STZ-induced DM in rats (p < 0.002). The lCC also proved better efficacy on MMP-2, and -9 plasma levels as compared to CC (p < 0.003, excepting STZ + CC2 vs. STZ + lCC1 comparison with p = 0.0553). The lCC demonstrated significantly better efficacy as compared to curcumin solution on all serum levels of the investigated markers, sustaining its possible use as adjuvant therapy in DM.
Collapse
|
14
|
Plasma citrulline is a sensitive safety biomarker for small intestinal injury in rats. Toxicol Lett 2018; 295:416-423. [DOI: 10.1016/j.toxlet.2018.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 05/22/2018] [Accepted: 07/06/2018] [Indexed: 11/21/2022]
|
15
|
Bothe MK, Meyer C, Mueller U, Queudot JC, Roger V, Harleman J, Westphal M. Characterization of a rat model of moderate liver dysfunction based on alpha-naphthylisothiocyanate-induced cholestasis. J Toxicol Sci 2018; 42:715-721. [PMID: 29142170 DOI: 10.2131/jts.42.715] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Plasma amino acid level changes occur in mild, moderate and severe stages of liver injury in human patients. In animal models, however, data are mainly restricted to severe liver injury models in rats. Here we present the characterization of a rat model of moderate liver dysfunction secondary to alpha-napthylisothiocyanate (ANIT)-induced cholestasis. Rats treated with 30 mg/kg/day ANIT for 3 weeks exhibited a time-dependent increase in plasma alanine aminotransferase (ALT), aspartate aminotransferase (AST) and bilirubin levels and a decrease in albumin concentration. According to a liver dysfunction evaluation based on the human Child-Pugh-Score, animals developed a moderate liver dysfunction in the first two weeks of ANIT treatment, while only a mild dysfunction was observed at the end of week 3 despite ongoing ANIT administration. Univariate analysis of branched-chain amino acid plasma levels indicated that reduced levels of branched chain amino acids were associated with the ANIT treatment. These data may set the stage for further research of amino acid disturbances and requirements in non-severe cholestasis.
Collapse
|
16
|
Bailey WJ, Glaab W. Derisking drug-induced liver injury from bench to bedside. CURRENT OPINION IN TOXICOLOGY 2017. [DOI: 10.1016/j.cotox.2017.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
17
|
Maliver P, Festag M, Bennecke M, Christen F, Bánfai B, Lenz B, Winter M. Assessment of Preclinical Liver and Skeletal Muscle Biomarkers Following Clofibrate Administration in Wistar Rats. Toxicol Pathol 2017; 45:506-525. [PMID: 28485676 DOI: 10.1177/0192623317707271] [Citation(s) in RCA: 4] [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
Clofibrate is a known rodent hepatotoxicant classically associated with hepatocellular hypertrophy and increased serum activities of cellular alanine aminotransferase/aspartate aminotransferase (ALT/AST) in the absence of microscopic hepatocellular degeneration. At toxic dose, clofibrate induces liver and skeletal muscle injury. The objective of this study was to assess novel liver and skeletal muscle biomarkers following clofibrate administration in Wistar rats at different dose levels for 7 days. In addition to classical biomarkers, liver injury was assessed by cytokeratin 18 (CK18) cleaved form, high-mobility group box 1, arginase 1 (ARG1), microRNA 122 (miR-122), and glutamate dehydrogenase. Skeletal muscle injury was evaluated with fatty acid binding protein 3 (Fabp3) and myosin light chain 3 (Myl3). Clofibrate-induced hepatocellular hypertrophy and skeletal muscle degeneration (type I rich muscles) were noted microscopically. CK, Fabp3, and Myl3 elevations correlated to myofiber degeneration. Fabp3 and Myl3 outperformed CK for detection of myofiber degeneration of minimal severity. miR-122 and ARG1 results were significantly correlated and indicated the absence of liver toxicity at low doses of clofibrate, despite increased ALT/AST activities. Moreover, combining classical and novel biomarkers (Fabp3, Myl3, ARG1, and miR-122) can be considered a valuable strategy for differentiating increased transaminases due to liver toxicity from skeletal muscle toxicity.
Collapse
Affiliation(s)
- Pierre Maliver
- 1 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Matthias Festag
- 1 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Moritz Bennecke
- 2 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Penzberg, Germany
| | - Francois Christen
- 1 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Balázs Bánfai
- 1 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland.,3 Soladis GmbH, Basel, Switzerland
| | - Barbara Lenz
- 1 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| | - Michael Winter
- 1 Pharmaceutical Sciences, Roche Pharmaceutical Research and Early Development, Roche Innovation Center, F. Hoffmann-La Roche Ltd., Basel, Switzerland
| |
Collapse
|
18
|
Hepatocyte-secreted extracellular vesicles modify blood metabolome and endothelial function by an arginase-dependent mechanism. Sci Rep 2017; 7:42798. [PMID: 28211494 PMCID: PMC5314384 DOI: 10.1038/srep42798] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 01/13/2017] [Indexed: 02/06/2023] Open
Abstract
Hepatocytes release extracellular vesicles (EVs) loaded with signaling molecules and enzymes into the bloodstream. Although the importance of EVs in the intercellular communication is already recognized, the metabolic impact of the enzymes carried by these vesicles is still unclear. We evaluated the global effect of the enzymatic activities of EVs by performing untargeted metabolomic profiling of serum samples after their exposure to EVs. This approach revealed a significant change in the abundance of 94 serum metabolic signals. Our study shows that these vesicles modify the concentration of metabolites of different chemical nature including metabolites related to arginine metabolism, which regulates vascular function. To assess the functional relevance of this finding, we examined the levels of arginase-1 protein and its activity in the hepatic EVs carrying the exosomal markers CD81 and CD63. Remarkably, the arginase activity was also detected in EVs isolated from the serum in vivo, and this vesicular activity significantly increased under liver-damaging conditions. Finally, we demonstrated that EVs secreted by hepatocytes inhibited the acetylcholine-induced relaxation in isolated pulmonary arteries, via an arginase-dependent mechanism. In summary, our study demonstrates that the hepatocyte-released EVs are metabolically active, affecting a number of serum metabolites involved in oxidative stress metabolism and the endothelial function.
Collapse
|
19
|
Gao D, Pang JY, Zhang CE, Li CY, Tu C, Zhang HZ, Niu M, Xiong Y, Xiao XH, Zhao KJ, Gao WW, Wang JB. Poria Attenuates Idiosyncratic Liver Injury Induced by Polygoni Multiflori Radix Praeparata. Front Pharmacol 2016; 7:386. [PMID: 27803670 PMCID: PMC5067826 DOI: 10.3389/fphar.2016.00386] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 10/03/2016] [Indexed: 12/31/2022] Open
Abstract
The hepatotoxicity induced by Polygoni Multiflori Radix Praeparata (PM) has aroused great concern throughout the world. Hence, it is worthwhile to perform studies on the detoxification with the combined use of medicinal herbs based on the compatibility theory of traditional Chinese medicine. In this work, the rat model of PM/LPS-induced idiosyncratic liver injury was used. The effects of Poria, Licorice, and Panax notoginseng on rats of PM/LPS-induced liver injury were investigated respectively, hoping to find the most effective herbal medicine to reduce the hepatotoxicity. According to results of biochemical and histological tests, PM could induce the idiosyncratic hepatotoxicity of rats which presented modest inflammation triggered by non-injurious dose of lipopolysaccharide (LPS). We also found that the combined use of Poria and PM in the ratio of 1:2 could significantly ameliorate the PM/LPS-induced liver injury and systemic inflammation. Furthermore, UPLC/QTOF-MS-based metabolomics was performed to identify possible biomarkers and underlying biological pathways. Ten metabolites were expressed differentially among LPS, PM/LPS, and detoxification-treated groups in terms of PCA and OPLS-DA analysis, which could be potential biomarkers. MetaboAnalyst and pathway enrichment analysis revealed that alterations of these metabolites were primarily involved in three pathways: arginine and proline metabolism, primary bile acid biosynthesis and sphingolipid metabolism. This research provides systematic experimental evidences for the hepatoprotective effect of Poria against PM/LPS-induced liver injury for the first time. And these findings may help better understand the underlying mechanisms of pathophysiologic changes in PM/LPS-induced liver injury.
Collapse
Affiliation(s)
- Dan Gao
- Institute of Medicinal Plant Development, Chinese Academy of Medical SciencesBeijing, China; China Military Institute of Chinese Medicine, 302 Military HospitalBeijing, China
| | - Jing-Yao Pang
- Pharmacy Department, Beijing Luhe Hospital Affiliated to Capital Medical UniversityBeijing, China; Department of Traditional Chinese Medicine, Beijing Friendship Hospital Affiliated to Capital Medical UniversityBeijing, China
| | - Cong-En Zhang
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Chun-Yu Li
- Institute of Medicinal Plant Development, Chinese Academy of Medical SciencesBeijing, China; China Military Institute of Chinese Medicine, 302 Military HospitalBeijing, China
| | - Can Tu
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Hai-Zhu Zhang
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Ming Niu
- China Military Institute of Chinese Medicine, 302 Military Hospital Beijing, China
| | - Yin Xiong
- Kunming University of Science and Technology Kunming, China
| | - Xiao-He Xiao
- Integrative Medicine Center, 302 Military Hospital Beijing, China
| | - Kui-Jun Zhao
- Department of Traditional Chinese Medicine, Beijing Friendship Hospital Affiliated to Capital Medical University Beijing, China
| | - Wei-Wei Gao
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences Beijing, China
| | - Jia-Bo Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical SciencesBeijing, China; China Military Institute of Chinese Medicine, 302 Military HospitalBeijing, China
| |
Collapse
|