1
|
Bai G, Chen W, Ji C, Yang Y, Shen J, Li F, Wen Y, Tan DJ, Jiang X, Xiao Y, Chen J. Metabolomic profiles in serum uncover novel biomarkers in children with Williams-Beuren syndrome. Sci Rep 2025; 15:9437. [PMID: 40108238 PMCID: PMC11923248 DOI: 10.1038/s41598-025-94018-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/11/2025] [Indexed: 03/22/2025] Open
Abstract
Williams-Beuren syndrome (WBS, OMIM-no.194050) is a rare congenital genetic disorder primarily marked by developmental delays and cardiovascular anomalies, with potential involvement of metabolic dysregulation. Despite this, the metabolic features of WBS have not been extensively studied. Thus, our objective was to examine the serum metabolome profile in children with WBS, elucidating metabolic changes and associated pathways in the disorder. We recruited 25 children with WBS (mean age 5.0 ± 2.6 years, 40% female) from the Children's Hospital affiliated to Zhejiang University between 2020 and 2023. An age and sex matched healthy control group (N = 25) were recruited from the Health Management Center in the same hospital. Clinical information of WBS were extracted from the medical records. Blood samples were obtained for untargeted metabolomics analysis using UPLC-MS/MS. The metabolomic profiles of WBS patients were compared to those of healthy controls to identify metabolites with differential abundance. Enrichment analysis was conducted to identify potentially impacted KEGG pathways. Associations between metabolites and phenotypes were evaluated. Children with WBS exhibited a unique metabolic profile compared to healthy controls, as evidenced by the identification of 465 untargeted metabolites in serum. Of these metabolites, 169 showed differential abundance in WBS children. The top enriched KEGG pathways in WBS children included nicotine addiction, cholesterol metabolism, arginine biosynthesis, retrograde endocannabinoid signaling. Additionally, there were indications of potential metabolic alterations in the L-tryptophan pathway, with a shift from serotonin to L-kynurenine, as well as disruptions in bile acid metabolism. Metabolome data in children with WBS showed neurological and amino acid metabolism changes, indicating multisystem involvement and developmental delay. This data can help monitor and manage the disease, but further studies are needed to understand the underlying mechanisms and consequences.
Collapse
Affiliation(s)
- Guannan Bai
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang Province, China
| | - Weijun Chen
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang Province, China
| | - Chai Ji
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang Province, China
| | - Yang Yang
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming, Yunnan Province, People's Republic of China
| | - Jiyang Shen
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang Province, China
| | - Fangfang Li
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang Province, China
| | - Yang Wen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | | | - Xiaoling Jiang
- Department of Child Health Care, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, Zhejiang Province, China
| | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, China.
| | - Jinluan Chen
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
2
|
Guo Q, Wu Z, Wang K, Shi J, Wei M, Lu B, Huang Z, Ji L. Forsythiaside-A improved bile-duct-ligation-induced liver fibrosis in mice: The involvement of alleviating mitochondrial damage and ferroptosis in hepatocytes via activating Nrf2. Free Radic Biol Med 2024; 222:27-40. [PMID: 38815774 DOI: 10.1016/j.freeradbiomed.2024.05.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/01/2024]
Abstract
Liver fibrosis is a key and reversible stage in the progression of many chronic liver diseases to cirrhosis or hepatocellular carcinoma. Forsythiaside-A (FTA), a main compound isolated from Forsythiae Fructus, has an excellent liver protective activity. This study aims to investigate the efficacy of FTA in improving cholestatic liver fibrosis. Bile-duct-ligation (BDL) was conducted to induce liver fibrosis in mice. Hepatic collagen deposition was evaluated by Masson and Sirus red staining. The bile acid spectrum in the liver and serum was analyzed by mass spectrometry. Liver oxidative stress injury and mitochondria damage were observed by using Mito-Tracker Red fluorescence staining, transmission electron microscopy, etc. The level of ferrous iron (Fe2+) and the expression of ferroptosis-associated molecules were detected. The binding between FTA and its target protein was confirmed by Co-immunoprecipitation (Co-IP), cellular thermal shift assay (CETSA), drug affinity responsive target stability (DARTS) and surface plasmon resonance (SPR). Our results demonstrated that FTA alleviated BDL-induced liver fibrosis in mice. FTA did not decrease the elevated amount of bile acids in BDL-treated mice, but reduced the bile acid-induced mitochondrial damage, oxidative stress and ferroptosis in hepatocytes, and also induced nuclear factor erythroid 2-related factor-2 (Nrf2) activation. In Nrf2 knock-out mice, the FTA-provided protection against BDL-induced liver fibrosis was disappeared, and FTA's inhibition on mitochondrial damage, oxidative stress and ferroptosis were lowered. Further results displayed that FTA could directly bind to Kelch-like ECH-associated protein-1 (Keap1), thereby activating Nrf2. Moreover, the BDL-induced liver fibrosis was markedly weakened in liver-specific Keap1 knockout mice. Hence, this study suggests that FTA alleviated the BDL-induced liver fibrosis through attenuating mitochondrial damage and ferroptosis in hepatocytes by activating Nrf2 via directly binding to Keap1.
Collapse
Affiliation(s)
- Qian Guo
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Zeqi Wu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Keke Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Jionghua Shi
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Mengjuan Wei
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Bin Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China
| | - Zhenlin Huang
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| | - Lili Ji
- The MOE Key Laboratory for Standardization of Chinese Medicines, Shanghai Key Laboratory of Compound Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai, 201203, China.
| |
Collapse
|
3
|
Xiong A, Lu L, Jiang K, Wang X, Chen Y, Wang X, Zhang W, Zhuge Y, Huang W, Li L, Liao Q, Yang F, Liu P, Ding L, Wang Z, Yang L. Functional metabolomics characterizes the contribution of farnesoid X receptor in pyrrolizidine alkaloid-induced hepatic sinusoidal obstruction syndrome. Arch Toxicol 2024; 98:2557-2576. [PMID: 38703205 DOI: 10.1007/s00204-024-03762-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 04/10/2024] [Indexed: 05/06/2024]
Abstract
Consumption of herbal products containing pyrrolizidine alkaloids (PAs) is one of the major causes for hepatic sinusoidal obstruction syndrome (HSOS), a deadly liver disease. However, the crucial metabolic variation and biomarkers which can reflect these changes remain amphibious and thus to result in a lack of effective prevention, diagnosis and treatments against this disease. The aim of the study was to determine the impact of HSOS caused by PA exposure, and to translate metabolomics-derived biomarkers to the mechanism. In present study, cholic acid species (namely, cholic acid, taurine conjugated-cholic acid, and glycine conjugated-cholic acid) were identified as the candidate biomarkers (area under the ROC curve 0.968 [95% CI 0.908-0.994], sensitivity 83.87%, specificity 96.55%) for PA-HSOS using two independent cohorts of patients with PA-HSOS. The increased primary bile acid biosynthesis and decreased liver expression of farnesoid X receptor (FXR, which is known to inhibit bile acid biosynthesis in hepatocytes) were highlighted in PA-HSOS patients. Furtherly, a murine PA-HSOS model induced by senecionine (50 mg/kg, p.o.), a hepatotoxic PA, showed increased biosynthesis of cholic acid species via inhibition of hepatic FXR-SHP singling and treatment with the FXR agonist obeticholic acid restored the cholic acid species to the normal levels and protected mice from senecionine-induced HSOS. This work elucidates that increased levels of cholic acid species can serve as diagnostic biomarkers in PA-HSOS and targeting FXR may represent a therapeutic strategy for treating PA-HSOS in clinics.
Collapse
Affiliation(s)
- Aizhen Xiong
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
- Shanghai R & D Center for Standardization of Traditional Chinese Medicines, Shanghai, 201210, China.
| | - Longhui Lu
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Kaiyuan Jiang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Xiaoning Wang
- E-Institute of Shanghai Municipal Education Committee, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yan Chen
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Xunjiang Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Wei Zhang
- Department of Gastroenterology, The Drum Tower Hospital of Nanjing, Affiliated to Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Yuzheng Zhuge
- Department of Gastroenterology, The Drum Tower Hospital of Nanjing, Affiliated to Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Lujin Li
- Center for Drug of Clinical Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Qi Liao
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China
| | - Fan Yang
- Department of Obstetrics and Gynecology, and Shanghai Key Laboratory of Gynecologic Oncology Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ping Liu
- E-Institute of Shanghai Municipal Education Committee, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lili Ding
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
- Shanghai R & D Center for Standardization of Traditional Chinese Medicines, Shanghai, 201210, China.
| | - Zhengtao Wang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
- Shanghai R & D Center for Standardization of Traditional Chinese Medicines, Shanghai, 201210, China.
| | - Li Yang
- The MOE Key Laboratory for Standardization of Chinese Medicines and the SATCM Key Laboratory for New Resources and Quality Evaluation of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 201210, China.
- Shanghai R & D Center for Standardization of Traditional Chinese Medicines, Shanghai, 201210, China.
| |
Collapse
|
4
|
Custers E, Vreeken D, Schuren F, van den Broek TJ, van Dongen L, Geenen B, de Blaauw I, Wiesmann M, Hazebroek EJ, Kleemann R, Kiliaan AJ. Impact of Microbiota and Metabolites on Intestinal Integrity and Inflammation in Severe Obesity. Pharmaceuticals (Basel) 2024; 17:918. [PMID: 39065768 PMCID: PMC11279642 DOI: 10.3390/ph17070918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/03/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Obesity is a multifactorial disease associated with low-grade inflammation. The gut is thought to be involved in obesity-related inflammation, as it is continuously exposed to antigens from food, microbiota and metabolites. However, the exact underlying mechanisms are still unknown. Therefore, we examined the relation between gut pathology, microbiota, its metabolites and cytokines in adults with severe obesity. Individuals eligible for bariatric surgery were included. Fecal and plasma samples were collected at surgery timepoint, to assess microbiota and metabolite composition. Jejunal biopsies were collected during surgery and stained for cytotoxic T cells, macrophages, mast cells and tight junction component zonula occludens-1. Based on these stainings, the cohort was divided into four groups: high versus low intestinal inflammation and high versus low intestinal integrity. We found no significant differences in microbiota diversity between groups, nor for individual bacterial species. No significant differences in metabolites were observed between the intestinal inflammatory groups. However, some metabolites and cytokines differed between the intestinal integrity groups. Higher plasma levels of interleukin-8 and tauro-chenodeoxycholic acid were found, whereas isovaleric acid and acetic acid were lower in the high intestinal integrity group. As the results were very subtle, we suggest that our cohort shows very early and minor intestinal pathology.
Collapse
Affiliation(s)
- Emma Custers
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, 6500 HB Nijmegen, The Netherlands; (E.C.); (B.G.); (M.W.)
- Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands;
| | - Debby Vreeken
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, 6500 HB Nijmegen, The Netherlands; (E.C.); (B.G.); (M.W.)
- Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands;
| | - Frank Schuren
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands; (F.S.); (R.K.)
| | - Tim J. van den Broek
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands; (F.S.); (R.K.)
| | - Lieke van Dongen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, 6500 HB Nijmegen, The Netherlands; (E.C.); (B.G.); (M.W.)
| | - Bram Geenen
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, 6500 HB Nijmegen, The Netherlands; (E.C.); (B.G.); (M.W.)
| | - Ivo de Blaauw
- Division of Pediatric Surgery, Department of Surgery, Radboudumc-Amalia Children’s Hospital, 6525 GA Nijmegen, The Netherlands;
| | - Maximilian Wiesmann
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, 6500 HB Nijmegen, The Netherlands; (E.C.); (B.G.); (M.W.)
| | - Eric J. Hazebroek
- Department of Bariatric Surgery, Vitalys, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands;
- Division of Human Nutrition and Health, Wageningen University and Research, 6708 WE Wageningen, The Netherlands
| | - Robert Kleemann
- Department of Metabolic Health Research, The Netherlands Organization for Applied Scientific Research (TNO), 2333 BE Leiden, The Netherlands; (F.S.); (R.K.)
| | - Amanda J. Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Radboud Alzheimer Center, Donders Institute for Brain Cognition and Behaviour, Center for Medical Neuroscience, 6500 HB Nijmegen, The Netherlands; (E.C.); (B.G.); (M.W.)
| |
Collapse
|
5
|
Li M, Zhang D, Yang Q, Zhao Z, Zhang C, Zhou Y, Bai Y, Chen L, Tang X, Liu C, Zhou J, Chen X, Ying B. Longitudinal metabolomics of human plasma reveal metabolic dynamics and predictive markers of antituberculosis drug-induced liver injury. Respir Res 2024; 25:254. [PMID: 38907347 PMCID: PMC11193241 DOI: 10.1186/s12931-024-02837-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 05/04/2024] [Indexed: 06/23/2024] Open
Abstract
Tuberculosis (TB) remains the second leading cause of death from a single infectious agent and long-term medication could lead to antituberculosis drug-induced liver injury (ATB-DILI). We established a prospective longitudinal cohort of ATB-DILI with multiple timepoint blood sampling and used untargeted metabolomics to analyze the metabolic profiles of 107 plasma samples from healthy controls and newly diagnosed TB patients who either developed ATB-DILI within 2 months of anti-TB treatment (ATB-DILI subjects) or completed their treatment without any adverse drug reaction (ATB-Ctrl subjects). The untargeted metabolome revealed that 77 metabolites (of 895 total) were significantly changed with ATB-DILI progression. Among them, levels of multiple fatty acids and bile acids significantly increased over time in ATB-DILI subjects. Meanwhile, metabolites of the same class were highly correlated with each other and pathway analysis indicated both fatty acids metabolism and bile acids metabolism were up-regulated with ATB-DILI progression. The targeted metabolome further validated that 5 fatty acids had prediction capability at the early stage of the disease and 6 bile acids had a better diagnostic performance when ATB-DILI occurred. These findings provide evidence indicating that fatty acids metabolism and bile acids metabolism play a vital role during ATB-DILI progression. Our report adds a dynamic perspective better to understand the pathological process of ATB-DILI in clinical settings.
Collapse
Affiliation(s)
- Mengjiao Li
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Zhang
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qingxin Yang
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenzhen Zhao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chunying Zhang
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yanbing Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yangjuan Bai
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Chen
- Department of Laboratory Medicine, State Key Laboratory of Biotherapy and Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyan Tang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Cuihua Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.
| | - Juan Zhou
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.
| | - Xuerong Chen
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China.
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
6
|
Tao W, Fan Q, Wei J. Gut-Liver Axis as a Therapeutic Target for Drug-Induced Liver Injury. Curr Issues Mol Biol 2024; 46:1219-1236. [PMID: 38392196 PMCID: PMC10887627 DOI: 10.3390/cimb46020078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/24/2024] Open
Abstract
Drug-induced liver injury (DILI) is a liver disease that remains difficult to predict and diagnose, and the underlying mechanisms are yet to be fully clarified. The gut-liver axis refers to the reciprocal interactions between the gut and the liver, and its homeostasis plays a prominent role in maintaining liver health. It has been recently reported that patients and animals with DILI have a disrupted gut-liver axis, involving altered gut microbiota composition, increased intestinal permeability and lipopolysaccharide translocation, decreased short-chain fatty acids production, and impaired bile acid metabolism homeostasis. The present review will summarize the evidence from both clinical and preclinical studies about the role of the gut-liver axis in the pathogenesis of DILI. Moreover, we will focus attention on the potential therapeutic strategies for DILI based on improving gut-liver axis function, including herbs and phytochemicals, probiotics, fecal microbial transplantation, postbiotics, bile acids, and Farnesoid X receptor agonists.
Collapse
Affiliation(s)
- Wenjing Tao
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Qiwen Fan
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| | - Jintao Wei
- Hubei Key Laboratory of Animal Embryo and Molecular Breeding, Institute of Animal Husbandry and Veterinary, Hubei Academy of Agricultural Sciences, Wuhan 430064, China
| |
Collapse
|
7
|
Kovacevic B, Jones M, Wagle SR, Ionescu CM, Foster T, Đanić M, Mikov M, Mooranian A, Al-Salami H. Influence of poly-L-ornithine-bile acid nano hydrogels on cellular bioactivity and potential pharmacological applications. Ther Deliv 2023. [PMID: 37667908 DOI: 10.4155/tde-2023-0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/06/2023] Open
Abstract
Aim: Cellular bioactivity and pathophysiological changes associated with chronic disorders are considered pivotal detrimental factors when developing novel formulations for biomedical applications. Methods: This paper investigates the use of bile acids and synthetic polypeptide poly-L-ornithine (PLO) in formulations and their impacts on a variety of cell lines, with a particular focus on their cellular bioactivity. Results: The hepatic cell line was the most negatively affected by the presence of PLO, while the muscle and beta-pancreatic cell lines did not show as profound of a negative impact of PLO on cellular viability. PLO was the least disruptive regarding mitochondrial function for muscle and beta cells. Conclusion: The addition of bile acids generally decreased mitochondrial respiration and altered bioenergetic parameters in all cell lines.
Collapse
Affiliation(s)
- Bozica Kovacevic
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Melissa Jones
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Susbin Raj Wagle
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Corina Mihaela Ionescu
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Thomas Foster
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
| | - Maja Đanić
- Department of Pharmacology, Toxicology & Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, 21101, Serbia
| | - Momir Mikov
- Department of Pharmacology, Toxicology & Clinical Pharmacology, Faculty of Medicine, University of Novi Sad, Novi Sad, 21101, Serbia
| | - Armin Mooranian
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
- School of Pharmacy, University of Otago, Dunedin, Otago, 9016, New Zealand
| | - Hani Al-Salami
- The Biotechnology & Drug Development Research Laboratory, Curtin Medical School & Curtin Health Innovation Research Institute, Curtin University, Bentley, Perth, WA 6102, Australia
- Medical School, University of Western Australia, Perth, 6000, Australia
| |
Collapse
|
8
|
Harnisch LO, Mihaylov D, Bein T, Apfelbacher C, Moerer O, Quintel M. A reduced glycine-to-taurine ratio of conjugated serum bile acids signifies an adaptive mechanism and is an early marker of outcome in acute respiratory distress syndrome. Intern Emerg Med 2023; 18:607-615. [PMID: 36378472 DOI: 10.1007/s11739-022-03152-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/03/2022] [Indexed: 11/16/2022]
Abstract
The accumulation of Bile Acids (BA) in serum is a common finding in critically ill patients and has been found in patients with Acute Respiratory Distress Syndrome (ARDS), where liver and biliary function could be essentially affected by the underlying disease process and subsequent therapeutic measures. We hypothesized that the glycine-to-taurine conjugation ratio (G/T-ratio) is predictive of outcome in ARDS patients and would support our previously published hypothesis that the BA profile reflects a (mal-) adaptive response of bile acid production when suffering from a disease or syndrome such as ARDS. In 70 patients with ARDS, we determined conjugated BA fractions from protein precipitated serum samples using a LC-MS/MS method and calculated the G/T-ratios, which were then compared with a healthy control group. In patients with ARDS, the G/T-ratio was markedly lower compared to the control group, due to an increase in taurine-conjugated BA. The G/T ratio was lowest on the day of diagnosis and increased steadily during the following days (control = 3.80 (2.28-4.44); day 0 = 1.79 (1.31-3.86); day 3 = 2.91 (1.71-5.68); day 5 = 2.28 (1.25-7.85), significant increases were found between day 0 and day 3 (p = 0.019) and between day 0 and day 5 (p = 0.031). G/T-ratio was significantly correlated with SAPS II score on day 0 (p = 0.009) and day 3 (p = 0.036) and with survival (p = 0.006). Regarding survival, the receiver-operator characteristic revealed an area-under-the-curve of 0.713 (CI 0.578-0.848), the Youden index revealed a G/T-ratio cut-off level of 2.835 (sensitivity 78.4%, specificity 63.2%). Our findings further support our previously published hypothesis that alterations in BA profiles represent adaptive mechanisms in states of severe disease. Our current study adds the finding of an increase in taurine-conjugated BA expressed by a decrease in the G/T-ratio of conjugated BA in serum. The G/T-ratio on day 3 using a threshold G/T-ratio of 2.8 was even associated with survival (p = 0.006); these results are yet to be confirmed by subsequent studies.
Collapse
Affiliation(s)
- Lars-Olav Harnisch
- Department of Anaesthesiology, University of Göttingen Medical Center, Robert-Koch-Street 40, 37099, Göttingen, Germany.
| | - Diana Mihaylov
- Institute of Clinical Chemistry and Laboratory Medicine of the University Hospital Jena, Am Klinikum 1, 07747, Jena, Germany
| | - Thomas Bein
- University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053, Regensburg, Germany
| | - Christian Apfelbacher
- Institute for Social Medicine and Health Economics, University of Magdeburg, Leipziger Street 44, 39120, Magdeburg, Germany
| | - Onnen Moerer
- Department of Anaesthesiology, University of Göttingen Medical Center, Robert-Koch-Street 40, 37099, Göttingen, Germany
| | - Michael Quintel
- Department of Anaesthesiology, University of Göttingen Medical Center, Robert-Koch-Street 40, 37099, Göttingen, Germany
| |
Collapse
|
9
|
Shen L, Wang C, Wang R, Hu X, Liao S, Liu W, Du A, Ji S, Galon EM, Li H, Xuan X, Xiao J, Liu M. Serum metabolomic profiles in BALB/c mice induced by Babesia microti infection. Front Cell Infect Microbiol 2023; 13:1179967. [PMID: 37187473 PMCID: PMC10176453 DOI: 10.3389/fcimb.2023.1179967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
Introduction The protozoan parasite Babesia microti is the primary cause of human babesiosis. This parasite invades and multiplies inside red blood cells (RBCs), and infections differ significantly based on the age and immune competency of the host. The aim of this study was to investigate the use of serum metabolic profiling to identify systemic metabolic variations between B. microti-infected mice and noninfected controls. Methods A serum metabolomics analysis of BALB/c mice that had been intraperitoneally injected with 107 B. microti-infected RBCs was performed. Serum samples from the early infected group (2 days postinfection), the acutely infected group (9 days postinfection), and the noninfected group were collected and evaluated using a liquid chromatography-mass spectrometry (LC-MS) platform. Principal component analysis (PCA), partial least squares discriminant analysis (PLS-DA), and orthogonal partial least squares discriminant analysis (OPLS-DA) identified metabolomic profiles that differentiated the B. microti-infected and noninfected groups. Results Our results confirm that the serum metabolome is significantly influenced by acute B. microti infection and show that infection results in dysregulation of metabolic pathways and perturbation of metabolites. Acutely infected mice displayed perturbations in metabolites associated with taurine and hypotaurine metabolism, histidine metabolism, and arachidonic acid metabolism. Taurocholic acid, anserine, and arachidonic acid may be potential candidates as serological biomarkers for diagnosing B. microti infection at the acute stage. These metabolites could be further examined for their role in disease complexity. Discussion Our findings demonstrate that the acute stage of B. microti infection induces abnormalities in the metabolites present in mouse serum and provide new insight into the mechanisms involved in systemic metabolic changes that occur during B. microti infection.
Collapse
Affiliation(s)
- Liang Shen
- Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Chunhua Wang
- Central Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Ruilin Wang
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
| | - Xue Hu
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
| | - Shiying Liao
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
| | - Wentong Liu
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
| | - Aoling Du
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
| | - Shengwei Ji
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Eloiza May Galon
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Hang Li
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Xuenan Xuan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Juan Xiao
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
- *Correspondence: Mingming Liu, ; Juan Xiao,
| | - Mingming Liu
- School of Basic Medicine, Hubei University of Arts and Science, Xiangyang, China
- *Correspondence: Mingming Liu, ; Juan Xiao,
| |
Collapse
|
10
|
Woo SM, Alhaqqan DM, Gildea DT, Patel PA, Cundra LB, Lewis JH. Highlights of the drug-induced liver injury literature for 2021. Expert Rev Gastroenterol Hepatol 2022; 16:767-785. [PMID: 35839342 DOI: 10.1080/17474124.2022.2101996] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
INTRODUCTION In 2021, over 3,000 articles on Drug-Induced Liver Injury (DILI) were published, nearly doubling the annual number compared to 2011. This review selected DILI articles from 2021 we felt held the greatest interest and clinical relevance. AREAS COVERED A literature search was conducted using PubMed between 1 March 2021 and 28 February 2022. 86 articles were included. This review discusses new and established cases of hepatotoxins, including new FDA approvals and COVID-19 therapeutics. Developments in biomarkers and causality assessment methods are discussed. Updates from registries are also explored. EXPERT OPINION DILI diagnosis and prognostication remain challenging. Roussel Uclaf Causality Assessment Method (RUCAM) is the best option for determining causality and has been increasingly accepted by clinicians. Revised Electronic Causality Assessment Method (RECAM) may be more user-friendly and accurate but requires further validation. Quantitative systems pharmacology methods, such as DILIsym, are increasingly used to predict hepatotoxicity. Oncotherapeutic agents represent many newly approved and described causes of DILI. Such hepatotoxicity is deemed acceptable relative to the benefit these drugs offer. Drugs developed for non-life-threatening disorders may not show a favorable benefit-to-risk ratio and will be more difficult to approve. As the COVID-19 landscape evolves, its effect on DILI deserves further investigation.
Collapse
Affiliation(s)
- Stephanie M Woo
- Department of Internal Medicine, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Dalal M Alhaqqan
- Department of Gastroenterology, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Daniel T Gildea
- Department of Internal Medicine, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Palak A Patel
- Department of Internal Medicine, MedStar Georgetown University Hospital, Washington, DC, USA
| | - Lindsey B Cundra
- Department of Internal Medicine, MedStar Georgetown University Hospital, Washington, DC, USA
| | - James H Lewis
- Department of Gastroenterology, MedStar Georgetown University Hospital, Washington, DC, USA
| |
Collapse
|
11
|
Wang L, Dong XL, Qin XM, Li ZY. Investigating the inter-individual variability of Astragali Radix against cisplatin-induced liver injury via 16S rRNA gene sequencing and LC/MS-based metabolomics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 101:154107. [PMID: 35561503 DOI: 10.1016/j.phymed.2022.154107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 03/31/2022] [Accepted: 04/14/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Cisplatin (CDDP), one of the widely used chemotherapeutic drugs, can induce a series of side effects, such as hepatotoxicity and gastrointestinal toxicity. Astragali Radix (AR) is widely used as the tonic herbal medicine in traditional Chinese medicine (TCM). However, there was no report about the hepatoprotective effect of AR against the cisplatin-induced hepatic damage. PURPOSE This study aimed to investigate the protective effect and potential mechanism of AR water extract against the cisplatin-induced liver injury. METHODS Cisplatin was utilized to induce the liver injury using ICR mice, and the protective effect of AR was evaluated by serum biochemistry indices and liver histopathology. Then UHPLC Q-TOF-MS/MS-based untargeted serum metabolomics approach combined with 16S rRNA-based microbiota analysis was used to explore the underlying biomarkers and mechanism about the liver-protective effect of AR. RESULTS AR could decrease the serum AST and ALT, ameliorate hepatic pathological damages caused by cisplatin. Serum metabolomics indicated AR could regulate the biosynthesis of unsaturated fatty acids, arachidonic acid metabolism, purine metabolism, and fatty acid biosynthesis. In addition, 16S rRNA gene sequencing analysis showed that AR could regulate cisplatin-induced gut microbiota disorder, especially the inflammation-related bacteria (p_Deferribacteres, g_Enterococcus, and g_Alistipes, etc.), and the short chain fatty acids (SCFAs)-producing bacteria (g_Alloprevotella, g_Intestinimoas, and g_Flavonifractor). Moreover, 7 mice (AR-7) showed better liver protective effect than the other 3 mice (AR-3), and their regulatory effect on the gut microbiota and serum metabolites were also different, indicating the presence of inter-individual variability for the liver protective effect of AR. CONCLUSIONS This study revealed the protective effect and the potential mechanisms of AR against cisplatin-induced liver injury, and found that inter-individual variability of the liver protective effect of AR was related to the host microbiome and metabolome. These findings provided new insight into the health effect of dietary AR as a functional food for cisplatin-based chemotherapy.
Collapse
Affiliation(s)
- Ling Wang
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No.92, Wucheng Road, Taiyuan, Shanxi 030006, China
| | - Xian-Long Dong
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No.92, Wucheng Road, Taiyuan, Shanxi 030006, China
| | - Xue-Mei Qin
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No.92, Wucheng Road, Taiyuan, Shanxi 030006, China
| | - Zhen-Yu Li
- Modern Research Center for Traditional Chinese Medicine, the Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Shanxi University, No.92, Wucheng Road, Taiyuan, Shanxi 030006, China.
| |
Collapse
|
12
|
Wang CY, Deng Y, Li P, Zheng S, Chen G, Zhou G, Xu J, Chen YP, Wang Z, Jin X, Tang JM, Hu KP, Bi JF, Zhang P, Li CX, Huang A, Lv GJ, Xiao XH, Zou Z, Ji D. Prediction of biochemical nonresolution in patients with chronic drug-induced liver injury: A large multicenter study. Hepatology 2022; 75:1373-1385. [PMID: 34919746 DOI: 10.1002/hep.32283] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 11/23/2021] [Accepted: 12/12/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS To clarify high-risk factors and develop a nomogram model to predict biochemical resolution or biochemical nonresolution (BNR) in patients with chronic DILI. APPROACH AND RESULTS Retrospectively, 3655 of 5326 patients with chronic DILI were enrolled from nine participating hospitals, of whom 2866 underwent liver biopsy. All of these patients were followed up for over 1 year and their clinical characteristics were retrieved from electronic medical records. The endpoint was BNR, defined as alanine aminotransferase or aspartate aminotransferase >1.5× upper limit of normal or alkaline phosphatase >1.1× ULN, at 12 months from chronic DILI diagnosis. The noninvasive high-risk factors for BNR identified by multivariable logistic regression were used to establish a nomogram, which was validated in an independent external cohort. Finally, 19.3% (707 of 3655) patients presented with BNR. Histologically, with the increase in liver inflammation grades and fibrosis stages, the proportion of BNR significantly increased. The risk of BNR was increased by 21.3-fold in patients with significant inflammation compared to none or mild inflammation (p < 0.001). Biochemically, aspartate aminotransferase and total bilirubin, platelets, prothrombin time, sex, and age were associated with BNR and incorporated to construct a nomogram model (BNR-6) with a concordance index of 0.824 (95% CI, 0.798-0.849), which was highly consistent with liver histology. These results were successfully validated both in the internal cohort and external cohort. CONCLUSIONS Significant liver inflammation is a robust predictor associated with biochemical nonresolution. The established BNR-6 model provides an easy-to-use approach to assess the outcome of chronic DILI.
Collapse
Affiliation(s)
- Chun-Yan Wang
- Senior Department of HepatologyFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Ya Deng
- The Second School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
| | - Ping Li
- Department of HepatologyTianjin Second People's HospitalTianjinChina
| | - Sujun Zheng
- First Department of Liver DiseaseBeijing YouAn HospitalCapital Medical UniversityBeijingChina
| | - Guofeng Chen
- Senior Department of HepatologyFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Guangde Zhou
- Department of PathologyThird People's Hospital of ShenzhenShenzhenGuangdongChina
| | - Jing Xu
- Department of Liver DiseaseThe Second People's Hospital of Fuyang CityFuyangAnhuiChina
| | - Yan-Ping Chen
- Department of Infectious DiseasesYan'an Second People's HospitalYan'anShanxiChina
- Department of Infectious DiseasesYan'an University Affiliated HospitalYan'anShanxiChina
| | - Zheng Wang
- First Department of Liver DiseaseBeijing YouAn HospitalCapital Medical UniversityBeijingChina
| | - Xueyuan Jin
- Quality Control DepartmentFifth Medical Center of ChinesePLA General HospitalBeijingChina
| | - Jin-Mo Tang
- Department of Infectious DiseasesXiamen Hospital of Traditional Chinese MedicineXiamenFujianChina
| | - Kun-Peng Hu
- Department of General SurgeryThe Third Affiliated Hospital of Sun Yat-sen UniversityGuangzhouChina
| | - Jing-Feng Bi
- Epidemiology Research OfficeFifth Medical Center of ChinesePLA General HospitalBeijingChina
| | - Ping Zhang
- Third Department of Liver DiseasesLiaoyang Infection HospitalLiaoyangLiaoningChina
| | - Chun-Xia Li
- Department of Infectious DiseasesYan'an Second People's HospitalYan'anShanxiChina
| | - Ang Huang
- Senior Department of HepatologyFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Gui-Ji Lv
- Peking University 302 Clinical Medical SchoolBeijingChina
| | - Xiao-He Xiao
- Senior Department of HepatologyFifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Zhengsheng Zou
- Senior Department of HepatologyFifth Medical Center of Chinese PLA General HospitalBeijingChina
- Peking University 302 Clinical Medical SchoolBeijingChina
| | - Dong Ji
- Senior Department of HepatologyFifth Medical Center of Chinese PLA General HospitalBeijingChina
- The Second School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
- Peking University 302 Clinical Medical SchoolBeijingChina
| |
Collapse
|
13
|
Zhao S, Fu H, Zhou T, Cai M, Huang Y, Gan Q, Zhang C, Qian C, Wang J, Zhang Z, Wang X, Xiang X, Xie Q. Alteration of Bile Acids and Omega-6 PUFAs Are Correlated With the Progression and Prognosis of Drug-Induced Liver Injury. Front Immunol 2022; 13:772368. [PMID: 35493499 PMCID: PMC9041619 DOI: 10.3389/fimmu.2022.772368] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 03/21/2022] [Indexed: 11/13/2022] Open
Abstract
Background & Aims Drug-induced liver injury (DILI) is one of the leading causes of liver failure with some of the patients progressed to chronic DILI. The mechanisms underlying the severity and chronicity of DILI are poorly elucidated and the biomarkers are limited. Metabolites and gut microbiota played a crucial role in the development of various liver diseases. Herein, a systematic analysis of serum metabolites and gut microbiota was performed in DILI patients, aiming to identify metabolites correlated with the progression and clinical prognosis of DILI. Methods Various serum metabolites were quantitated using a metabolite array technology in this prospective study. Gut microbiome compositions and the expression profiles of liver genes were determined in patients with DILI and healthy controls. Results Metabolomic analysis revealed that bile acids (BAs) and polyunsaturated fatty acids (PUFAs) were closely related to DILI severity and chronicity respectively. The ratios of serum primary/secondary BAs and omega-6/omega-3 PUFAs were elevated in DILI patients. A model established by adrenic acid (AdA) and aspartic acid (Asp) exerts good performance for predicting the chronicity of DLIL. Hepatic transcriptome revealed enhanced expression of PUFA peroxidation and supressed expression of BA synthesis related genes in DILI patients. In addition, Lactic acid bacteria and BA converting bacteria were increased in gut of DILI patients. Besides, elevated serum malondialdehyde (MDA) and fibroblast growth factor 19 (FGF19) was observed in DILI patients. Conclusion BAs and PUFAs could be potent markers for the severity and chronicity of DILI respectively. The panel of AdA and Asp could be ideal predictive model for the risk of chronicity at the acute stage of DILI. Gut microbiota might act as a negative feedback mechanism to maintain the homeostasis of BAs and PUFAs via FGF19 signalling and PUFA saturation, respectively. Our study revealed novel biomarkers for severe and chronic DILI and provided new therapeutic targets for DILI.
Collapse
Affiliation(s)
- Shuang Zhao
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoshuang Fu
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianhui Zhou
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Minghao Cai
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Huang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qinyi Gan
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenxi Zhang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cong Qian
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiexiao Wang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhenglan Zhang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolin Wang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaogang Xiang
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing Xie
- Department of Infectious Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Shan Q, Tian G, Han X, Hui H, Yamamoto M, Hao M, Wang J, Wang K, Sang X, Qin L, Chen G, Cao G. Toxicity of Tetradium ruticarpum: Subacute Toxicity Assessment and Metabolomic Identification of Relevant Biomarkers. Front Pharmacol 2022; 13:803855. [PMID: 35295336 PMCID: PMC8918793 DOI: 10.3389/fphar.2022.803855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 01/12/2022] [Indexed: 11/19/2022] Open
Abstract
Tetradium ruticarpum (TR) is widely used in Asia to treat gastrointestinal disorders and pain. Stir-frying with licorice aqueous extract is a traditional processing procedure of TR formed in a long-term practice and performed before clinical application, and believed to reduce TR’s toxicity. However, its toxicity and possible toxicity attenuation approach are yet to be well investigated. Subacute toxicity and metabolomics studies were conducted to help understand the toxicity of TR. The subacute toxicity assessment indicated that 3 fold of the recommended therapeutic dose of TR did not show obvious subacute toxicity in rats. Although an extremely high dose (i.e., 60 fold of the recommended dose) may cause toxicity in rats, it reversed to normal after 2 weeks of recovery. Hepatocellular injury was the major toxic phenotype of TR-induced liver damage, indicating as aspartate aminotransferase (AST) and liver index increasing, with histopathologic findings as local hepatocyte necrosis, focal inflammatory cell infiltration, slightly bile duct hyperplasia, and partial hepatocyte vacuolation. Moreover, we evaluated the impact of processing in toxicity. TR processed with licorice could effectively reduce drug-induced toxicity, which is a valuable step in TR pretreatment before clinical application. Metabolomics profiling revealed that primary bile acid biosynthesis, steroid biosynthesis, and arachidonic acid metabolism were mainly involved in profiling the toxicity metabolic regulatory network. The processing procedure could back-regulate these three pathways, and may be in an Aryl hydrocarbon Receptor (AhR) dependent manner to alleviate the metabolic perturbations induced by TR. 7α-hydroxycholesterol, calcitriol, and taurocholic acid were screened and validated as the toxicity biomarkers of TR for potential clinical translation. Overall, the extensive subacute toxicity evaluation and metabolomic analysis would not only expand knowledge of the toxicity mechanisms of TR, but also provide scientific insight of traditional processing theory, and support clinical rational use of TR.
Collapse
Affiliation(s)
- Qiyuan Shan
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Gang Tian
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xin Han
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Hui Hui
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Mai Yamamoto
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
| | - Min Hao
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingwei Wang
- The Public Platform of Medical Research Center, Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Kuilong Wang
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xianan Sang
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Luping Qin
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Luping Qin, ; Guanqun Chen, ; Gang Cao,
| | - Guanqun Chen
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- *Correspondence: Luping Qin, ; Guanqun Chen, ; Gang Cao,
| | - Gang Cao
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
- *Correspondence: Luping Qin, ; Guanqun Chen, ; Gang Cao,
| |
Collapse
|
15
|
Xie Z, Zhang L, Chen E, Lu J, Xiao L, Liu Q, Zhu D, Zhang F, Xu X, Li L. Targeted Metabolomics Analysis of Bile Acids in Patients with Idiosyncratic Drug-Induced Liver Injury. Metabolites 2021; 11:852. [PMID: 34940610 PMCID: PMC8706581 DOI: 10.3390/metabo11120852] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/26/2021] [Accepted: 12/04/2021] [Indexed: 12/12/2022] Open
Abstract
Drug-induced liver injury (DILI) is rare but clinically important due to a high rate of mortality. However, specific biomarkers for diagnosing and predicting the severity and prognosis of DILI are lacking. Here, we used targeted metabolomics to identify and quantify specific types of bile acids that can predict the severity of DILI. A total of 161 DILI patients were enrolled in this prospective cohort study, as well as 31 health controls. A targeted metabolomics method was used to identify 24 types of bile acids. DILI patients were divided into mild, moderate, and severe groups according to disease severity. A multivariate analysis was performed to identify characteristic bile acids. Then the patients were divided into severe and non-severe groups, and logistic regression was used to identify bile acids that could predict DILI severity. Among the enrolled DILI patients, 32 were in the mild group, 90 were in the moderate group, and 39 were in the severe group. Orthogonal partial least squares-discriminant analysis (OPLS-DA) modeling clearly discriminated among the different groups. Among the four groups, glycochenodeoxycholate (GCDCA), taurochenodeoxycholate (TCDCA), deoxycholic acid (DCA), Nor Cholic acid (NorCA), glycocholic acid (GCA), and taurocholic acid (TCA) showed significant differences in concentration between at least two groups. NorCA, GCDCA, and TCDCA were all independent risk factors that differentiated severe DILI patients from the other groups. The area under the receiver operating characteristic curve (AUROC) of GCDCA, TCDCA, and NorCA was 0.856, 0.792, and 0.753, respectively. Together, these three bile acids had an AUROC of 0.895 for predicting severe DILI patients. DILI patients with different disease severities have specific bile acid metabolomics. NorCA, GCDTA, and TCDCA were independent risk factors for differentiating severe DILI patients from less-severe patients and have the potential to predict DILI severity.
Collapse
Affiliation(s)
- Zhongyang Xie
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; (Z.X.); (L.Z.); (J.L.); (L.X.); (Q.L.); (D.Z.); (F.Z.)
| | - Lingjian Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; (Z.X.); (L.Z.); (J.L.); (L.X.); (Q.L.); (D.Z.); (F.Z.)
| | - Ermei Chen
- Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen 361004, China;
| | - Juan Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; (Z.X.); (L.Z.); (J.L.); (L.X.); (Q.L.); (D.Z.); (F.Z.)
| | - Lanlan Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; (Z.X.); (L.Z.); (J.L.); (L.X.); (Q.L.); (D.Z.); (F.Z.)
| | - Qiuhong Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; (Z.X.); (L.Z.); (J.L.); (L.X.); (Q.L.); (D.Z.); (F.Z.)
| | - Danhua Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; (Z.X.); (L.Z.); (J.L.); (L.X.); (Q.L.); (D.Z.); (F.Z.)
| | - Fen Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; (Z.X.); (L.Z.); (J.L.); (L.X.); (Q.L.); (D.Z.); (F.Z.)
| | - Xiaowei Xu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; (Z.X.); (L.Z.); (J.L.); (L.X.); (Q.L.); (D.Z.); (F.Z.)
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310009, China; (Z.X.); (L.Z.); (J.L.); (L.X.); (Q.L.); (D.Z.); (F.Z.)
| |
Collapse
|
16
|
Thangamani S, Monasky R, Lee JK, Antharam V, HogenEsch H, Hazbun TR, Jin Y, Gu H, Guo GL. Bile Acid Regulates the Colonization and Dissemination of Candida albicans from the Gastrointestinal Tract by Controlling Host Defense System and Microbiota. J Fungi (Basel) 2021; 7:jof7121030. [PMID: 34947012 PMCID: PMC8708873 DOI: 10.3390/jof7121030] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/23/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
Candida albicans (CA), a commensal and opportunistic eukaryotic organism, frequently inhabits the gastrointestinal (GI) tract and causes life-threatening infections. Antibiotic-induced gut dysbiosis is a major risk factor for increased CA colonization and dissemination from the GI tract. We identified a significant increase of taurocholic acid (TCA), a major bile acid in antibiotic-treated mice susceptible to CA infection. In vivo findings indicate that administration of TCA through drinking water is sufficient to induce colonization and dissemination of CA in wild-type and immunosuppressed mice. Treatment with TCA significantly reduced mRNA expression of immune genes ang4 and Cxcr3 in the colon. In addition, TCA significantly decreased the relative abundance of three culturable species of commensal bacteria, Turicibacter sanguinis, Lactobacillus johnsonii, and Clostridium celatum, in both cecal contents and mucosal scrapings from the colon. Taken together, our results indicate that TCA promotes fungal colonization and dissemination of CA from the GI tract by controlling the host defense system and intestinal microbiota that play a critical role in regulating CA in the intestine.
Collapse
Affiliation(s)
- Shankar Thangamani
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA;
- Purdue Institute for Immunology, Inflammation and Infectious Diseases (PI4D), West Lafayette, IN 47906, USA
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA; (R.M.); (J.K.L.)
- Correspondence: ; Tel.: +1-765-494-0763
| | - Ross Monasky
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA; (R.M.); (J.K.L.)
| | - Jung Keun Lee
- College of Veterinary Medicine, Midwestern University, Glendale, AZ 85308, USA; (R.M.); (J.K.L.)
| | - Vijay Antharam
- Department of Chemistry, College of Arts, Humanities and Sciences, Methodist University, Fayetteville, NC 28311, USA;
| | - Harm HogenEsch
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN 47906, USA;
- Purdue Institute for Immunology, Inflammation and Infectious Diseases (PI4D), West Lafayette, IN 47906, USA
| | - Tony R. Hazbun
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, IN 47906, USA;
| | - Yan Jin
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (Y.J.); (H.G.)
| | - Haiwei Gu
- Arizona Metabolomics Laboratory, College of Health Solutions, Arizona State University, Phoenix, AZ 85004, USA; (Y.J.); (H.G.)
- Center for Translational Science, Department of Environmental Health Sciences, Robert Stempel College of Public Health and Social Work, Florida International University, Port St. Lucie, FL 33199, USA
| | - Grace L. Guo
- Department of Pharmacology and Toxicology, Earnest Mario School of Pharmacy, Rutgers University, Piscataway, NJ 08854, USA;
- Department of Veterans Affairs New Jersey Health Care System, East Orange, NJ 07018, USA
| |
Collapse
|
17
|
Liu N, Chen X, Song J, Chen M, Gong P, Jia W, Li G. Hypoglycemic effects of Auricularia auricula polysaccharides on high fat diet and streptozotocin-induced diabetic mice using metabolomics analysis. Food Funct 2021; 12:9994-10007. [PMID: 34505615 DOI: 10.1039/d1fo02022f] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
This study evaluated the hypoglycemic effect of Auricularia auricula polysaccharides (AAPs) on streptozotocin-induced type 2 diabetes mellitus (T2DM) mice using metabolomic analysis. The results of fasting blood glucose, oral glucose tolerance test, fasting serum insulin level, Homeostatic Model Assessment of Insulin Resistance index, TC, TG, HDL-C, LDL-C, and histopathological observation demonstrated that 200 mg per kg body weight per day AAP led to significant hypoglycemic activities. The metabolic profile of the mice was significantly changed after AAP intervention. 45 differential metabolites were screened as biomarkers for AAP adjuvant treatment, and AAPs' effects on the metabolism of amino acids, unsaturated fatty acids, bile acids, and glycerophospholipids were analyzed. Thus, the current results elucidated the metabolic pathway of AAPs for T2DM alleviation and provided guidance for functional food adjuvant development for T2DM treatment.
Collapse
Affiliation(s)
- Nannan Liu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China. .,College of Chemistry and Materials Science, Weinan Normal University, Weinan 714000, China
| | - Xuefeng Chen
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Juanna Song
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Mengyin Chen
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Pin Gong
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Wei Jia
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| | - Guoliang Li
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi'an 710021, China.
| |
Collapse
|