1
|
Cao Q, Liu L, Ma X, Zhong C, Tang M, Liu M, Qu LB, Wei B, Xu X. 1, 8-Cineole Ameliorated Staphylococcus aureus-Induced Pneumonia through Modulation of TRP-KYN and Arginine-NO Reprogramming. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:11670-11683. [PMID: 40314286 DOI: 10.1021/acs.jafc.4c10860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
1, 8-Cineole (Cin), a cyclic monoterpenoid derived from tea trees and eucalyptus species, exhibits diverse pharmacological properties. Yet, its therapeutic impact and underlying mechanism against Staphylococcus aureus (S. aureus) pneumonia remain to be elucidated. In this study, metabolomics based on UPLC-MS/MS was integrated with network pharmacology, molecular biology, and molecular docking to investigate the effects of Cin. The findings demonstrated that Cin markedly reduced mortality and lung bacterial load, lessened pulmonary damage while suppressing the levels of proinflammatory factors, including tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) in the bronchoalveolar lavage fluid (BALF) of infected mice. Additionally, 19 metabolites, primarily involved in tryptophan metabolism and arginine biosynthesis, were notably modified by Cin via suppressing the enzymatic activity of indoleamine 2, 3-dioxygenase 1 (IDO1) and inducible nitric oxide synthase (iNOS), thereby attenuating the inflammatory response. Notably, knockdown of IDO1 or iNOS significantly diminished the anti-inflammation effect of Cin. In conclusion, our study validates the therapeutic potential of Cin against S. aureus pneumonia via anti-inflammation by downregulating IDO1 and iNOS. Our results provide a theoretical basis of natural substances applied in bacterial pneumonia treatment.
Collapse
Affiliation(s)
- Qianwen Cao
- School of Pharmaceutical Science, Food Laboratory of Zhongyuan Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Luyao Liu
- School of Pharmaceutical Science, Food Laboratory of Zhongyuan Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xiaoge Ma
- School of Pharmaceutical Science, Food Laboratory of Zhongyuan Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Chaomin Zhong
- School of Pharmaceutical Science, Food Laboratory of Zhongyuan Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Mengqi Tang
- School of Pharmaceutical Science, Food Laboratory of Zhongyuan Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Mengge Liu
- School of Pharmaceutical Science, Food Laboratory of Zhongyuan Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Ling-Bo Qu
- School of Pharmaceutical Science, Food Laboratory of Zhongyuan Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Bo Wei
- School of Pharmaceutical Science, Food Laboratory of Zhongyuan Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xia Xu
- School of Pharmaceutical Science, Food Laboratory of Zhongyuan Zhengzhou University, Zhengzhou, Henan 450001, China
| |
Collapse
|
2
|
Chen Y, Jiang Y, Huang L, Li Z, Zhu M, Luo L, Zhou K, Chen M. Urate-lowering effect of delphinidin-3-glucoside in red kidney beans via binding to the FAD site of the XO enzyme. J Adv Res 2025:S2090-1232(25)00266-8. [PMID: 40254219 DOI: 10.1016/j.jare.2025.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 03/20/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND The incidence of hyperuricemia (HUA) is increasing globally, posing serious health risks. The discovery of natural urate-lowering agents is urgently needed. OBJECTIVE To discover natural urate-lowering agents and investigate their effect and action mechanisms for ameliorating HUA. METHODS Our study comprehensively explored the association between the intake of 13 specific legume varieties in the US population. A composition-target-metabolic (C-T-M) pathway network was constructed to identify key agents and their interactions with key proteins, which were verified by molecular dynamics simulations (MD) and surface plasmon resonance (SPR). Biochemical, in vitro, and in vivo metabolomic studies in male ICR mice were conducted to examine the effects of the key agent in red kidney beans on uric acid production and other metabolisms. RESULTS We found that consuming red kidney beans was robustly negatively associated with the risk of HUA. Based on the C-T-M network, delphinidin-3-glucoside (Dp-3G) was identified as the key agent in red kidney beans, focusing on its binding to xanthine oxidase (XO) enzyme. This interaction was subsequently verified by MD and SPR, revealing that Dp-3G binds to the FAD site of the XO enzyme, thereby blocking electron transfer during enzyme catalysis involving Moco, [2Fe-2S], and FAD. Dp-3G consistently reduces uric acid production under biochemical, in vitro, and in vivo conditions and reverses metabolic abnormalities related to HUA in mice, including methionine, proline, and folate. CONCLUSIONS This study identifies Dp-3G, a novel natural agent enriched in red kidney beans, as capable of occupying the FAD site of the XO enzyme, thereby interfering with uric acid synthesis, and suggesting its potential for preventing and treating HUA.
Collapse
Affiliation(s)
- Yanling Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Yingtong Jiang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Lei Huang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Ziyi Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Mengyuan Zhu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Lu Luo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Kun Zhou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Minjian Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China; Department of Occupational Medicine and Environmental Health, School of Public Health, Key Laboratory of Public Health Safety and Emergency Prevention and Control Technology of Higher Education Institutions in Jiangsu Province, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
3
|
Chuang YT, Yen CY, Liu W, Chien TM, Chang FR, Tsai YH, Tang JY, Chang HW. The protection of bisphenol A-modulated miRNAs and targets by natural products. ENVIRONMENT INTERNATIONAL 2025; 196:109299. [PMID: 39884249 DOI: 10.1016/j.envint.2025.109299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/04/2024] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Bisphenol A (BPA) is a ubiquitous environmental pollutant with endocrine-disrupting functions. Identifying protective drugs and exploring the mechanisms against BPA are crucial in healthcare. Natural products exhibiting antioxidant properties are considered to be able to protect against BPA toxicity. Although BPA-modulated targets and miRNAs have been individually reported, their connections to natural products were rarely organized. With the help of a protein-protein interaction database (STRING), the relationship between individual BPA-modulated targets was interconnected to provide a systemic view. In this review, BPA-downregulated and -upregulated targets are classified, and their interactive network was innovatively analyzed using the bioinformatic database (STRING). BPA-modulated miRNAs were also retrieved and ingeniously connected to BPA-modulated targets. Moreover, a novel connection between BPA-countering natural products was integrated into BPA-modulated miRNAs and targets. All these targets-associated natural products and/or miRNAs were incorporated into the STRING network, providing systemic relationships. Overall, the BPA-modulated target-miRNA-protecting natural product axis was innovatively constructed, providing a straightforward direction for exploring the integrated BPA-countering effects and mechanisms of natural products.
Collapse
Affiliation(s)
- Ya-Ting Chuang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Ching-Yu Yen
- School of Dentistry, Taipei Medical University, Taipei 11031, Taiwan; Department of Oral and Maxillofacial Surgery, Chi-Mei Medical Center, Tainan 71004, Taiwan.
| | - Wangta Liu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Tsu-Ming Chien
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Urology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Urology, Kaohsiung Gangshan Hospital, Kaohsiung Medical University, Kaohsiung 820111, Taiwan.
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Yi-Hong Tsai
- Department of Pharmacy and Master Program, College of Pharmacy and Health Care, Tajen University, Pingtung 907101, Taiwan.
| | - Jen-Yang Tang
- School of Post-Baccalaureate Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Radiation Oncology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Hsueh-Wei Chang
- Department of Biomedical Science and Environmental Biology, PhD Program in Life Sciences, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan.
| |
Collapse
|
4
|
Cui W, Cao Q, Liu L, Yin X, Wang X, Zhao Y, Wang Y, Wei B, Xu X, Tang Y. Artemisia Argyi essential oil ameliorates acetaminophen-induced hepatotoxicity via CYP2E1 and γ-glutamyl cycle reprogramming. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156106. [PMID: 39366156 DOI: 10.1016/j.phymed.2024.156106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 09/22/2024] [Accepted: 09/28/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND The hepatotoxicity induced by acetaminophen (APAP), a commonly used antipyretic, analgesic and anti-inflammatory drug in clinical practice, has received accumulated attention. Artemisia argyi essential oil (AAEO), a volatile oil component extracted from traditional Chinese medicine Artemisia argyi H.Lév. & Vaniot, has great hepatoprotective effects. However, the potential role of AAEO in APAP-induced hepatotoxicity has not been characterized. The present study aimed to investigate the effects of AAEO on hepatic metabolic changes in mice exposed to APAP. METHODS In this study, 300.00 mg/kg acetaminophen was used to establish liver injury model in C57BL/6 J mice. Hepatoprotective effect of AAEO on APAP-induced hepatotoxicity in mice was investigated by detecting liver function enzymes and histopathological examination. Secondly, UPLC-MS/MS was used to analyze the to analyze the small molecule metabolites and metabolic pathways induced by AAEO treatment; In addition, the effect of AAEO on APAP-induced oxidative stress and inflammation were evaluated by detecting the levels of glutathione peroxidase 4, malondialdehyde, reactive oxygen species and inflammatory factors. Finally, the active components of AAEO were preliminarily screened by cellular assays. The hepatoprotective effect of AAEO against APAP-induced hepatotoxicity was examined through the Western blotting, after the CYP2E1 gene was knocked down in AML12 cells by siRNA transfection. RESULTS Compared with the APAP group, AAEO could reduce the abnormal increase in the levels of liver function enzymes caused by APAP. AAEO could enhance the antioxidant capacity by down-regulating the biosynthesis pathway of unsaturated fatty acids and promoting the activity of antioxidant enzymes SOD and CAT in liver tissue induced by APAP. Our study revealed that AAEO promoted GSH synthesis and covalently combined to form APAP-GSH conjugates to reduce the accumulation of APAP in liver tissue. In addition, the chemical constituents in AAEO were analyzed by GC-MS/MS, and it was determined to identify that dihydro-beta-ionone and (-)-verbenone in AAEO might have a significant protective effect on hepatocyte survival after APAP exposure. Further studies on the hepatoprotective mechanism of AAEO indicated that it might reduce the production of toxic metabolites by regulating CYP2E1 levels. CONCLUSION AAEO exerted hepatoprotective effects on acetaminophen-induced hepatotoxicity in mice via regulating the activity of CYP2E1 and regulating the γ-glutamyl cycle pathway.
Collapse
Affiliation(s)
- Weiqi Cui
- Henan Key Laboratory of Rehabilitation Medicine, Henan Joint International Research Laboratory of Chronic Liver Injury, Henan Workshop of Chronic Liver Injury for Outstanding Overseas Scientists, Zhengzhou Key Laboratory of Metabolic-dysfunction-associated Fatty Liver Disease, Department of Pharmacy, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Qianwen Cao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Luyao Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China
| | - Xuecui Yin
- Henan Key Laboratory of Rehabilitation Medicine, Henan Joint International Research Laboratory of Chronic Liver Injury, Henan Workshop of Chronic Liver Injury for Outstanding Overseas Scientists, Zhengzhou Key Laboratory of Metabolic-dysfunction-associated Fatty Liver Disease, Department of Pharmacy, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xiaohan Wang
- Henan Key Laboratory of Rehabilitation Medicine, Henan Joint International Research Laboratory of Chronic Liver Injury, Henan Workshop of Chronic Liver Injury for Outstanding Overseas Scientists, Zhengzhou Key Laboratory of Metabolic-dysfunction-associated Fatty Liver Disease, Department of Pharmacy, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yang Zhao
- Henan Key Laboratory of Rehabilitation Medicine, Henan Joint International Research Laboratory of Chronic Liver Injury, Henan Workshop of Chronic Liver Injury for Outstanding Overseas Scientists, Zhengzhou Key Laboratory of Metabolic-dysfunction-associated Fatty Liver Disease, Department of Pharmacy, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yanhong Wang
- Henan Key Laboratory of Rehabilitation Medicine, Henan Joint International Research Laboratory of Chronic Liver Injury, Henan Workshop of Chronic Liver Injury for Outstanding Overseas Scientists, Zhengzhou Key Laboratory of Metabolic-dysfunction-associated Fatty Liver Disease, Department of Pharmacy, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Bo Wei
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Xia Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan 450001, China.
| | - Youcai Tang
- Henan Key Laboratory of Rehabilitation Medicine, Henan Joint International Research Laboratory of Chronic Liver Injury, Henan Workshop of Chronic Liver Injury for Outstanding Overseas Scientists, Zhengzhou Key Laboratory of Metabolic-dysfunction-associated Fatty Liver Disease, Department of Pharmacy, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
5
|
Rong Y, Tang M, Liu L, Ma X, Liu M, Qu L, Liao X, Jiang Q, Zhang N, Xu X. Artemisia argyi essential oil alleviates asthma by regulating 5-LOX-CysLTs and IDO-1-KYN pathways: Insights from metabolomics. JOURNAL OF ETHNOPHARMACOLOGY 2024; 333:118458. [PMID: 38871010 DOI: 10.1016/j.jep.2024.118458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/05/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Artemisia argyi essential oil (AAEO) is a traditional herbal remedy for asthma. However, the potential effect of AAEO on asthma has not been elucidated. AIM OF THE STUDY To investigate the protective properties of AAEO upon asthma and elucidate its mechanism. MATERIALS AND METHODS The effects of AAEO in asthma were assessed by histology and biochemical analysis. Then, we integrated real-time reverse transcription-quantitative polymerase chain reaction, enzyme-linked immunosorbent assay, immunohistochemistry and metabolomics analysis to reveal its mechanism. RESULTS In vivo, AAEO reduced the counts of white blood cells (WBCs) and cytokines in bronchoalveolar lavage fluid (BALF), ameliorated pathologic alterations in lung tissues, and inhibited secretion of OVA-sIgE and muc5ac. Metabolomics results showed that AAEO can exert therapeutic effects on asthmatic mice by regulating disordered arachidonic acid metabolism and tryptophan metabolism. Further studies shown that AAEO inhibited the expression of 5-LOX and reduced the accumulation of CysLTs in mice. Meanwhile, AAEO promoted the activity of IDO-1, facilitated the conversion of tryptophan to kynurenine, and regulated the imbalance of Treg/Th17 immunity. Immunohistochemical results showed that AAEO promoted the expression of IDO-1. RT-qPCR results showed that AAEO promoted the expression of IL-10 and Foxp3 mRNA, and inhibited the expression of IL-17A and RORγt mRNA, thus regulated the imbalance of Treg/Th17 immunity and exerted its therapeutic effects. CONCLUSION AAEO treatment not only attenuates the clinical symptoms of asthma but is also involved in regulating lung tissue metabolism. The anti-asthmatic activity of AAEO may be achieved by reprogramming 5-LOX-CysLTs and IDO-1-KYN pathways.
Collapse
Affiliation(s)
- Ying Rong
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Mengqi Tang
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Luyao Liu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Xiaoge Ma
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Mengge Liu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Lingbo Qu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| | - Xinglin Liao
- Nanyang LANHAISENYUAN Medical Technology Ltd.,CO, Nanyang, Henan, 473000, PR China
| | - Qiman Jiang
- Nanyang LANHAISENYUAN Medical Technology Ltd.,CO, Nanyang, Henan, 473000, PR China
| | - Nan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China.
| | - Xia Xu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, PR China
| |
Collapse
|
6
|
Song S, Qiu R, Huang Y, Zhou Z, Yan J, Ou Q, Wei D, He J, Liang Y, Du X, Yao W, Lu T. Study on the mechanism of hepatotoxicity of Aucklandiae radix through liver metabolomics and network pharmacology. Toxicol Res (Camb) 2024; 13:tfae123. [PMID: 39119266 PMCID: PMC11303830 DOI: 10.1093/toxres/tfae123] [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: 02/08/2024] [Revised: 07/15/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024] Open
Abstract
Background Aucklandiae Radix (CAR) and its roasted processed products (PAR) are extensively used in various Chinese patent medicines due to their diverse pharmacological activities. However, numerous side effects of CAR have been reported and the hepatotoxicity and the corresponding mechanisms have not been thoroughly investigated. Our study aims to explore the underlying mechanism of the hepatotoxic impacts of CAR. Methods In this study, metabolomic analysis was performed using liver tissue from the mice administered with different dosages of CAR/PAR extracts to examine the hepatotoxic impacts of CAR and elucidate the underlying mechanism. Network pharmacology was employed to predict the potential molecular targets and associated signaling pathways based on the distinctive compounds between CAR and PAR. A composition-target-GO-Bio process-metabolic pathway network was constructed by integrating the hepatotoxicity-related metabolic pathways. Finally, the target proteins related with the hepatotoxic effect of CAR were identified and validated in vivo. Results The metabolomics analysis revealed that 33 related metabolic pathways were significantly altered in the high-dose CAR group, four of which were associated with the hepatotoxicity and could be alleviated by PAR. The network identified NQO1 as the primary target of the hepatotoxic effect induced by CAR exposure, which was subsequently verified by Western Blotting. Further evidence in vivo demonstrated that Nrf2 and HO-1, closely related to NQO1, were also the main targets through which CAR induced the liver injury, and that oxidative stress should be the primary mechanism for the CAR-induced hepatotoxicity. Conclusions This preliminary study on the hepatic toxic injury of CAR provides a theoretical basis for the rational and safe use of CAR rationally and safely in clinical settings.
Collapse
Affiliation(s)
- Shen Song
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, China
| | - Rongli Qiu
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, China
| | - Yan Huang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, China
| | - Zhuxiu Zhou
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, China
| | - Jin Yan
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, China
| | - Qiaochan Ou
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, China
| | - Donghui Wei
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, China
| | - Jingxuan He
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, China
| | - Yi Liang
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, China
| | - Xingyue Du
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, China
| | - Weifeng Yao
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, China
| | - Tulin Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Xianlin Road 138, Nanjing 210023, China
| |
Collapse
|
7
|
Rong Y, Liu F, Zhou H, Yu T, Qin Z, Cao Q, Liu L, Ma X, Qu L, Xu P, Liao X, Jiang Q, Zhang N, Xu X. Reprogramming of arachidonic acid metabolism using α-terpineol to alleviate asthma: insights from metabolomics. Food Funct 2024; 15:4292-4309. [PMID: 38526853 DOI: 10.1039/d3fo04078j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Asthma is a chronic inflammatory disorder in airways with typical pathologic features of airway inflammation and mucus hypersecretion. α-Terpineol is a monocyclic terpene found in many natural plants and foods. It has been reported to possess a wide range of pharmacological activities including anti-inflammatory and expectorant effects. However, the role of α-terpineol in asthma and its potential protective mechanism have not been well elucidated. This study is designed to investigate the pharmacological effect and mechanism of α-terpineol on asthmatic mice using the metabolomics platform. A murine model of asthma was established using ovalbumin (OVA) sensitization and then challenged for one week. The leukocyte count and inflammatory cytokines in the bronchoalveolar lavage fluid (BALF), lung histopathology, inflammatory infiltrate and mucus secretion were evaluated. An ultra-high performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS)-based metabolomics study was performed on lung tissues and serum to explore endogenous small molecule metabolites affected by α-terpineol in asthmatic mice. After α-terpineol treatment, leukocyte count, inflammatory cytokines in the BALF, and peribronchial inflammation infiltration were significantly downregulated. Goblet cell hyperplasia and mucus secretion were attenuated, with the level of Muc5ac in BALF decreased. These results proved the protective effect of α-terpineol against airway inflammation, mucus hypersecretion and Th1/Th2 immune imbalance. To further investigate the underlying mechanisms of α-terpineol in asthma treatment, UPLC-MS/MS-based metabolomics analysis was performed. 26 and 15 identified significant differential metabolites were found in the lung tissues and serum of the control, model and α-terpineol groups, respectively. Based on the above differential metabolites, enrichment analysis showed that arachidonic acid (AA) metabolism was reprogrammed in both mouse lung tissues and serum. 5-Lipoxygenase (5-LOX) and cysteinyl leukotrienes (CysLTs) are the key enzyme and the end product of AA metabolism, respectively. In-depth studies have shown that pretreatment with α-terpineol can alleviate asthma by decreasing the AA level, downregulating the expression of 5-LOX and reducing the accumulation of CysLTs in mouse lung tissues. In summary, this study demonstrates that α-terpineol is a potential agent that can prevent asthma via regulating disordered AA metabolism.
Collapse
Affiliation(s)
- Ying Rong
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Fanglin Liu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Hui Zhou
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Tong Yu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Zhaolong Qin
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Qianwen Cao
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Luyao Liu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Xiaoge Ma
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Lingbo Qu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Peirong Xu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Xinglin Liao
- Nanyang LANHAISENYUAN Medical Technology Ltd, CO. Nanyang, Henan, 473000, China
| | - Qiman Jiang
- Nanyang LANHAISENYUAN Medical Technology Ltd, CO. Nanyang, Henan, 473000, China
| | - Nan Zhang
- Department of Pharmaceutics, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Xia Xu
- Department of Medical Analysis, School of Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
8
|
Charles DA, Prince SE. Deciphering the molecular mechanism of NLRP3 in BPA-mediated toxicity: Implications for targeted therapies. Heliyon 2024; 10:e28917. [PMID: 38596095 PMCID: PMC11002687 DOI: 10.1016/j.heliyon.2024.e28917] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/26/2024] [Accepted: 03/27/2024] [Indexed: 04/11/2024] Open
Abstract
Bisphenol-A (BPA), a pervasive industrial chemical used in polymer synthesis, is found in numerous consumer products including food packaging, medical devices, and resins. Detectable in a majority of the global population, BPA exposure occurs via ingestion, inhalation, and dermal routes. Extensive research has demonstrated the adverse health effects of BPA, particularly its disruption of immune and endocrine systems, along with genotoxic potential. This review focuses on the complex relationship between BPA exposure and the NOD-like receptor protein 3 (NLRP3) inflammasome, a multiprotein complex central to inflammatory disease processes. We examine how BPA induces oxidative stress through the generation of intracellular free radicals, subsequently activating NLRP3 signaling. The mechanistic details of this process are explored, including the involvement of signaling cascades such as PI3K/AKT, JAK/STAT, AMPK/mTOR, and ERK/MAPK, which are implicated in NLRP3 inflammasome activation. A key focus of this review is the wide-ranging organ toxicities associated with BPA exposure, including hepatic, renal, gastrointestinal, and cardiovascular dysfunction. We investigate the immunopathogenesis and molecular pathways driving these injuries, highlighting the interplay among BPA, oxidative stress, and the NLRP3 inflammasome. Finally, this review explores the emerging concept of targeting NLRP3 as a potential therapeutic strategy to mitigate the organ toxicities stemming from BPA exposure. This work integrates current knowledge, emphasizes complex molecular mechanisms, and promotes further research into NLRP3-targeted interventions.
Collapse
Affiliation(s)
- Doveit Antony Charles
- Department of Biotechnology, School of Biosciences and Technology, VIT, Vellore, Tamil Nadu, India
| | - Sabina Evan Prince
- Department of Biotechnology, School of Biosciences and Technology, VIT, Vellore, Tamil Nadu, India
| |
Collapse
|
9
|
Zhang L, Yu Y, Wang Q, Huang X, Feng Z, Li Z. Oridonin loaded peptide nanovesicles alleviate nonalcoholic fatty liver disease in mice. Pharm Dev Technol 2024; 29:123-130. [PMID: 38327230 DOI: 10.1080/10837450.2024.2315460] [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: 09/30/2023] [Accepted: 02/03/2024] [Indexed: 02/09/2024]
Abstract
This study was to construct a nanovesicle delivery system to improve the loading efficiency and stability of ORI for the treatment of nonalcoholic fatty liver disease (NAFLD). This nanovesicles (NVs) exerted a narrow size distribution (195.6 ± 11.49 nm) and high entrapment efficiency (84.46 ± 1.34%). In vitro cell studies demonstrated that the NVs treatment enhanced the cellular uptake of ORI and reduced lipid over-accumulation and total cholesterol levels in NAFLD cell model. At the same time, in vivo study proved that, compared with the normal group, the model group mice showed a decrease in body weight, a significant increase in liver index (6.71 ± 0.62, p < 0.01), and symptoms of liver lipid accumulation, lipid vesicles, and liver tissue fibrosis. Compared with the model group, after high-dose ORI NVs intervention, mice gained weight, decreased liver index (4.69 ± 0.55, p < 0.01), reduced hepatic lipid droplet vacuoles, reduced lipid accumulation (reduced oil red area, p < 0.001), and alleviated the degree of liver fibrosis (reduced blue collagen area, p < 0.001). In conclusion, ORI/HP-β-CD/H9-HePC NVs showed specific liver accumulation and improved therapeutic effects, the nano drug loading system provides a promising strategy for the encapsulation of ORI to effectively alleviate the process of NAFLD.
Collapse
Affiliation(s)
- Lifen Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Yao Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Qi Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xi Huang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zheng Feng
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Zhi Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
- Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, Zhengzhou, China
| |
Collapse
|
10
|
Cui W, Zhou H, Zhang J, Zhang J, Wu D, Rong Y, Liu F, Liu J, Liu H, Wei B, Tang Y, Liao X, Xu X. Hepatoprotective effect of Artemisia Argyi essential oil on bisphenol A-induced hepatotoxicity via inhibition of ferroptosis in mice. ENVIRONMENTAL TOXICOLOGY 2023; 38:2416-2428. [PMID: 37347548 DOI: 10.1002/tox.23877] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/19/2023] [Accepted: 06/11/2023] [Indexed: 06/24/2023]
Abstract
The environmental pollutant bisphenol A (BPA), used in the manufacture of plastic packaging materials for various diets, is widely distributed in the environment and causes severe hepatotoxicity by inducing oxidative stress. Artemisia argyi essential oil (AAEO), a volatile oil component isolated from Artemisia argyi H.Lév. & Vaniot, has pharmacological effects, especially for hepatoprotective actions. However, the potential effect of AAEO in BPA induced hepatotoxicity has not been characterized. First, we analyzed the chemical composition in AAEO by gas chromatography-mass spectrometry. Herein, we investigated the effect of AAEO on hepatic metabolic changes in mice exposed to BPA. Results showed that compared with the BPA group, AAEO could reduce the level of liver function enzymes in BPA mice serum, and ameliorate hepatic lesions and fibrosis. Additionally, 20 differential metabolites screened by metabolomics were mainly involved in the reprogramming of glutathione metabolism, purine metabolism, and polyunsaturated fatty acid synthesis. Moreover, AAEO could reduce hepatic ferroptosis induced by BPA, as demonstrated by reducing xanthine oxidase activity, up-regulating the activities of glutathione peroxidase 4 (GPX4), superoxide dismutase, and catalase and the expression of SLC7A11 to promote the glutathione synthetic, while inhibiting transferrin receptor 1 (TFR1) expression to reduce the accumulation of Fe2+ in cells. Therefore, our study identified AAEO as a hepatic protectant against BPA-induced hepatotoxicity by reversing the occurrence of ferroptosis.
Collapse
Affiliation(s)
- Weiqi Cui
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Hui Zhou
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jingxian Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junwei Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Deqiao Wu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ying Rong
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Fanglin Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Junhui Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Haiyan Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Bo Wei
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Youcai Tang
- Henan Joint International Research Laboratory of Chronic Liver Injury, Henan Key Laboratory of Rehabilitation Medicine, Department of Pediatrics, the Fifth Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xinglin Liao
- Nanyang Lanhaisenyuan Medical Technology Ltd, Co, Nanyang, China
| | - Xia Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Liu FL, Rong Y, Zhou H, Yu T, Liu L, Cao Q, Qin Z, Qu L, Liao X, Jiang Q, Zhang N, Xu X. Cineole inhibits the biosynthesis of leukotrienes and prostaglandins to alleviate allergic rhinitis: Insights from metabolomics. J Pharm Biomed Anal 2023; 234:115574. [PMID: 37481900 DOI: 10.1016/j.jpba.2023.115574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/25/2023]
Abstract
Allergic rhinitis (AR) is a common allergic disease characterized by nasal congestion, rhinorrhoea, and sneezing. Cineole, a monoterpenoid compound widely present in various volatile oils, has a wide range of pharmacological activities and is of interest in allergic airway diseases for its anti-inflammatory and anti-mucus production abilities. However, the protective effects of cineole in mice with allergic rhinitis and its mechanisms have not been well investigated. In this study, the protective effect of cineole against ovalbumin-induced (OVA-induced) allergic rhinitis and its molecular mechanism is investigated by metabolomic analysis based on ultra-high performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS). OVA combined with aluminum hydroxide adjuvant is used to sensitize and establish the allergic rhinitis (AR) mouse model. The mice are randomly divided into groups of control, AR, cineole (30 mg/kg), and budesonide (38.83 μg/kg). The pharmacodynamic results show that cineole significantly reduces the levels of Th2-type cytokines and OVA-specific IgE (OVA-sIgE) in AR mice, improves nasal mucosal tissue damage and alleviates nasal symptoms compared to the untreated AR group. Metabolomic results show that arachidonic acid (AA) metabolism and tryptophan (Trp) metabolism are reprogrammed on the basis of 27 significantly altered metabolites. Further studies show that cineole inhibits the biosynthesis of pro-inflammatory lipid mediators leukotrienes (LTs) and prostaglandins (PGs) in mice by inhibiting the activity of 5-lipoxygenase (5-LOX) and cyclooxygenase-2 (COX-2) in the arachidonic acid metabolic (AA metabolic) pathway. It also inhibits the production of Th2 cytokines and inflammatory cell infiltration, thereby alleviating symptoms such as nasal congestion and nasal leakage. These results reveal the action and molecular mechanism of cineole in alleviating AR and provide a theoretical basis for the clinical application of cineole in treating AR.
Collapse
Affiliation(s)
- Fang-Lin Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ying Rong
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Hui Zhou
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Tong Yu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Luyao Liu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Qianwen Cao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Zhaolong Qin
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Lingbo Qu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Xinglin Liao
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Qiman Jiang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Nan Zhang
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Xia Xu
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Co-innovation Center of Henan Province for New Drug R&D and Preclinical Safety, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
12
|
Baralić K, Pavić A, Javorac D, Živančević K, Božić D, Radaković N, Antonijević Miljaković E, Buha Djordjevic A, Ćurčić M, Bulat Z, Antonijević B, Đukić-Ćosić D. Comprehensive investigation of hepatotoxicity of the mixture containing phthalates and bisphenol A. JOURNAL OF HAZARDOUS MATERIALS 2023; 445:130404. [PMID: 36455319 DOI: 10.1016/j.jhazmat.2022.130404] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 10/23/2022] [Accepted: 11/13/2022] [Indexed: 06/17/2023]
Abstract
Connections between the mixture containing bis(2- ethylhexyl) phthalate (DEHP), dibutyl phthalate (DBP) and bisphenol A (BPA) and liver injury were explored through in silico investigation and 2 in vivo models. Comparative Toxicogenomics Database (CTD), ShinyGO, ToppCluster and Cytoscape were used for bioinformatic analysis. In vivo subacute study was performed on rats - five groups (n = 6): (1) Control: corn oil, (2) DEHP: 50 mg/kg b.w./day, (3) DBP: 50 mg/kg b.w./day, (4) BPA: 25 mg/kg b.w./day, (5) MIX: DEHP + DBP + BPA. Zebrafish embryos were exposed to the investigated substances in different doses, singularly and combined (binary and ternary mixtures). Liver injury was linked to 75 DEHP, DBP, and BPA genes, mostly connected to inflammation/oxidative stress. In rats, significant alterations in redox status/bioelements and pathohistology were most notable or exclusively present in MIX (probable additive effects). BPA decreased liver area (LA) index in dose-dependent manner. DEHP (< 2 µg/mL) and DBP (≤ 5 µg/mL) reduced LA values, while their higher doses increased LA index. The effect of DBP in binary mixtures led to a lethal outcome at the two highest concentrations, while the hepatotoxicity of DEHP/DBP/BPA mixture was dictated by BPA (confirmed by the benchmark dose analysis).
Collapse
Affiliation(s)
- Katarina Baralić
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia.
| | - Aleksandar Pavić
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11000 Belgrade, Serbia
| | - Dragana Javorac
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Katarina Živančević
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia; University of Belgrade - Faculty of Biology, Institute of Physiology and Biochemistry "Ivan Djaja", Studentski trg, 3, Belgrade, Serbia
| | - Dragica Božić
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Nataša Radaković
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11000 Belgrade, Serbia
| | - Evica Antonijević Miljaković
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Aleksandra Buha Djordjevic
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Marijana Ćurčić
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Zorica Bulat
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Biljana Antonijević
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia
| | - Danijela Đukić-Ćosić
- Department of Toxicology "Akademik Danilo Soldatović", University of Belgrade - Faculty of Pharmacy, Vojvode Stepe 450, 11221 Belgrade, Serbia
| |
Collapse
|
13
|
Liu R, Jin Y, Liu B, Zhang Q, Li X, Cai D, Tian L, Jiang X, Zhang W, Sun J, Bai W. Untargeted Lipidomics Revealed the Protective Effects of Cyanidin-3- O-glucoside on Bisphenol A-Induced Liver Lipid Metabolism Disorder in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:1077-1090. [PMID: 36597173 DOI: 10.1021/acs.jafc.2c06849] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Bisphenol A (BPA) is an estrogenic endocrine disruptor that induces metabolic disorders. Cyanidin-3-O-glucoside (C3G) has multiple functional activities and is the most abundant anthocyanin belonging to the flavonoid subgroup. This study aimed to investigate the protective effect of C3G on BPA-induced liver lipid metabolism disorder and explore its mechanism via lipidomics analysis. The results showed that C3G supplementation significantly ameliorated the serum levels of low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, total cholesterol, triacylglycerols (TG), and alanine and aspartate aminotransferase (ALT and AST). Furthermore, liver lipidomics indicated that C3G effectively facilitated the recovery of differential lipid metabolites, including TGs, phosphatidylethanolamines, phosphatidylcholines, lysophosphatidylcholines, phosphatidylinositol, cholesteryl esters, and phosphatidylserine, and reversed the levels of hepatic lipid synthesis-related genes. Our results suggest that C3G has an effective regulatory effect on BPA-induced disorders of lipid metabolism.
Collapse
Affiliation(s)
- Ruijing Liu
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
- Key Laboratory for Bio-Based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Yulong Jin
- Key Laboratory for Bio-Based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Boping Liu
- Key Laboratory for Bio-Based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, P. R. China
| | - Qing Zhang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| | - Xusheng Li
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| | - Dongbao Cai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| | - Lingmin Tian
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| | - Xinwei Jiang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| | - Wenbao Zhang
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| | - Jianxia Sun
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, P. R. China
| | - Weibin Bai
- Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, Guangzhou 510632, P. R. China
| |
Collapse
|
14
|
WU S, ZHOU W. Antimicrobial activity of oridonin. FOOD SCIENCE AND TECHNOLOGY 2023. [DOI: 10.1590/fst.110222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Saile WU
- Henan Institute of Science and Technology, China
| | - Wei ZHOU
- Henan Institute of Science and Technology, China
| |
Collapse
|
15
|
Yu D, Li J, Wang Y, Guo D, Zhang X, Chen M, Zhou Z. Oridonin ameliorates acetaminophen-induced acute liver injury through ATF4/PGC-1α pathway. Drug Dev Res 2022; 84:211-225. [PMID: 36567664 DOI: 10.1002/ddr.22024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 11/26/2022] [Accepted: 12/04/2022] [Indexed: 12/27/2022]
Abstract
Acetaminophen (APAP) overdose-induced acute liver injury (ALI) causes hepatocyte cell death, oxidative stress, and inflammation. Oridonin (Ori), a covalent NLRP3-inflammasome inhibitor, ameliorates APAP-induced ALI through an unclear molecular mechanism. This study found that Ori decreased hepatic cytochrome P450 2E1 level and increased glutathione content to prevent APAP metabolism, and then reduced the necrotic area, improved liver function, and inhibited APAP-induced proinflammatory cytokines and oxidative stress. Ori also decreased activating transcription factor 4 (ATF4) protein levels and increased peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) to reduce APAP-induced endoplasmic reticulum stress activation and mitochondrial dysfunction. Furthermore, western blot and luciferase assay found that ATF4 inhibited transcription in the PGC-1α promoter -507 to -495 region to reduce PGC-1α levels, while ATF4 knockdown neutralized the hepatoprotective effect of Ori. Molecular docking showed that Ori bound to ATF4's amino acid residue glutamate 302 through 6, 7, and 18 hydroxyl bands. Our findings demonstrated that Ori prevented metabolic activation of APAP and further inhibited the ATF4/PGC-1α pathway to alleviate APAP overdose-induced hepatic toxicity, which illuminated its potential therapeutic effects on ALI.
Collapse
Affiliation(s)
- Dongsheng Yu
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiye Li
- Henan Research Centre for Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
| | - Yu Wang
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Danfeng Guo
- Henan Research Centre for Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
| | - Xiaodan Zhang
- Henan Research Centre for Organ Transplantation, Zhengzhou, China.,Henan Key Laboratory for Digestive Organ Transplantation, Zhengzhou, China
| | - Mingming Chen
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.,Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Zheng Zhou
- Department of Chinese Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
16
|
Satyanarayana N, Chinni SV, Gobinath R, Sunitha P, Uma Sankar A, Muthuvenkatachalam BS. Antidiabetic activity of Solanum torvum fruit extract in streptozotocin-induced diabetic rats. Front Nutr 2022; 9:987552. [PMID: 36386935 PMCID: PMC9650639 DOI: 10.3389/fnut.2022.987552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 08/24/2022] [Indexed: 12/06/2022] Open
Abstract
Background Solanum torvum Swartz, a medicinal plant belonging to the family Solanaceae, is an important medicinal plant widely distributed throughout the world and used as medicine to treat diabetes, hypertension, tooth decay, and reproductive problems in traditional systems of medicine around the world including Malaysia. The objective of this study was to investigate hypoglycemic, antilipidemic, and hepatoprotective activities, histopathology of the pancreas, and specific glucose regulating gene expression of the ethanolic extract of S. torvum fruit in streptozotocin-induced diabetic Sprague–Dawley rats. Materials and methods Acute toxicity study was done according to OECD-423 guidelines. Diabetes was induced by intraperitoneal (i.p.) injection of streptozotocin (55 mg/kg) in male Sprague–Dawley rats. Experimental diabetic rats were divided into six different groups; normal, diabetic control, and glibenclamide at 6 mg/kg body weight, and the other three groups of animals were treated with oral administration of ethanolic extract of S. torvum fruit at 120, 160, and 200 mg/kg for 28 days. The effect of ethanolic extract of S. torvum fruit on body weight, blood glucose, lipid profile, liver enzymes, histopathology of pancreas, and gene expression of glucose transporter 2 (slc2a2), and phosphoenolpyruvate carboxykinase (PCK1) was determined by RT-PCR. Results Acute toxicity studies showed LD50 of ethanolic extract of S. torvum fruit to be at the dose of 1600 mg/kg body weight. Blood glucose, total cholesterol, triglycerides, low-density lipoproteins, very low-density lipoproteins, serum alanine aminotransferase, and aspartate aminotransferase were significantly reduced, whereas high-density lipoproteins were significantly increased in S. torvum fruit (200 mg/kg)-treated rats. Histopathological study of the pancreas showed an increase in number, size, and regeneration of β-cell of islets of Langerhans. Gene expression studies revealed the lower expression of slc2a2 and PCK1 in treated animals when compared to diabetic control. Conclusion Ethanolic extract of S. torvum fruits showed hypoglycemic, hypolipidemic, and hepatoprotective activity in streptozocin-induced diabetic rats. Histopathological studies revealed regeneration of β cells of islets of Langerhans. Gene expression studies indicated lower expression of slc2a2 and PCK1 in treated animals when compared to diabetic control, indicating that the treated animals prefer the gluconeogenesis pathway.
Collapse
Affiliation(s)
- Namani Satyanarayana
- Department of Anatomy, Saint James School of Medicine, Saint Vincent, Saint Vincent and the Grenadines
| | - Suresh V. Chinni
- Department of Biochemistry, Faculty of Medicine, Bioscience, and Nursing, MAHSA University, Jenjarom, Selangor, Malaysia
- Department of Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
- *Correspondence: Suresh V. Chinni, ,
| | - Ramachawolran Gobinath
- Department of Foundation, RCSI and UCD Malaysia Campus, Georgetown, Pulau Pinang, Malaysia
| | - Paripelli Sunitha
- Department of Physiology, Saint James School of Medicine, Saint Vincent, Saint Vincent and the Grenadines
| | - Akula Uma Sankar
- Faculty of Medicine, Biochemistry Unit, AIMST University, Bedong, Kedah, Malaysia
| | | |
Collapse
|
17
|
Changes in physiological activities are responsible for homoyessotoxin-induced toxicity in abalone Haliotis discus hannai. Toxicology 2022; 477:153270. [PMID: 35870676 DOI: 10.1016/j.tox.2022.153270] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/22/2022]
Abstract
Homoyessotoxin (homo-YTX) is a lipid-soluble toxin produced by toxic dinoflagellates. It is widely distributed in marine ecosystems worldwide, and it poses a threat to the survival of aquatic animals. The tissues of the abalone Haliotis discus hannai are easily damaged by homo-YTX during harmful algal blooms. In this study, H. discus hannai was exposed to homo-YTX (0, 2, 5, and 10 µg L-1) to evaluate the rates of survival (S) and death (D) and the antioxidative, metabolic, and digestive physiological responses in the gills and digestive gland of abalone. Homo-YTX decreased S and the activities of Na+/K+-adenosine triphosphatase, Ca2+/Mg2+-adenosine triphosphatase, superoxide dismutase, catalase, alkaline phosphatase, xanthine oxidase, lactate dehydrogenase, amylase, protease, and lipase. Meanwhile, D, the reactive oxygen species level, and the malondialdehyde content increased with increasing concentrations of homo-YTX. In addition, homo-YTX induced oxidative stress, enhanced the lipid peroxidation reaction, reduced the energy supply, and inhibited the metabolic and digestive physiological activities in the gills and digestive gland of abalone. Oxidative stress-mediated insufficient energy supply and physiological activity reduction caused the death of abalone.
Collapse
|
18
|
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
|
19
|
Zhan Z, Dai F, Zhang T, Chen Y, She J, Jiang H, Liu S, Gu T, Tang L. Oridonin alleviates hyperbilirubinemia through activating LXRα-UGT1A1 axis. Pharmacol Res 2022; 178:106188. [PMID: 35338002 DOI: 10.1016/j.phrs.2022.106188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 10/18/2022]
Abstract
Hyperbilirubinemia is a serious hazard to human health due to its neurotoxicity and lethality. So far, successful therapy for hyperbilirubinemia with fewer side effects is still lacking. In this study, we aimed to clarify the effects of oridonin (Ori), an active diterpenoid extracted from Rabdosia rubescens, on hyperbilirubinemia and revealed the underlying molecular mechanism in vivo and in vitro. Here, we showed that liver X receptor alpha (LXRα) deletion eliminated the protective effect of Ori on phenylhydrazine hydrochloride-induced hyperbilirubinemia mice, indicating that LXRα acted as a key target for Ori treatment of hyperbilirubinemia. Ori significantly increased the expression of LXRα and UDP-glucuronosyltransferase 1A1 (UGT1A1) in the liver of wild-type (WT) mice, which were lost in LXRα-/- mice. Ori or LXR agonist GW3965 also reduced lipopolysaccharide/D-galactosamine-induced hyperbilirubinemia via activating LXRα/UGT1A1 in WT mice. Liver UGT1A1 enzyme activity was elevated by Ori or GW3965 in WT mice. Further, Ori up-regulated LXRα gene expression, increased its nuclear translocation and stimulated UGT1A1 promoter activity in HepG2 cells. After silencing LXRα by siRNA, Ori-induced UGT1A1 expression was markedly reduced in HepG2 cells and primary mouse hepatocytes. Taken together, Ori stimulated the transcriptional activity of LXRα, resulting in the up-regulation of UGT1A1. Therefore, Ori or its analogs might have the potential to treat hyperbilirubinemia-related diseases through modulating LXRα-UGT1A1 signaling.
Collapse
Affiliation(s)
- Zhikun Zhan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Fahong Dai
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tao Zhang
- Department of Pharmaceutical, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, China
| | - Yulian Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jianglian She
- CAS Key Laboratory of Tropical Marine Bioresources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Huanguo Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shuwen Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou 510515, China
| | - Tanwei Gu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lan Tang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
20
|
Sun J, Fang R, Wang H, Xu DX, Yang J, Huang X, Cozzolino D, Fang M, Huang Y. A review of environmental metabolism disrupting chemicals and effect biomarkers associating disease risks: Where exposomics meets metabolomics. ENVIRONMENT INTERNATIONAL 2022; 158:106941. [PMID: 34689039 DOI: 10.1016/j.envint.2021.106941] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/03/2021] [Accepted: 10/12/2021] [Indexed: 05/27/2023]
Abstract
Humans are exposed to an ever-increasing number of environmental toxicants, some of which have gradually been elucidated to be important risk factors for metabolic diseases, such as diabetes and obesity. These metabolism-sensitive diseases typically occur when key metabolic and signaling pathways were disrupted, which can be influenced by the exposure to contaminants such as endocrine disrupting chemicals (EDCs), along with genetic and lifestyle factors. This promotes the concept and research on environmental metabolism disrupting chemicals (MDCs). In addition, identifying endogenous biochemical markers of effect linked to disease states is becoming an important tool to screen the biological targets following environmental contaminant exposure, as well as to provide an overview of toxicity risk assessment. As such, the current review aims to contribute to the further understanding of exposome and human health and disease by characterizing environmental exposure and effect metabolic biomarkers. We summarized MDC-associated metabolic biomarkers in laboratory animal and human cohort studies using high throughput targeted and nontargeted metabolomics techniques. Contaminants including heavy metals and organohalogen compounds, especially EDCs, have been repetitively associated with metabolic disorders, whereas emerging contaminants such as perfluoroalkyl substances and microplastics have also been found to disrupt metabolism. In addition, we found major limitations in the effective identification of metabolic biomarkers especially in human studies, toxicological research on the mixed effect of environmental exposure has also been insufficient compared to the research on single chemicals. Thus, it is timely to call for research efforts dedicated to the study of combined effect and metabolic alterations for the better assessment of exposomic toxicology and health risks. Moreover, advanced computational and prediction tools, further validation of metabolic biomarkers, as well as systematic and integrative investigations are also needed in order to reliably identify novel biomarkers and elucidate toxicity mechanisms, and to further utilize exposome and metabolome profiling in public health and safety management.
Collapse
Affiliation(s)
- Jiachen Sun
- College of Marine Life Science, Ocean University of China, Qingdao, China
| | - Runcheng Fang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China
| | - Jing Yang
- State Environmental Protection Key Laboratory of Quality Control in Environmental, Monitoring, China National Environmental Monitoring Center, Beijing, China
| | - Xiaochen Huang
- School of Agriculture, Sun Yat-sen University, Guangzhou, China
| | - Daniel Cozzolino
- ARC Industrial Transformation Training Centre for Uniquely Australian Foods, Centre for Nutrition and Food Sciences, Queensland Alliance for Agriculture and Food Innovation, The University of Queensland, Coopers Plans, Australia
| | - Mingliang Fang
- School of Civil and Environmental Engineering, Nanyang Technological University, Singapore
| | - Yichao Huang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Hefei, China.
| |
Collapse
|
21
|
Liang Y, Zhou Y, Wang Y, Liu R, Qi J, Lin Y, Zhang T, Jiang Q. Use of physiological activities to estimate the population growth of rotifer (Brachionus calyciflorus) under the stress of toxic Microcystis and nitrite. CHEMOSPHERE 2021; 285:131419. [PMID: 34246096 DOI: 10.1016/j.chemosphere.2021.131419] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 06/13/2023]
Abstract
Microcystis blooms disrupt aquatic systems and adversely affect zooplankton growth. Brachionus calyciflorus Pallas (rotifer) was introduced to different combinations of toxic Microcystis aeruginosa (0, 2 × 105, 2 × 106, and 2 × 107 cells mL-1) and nitrite (0, 2, 4, and 6 mg L-1) to evaluate their physiological activities and population growth under stress. Survival rate (S), population growth rate (r), grazing rate (G), antioxidant response, and metabolic and digestive enzyme activities were determined. Results revealed that G declined with the increasing nitrite doses and grazing time upon exposure to a certain Microcystis concentration. Toxic M. aeruginosa and nitrite inhibited the S, r, glutathione content, total antioxidant capacity level, and activities of alkaline phosphatase, xanthine oxidase, lactate dehydrogenase, and cellulase (p < 0.05) but increased the reactive oxygen species level, malondialdehyde content, and amylase activity (p < 0.05). The activities of superoxide dismutase, catalase, and pepsase were also increased in single low doses of nitrite solutions (p < 0.05). Therefore, the grazing intensity of rotifers affected B. calyciflorus physiological activities, which are useful in the estimation of its population growth in eutrophic water environments.
Collapse
Affiliation(s)
- Ye Liang
- School of Marine Sciences, Nanjing University of Information Science & Technology, No. 219 Ningliu Road, Nanjing, 210044, PR China; Fujian Provincial Key Laboratory of Marine Ecological Conservation and Restoration, Xiamen, No. 178 Daxue Road, 361005, PR China.
| | - Yang Zhou
- School of Marine Science and Engineering, Nanjing Normal University, No.1 Wenyuan Road, Nanjing, 210023, PR China
| | - Yishan Wang
- School of Marine Sciences, Nanjing University of Information Science & Technology, No. 219 Ningliu Road, Nanjing, 210044, PR China
| | - Rui Liu
- School of Marine Sciences, Nanjing University of Information Science & Technology, No. 219 Ningliu Road, Nanjing, 210044, PR China
| | - Jun Qi
- School of Marine Sciences, Nanjing University of Information Science & Technology, No. 219 Ningliu Road, Nanjing, 210044, PR China
| | - Yangjie Lin
- School of Marine Sciences, Nanjing University of Information Science & Technology, No. 219 Ningliu Road, Nanjing, 210044, PR China
| | - Tongqing Zhang
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, 210017, PR China
| | - Qichen Jiang
- Freshwater Fisheries Research Institute of Jiangsu Province, Nanjing, 210017, PR China
| |
Collapse
|
22
|
5'-Nucleotidase Plays a Key Role in Uric Acid Metabolism of Bombyx mori. Cells 2021; 10:cells10092243. [PMID: 34571893 PMCID: PMC8468349 DOI: 10.3390/cells10092243] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 07/06/2021] [Indexed: 01/26/2023] Open
Abstract
Uric acid (UA) is the end-product in the human purine metabolism pathway. The UA that accumulates in silkworm tissues is excreted as a nitrogen waste product. Here, we first validated that Bombyx mori has a homolog of the human gene that encodes the 5′-nucleotidase (5′N) involved in purine metabolism. The B. mori gene, Bm5′N, is located upstream of other genes involved in UA metabolism in the silkworm. Disruption of Bm5′N via the CRISPR/Cas9 system resulted in decreased UA levels in the silkworm epidermis and caused a translucent skin phenotype. When Bm5′N mutant silkworms were fed with the uric acid precursor inosine, the UA levels in the epidermis increased significantly. Furthermore, the metabolomic and transcriptomic analyses of Bm5′N mutants indicated that loss of the Bm5′N affected purine metabolism and the ABC transport pathway. Taken together, these results suggest that the UA pathway is conserved between the silkworm and humans and that the Bm5′N gene plays a crucial role in the uric acid metabolism of the silkworm. Thus, the silkworm may be a suitable model for the study of UA metabolism pathways relevant to human disease.
Collapse
|
23
|
Shen Y, Zhang Y, Li W, Chen K, Xiang M, Ma H. Glutamine metabolism: from proliferating cells to cardiomyocytes. Metabolism 2021; 121:154778. [PMID: 33901502 DOI: 10.1016/j.metabol.2021.154778] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/16/2021] [Accepted: 04/19/2021] [Indexed: 02/06/2023]
Abstract
Glutamine is a major energy source for rapidly dividing cells, such as hematopoietic stem cells and cancer cells. Reliance on glutamine is therefore regarded as a metabolic hallmark of proliferating cells. Moreover, reprogramming glutamine metabolism by various factors, including tissue type, microenvironment, pro-oncogenes, and tumor suppressor genes, can facilitate stem cell fate decisions, tumor recurrence, and drug resistance. However, the significance of glutamine metabolism in cardiomyocytes, an end-differentiated cell type, is not fully understood. Existing evidence suggests important roles of glutamine metabolism in the development of cardiovascular diseases. In this review, we have focused on glutaminolysis and its regulatory network in proliferating cells. We have summarized current findings about the role of glutamine utilization in cardiomyocytes and have discussed possibilities of targeting glutamine metabolism for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Yimin Shen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Yuhao Zhang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Wudi Li
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Kaijie Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.
| | - Hong Ma
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China.
| |
Collapse
|
24
|
Mao L, Fang S, Zhao M, Liu W, Jin H. Effects of Bisphenol A and Bisphenol S Exposure at Low Doses on the Metabolome of Adolescent Male Sprague-Dawley Rats. Chem Res Toxicol 2021; 34:1578-1587. [PMID: 34019419 DOI: 10.1021/acs.chemrestox.1c00018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Toxic effects induced upon exposure to low-dose bisphenol A (BPA) or bisphenol S (BPS) remains controversial. In this study, metabolomics was used to examine the metabolomic perturbation arising from 28 days of exposure to BPA or BPS at 50 μg/kg bw/day in Sprague-Dawley (SD) rats. Endogenous metabolite profiling revealed a clear discrimination of metabolome in the rat plasma among BPA-treatment, BPS-treatment, and control groups. BPA exposure induced the up-regulation of 19 metabolites and down-regulation of 32 metabolites in plasma of SD rats, compared with the control. BPS exposure induced the up-regulation of 15 metabolites and the down-regulation of 33 metabolites in the plasma of SD rats, compared with the control. Joint pathway analysis suggested marked perturbations in the citrate cycle, butanoate metabolism, and alanine, aspartate, and glutamate metabolism for BPA-exposed rats as well as glycerophospholipid metabolism for BPS-exposed rats. These findings provide novel insights into associations between the metabolomic perturbation and phenotypic changes arising from BPA and BPS exposure.
Collapse
Affiliation(s)
- Lingling Mao
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310032, P. R. China
| | - Shuhong Fang
- College of Resources and Environment, Chengdu University of Information Technology, Chengdu 610225, P. R. China
| | - Meirong Zhao
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310032, P. R. China
| | - Weiping Liu
- Ministry of Education Key Laboratory of Environmental Remediation and Ecosystem Health, Institution of Environmental Health, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou 310058, P. R. China
| | - Hangbiao Jin
- Key Laboratory of Microbial Technology for Industrial Pollution Control of Zhejiang Province, College of Environment, Zhejiang University of Technology, Hangzhou 310032, P. R. China
| |
Collapse
|