1
|
Wei J, Chen H, Xu M, Zhang Z, Wang J, Jiang W, Zhou W, Xiong M. Prediction of potential targets of aloe-emodin in the treatment of hepatocellular carcinoma using network pharmacology combined with bioinformatics. Discov Oncol 2025; 16:464. [PMID: 40186059 PMCID: PMC11971119 DOI: 10.1007/s12672-025-02215-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/24/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma is one of the most common and malignant tumors worldwide. Although aloe-emodin (AE), a pure natural drug, can effectively kill hepatocellular carcinoma cells, its internal molecular mechanism has not been fully elucidated. In this study, the anti-hepatocellular carcinoma targets of AE were predicted using network pharmacology and bioinformatics. METHODS The differentially expressed genes between hepatocellular carcinoma and normal tissues were first identified and then further intersected with the potential pharmacological target genes of AE for subsequent analysis. Moreover, the potential targets of AE were enriched and analyzed to identify potential downstream pathways. The binding ability and interaction between the above drug targets and AE were analyzed by molecular docking. The prognostic model of hepatocellular carcinoma was subsequently constructed via univariate Cox regression analysis, LASSO regression analysis and multivariate Cox regression analysis. Finally, the potential targets that can stably bind to AE were further screened through molecular dynamics simulation. Finally, we validated the potential utility of AE in treating hepatocellular carcinoma through in vitro experiments. RESULTS After 90 target genes related to AE were crossed with hepatocellular carcinoma differential genes, 13 cross genes were obtained. The above 13 genes might act on hepatocellular carcinoma through the following pathways: p53 signaling pathway, cell cycle, cellular sense, mismatch repair, apoptosis-multiple specifications, base example repair and DNA replication. Molecular docking revealed that the combination of the BAX, FASN, CDK1, PCNA, CLIC1, VWF, RAN, FOXM1, TGM3, CANT1, and NSMCE2 proteins with AE was relatively stable. A 4-gene prognostic model was further constructed. The area under the curve (AUC) values of the 1-year, 3-year and 5-year survival rates from the ROC curve were 0.809, 0.673 and 0.641, respectively. Molecular dynamics analysis revealed that CDK1 and PCNA were the most stable binding targets among the above proteins. CCK8 and wound healing assays revealed that AE inhibited the proliferation and migration of hepatocellular carcinoma cells at increasing concentrations. Western blot experiments revealed that AE achieved therapeutic effects on hepatocellular carcinoma by promoting apoptosis of hepatocellular carcinoma cells. CONCLUSIONS Based on network pharmacology, bioinformatics, molecular dynamics simulation, and in vitro experimental verification, we found that AE achieved a therapeutic effect on hepatocellular carcinoma by promoting apoptosis of hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Jinlong Wei
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
- Department of Third Surgery, Wuhu City Hospital of Traditional Chinese Medicine, Wuhu, 2461002, People's Republic of China
| | - Haosong Chen
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Maoqi Xu
- Department of Third Surgery, Wuhu City Hospital of Traditional Chinese Medicine, Wuhu, 2461002, People's Republic of China
| | - Zhenglin Zhang
- Department of Third Surgery, Wuhu City Hospital of Traditional Chinese Medicine, Wuhu, 2461002, People's Republic of China
| | - Jin Wang
- Department of Third Surgery, Wuhu City Hospital of Traditional Chinese Medicine, Wuhu, 2461002, People's Republic of China
| | - Wen Jiang
- Department of Third Surgery, Wuhu City Hospital of Traditional Chinese Medicine, Wuhu, 2461002, People's Republic of China
| | - Weiguo Zhou
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Maoming Xiong
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| |
Collapse
|
2
|
Cheng CY, Hsu SH, Chokkalingam U, Dai YS, Shih PC, Ekambaranellore P, Lin WW. Aloe polysaccharide promotes keratinocyte proliferation, migration, and differentiation by upregulating the EGFR/PKC-dependent signaling pathways. Sci Rep 2025; 15:8196. [PMID: 40064981 PMCID: PMC11893877 DOI: 10.1038/s41598-025-91201-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
Aloe vera is a popular medicinal plant in the cosmetic, pharmaceutical, and food industries. Acemannan (ACE), a β-(1,4)-acetylated mannan, is one of the bioactive compounds isolated from the A. vera gel. The pharmacological effects of ACE have been reported regarding digestive disease protection, antimicrobia, and prebiotic activity. Here, we used human HaCaT cells as a model to uncover the potential biological functions of ACE in keratinocytes. ACE increased cell growth in a concentration-dependent manner, and a higher incorporation of BrdU was detected in ACE-treated cells than in vehicle-treated cells, indicating ACE promotes cell proliferation. Furthermore, ACE concentration-dependently promoted cell migration in the wound scratch model. ACE regulated cell differentiation by transiently decreasing p63α expression, but increasing the expression of involucrin, loricrin, and transglutaminase 1 (TGase 1). These effects were non-additive to those induced by phorbol myristate acetate (PMA), but additive to epidermal growth factor (EGF), which are complete and incomplete differentiation agents of keratinocytes, respectively. Moreover, ACE activated EGF receptor (EGFR), protein kinase C (PKC), and protein kinase B (AKT/PKB). PKC inhibitor Ro320432 enhanced cell growth and migration, while EGFR inhibitor osimertinib blocked both responses. In summary, ACE is a potential therapeutic agent in wound healing. ACE activates PKC, leading to keratinocyte differentiation and activates EGFR, contributing to keratinocyte proliferation and migration.
Collapse
Affiliation(s)
- Ching-Yuan Cheng
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, 11031, Taiwan
| | - Shao-Hsuan Hsu
- Department of Dermatology, Yunlin Branch, National Taiwan University Hospital, Zhongshan, 640203, Taiwan
| | | | - Yang-Shia Dai
- Department of Dermatology, National Taiwan University Hospital, Taipei, 100225, Taiwan
| | - Pei-Chun Shih
- Dazzeon Biotechnology Co., Ltd, New Taipei City, 248022, Taiwan
| | | | - Wan-Wan Lin
- Department of Pharmacology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan.
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, 11031, Taiwan.
| |
Collapse
|
3
|
Pan J, Wang J, Lei Z, Wang H, Zeng N, Zou J, Zhang X, Sun J, Guo D, Luan F, Shi Y. Therapeutic Potential of Chinese Herbal Medicine and Underlying Mechanism for the Treatment of Myocardial Infarction. Phytother Res 2025; 39:189-232. [PMID: 39523856 DOI: 10.1002/ptr.8368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 11/16/2024]
Abstract
Myocardial infarction (MI) is a prevalent disease with high mortality rates worldwide. The course of MI is intricate and variable, necessitating personalized treatment strategies based on different mechanisms. However, variety of postoperative complications and rejections, such as heart failure, arrhythmias, cardiac rupture, and left ventricular thrombus, contribute to a poor prognosis. Despite the inclusion of antiplatelet agents and statins in the conventional treatment regimen, their clinical applicability is constrained by potential adverse effects and limited efficacy. Additionally, the mechanisms leading to MI are complex and diverse. Therefore, the development of novel compounds for MI treatment. The use of traditional Chinese medicine (TCM) in the prevention and treatment of cardiovascular diseases, including MI, is grounded in its profound historical background, comprehensive theoretical system, and accumulated knowledge. An increasing number of contemporary evidence-based medical studies have demonstrated that TCM plays a significant role in alleviating symptoms and improving the quality of life for MI patients. Chinese herbal formulations and active ingredients can intervene in the pathological process of MI through key factors such as inflammation, oxidative stress, apoptosis, ferroptosis, pyroptosis, myocardial fibrosis, angiogenesis, and autophagy. This article critically reviews existing herbal formulations from an evidence-based medicine perspective, evaluating their research status and potential clinical applications. Additionally, it explores recent advancements in the use of herbal medicines and their components for the prevention and treatment of MI, offering detailed insights into their mechanisms of action.
Collapse
Affiliation(s)
- Jiaojiao Pan
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Jinhui Wang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Ziwen Lei
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - He Wang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Nan Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, People's Republic of China
| | - Junbo Zou
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Xiaofei Zhang
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Jing Sun
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Dongyan Guo
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Fei Luan
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| | - Yajun Shi
- Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, People's Republic of China
| |
Collapse
|
4
|
Zhao A, Liu X, Chen X, Na S, Wang H, Peng X, Kong P, Li L. Aqueous Extract of Rhubarb Promotes Hepatotoxicity via Facilitating PKM2-Mediated Aerobic Glycolysis in a Rat Model of Diethylnitrosamine-Induced Liver Cancer. Drug Des Devel Ther 2024; 18:4497-4510. [PMID: 39403095 PMCID: PMC11471889 DOI: 10.2147/dddt.s476273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/21/2024] [Indexed: 10/19/2024] Open
Abstract
Objective To identify the polar parts in Rhubarb that cause hepatotoxicity and explore the underlying mechanisms. Methods The rat model of liver cancer was established by gavage of diethylnitrosamine (DEN; 0.002 g/rat) for 14 weeks. Starting from the 11th week, Rhubarb granule (4 g/kg), aqueous, ethyl acetate and n-butanol extract of Rhubarb or Rhein equivalent to a dose of 4 g/kg Rhubarb granule were administered intragastrically for 4 consecutive weeks. Liver tissues from rats treated with DEN and Rhubarb granules were used for non-targeted metabolomics analysis. The correlation between pyruvate kinase isozyme type M2 (PKM2) expression level and the progress and prognosis of hepatocellular carcinoma (HCC) was evaluated through bioinformatics analysis based on TCGA database. Liver tissues and blood samples from rats treated with DEN and aqueous, ethyl acetate and n-butanol extract of Rhubarb were used for the screening of hepatotoxic polar parts of Rhubarb. The liver injuries were evaluated by the changes in pathology, liver function, and the expression levels of proliferating cell nuclear antigen (PCNA) and transforming growth factor beta1 (TGF-β1). The mechanism studies focus on PKM2 expression, and the metabolic reprogramming via detecting the activities of lactate dehydrogenase A (LDHA) and isocitrate dehydrogenase (ICDH). Furthermore, molecular docking analysis was performed to validate the target interaction between Rhein and PKM2, and the hepatotoxicity of Rhein was evaluated by testing liver function in the DEN-induced liver cancer model. Results The non-targeted metabolomics analysis revealed that Rhubarb promoted aerobic glycolysis in the rat model of DEN-induced liver cancer. And bioinformatics analysis revealed that high PKM2 expression was closely related to the progression and poor prognosis of HCC. In vivo studies indicated that the aqueous extract of Rhubarb, but not ethyl acetate and n-butanol extract, promoted the liver injuries induced by DEN. The mechanism study showed that the aqueous extract of Rhubarb increased the expression of PKM2 and promoted aerobic glycolysis. Moreover, Rhein had a strong binding affinity for PKM2 and aggravated liver injury in the DEN-induced liver cancer model. Conclusion Aqueous extract of Rhubarb promoted hepatotoxicity via facilitating PKM2-mediated aerobic glycolysis in the rat model of DEN-induced liver cancer.
Collapse
Affiliation(s)
- Anni Zhao
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People’s Republic of China
| | - Xiaomei Liu
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People’s Republic of China
| | - Xiping Chen
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People’s Republic of China
| | - Sha Na
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People’s Republic of China
| | - Hui Wang
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People’s Republic of China
| | - Xuan Peng
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People’s Republic of China
| | - Peizhong Kong
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People’s Republic of China
| | - Lu Li
- Department of Biochemistry and Molecular Biology, School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, People’s Republic of China
- Institute of Integrated Chinese and Western Medicine, Anhui Academy of Chinese Medicine, Hefei, 230012, People’s Republic of China
| |
Collapse
|
5
|
Guo Q, Wang J, Ni C, Pan J, Zou J, Shi Y, Sun J, Zhang X, Wang D, Luan F. Research progress on the natural products in the intervention of myocardial infarction. Front Pharmacol 2024; 15:1445349. [PMID: 39239656 PMCID: PMC11374734 DOI: 10.3389/fphar.2024.1445349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Coronary heart disease is a prevalent cardiovascular ailment globally, with myocardial infarction (MI) being one of its most severe manifestations. The morbidity and mortality of MI are escalating, showing an increasing trend among younger, highly educated individuals, thereby posing a serious threat to public health. Currently, thrombolysis, percutaneous coronary intervention, and coronary artery bypass grafting are the primary clinical treatments for MI. Although these methods significantly reduce patient mortality, complications often result in poor prognoses. Due to limitations in chemical synthetic drug research, the focus has shifted towards developing herbs based on natural substances. Natural medicines represent a novel approach for safer and more effective MI management and treatment. They can control multiple pathogenic variables by targeting various pathways and systems. This paper investigates the molecular mechanisms of MI and evaluates the application of natural products and medicinal plants in MI treatment over the past 5 years, demonstrating their specific good therapeutic potential and superior tolerance. These natural therapies have been shown to mitigate myocardial cell damage caused by MI through mechanisms such as oxidative stress, inflammation, apoptosis, angiogenesis, myocardial fibrosis, autophagy, endoplasmic reticulum stress, mitophagy, and pyroptosis. This review offers the latest insights into the application of natural products and medicinal plants in MI treatment, elucidating their mechanisms of action and serving as an important reference for MI prevention.
Collapse
Affiliation(s)
- Qiuting Guo
- College of Pharmacy, Xianyang Polytechnic Institute, Xianyang, China
| | - Jinhui Wang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Caixia Ni
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu, Sichuan, China
| | - Jiaojiao Pan
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Junbo Zou
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Yajun Shi
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Jing Sun
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Xiaofei Zhang
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| | - Deng Wang
- Department of Pharmacy, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi, China
| | - Fei Luan
- Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, School of Pharmacy, Shaanxi University of Chinese Medicine, Xi'an, Shaanxi, China
| |
Collapse
|
6
|
Wang Y, Zhao M, Li B, Geng X. Advances in the mechanism of emodin-induced hepatotoxicity. Heliyon 2024; 10:e33631. [PMID: 39027614 PMCID: PMC11255441 DOI: 10.1016/j.heliyon.2024.e33631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 06/21/2024] [Accepted: 06/24/2024] [Indexed: 07/20/2024] Open
Abstract
Emodin is a naturally occurring anthraquinone derivative and serves as an active component in various traditional Chinese herbal medicines. It is widely known for its broad pharmacological effects, including anti-inflammatory, antioxidant, and anticancer properties. However, high doses and long-term use of emodin can also lead to liver toxicity. Nevertheless, the mechanism of emodin-induced liver toxicity remains unclear at present. This article aims to summarize the toxicological research progress on emodin, with a particular focus on elucidating the mechanisms underlying emodin-induced hepatocyte injury. By providing essential information, the study intends to facilitate further research and safe usage of emodin for researchers and clinical practitioners.
Collapse
Affiliation(s)
- Yupeng Wang
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control. Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, China
| | - Mengchao Zhao
- Department of Pharmacy, General Hospital of Ningxia Medical University, 804 Shengli Street, Xingqing District, Ningxia, 750004, China
| | - Bo Li
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control. Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, China
| | - Xingchao Geng
- National Center for Safety Evaluation of Drugs, National Institutes for Food and Drug Control. Chinese Academy of Medical Sciences & Peking Union Medical College, 100050, China
| |
Collapse
|
7
|
Catalano A, Ceramella J, Iacopetta D, Marra M, Conforti F, Lupi FR, Gabriele D, Borges F, Sinicropi MS. Aloe vera-An Extensive Review Focused on Recent Studies. Foods 2024; 13:2155. [PMID: 38998660 PMCID: PMC11241682 DOI: 10.3390/foods13132155] [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: 05/13/2024] [Revised: 06/16/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024] Open
Abstract
Since ancient times, Aloe vera L. (AV) has attracted scientific interest because of its multiple cosmetic and medicinal properties, attributable to compounds present in leaves and other parts of the plant. The collected literature data show that AV and its products have a beneficial influence on human health, both by topical and oral use, as juice or an extract. Several scientific studies demonstrated the numerous biological activities of AV, including, for instance, antiviral, antimicrobial, antitumor, and antifungal. Moreover, its important antidepressant activity in relation to several diseases, including skin disorders (psoriasis, acne, and so on) and prediabetes, is a growing field of research. This comprehensive review intends to present the most significant and recent studies regarding the plethora of AV's biological activities and an in-depth analysis exploring the component/s responsible for them. Moreover, its morphology and chemical composition are described, along with some studies regarding the single components of AV available in commerce. Finally, valorization studies and a discussion about the metabolism and toxicological aspects of this "Wonder Plant" are reported.
Collapse
Affiliation(s)
- Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Via Orabona 4, 70126 Bari, Italy
| | - Jessica Ceramella
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Domenico Iacopetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Maria Marra
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Filomena Conforti
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| | - Francesca R Lupi
- Department of Information, Modeling, Electronics and System Engineering, (D.I.M.E.S.), University of Calabria, Via P. Bucci, Cubo 39C, CS, 87036 Rende, Italy
| | - Domenico Gabriele
- Department of Information, Modeling, Electronics and System Engineering, (D.I.M.E.S.), University of Calabria, Via P. Bucci, Cubo 39C, CS, 87036 Rende, Italy
| | - Fernanda Borges
- CIQUP-IMS-Centro de Investigação em Química da Universidade do Porto, Institute of Molecular Sciences, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, Rua do Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
8
|
Qiu Q, Fu F, Wu Y, Han C, Pu W, Wen L, Xia Q, Du D. Rhei Radix et Rhizoma and its anthraquinone derivatives: Potential candidates for pancreatitis treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155708. [PMID: 38733906 DOI: 10.1016/j.phymed.2024.155708] [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: 03/01/2024] [Revised: 04/22/2024] [Accepted: 05/02/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Pancreatitis is a common exocrine inflammatory disease of the pancreas and lacks specific medication currently. Rhei Radix et Rhizoma (RR) and its anthraquinone derivatives (AQs) have been successively reported for their pharmacological effects and molecular mechanisms in experimental and clinical pancreatitis. However, an overview of the anti-pancreatitis potential of RR and its AQs is limited. PURPOSE To summarize and analyze the pharmacological effects of RR and its AQs on pancreatitis and the underlying mechanisms, and discuss their drug-like properties and future perspectives. METHODS The articles related to RR and its AQs were collected from the Chinese National Knowledge Infrastructure, Wanfang data, PubMed, and the Web of Science using relevant keywords from the study's inception until April first, 2024. Studies involving RR or its AQs in cell or animal pancreatitis models as well as structure-activity relationship, pharmacokinetics, toxicology, and clinical trials were included. RESULTS Most experimental studies are based on severe acute pancreatitis rat models and a few on chronic pancreatitis. Several bioactive anthraquinone derivatives of Rhei Radix et Rhizoma (RRAQs) exert local protective effects on the pancreas by maintaining pancreatic acinar cell homeostasis, inhibiting inflammatory signaling, and anti-fibrosis, and they improve systemic organ function by alleviating intestinal and lung injury. Pharmacokinetic and toxicity studies have revealed the low bioavailability and wide distribution of RRAQs, as well as hepatotoxicity and nephrotoxicity. However, there is insufficient research on the clinical application of RRAQs in pancreatitis. Furthermore, we propose effective strategies for subsequent improvement in terms of balancing effectiveness and safety. CONCLUSION RRAQs can be developed as either candidate drugs or novel lead structures for pancreatitis treatment. The comprehensive review of RR and its AQs provides references for optimizing drugs, developing therapies, and conducting future studies on pancreatitis.
Collapse
Affiliation(s)
- Qi Qiu
- West China Center of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Fei Fu
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610213, China
| | - Yaling Wu
- Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610213, China
| | - Chenxia Han
- West China Center of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weiling Pu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Li Wen
- State Key Laboratory of Complex, Severe, and Rare Diseases, Center for Biomarker Discovery and Validation, National Infrastructures for Translational Medicine (PUMCH), Institute of Clinical Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100073, China
| | - Qing Xia
- West China Center of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Dan Du
- West China Center of Excellence for Pancreatitis, Institute of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu 610041, China; Advanced Mass Spectrometry Center, Research Core Facility, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610213, China.
| |
Collapse
|
9
|
Luo S, Huang M, Lu X, Zhang M, Xiong H, Tan X, Deng X, Zhang W, Ma X, Zeng J, Efferth T. Optimized therapeutic potential of Yinchenhao decoction for cholestatic hepatitis by combined network meta-analysis and network pharmacology. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155573. [PMID: 38583348 DOI: 10.1016/j.phymed.2024.155573] [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: 12/17/2023] [Revised: 03/13/2024] [Accepted: 03/27/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Cholestatic hepatitis is recognized as a significant contributor to the development of liver fibrosis and cirrhosis. As a well-known classic formula for the treatment of cholestatic hepatitis, Yinchenhao decoction (YCHD) is widely used in countries in Asia, including China, Japan, and Korea. However, in recent years, a risk of liver injury has been reported from Rheum palmatum L. and Gardenia jasmonoides J.Ellis which are the main ingredients of YCHD. Therefore, the question arises whether YCHD is still safe enough for the treatment of cholestatic hepatitis or whether an optimized ratio of ingredients should be applied. These is inevitable questions for the clinical application of YCHD. PURPOSE To provide a scientific basis for the clinical application of YCHD through a combination of meta-analysis and network pharmacology and to find the best ratio of components to ensure optimal therapeutic efficacy and safety. At the same time, a deeper understanding of the mechanisms of YCHD was explored. METHODS We retrieved relevant trials from various databases including PubMed, Web of Science, EMBASE, Cochrane Library, China National Knowledge Infrastructure (CNKI), VIP and Wanfang databases up to August 2023. After screening for inclusion and exclusion criteria, we assessed efficiency, ALT, AST, and TBIL as outcome parameters. The relevant data underwent a network meta-analysis using STATA 16.0 software. Based on network pharmacology, we screened the disease targets, active ingredients, and targets related to YCHD. The targets were visualized using Cytoscape 3.9.1. Then, potential mechanisms were explored based on bioinformatic techniques. RESULTS Twenty eligible studies were finally screened and a total of 1,591 patients who fulfilled the inclusion criteria were enrolled in the study. The meta-analysis results indicated that TG-c (treatment group c) [(Artemisia capillaris Thunb. : Gardenia jasminoides J.Ellis : Rheum palmatum L. = 10:5:2-10:5:3) + CT] was the most promising therapeutic approach, demonstrating superior efficacy and notable improvements in both AST and TBIL levels. For ALT, TG-d [(Artemisia capillaris : Gardenia jasminoides : Rheum palmatum = 5:1:1-5:2:1) + CT] exhibited the greatest potential as optimal therapy option. Based on the surface under the cumulative ranking curve (SUCRA) values, TG-c was the best therapy in terms of efficiency and improvement in TBIL levels, while TG-d was the most effective in reducing ALT levels. For AST levels, TG-e [(Artemisia capillaris : Gardenia jasminoides : Rheum palmatum = 5:2:2-5:3:3) + CT] was the most effective therapy. The comprehensive analysis revealed that TG-c exhibited the most pronounced efficacy. Combined network pharmacology, GO enrichment analysis and KEGG pathway enrichment analysis displayed that the key target genes of Artemisia capillaris, Rheum palmatum, and Gardenia jasminoides were closely involved in inflammation response, bile transport, apoptosis, oxidative stress, and regulation of leukocyte migration. Notably, bile secretion dominated the common pathway of the three herbs. On the other hand, Artemisia capillaris exhibited a unique mode of action by regulating the IL-17 signaling pathway, which may play a crucial role in its effectiveness. CONCLUSION Based on our findings, the optimal TG-C demonstrated the most favorable overall therapeutic efficacy by increasing the dosage of Artemisia capillaris while reducing the dosage of Gardenia jasminoides and Rheum palmatum. This is attributed to the potent ability of Artemisia capillaris. to effectively modulate the IL-17 signaling pathway, thereby exerting a beneficial therapeutic effect. Conversely, Gardenia jasminoides and Rheum palmatum may potentially enhance the activation of the NF-кB signaling pathway, thereby elevating the risk of hepatotoxicity.
Collapse
Affiliation(s)
- Shiman Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Meilan Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Mingming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Huiling Xiong
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiyue Tan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xinyu Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Wenwen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China; Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
10
|
Li Y, Ye Y, Yuan H, Rihan N, Han M, Liu X, Zhu T, Zhao Y, Che X. Exposure to polystyrene nanoplastics induces apoptosis, autophagy, histopathological damage, and intestinal microbiota dysbiosis of the Pacific whiteleg shrimp (Litopenaeus vannamei). THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 919:170924. [PMID: 38360329 DOI: 10.1016/j.scitotenv.2024.170924] [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: 01/18/2024] [Revised: 02/10/2024] [Accepted: 02/10/2024] [Indexed: 02/17/2024]
Abstract
Nanoplastics (NPs) are widely distributed environmental pollutants that can disrupt intestinal immunity of crustaceans. In this study, the effects of NPs on gut immune enzyme activities, cell morphology, apoptosis, and microbiota diversity of Litopenaeus vannamei were investigated. L. vannamei was exposed to five concentrations of NPs (0, 0.1, 1, 5, and 10 mg/L) for 28 days. The results showed that higher concentrations of NPs damaged the intestinal villi, promoted formation of autophagosomes, increased intestinal non-specific immunoenzyme activities, and significantly increased apoptosis at 10 mg/L. In response to exposure to NPs, the expression levels of ATG3, ATG4, ATG12, Caspase-3, p53, and TNF initially increased and then decreased. In addition, the concentration of NPs was negatively correlated to the expression levels of the genes of interest and intestinal enzyme activities, suggesting that exposure to NPs inhibited apoptosis and immune function. The five dominant phyla of the gut microbiota (Proteobacteria, Firmicutes, Bacteroidetes, Acidobacteria, and Actinomycetes) were similar among groups exposed to different concentrations of NPs, but the abundances tended to differ. Notably, exposure to NPs increased the abundance of pathogenic bacteria. These results confirm that exposure to NPs negatively impacted intestinal immune function of L. vannamei. These findings provide useful references for efficient breeding of L. vannamei.
Collapse
Affiliation(s)
- Yiming Li
- Fishery Machinery and Instrument Research Institute, Chinese Academy of Fisheries Sciences, Shanghai 200092, China
| | - Yucong Ye
- School of Life Science, East China Normal University, Shanghai 200241, China
| | - Haojuan Yuan
- School of Life Science, East China Normal University, Shanghai 200241, China
| | - Na Rihan
- School of Life Science, East China Normal University, Shanghai 200241, China
| | - Mingming Han
- Centre for Marine and Coastal Studies, University Sains Malaysia, Minden, Penang 11800, Malaysia
| | - Xingguo Liu
- Fishery Machinery and Instrument Research Institute, Chinese Academy of Fisheries Sciences, Shanghai 200092, China
| | - Tian Zhu
- Centre for Marine and Coastal Studies, University Sains Malaysia, Minden, Penang 11800, Malaysia
| | - Yunlong Zhao
- School of Life Science, East China Normal University, Shanghai 200241, China.
| | - Xuan Che
- Fishery Machinery and Instrument Research Institute, Chinese Academy of Fisheries Sciences, Shanghai 200092, China.
| |
Collapse
|
11
|
Gao T, Lin L, Yang Q, Zhu Z, Wang S, Xie T, Liao W. The raw and vinegar-processed Curcuma phaeocaulis Val. ameliorate TAA-induced zebrafish liver injury by inhibiting TLR4/MyD88/NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117246. [PMID: 37778523 DOI: 10.1016/j.jep.2023.117246] [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: 07/31/2023] [Revised: 09/27/2023] [Accepted: 09/27/2023] [Indexed: 10/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liver injury, the main factor in the pathogenesis of most liver diseases, is a known contributor to acute liver failure, liver fibrosis, or liver cancer. Curcuma phaeocaulis Val. (PEZ) has been broadly used in treating liver injury with satisfying therapeutic effects; however, the mechanism is still unclear. AIM OF THE STUDY This study aimed to explore the mechanism of PEZ in ameliorating thioacetamide (TAA)-induced zebrafish liver injury based on a comprehensive method integrating network-based computational prediction and experimental validations. MATERIALS AND METHODS Ultrahigh-performance liquid chromatography-quadrupole exactive mass spectrometry/mass spectrometry (UPLC-Q-Exactive MS/MS) analysis was used to analyze components in raw and vinegar-processed PEZ (VPEZ). Network pharmacology was used to construct a compound-target network for liver injury to predict the possible biological targets of PEZ along with potential signaling pathways. TAA-induced zebrafish larvae liver injury model was established, and the anti-liver injury effect of PEZ by a series of indexes was measured, including liver phenotype analysis, histopathological analysis of liver tissues, and biochemical indexes analysis. Remarkably, the predicted pathway by network pharmacology was further validated using RT-qPCR and Western blotting analyzes in animal experiments. RESULTS 40 chemical constituents derived from PEZ were identified, while 45 chemical components derived from VPEZ were identified. Based on it, 565 genes related to these identified compounds in PEZ and 1023 genes linked to liver injury were collected by network pharmacology. Critically, KEGG analysis indicated that the TLR4/MyD88/NF-κB signaling pathway was recommended as one of the main pathways related to the anti-liver injury effect of PEZ. Experimentally, PEZ could alleviate TAA-induced liver injury. Compared to the liver injury model group without any treatment, the treatment of PEZ significantly reduced the expression of both mRNA and protein targets in the TLR4/MyD88/NF-κB signaling pathway. In addition, the effect of VPEZ was more significant than that of the raw one. CONCLUSION The raw and vinegar-processed PEZ could ameliorate TAA-induced zebrafish liver injury through TLR4/MyD88/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Tianhui Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| | - Liting Lin
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| | - Qingsong Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| | - Zongping Zhu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| | - Shuyi Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| | - Tian Xie
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| | - Wan Liao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China; Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.
| |
Collapse
|
12
|
Luo H, Ji X, Zhang M, Ren Y, Tan R, Jiang H, Wu X. Aloe-emodin: Progress in Pharmacological Activity, Safety, and Pharmaceutical Formulation Applications. Mini Rev Med Chem 2024; 24:1784-1798. [PMID: 38639277 DOI: 10.2174/0113895575298364240409064833] [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: 02/04/2024] [Revised: 03/07/2024] [Accepted: 03/12/2024] [Indexed: 04/20/2024]
Abstract
Aloe-emodin (AE) is an anthraquinone derivative and a biologically active component sourced from various plants, including Rheum palmatum L. and Aloe vera. Known chemically as 1,8-dihydroxy-3-hydroxymethyl-anthraquinone, AE has a rich history in traditional medicine and is esteemed for its accessibility, safety, affordability, and effectiveness. AE boasts multiple biochemical and pharmacological properties, such as strong antibacterial, antioxidant, and antitumor effects. Despite its array of benefits, AE's identity as an anthraquinone derivative raises concerns about its potential for liver and kidney toxicity. Nevertheless, AE is considered a promising drug candidate due to its significant bioactivities and cost efficiency. Recent research has highlighted that nanoformulated AE may enhance drug delivery, biocompatibility, and pharmacological benefits, offering a novel approach to drug design. This review delves into AE's pharmacological impacts, mechanisms, pharmacokinetics, and safety profile, incorporating insights from studies on its nanoformulations. The goal is to outline the burgeoning research in this area and to support the ongoing development and utilization of AE-based therapies.
Collapse
Affiliation(s)
- Haimeng Luo
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiaoyun Ji
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Mengyu Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Yaoyao Ren
- School of Chemistry, Southwest Jiaotong University, Chengdu 610031, P.R. China
| | - Rui Tan
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Hezhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiaoqing Wu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
13
|
Xia W, Li S, Li L, Zhang S, Wang X, Ding W, Ding L, Zhang X, Wang Z. Role of anthraquinones in combating insulin resistance. Front Pharmacol 2023; 14:1275430. [PMID: 38053837 PMCID: PMC10694622 DOI: 10.3389/fphar.2023.1275430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
Insulin resistance presents a formidable public health challenge that is intricately linked to the onset and progression of various chronic ailments, including diabetes, cardiovascular disease, hypertension, metabolic syndrome, nonalcoholic fatty liver disease, and cancer. Effectively addressing insulin resistance is paramount in preventing and managing these metabolic disorders. Natural herbal remedies show promise in combating insulin resistance, with anthraquinone extracts garnering attention for their role in enhancing insulin sensitivity and treating diabetes. Anthraquinones are believed to ameliorate insulin resistance through diverse pathways, encompassing activation of the AMP-activated protein kinase (AMPK) signaling pathway, restoration of insulin signal transduction, attenuation of inflammatory pathways, and modulation of gut microbiota. This comprehensive review aims to consolidate the potential anthraquinone compounds that exert beneficial effects on insulin resistance, elucidating the underlying mechanisms responsible for their therapeutic impact. The evidence discussed in this review points toward the potential utilization of anthraquinones as a promising therapeutic strategy to combat insulin resistance and its associated metabolic diseases.
Collapse
Affiliation(s)
- Wanru Xia
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shuqian Li
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - LinZehao Li
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Shibo Zhang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaolei Wang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Wenyu Ding
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Lina Ding
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Xiandang Zhang
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Zhibin Wang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
14
|
Liu R, Huang Y, Chen Y, Huang D, Zhao Z, He T, Shi Y, Chen X. Lactobacillus plantarum E2 regulates intestinal microbiota and alleviates Pseudomonas plecoglossicida induced inflammation and apoptosis in zebrafish (Danio rerio). FISH & SHELLFISH IMMUNOLOGY 2023; 142:109170. [PMID: 37852511 DOI: 10.1016/j.fsi.2023.109170] [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: 07/07/2023] [Revised: 10/11/2023] [Accepted: 10/16/2023] [Indexed: 10/20/2023]
Abstract
Pseudomonas plecoglossicida infection is a highly contagious epidemic in aquaculture, causing significant mortality among teleost. Our previous research has demonstrated that Lactobacillus plantarum E2 is beneficial for large yellow croaker in resisting infections caused by P. plecoglossicida. However, the relevant mechanisms remain largely unclear. In the present study, we used zebrafish (Danio rerio) to further explore the function of L. plantarum E2 and its mechanisms for resisting P. plecoglossicida infection. E2 supplementation diet significantly improved the growth rates and α-amylase and trypsin activities of the liver in zebrafish. After challenge with P. plecoglossicida strain PQLYC4, the survival rates of zebrafish were improved, and immune-related genes expression (IL-1β, TNF-α, IL-8, Ig-Z, TLR-22 and IL-12α) were down-regulated. Histological analysis showed that E2 group had a longer intestinal villus and thicker intestinal walls after 30 days of feeding and healthier intestinal structure after challenge with P. plecoglossicida strain PQLYC4. Furthermore, co-incubation of zebrafish embryo fibroblast (ZF-4 cells) with L. plantarum E2 reduced apoptosis of ZF-4 cells after exposed to P. plecoglossicida. Intestinal microbiota analysis showed that E2 strain significantly increased the relative abundance of Lactobacillus and Pseudomonas, and PCoA analysis revealed a noticeable divergence in the intestinal microbial communities after E2 supplement. Together, our results suggested that E2 strain may promote zebrafish survival against P. plecoglossicida infection by regulating the intestinal microbiota and alleviating inflammatory response and apoptosis, thus exhibiting the potential as a probiotic.
Collapse
Affiliation(s)
- Ruizhe Liu
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yulu Huang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - You Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Dongliang Huang
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Zhexu Zhao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Tianliang He
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Yuan Shi
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xinhua Chen
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou, 350002, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, 519000, China; Fuzhou Institute of Oceanography, Fuzhou, 350108, China.
| |
Collapse
|
15
|
Wu P, Wang X. Natural Drugs: A New Direction for the Prevention and Treatment of Diabetes. Molecules 2023; 28:5525. [PMID: 37513397 PMCID: PMC10385698 DOI: 10.3390/molecules28145525] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Insulin resistance, as a common pathological process of many metabolic diseases, including diabetes and obesity, has attracted much attention due to its relevant influencing factors. To date, studies have mainly focused on the shared mechanisms between mitochondrial stress and insulin resistance, and they are now being pursued as a very attractive therapeutic target due to their extensive involvement in many human clinical settings. In view of the complex pathogenesis of diabetes, natural drugs have become new players in diabetes prevention and treatment because of their wide targets and few side effects. In particular, plant phenolics have received attention because of their close relationship with oxidative stress. In this review, we briefly review the mechanisms by which mitochondrial stress leads to insulin resistance. Moreover, we list some cytokines and genes that have recently been found to play roles in mitochondrial stress and insulin resistance. Furthermore, we describe several natural drugs that are currently widely used and give a brief overview of their therapeutic mechanisms. Finally, we suggest possible ideas for future research related to the unique role that natural drugs play in the treatment of insulin resistance through the above targets.
Collapse
Affiliation(s)
- Peishan Wu
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250001, China
| | - Xiaolei Wang
- Endocrine and Metabolic Diseases Hospital of Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250001, China
| |
Collapse
|
16
|
Chang C, Li H, Zhang R. Zebrafish facilitate non-alcoholic fatty liver disease research: Tools, models and applications. Liver Int 2023; 43:1385-1398. [PMID: 37122203 DOI: 10.1111/liv.15601] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 04/14/2023] [Accepted: 04/20/2023] [Indexed: 05/02/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become an increasingly epidemic metabolic disease worldwide. NAFLD can gradually deteriorate from simple liver steatosis, inflammation and fibrosis to liver cirrhosis and/or hepatocellular carcinoma. Zebrafish are vertebrate animal models that are genetically and metabolically conserved with mammals and have unique advantages such as high fecundity, rapid development ex utero and optical transparency. These features have rendered zebrafish an emerging model system for liver diseases and metabolic diseases favoured by many researchers in recent years. In the present review, we summarize a series of tools for zebrafish NAFLD research and the models established through different dietary feeding, hepatotoxic chemical treatments and genetic manipulations via transgenic or genome editing technologies. We also discuss how zebrafish models facilitate NAFLD studies by providing novel insights into NAFLD pathogenesis, toxicology research, and drug evaluation and discovery.
Collapse
Affiliation(s)
- Cheng Chang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Huicong Li
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Ruilin Zhang
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| |
Collapse
|
17
|
Sun Q, Tao Q, Ming T, Tang S, Zhao H, Liu M, Yang H, Ren S, Lei J, Liang Y, Peng Y, Wang M, Xu H. Berberine is a suppressor of Hedgehog signaling cascade in colorectal cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154792. [PMID: 37028248 DOI: 10.1016/j.phymed.2023.154792] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 03/01/2023] [Accepted: 03/28/2023] [Indexed: 06/19/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is a malignant affliction that burdens people globally. Overactivated Hedgehog signal is highly implicated in CRC pathogenesis. Phytochemical berberine exerts strong potency on CRC, with molecular mechanism elusive. PURPOSE We sought to study berberine's anti-CRC action and explore its underlying mechanism based on Hedgehog signaling cascade. METHODS In CRC HCT116 cells and SW480 cells treated with berberine, the proliferation, migration, invasion, clonogenesis, apoptosis and cell cycle were measured, with determination of Hedgehog signaling pathway activity. Following establishment of mouse model of HCT116 xenograft tumor, the efficacies of berberine on carcinogenesis, pathological manifestation and malignant phenotypes of CRC were examined, with analysis of Hedgehog signaling axis in HCT116 xenograft tumor tissues. Additionally, toxicological study of berberine was conducted on zebrafish. RESULTS Berberine was discovered to suppress the proliferation, migration, invasion and clonogenesis of HCT116 cells and SW480 cells. Furthermore, berberine caused cell apoptosis and blockaded cell cycle at phase G0/G1 in CRC cells, with dampened Hedgehog signaling cascade. In HCT116 xenograft tumor of nude mice, berberine inhibited tumor growth, alleviated pathological score, and promoted apoptosis and cell cycle arrest in tumor tissues, through constraining Hedgehog signaling. The toxicological study of berberine on zebrafish indicated that berberine incurred damage to the liver and heart of zebrafish at high dosage and prolonged administration. CONCLUSIONS Taken together, berberine may inhibit the malignant phenotypes of CRC through diminishing Hedgehog signaling cascade. However, the potential adverse reactions should be taken into account upon abuse of berberine.
Collapse
Affiliation(s)
- Qiang Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Qiu Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tianqi Ming
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shun Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Hui Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Maolun Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Han Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shan Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiarong Lei
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuanjing Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuhui Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Minmin Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Haibo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
18
|
Sun W, Zhang X, Qiao Y, Griffin N, Zhang H, Wang L, Liu H. Exposure to PFOA and its novel analogs disrupts lipid metabolism in zebrafish. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 259:115020. [PMID: 37201426 DOI: 10.1016/j.ecoenv.2023.115020] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/24/2023] [Accepted: 05/14/2023] [Indexed: 05/20/2023]
Abstract
Perfluorooctanoic acid (PFOA), a typical perfluoroalkyl group compound, has received worldwide attention due to its significant environmental toxicity. Following regulatory bans on the production and emission of PFOA, concerns have been raised about the potential health risks and the safety of novel perfluoroalkyl analogues. HFPO-DA (trade name Gen-X) and HFPO-TA are two perfluoroalkyl analogues known to be bioaccumulative, whose level of toxicity and whether they are safe alternatives to PFOA remain unclear. In the following study, the physiological and metabolic effects of exposure to PFOA and its novel analogues were explored in zebrafish using 1/3 LC50 (PFOA 100 μM, Gen-X 200 μM, HFPO-TA 30 μM). At the same LC50 toxicological effect, exposure to PFOA and HFPO-TA resulted in abnormal phenotypes such as spinal curvature, pericardial edema and aberrant body length, while Gen-X was little changed. Metabolically, PFOA, HFPO-TA and Gen-X all significantly increased total cholesterol in exposed zebrafish with PFOA and HFPO-TA also increasing total triglyceride levels. Transcriptome analysis showed that the number of differentially expressed genes in PFOA, Gen-X, and HFPO-TA treated conditions compared to control groups were 527, 572, and 3, 933, respectively. KEGG and GO analysis of differentially expressed genes revealed pathways and functions related to lipid metabolism as well as significant activation of the peroxisome proliferators-activated receptor (PPARs) pathway. Furthermore, RT-qPCR analysis identified significant dysregulation in the downstream target genes of PPARα, which is responsible for lipid oxidative catabolism, and the SREBP pathway, which is responsible for lipid synthesis. In conclusion, both perfluoroalkyl analogues HFPO-TA and Gen-X exhibit significant physiological and metabolic toxicity to aquatic organisms and their environmental accumulation should be closely regulated.
Collapse
Affiliation(s)
- Weiqiang Sun
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu 233030, PR China; Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu 233030, PR China; Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, School of Laboratory Medicine, Bengbu Medical College, Bengbu 233030, PR China
| | - Xuemin Zhang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu 233030, PR China; Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu 233030, PR China; Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, School of Laboratory Medicine, Bengbu Medical College, Bengbu 233030, PR China
| | - Ying Qiao
- School of Public Health, Bengbu Medical College, Bengbu 233030, PR China
| | - Nathan Griffin
- Department of Cell and Tissue Biology, University of California, San Francisco, CA, USA
| | - Hongxia Zhang
- State Key Laboratory of Integrated Management of Pest Insects and Rodents, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
| | - Li Wang
- School of Public Health, Bengbu Medical College, Bengbu 233030, PR China.
| | - Hui Liu
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu 233030, PR China; Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, Bengbu 233030, PR China; Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, School of Laboratory Medicine, Bengbu Medical College, Bengbu 233030, PR China.
| |
Collapse
|
19
|
Zhi G, Shao B, Zheng T, Mu J, Li J, Feng Y, Zhu S, Dang Y, Liu F, Wang D. Exploring the molecular mechanism of Gan Shuang granules for the treatment of non-alcoholic steatohepatitis using network pharmacology, molecular docking, and experimental verification. Front Pharmacol 2023; 14:1082451. [PMID: 36762105 PMCID: PMC9902723 DOI: 10.3389/fphar.2023.1082451] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/13/2023] [Indexed: 01/25/2023] Open
Abstract
Background: With the gradual increase in prevalence in recent years, non-alcoholic steatohepatitis (NASH) has become one of the significant health problems that urgently needs to be addressed worldwide. GanShuang Granules (GSG) is derived from the classical Chinese formula Xiaoyao San and mainly used in the clinical treatment of chronic liver diseases. Objective: In this study, we aim to gain a deeper insight into the inhibiting effects of GSG on non-alcoholic fatty liver disease (NAFLD) rats and preliminarily elucidate the underlying intervention mechanisms. Methods: First, High performance liquid chromatography (UHPLC-Q/Orbitrap-MS/MS) was used for the active compounds prediction in GSG. Then the data was mapped to mzCloud database. The targets corresponding to GSG compounds were collected from public databases, along with disease genes for NAFLD. The core targets and molecular mechanisms of GSG for NAFLD treatment were predicted by protein-protein interaction (PPI) network, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment analyses. Molecular docking of the core target-component interactions was simulated using AutoDock Vina software. The effect of GSG on NASH rats was evaluated by pathological staining and analysis of various index results. Finally, the candidate targets were further validated by ELISA and western blot (WB) analyses. Results: Combining UHPLC-Q/Orbitrap-MS/MS data analysis and public database data, a total of 346 cross-targets were obtained, corresponding to 81 compounds. The subnetwork with an MCODE score of 53.623 is a potential core target group for this study. GO and KEGG enrichment analyses showed that the targets of GSG in NAFLD were mostly related to oxidative stress, the NF-κB signaling pathway, and the apoptosis signaling pathway. By integrating the results of network pharmacology analysis, the core objectives of this study mainly include AKT1, CASP9, TNF, and CASP8. The core ingredients are related to resveratrol and fisetin. The molecular docking results indicated key binding activity between AKT1-fisetin, AKT1-Resveratrol, and CASP8-fisetin. Moreover, GSG could improve the inflammatory status and restore the abnormal lipid accumulation of NAFLD/NASH liver, and these levels are further verified by pathological staining and detection of related indicators. Mechanistically, GSG could regulate protein expression levels in the liver for P65, p-P65, IKB, p-IKB, IKK, caspase-3, -8, -9, and cytochrome C, etc. It reflects the inhibitory effect of GSG on the NF-κB/IκB signaling pathway. Conclusion: Our results suggested that GSG demonstrated therapeutic effects on NAFLD/NASH rats, and these may be mainly reflected in the inhibitory effects on the NF-κB/IκB signaling pathway and its downstream inflammation and apoptosis signals.
Collapse
Affiliation(s)
- Guoguo Zhi
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Bingjie Shao
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Tianyan Zheng
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jie Mu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Jingwei Li
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yiyuan Feng
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Sha Zhu
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yanni Dang
- Shanxi Buchang Pharmaceutical Company Limited, Xi’an, Shanxi, China
| | - Feng Liu
- Shanxi Buchang Pharmaceutical Company Limited, Xi’an, Shanxi, China,*Correspondence: Feng Liu, ; Dong Wang,
| | - Dong Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China,*Correspondence: Feng Liu, ; Dong Wang,
| |
Collapse
|
20
|
He G, Zhao Q, Zhao Y, Zong Y, Gu S, Li M, Li R, Sun J. Deer antler based active ingredients have protective effects on LPS/d-GalN-induced acute liver injury in mice through MAPK and NF-κB signalling pathways. PHARMACEUTICAL BIOLOGY 2022; 60:1077-1087. [PMID: 35645173 PMCID: PMC9154794 DOI: 10.1080/13880209.2022.2068617] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
CONTEXT Deer antler based active ingredients are known to have certain anti-inflammatory and antioxidant activities. However, its potential hepatoprotective effect remains unclear. OBJECTIVE This article reports the hepatoprotective effect of protein components in deer antler bases (R1) on lipopolysaccharide/d-galactosamine (LPS/d-GalN)-induced acute liver injury (ALI) in mice, and explores its possible mechanism. MATERIALS AND METHODS The four separated and purified protein components of deer antler bases were screened and verified by the RAW264.7 cell inflammation model. In the in vivo experiment of LPS/d-GalN-induced ALI in mice, ALT, AST, SOD, CAT, GSH and MDA were detected. The liver histopathology was analysed, the COX-2 and iNOS proteins were analysed by immunohistochemistry, and 4-HNE was analysed by immunofluorescence staining. In addition, the effects on the MAPK pathway and NF-κB/IκB-α pathway in liver proteins were explored. RESULTS With isolated RA protein fraction pre-treated RAW264.7 cells, NO production decreased by 35.3% compared with the model group. The experimental results of ALI in mice induced by LPS/d-GalN show that R1 protein components can protect mice from ALI through anti-inflammatory and anti-oxidative stress effects and reduce liver pathological damage in mice. The results also indicate that the R1 protein component may protect the liver by inhibiting the activation of the MAPK pathway and the NF-κB/IκB-α pathway induced by LPS/d-GalN. CONCLUSIONS The separated and purified R1 protein component of deer antler base has a good protective effect on LPS/d-GalN-induced liver injury, and may become a potential material for protecting against liver injury.
Collapse
Affiliation(s)
- Guixiang He
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Quanmin Zhao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
- CONTACT Quanmin Zhao
| | - Yan Zhao
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
- Yan Zhao College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun130118, Jilin, China
| | - Ying Zong
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Shigang Gu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Mengjie Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Renjie Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| | - Jiaxin Sun
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, China
| |
Collapse
|
21
|
The Synthesis and Biological Evaluation of Aloe-Emodin-Coumarin Hybrids as Potential Antitumor Agents. Molecules 2022; 27:molecules27196153. [PMID: 36234685 PMCID: PMC9571363 DOI: 10.3390/molecules27196153] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/15/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
A series of novel aloe-emodin–coumarin hybrids were designed and synthesized. The antitumor activity of these derivatives was evaluated against five human tumor cell lines (A549, SGC-7901, HepG2, MCF-7 and HCT-8). Some of the synthesized compounds exhibited moderate to good activity against one or more cell lines. Particularly, compound 5d exhibited more potent antiproliferative activity than the reference drug etoposide against all tested tumor cell lines, indicating that it had a broad spectrum of antitumor activity and that it may provide a promising lead compound for further development as an antitumor agent by structural modification. Furthermore, the structure–activity relationship study of the synthesized compounds was also performed.
Collapse
|
22
|
Zhang ZL, Li YZ, Wu GQ, Zhang DD, Deng C, Wang ZM, Song XM, Wang W. A comprehensive review of traditional uses, phytochemistry and pharmacology of Reynoutria genus. J Pharm Pharmacol 2022; 74:1718-1742. [DOI: 10.1093/jpp/rgac068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/22/2022] [Indexed: 11/14/2022]
Abstract
Abstract
Objectives
The genus Reynoutria belonging to the family Polygonaceae is widely distributed in the north temperate zone and used in folk medicine. It is administered as a sedative, tonic and digestive, also as a treatment for canities and alopecia. Herein, we reported a review on traditional uses, phytochemistry and pharmacology reported from 1985 up to early 2022. All the information and studies concerning Reynoutria plants were summarized from the library and digital databases (e.g. ScienceDirect, SciFinder, Medline PubMed, Google Scholar, and CNKI).
Key findings
A total of 185 articles on the genus Reynoutria have been collected. The phytochemical investigations of Reynoutria species revealed the presence of more than 277 chemical components, including stilbenoids, quinones, flavonoids, phenylpropanoids, phospholipids, lactones, phenolics and phenolic acids. Moreover, the compounds isolated from the genus Reynoutria possess a wide spectrum of pharmacology such as anti-atherosclerosis, anti-inflammatory, antioxidative, anticancer, neuroprotective, anti-virus and heart protection.
Summary
In this paper, the traditional uses, phytochemistry and pharmacology of genus Reynoutria were reviewed. As a source of traditional folk medicine, the Reynoutria genus have high medicinal value and they are widely used in medicine. Therefore, we hope our review can help genus Reynoutria get better development and utilization.
Collapse
Affiliation(s)
- Zi-Long Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine , Xian Yang, Shaanxi 712046 , China
| | - Yu-Ze Li
- School of Pharmacy, Shaanxi University of Chinese Medicine , Xian Yang, Shaanxi 712046 , China
| | - Guo-Qing Wu
- School of Pharmacy, Shaanxi University of Chinese Medicine , Xian Yang, Shaanxi 712046 , China
| | - Dong-Dong Zhang
- School of Pharmacy, Shaanxi University of Chinese Medicine , Xian Yang, Shaanxi 712046 , China
| | - Chong Deng
- School of Pharmacy, Shaanxi University of Chinese Medicine , Xian Yang, Shaanxi 712046 , China
| | - Zhi-Min Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences , BeiJing 100700 , China
| | - Xiao-Mei Song
- School of Pharmacy, Shaanxi University of Chinese Medicine , Xian Yang, Shaanxi 712046 , China
| | - Wei Wang
- School of Pharmacy, Shaanxi University of Chinese Medicine , Xian Yang, Shaanxi 712046 , China
| |
Collapse
|
23
|
Yang H, Gu X, Chen H, Zeng Q, Mao Z, Jin M, Li H, Ge Y, Zha J, Martyniuk CJ. Transcriptome profiling reveals toxicity mechanisms following sertraline exposure in the brain of juvenile zebrafish (Danio rerio). ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 242:113936. [PMID: 35930839 DOI: 10.1016/j.ecoenv.2022.113936] [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: 05/14/2022] [Revised: 07/24/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Sertraline (SER) is one of the most commonly detected antidepressants in the aquatic environment that can negatively affect aquatic organisms at low concentrations. Despite some knowledge on its acute toxicity to fish, the effects of chronic SER exposure remain poorly understood along with any underlying mechanisms of SER-induced toxicity. To address this knowledge gap, the effects of chronic exposure to three SER concentrations from low to high were investigated in zebrafish. Juvenile zebrafish were exposed to three concentrations of 1, 10, or 100 μg/L of SER for 28 d, after which indicators of oxidative stress and neurotoxicity in the brain were measured. Superoxide dismutase (SOD) activity was significantly enhanced by SER at 1 up to 100 μg/L, and catalase (CAT) activity was significantly induced by SER at 1 or 10 μg/L. The activity of acetylcholinesterase (AChE) was significantly induced by 10 and 100 μg/L of SER, and the serotonin (5-HT) level was significantly increased by all three concentrations of SER. To ascertain mechanisms of SER-induced toxicity, transcriptomics was conducted in the brain of zebrafish following 100 μg/L SER exposure. The molecular signaling pathways connected with circadian system and the immune system were significantly altered in the zebrafish brain. Based on transcriptomic data, the expression levels of six circadian clock genes were measured, and three genes were significantly altered in relative abundance in fish from all experimental treatments with SER, including cryptochrome circadian regulator 2 (cry2), period circadian clock 2 (per2), and period circadian clock 3 (per3). We hypothesize that the circadian system may be related to SER-induced neurotoxicity and oxidative stress in the central nervous system. This study reveals potential mechanisms and key events (i.e., oxidative stress and neurotoxicity) associated with SER-induced toxicity, and improves understanding of the molecular and biochemical pathways putatively perturbed by SER.
Collapse
Affiliation(s)
- Huiting Yang
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaohong Gu
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian 223300, China.
| | - Huihui Chen
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China.
| | - Qingfei Zeng
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China
| | - Zhigang Mao
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China
| | - Miao Jin
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongmin Li
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - You Ge
- State Key Laboratory of Lake Science and Environment, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing 210008, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinmiao Zha
- University of Chinese Academy of Sciences, Beijing 100049, China; State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Christopher J Martyniuk
- Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida Genetics Institute, Interdisciplinary Program in Biomedical Sciences Neuroscience, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611 USA
| |
Collapse
|
24
|
Zhao Y, Wang H, Duah PA, Retyunskiy V, Liu Y, Chen G. Zinc pyrithione (ZPT) -induced embryonic toxicogenomic responses reveal involvement of oxidative damage, apoptosis, endoplasmic reticulum (ER) stress and autophagy. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2022; 248:106195. [PMID: 35594629 DOI: 10.1016/j.aquatox.2022.106195] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/19/2022] [Accepted: 05/10/2022] [Indexed: 06/15/2023]
Abstract
Zinc pyrithione (ZPT) is a frequently used organometallic biocide, carrying potentially adverse consequences to multiple species in the environment. Previously we have demonstrated its embryonic, organ developmental and liver metabolic toxicity of zebrafish. However, details of ZPT toxicity during embryogenesis are still limited. The present study was designed to evaluate the effects and possible mechanisms of ZPT-induced embryonic toxicogenomic responses by morphological investigations, transcriptome and gene quantitative analysis, as well as biochemical assays. The results revealed that treatment with ZPT caused embryogenesis toxicity, specifically in irregular cell division and rearrangement, delayed differentiations of eyes and notochords, the epiboly and germ ring formation and somite segmentation defects. In addition, ZPT exposure altered gene expression during early embryonic development, especially related with morphological abnormities and metabolic dysfunctions including reduction of oxidoreductase activity. Activities of antioxidants and caspases examinations showed inductions of oxidative stress and apoptosis by ZPT and quantitative analysis of marker genes further indicated that ZPT also triggered endoplasmic reticulum (ER) stress and autophagy. Thus, we deduce here that ZPT-induced embryonic toxicogenomic responses reveal involvement of oxidative damage, apoptosis, endoplasmic reticulum (ER) stress and autophagy.
Collapse
Affiliation(s)
- Ye Zhao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211800, China.
| | - Huiling Wang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211800, China
| | | | - Vladimir Retyunskiy
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211800, China
| | - Yizheng Liu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211800, China
| | - Guoguang Chen
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing 211800, China.
| |
Collapse
|
25
|
Jiang H, Mao T, Liu Y, Tan X, Sun Z, Cheng Y, Han X, Zhang Y, Wang J, Shi L, Guo Y, Li J, Han H. Protective Effects and Mechanisms of Yinchen Linggui Zhugan Decoction in HFD-Induced Nonalcoholic Fatty Liver Disease Rats Based on Network Pharmacology and Experimental Verification. Front Pharmacol 2022; 13:908128. [PMID: 35721171 PMCID: PMC9202027 DOI: 10.3389/fphar.2022.908128] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a common chronic liver disease, characterized by excessive accumulation of hepatocyte fat. However, there is no exact and effective pharmacotherapy for NAFLD. Yinchen linggui zhugan decoction (YLZD) has been widely used to treat NAFLD. Nevertheless, its pharmacological and molecular mechanisms have not been clearly elucidated. This study was carried out to investigate the active components of YLZD and explore its potential mechanisms for treating NAFLD by network pharmacology and experimental verification. The results showed that a total of 120 active components of YLZD and 365 targets were retrieved through databases, and the main active ingredients of YLZD consisted of chlorogenic acid, emodin, aloe-emodin, rhein, and geniposide. KEGG enrichment analysis revealed fundamental roles of TNF, PI3K/AKT, HIF-1α, and insulin resistance signaling pathways in the treatment of NAFLD by YLZD. Moreover, our experimental verification results showed that YLZD improved the liver pathological and cholesterol level, and reduced the expressions of TNF-α, IL-1β, IL-6, NF-κB, CCL2, and CXCL10 in NAFLD rats, which all belonged to TNF signaling pathway. The molecular docking confirmed the correlation between the four core components (chlorogenic acid, emodin, rhein, and geniposide) and key factors (TNF-α, IL-6, and NF-κB) in TNF signaling pathway. In conclusion, the present study systematically clarified the protective mechanisms of YLZD against NAFLD through targeting the TNF signaling pathway, and provided new ideas for the drug research of this disease.
Collapse
Affiliation(s)
- Hui Jiang
- School of Graduate, Beijing University of Chinese Medicine, Beijing, China.,Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Tangyou Mao
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yuyue Liu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiang Tan
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Zhongmei Sun
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yuan Cheng
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao Han
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yang Zhang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jiali Wang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Lei Shi
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yi Guo
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Junxiang Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Haixiao Han
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
26
|
Wang Y, Wang J, Xiang H, Ding P, Wu T, Ji G. Recent update on application of dihydromyricetin in metabolic related diseases. Biomed Pharmacother 2022; 148:112771. [PMID: 35247719 DOI: 10.1016/j.biopha.2022.112771] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/22/2022] [Accepted: 02/27/2022] [Indexed: 11/02/2022] Open
Abstract
As a new type of natural flavonoids, dihydromyricetin (DMY) has attracted more and more attention. It has a series of pharmacological effects, such as anti-inflammatory, anti-tumor, anti-oxidation, antibacterial and so on, and it is almost no toxicity and with excellent safety. Therefore, even if the bioavailability is poor, it is often added to daily food, beverages and even medicines. In recent years, some researchers have found that DMY can treat some diseases by anti-oxidation, anti-inflammation, promoting cell death and regulate the activity of lipid and glucose metabolism. In addition, the mechanism of DMY on these diseases was also related to the signal pathway of AMPK, PI3K/Akt, PPAR and the participation of microRNAs. This review describes the mechanism of DMY in metabolic related diseases from three aspects: metabolic diseases, liver diseases, and cancers, hoping to provide some new ideas for clinical researches.
Collapse
Affiliation(s)
- Yirong Wang
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Junmin Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Peilun Ding
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
27
|
Liu F, Zhang Y, Wang F. Environmental relevant concentrations of triclosan affected developmental toxicity, oxidative stress, and apoptosis in zebrafish embryos. ENVIRONMENTAL TOXICOLOGY 2022; 37:848-857. [PMID: 34981884 DOI: 10.1002/tox.23448] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/16/2021] [Accepted: 12/27/2021] [Indexed: 06/14/2023]
Abstract
Triclosan (TCS), as a broad-spectrum antibacterial agent, is widely used in various pharmaceutical and personal care products. However, the details of ecological environmental health risks of TCS are not clear. In this study, zebrafish embryos/larval were exposed to environmentally relevant concentrations of TCS to evaluate the developmental toxicity. Four-hour post-fertilization (hpf) zebrafish embryos were exposed to 0, 2, 10, 50, and 250 μg/L TCS until 96 h. The heart beats at 72 hpf were significantly increased in 2 μg/L TCS group, while significantly decreased in 250 μg/L TCS treated group compared with control. The results of acridine orange staining, terminal deoxynucleotide transferase-mediated UTPnick end labeling assay, and detection of mitochondrial membrane potential showed that 50 and 250 μg/L TCS resulted in apoptosis. Meanwhile, reactive oxygen species (ROS) and DNA damage were induced, but SOD activity was significantly decreased in 250 μg/L TCS treated group. In addition, SOD(Mn) and GPx gene mRNA expressions were significantly down-regulated in 50 and 250 μg/L TCS treated groups, while Casp3, Casp9, Puma, Casp8, Apaf1, and Bid genes in 250 μg/L TCS and Mdm2 gene in 50 μg/L treated groups were significantly up-regulated. P53 protein was significantly up-regulated in 250 μg/L TCS treated group. The overall results showed that TCS can cause oxidative stress and result in apoptosis via the involvement of ROS-p53-caspase-dependent apoptotic pathway in zebrafish embryos. The present findings suggest the potential mechanisms of TCS-induced developmental toxicity appears to be the generation of ROS and the consequent triggering of apoptosis genes.
Collapse
Affiliation(s)
- Fei Liu
- School of Biological Science, Luoyang Normal University, Luoyang, China
| | - Ying Zhang
- School of Biological Science, Luoyang Normal University, Luoyang, China
| | - Fan Wang
- School of Biological Science, Luoyang Normal University, Luoyang, China
| |
Collapse
|
28
|
Zhou J, Peng C, Li Q, Yan X, Yang L, Li M, Cao X, Xie X, Chen D, Rao C, Huang S, Peng F, Pan X. Dopamine Homeostasis Imbalance and Dopamine Receptors-Mediated AC/cAMP/PKA Pathway Activation are Involved in Aconitine-Induced Neurological Impairment in Zebrafish and SH-SY5Y Cells. Front Pharmacol 2022; 13:837810. [PMID: 35370746 PMCID: PMC8971779 DOI: 10.3389/fphar.2022.837810] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/25/2022] [Indexed: 11/25/2022] Open
Abstract
Aconitine is one of the main bioactive and toxic ingredients of Aconitum species. Increasingly, aconitine has been reported to induce neurotoxicity. However, whether aconitine has effects on the dopaminergic nervous system remains unclear. In this study, zebrafish embryos at 6-days postfertilization were exposed to aconitine at doses of 0.5, 1, and 2 μM for 24 h, and SH-SY5Y cells were treated with 50, 100, and 200 μM of aconitine for 24 h. Results demonstrated that aconitine treatment induced deformities and enhanced the swimming behavior of zebrafish larvaes. Aconitine exposure suppressed cell proliferation and increased the number of reactive oxygen species and apoptosis in zebrafish larvaes and SH-SY5Y cells. Aconitine altered the levels of dopamine and its metabolites by regulating the expression of genes and proteins related to dopamine synthesis, storage, degradation, and reuptake in vivo and in vitro. Moreover, aconitine activated the AC/cAMP/PKA pathway by activating the dopamine D1 receptor (D1R) and inhibiting the dopamine D2 receptor (D2R) to disturb intracellular calcium homeostasis, eventually leading to the damage of nerve cells. Furthermore, the D1R antagonist SCH23390 and D2R agonist sumanirole pretreatment effectively attenuated the excitatory state of larvaes. Sumanirole and PKA antagonist H-89 pretreatment effectively decreased intracellular Ca2+ accumulation induced by aconitine in vivo. SCH23390 and sumanirole also reduced aconitine-induced cytotoxicity by inhibiting the AC/cAMP/PKA pathway in vitro. These results suggested that dopamine homeostasis imbalance and dopamine receptors (DRs)-mediated AC/cAMP/PKA pathway activation might be vital mechanisms underlying aconitine-induced neurological injury.
Collapse
Affiliation(s)
- Jie Zhou
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Cheng Peng
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiuju Li
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Yan
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liang Yang
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengting Li
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Cao
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Xie
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dayi Chen
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chaolong Rao
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sizhou Huang
- Development and Regeneration Key Laboratory of Sichuan Province, Department of Anatomy and Histology and Embryology, School of Basic Medicine, Chengdu Medical College, Chengdu, China
| | - Fu Peng
- West China School of Pharmacy, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Fu Peng, ; Xiaoqi Pan,
| | - Xiaoqi Pan
- Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Public Health, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Fu Peng, ; Xiaoqi Pan,
| |
Collapse
|
29
|
Tang X, Zhang Y, Liu X, Li X, Zhao H, Cui H, Shi Y, Chen Y, Xu H, Meng Z, Zhao L, Chen H, Wang Z, Zhu M, Lin Y, Yang B, Zhang Y. Aloe-emodin derivative produces anti-atherosclerosis effect by reinforcing AMBRA1-mediated endothelial autophagy. Eur J Pharmacol 2022; 916:174641. [PMID: 34800465 DOI: 10.1016/j.ejphar.2021.174641] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 10/15/2021] [Accepted: 11/12/2021] [Indexed: 11/29/2022]
Abstract
Atherosclerosis is an inflammatory disease of high lethality associated with endothelial dysfunction. Due to the pathophysiological complexity and our incomplete understanding of the mechanisms for the development and progression of atherosclerosis, effective means for the prevention and treatment of atherosclerosis still need further exploration. This study was designed to investigate the potential effects and underlying mechanisms of aloe-emodin derivative (AED) on atherosclerosis. High fat diet (HFD) treated ApoE-/- mice were used as an animal model of atherosclerosis. Intragastric administration of aloe-emodin (AE) or AED for 12 weeks markedly reduced the atherosclerotic plaque in aorta with decreased plaque area, lipid accumulation, macrophage infiltration, collagen content and metabolic abnormalities. By comparison, AED produced more potent anti-atherosclerosis effects than AE at the same dose. AED enhanced production of autophagy flux in cultured human aortic endothelial cells (HAECs). Moreover, AED increased the expression of activating molecule in Beclin1-regulated autophagy 1 (AMBRA1), a key protein involved in autophagosome formation. Furthermore, knockdown of AMBRA1 blocked the promotion effect of AED on autophagy in HAECs. Taken together, AED facilitates endothelial autophagy via AMBRA1 during the progression of atherosclerosis, suggesting the potential application of this compound for atherosclerosis treatment.
Collapse
Affiliation(s)
- Xueqing Tang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yue Zhang
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Xin Liu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Xiaohan Li
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Hongrui Zhao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Hao Cui
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yang Shi
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yongchao Chen
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Honglin Xu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Ziyu Meng
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Limin Zhao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Hui Chen
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Zhixia Wang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Mengying Zhu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Yuan Lin
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China
| | - Baofeng Yang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070, PR China; Department of Pharmacology and Therapeutics, Melbourne School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences University of Melbourne, Melbourne, Australia.
| | - Yong Zhang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, PR China; Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Harbin, PR China; Research Unit of Noninfectious Chronic Diseases in Frigid Zone, Chinese Academy of Medical Sciences, 2019RU070, PR China.
| |
Collapse
|
30
|
Advances in Understanding the Role of Aloe Emodin and Targeted Drug Delivery Systems in Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7928200. [PMID: 35087619 PMCID: PMC8789423 DOI: 10.1155/2022/7928200] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 12/06/2021] [Accepted: 12/18/2021] [Indexed: 12/20/2022]
Abstract
Cancer is one of the important causes of death worldwide. Despite remarkable improvements in cancer research in the past few decades, several cancer patients still cannot be cured owing to the development of drug resistance. Natural sources might have prominence as potential drug candidates. Among the several chemical classes of natural products, anthraquinones are characterized by their large structural variety, noticeable biological activity, and low toxicity. Aloe emodin, an anthraquinone derivative, is a natural compound found in the roots and rhizomes of many plants. This compound has proven its antineoplastic, anti-inflammatory, antiangiogenic, and antiproliferative potential as well as ability to prevent cancer metastasis and potential in reversing multidrug resistance of cancer cells. The anticancer property of aloe emodin, a broad-spectrum inhibitory agent of cancer cells, has been detailed in many biological pathways. In cancer cells, these molecular mechanisms consist of inhibition of cell growth and proliferation, cell cycle arrest deterioration, initiation of apoptosis, antimetastasis, and antiangiogenic effect. In accordance with the strategy of developing potential drug candidates from natural products, aloe emodin's low bioavailability has been tried to be overcome by structural modifications and nanocarrier systems. Consequently, this review summarizes the antiproliferative and anticarcinogenic properties of aloe emodin, as well as the enhanced activity of its derivatives and the advantages of drug delivery systems on bioavailability.
Collapse
|
31
|
Zhang Y, Xia Q, Wang J, Zhuang K, Jin H, Liu K. Progress in using zebrafish as a toxicological model for traditional Chinese medicine. JOURNAL OF ETHNOPHARMACOLOGY 2022; 282:114638. [PMID: 34530096 DOI: 10.1016/j.jep.2021.114638] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/25/2021] [Accepted: 09/11/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese medicine (TCM) has been applied for more than 2000 years. However, modern basic research on the safety of TCMs is limited. Establishing safety evaluation technology in line with the characteristics of TCM and conducting large-scale basic toxicity research are keys to comprehensively understand the toxicity of TCMs. In recent years, zebrafish has been used as a model organism for toxicity assessment and is increasingly utilized for toxicity research of TCMs. Yet, a comprehensive review in using zebrafish as a toxicological model for TCMs is lacked. AIM OF THE STUDY We aim to summarize the progress and limitation in toxicity evaluation of TCMs using zebrafish and put forward the future research ideas. MATERIALS AND METHODS The scientific databases, including Springer, Science Direct, Wiley, Pubmed and China Knowledge Resource Integrated (CNKI) were searched using the key words of zebrafish, toxicology, traditional Chinese medicine, acute toxicity, liver injury, cardiotoxicity, kidney toxicity, developmental toxicity, neurotoxicity, gastrointestinal irritation, immunotoxicity, ototoxicity, and osteotoxicity. RESULTS Zebrafish assays are low experimental cost and short cycle, easily achieving high-throughput toxicity screening, and exemption from ethical legislation up to 5 dpf. It has been widely used to evaluate the acute toxicity, liver toxicity, cardiotoxicity, nephrotoxicity, developmental toxicity, neurotoxicity, gastrointestinal irritation, immunotoxicity, and ototoxicity caused by TCMs, although some physiological difference limited its application. CONCLUSIONS Zebrafish is a powerful model for TCMs toxicity evaluation, but it is not flawless. The toxicity testing criterion and high throughput assays are urgent to be established. This review provides references for future studies.
Collapse
Affiliation(s)
- Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, China
| | - Qing Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, China
| | - Jiabo Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Kaiyan Zhuang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, China
| | - Hongtao Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan, China.
| |
Collapse
|
32
|
Hu YH, Quan ZY, Li DK, Wang CY, Sun ZX. Inhibition of CYP3A4 enhances aloe-emodin induced hepatocyte injury. Toxicol In Vitro 2021; 79:105276. [PMID: 34875353 DOI: 10.1016/j.tiv.2021.105276] [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: 09/08/2021] [Revised: 11/19/2021] [Accepted: 11/22/2021] [Indexed: 11/18/2022]
Abstract
Aloe-emodin (AE) is a natural hydroxyanthraquinone derivative that was found in many medicinal plants and ethnic medicines. AE showed a wide array of pharmacological activities including anticancer, antifungal, laxative, antiviral, and antibacterial effects. However, increasing number of published studies have shown that AE may have some hepatotoxicity effects but the mechanism is not fully understood. Studies have shown that the liver injury induced by some free hydroxyanthraquinone compounds is associated with the inhibition of some metabolic enzymes. In this study, the CYP3A4 and CYP3A1 were found to be the main metabolic enzymes of AE in human and rat liver microsomes respectively. And AE was metabolized by liver microsomes to produce hydroxyl metabolites and rhein. When CYP3A4 was knocked down in L02 and HepaRG cells, the cytotoxicity of AE was increased significantly. Furthermore, AE increased the rates of apoptosis of L02 and HepaRG cells, accompanied by Ca2+ elevation, mitochondrial membrane potential (MMP) loss and reactive oxygen species (ROS) overproduction. The mRNA expression of heme oxygenase-1 in L02 and HepaRG cells increased significantly in the high-dose of AE (40 μmol/L) group, and the mRNA expression of quinone oxidoreductase-1 was activated by AE in all concentrations. Taken together, the inhibition of CYP3A4 enhances the hepatocyte injury of AE. AE can induce mitochondrial injury and the imbalance of oxidative stress of hepatocytes, which results in hepatocyte apoptosis.
Collapse
Affiliation(s)
- Ying-Huan Hu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zheng-Yang Quan
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Deng-Ke Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Cheng-Yu Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhen-Xiao Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
33
|
Ma Y, Li J, Ju Z, Huang W, Wang Z, Yang L, Ding L. Danning tablets alleviate high fat diet-induced obesity and fatty liver in mice via modulating SREBP pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114320. [PMID: 34116189 DOI: 10.1016/j.jep.2021.114320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 06/01/2021] [Accepted: 06/06/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The traditional Chinese formula Danning tablets exhibit wide clinical applications in liver and gallbladder diseases, and currently it is reported to be effective on fatty liver disease in clinical trials. However, the underlying mechanisms remain elusive. AIM OF THE STUDY The purpose of the present study was to assess the effects and potential pharmacological mechanisms of Danning tablet against high fat diet (HFD)-induced obesity, fatty liver, and related metabolic disorders in mice. MATERIALS AND METHODS C57BL/6 J male mice were treated with HFD for 12 weeks to trigger obesity and fatty liver condition. Then those mice were randomly divided into 5 groups, namely HFD, Danning tablet (0.75, 1.5 or 3 g/kg bodyweight) or lovastatin (30 mg/kg bodyweight) for extra 6 weeks' treatment of HFD. Food intake and bodyweight were recorded each week. In the last week, before the mice were sacrificed, fasting blood glucose levels and insulin levels were measured. Furthermore, insulin and glucose tolerance tests were performed. Blood and hepatic lipid levels were examined, the lipid metabolism-associated gene expressions and protein levels in the liver or adipose tissues were assayed after sacrificing all mice. RESULTS Our results demonstrated that a high dose of Danning tablet (3 g/kg) treatment mitigated body weight gain, reduced blood and hepatic cholesterol and triglyceride levels. The morphology analysis showed that Danning tablets could reduce lipid accumulation in both liver and brown adipose tissue. Moreover, Danning tablets could improve fasting blood glucose levels and ameliorate glucose and insulin tolerance in HFD-induced obese mice. Furthermore, qRT-PCR analysis revealed that the mRNA expressions of SREBP-1 and SREBP-2 as well as their target genes were remarkedly down-regulated in the liver and adipose tissue of diet-induced obesity (DIO) mice after treating those mice with Danning tablets. CONCLUSION Our results indicated that Danning tablets could improve the obesity-induced metabolic associated fatty liver disease (MAFLD) and related metabolic disorders. The potential mechanism may probably involve the regulation of the SREBP pathway.
Collapse
Affiliation(s)
- Yujie Ma
- The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai 201203, China
| | - Jinmei Li
- The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai 201203, China
| | - Zhengcai Ju
- The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai 201203, China
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Institute of Diabetes Center, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Zhengtao Wang
- The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai 201203, China
| | - Li Yang
- The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai 201203, China
| | - Lili Ding
- The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The State Administration of TCM (SATCM) Key Laboratory for New Resources and Quality Evaluation of Chinese Medicine, Shanghai Key Laboratory of Complex Prescriptions, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai R&D Center for Standardization of Traditional Chinese Medicines, Shanghai 201203, China.
| |
Collapse
|
34
|
Systematic analysis of the mechanism of Xiaochaihu decoction in hepatitis B treatment via network pharmacology and molecular docking. Comput Biol Med 2021; 138:104894. [PMID: 34607274 DOI: 10.1016/j.compbiomed.2021.104894] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/18/2021] [Accepted: 09/22/2021] [Indexed: 12/18/2022]
Abstract
Hepatitis B (HB) is a globally prevalent infectious disease caused by the HB virus. Xiaochaihu decoction (XCHD) is a classic herbal formula with a long history of clinical application in treating HB. Although the anti-HB activity of XCHD has been reported, systematic research on the exact mechanism of action is lacking. Here, a network pharmacology-based approach was used to predict the active components, important targets, and potential mechanism of XCHD in HB treatment. Investigation included drug-likeness evaluation; absorption, distribution, metabolism, and elimination (ADME) screening; protein-protein interaction (PPI) network construction and cluster analysis; Gene Ontology (GO) analysis; and Kyoto Encyclopedia of Genes and Genomes (KEGG) annotation. Molecular docking was adopted to investigate the interaction between important target proteins and active components. Eighty-seven active components of XCHD and 155 anti-HB targets were selected for further analysis. The GO enrichment and similarity analysis results indicated that XCHD might perform similar or the same GO functions. Glycyrrhizae Radix (GR), one of the seven XCHD herbs, likely exerts some unique GO functions such as the regulation of interleukin-12 production, positive regulation of interleukin-1 beta secretion, and regulation of the I-kappaB/NF-kappaB complex. The PPI network and KEGG pathway analysis results showed that XCHD affects HB mainly through modulating pathways related to viral infection, immunity, cancer, signal transduction, and metabolism. Additionally, molecular docking verified that the active compounds (quercetin, chrysin, and capsaicin) could bind with the key targets. This work systematically explored the anti-HB mechanism of XCHD and provides a novel perspective for future pharmacological research.
Collapse
|
35
|
Yang F, Cao Y, Yu H, Guo Y, Cheng Y, Qian H, Yao W, Xie Y. Transformation and degradation of barbaloin in aqueous solutions and aloe powder under different processing conditions. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.101279] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
36
|
Chen J, Su Y, Lin R, Lin F, Shang P, Hussain R, Shi D. Effects of Acute Diquat Poisoning on Liver Mitochondrial Apoptosis and Autophagy in Ducks. Front Vet Sci 2021; 8:727766. [PMID: 34458360 PMCID: PMC8385319 DOI: 10.3389/fvets.2021.727766] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 07/12/2021] [Indexed: 12/21/2022] Open
Abstract
Diquat (DQ) is an effective herbicide and is widely used in agriculture. Due to persistent and frequent applications, it can enter into aquatic ecosystem and induce toxic effects to exposed aquatic animals. The residues of DQ via food chain accumulate in different tissues of exposed animals including humans and cause adverse toxic effects. Therefore, it is crucial and important to understand the mechanisms of toxic effects of DQ in exposed animals. We used ducks as test specimens to know the effects of acute DQ poisoning on mechanisms of apoptosis and autophagy in liver tissues. Results on comparison of various indexes of visceral organs including histopathological changes, apoptosis, autophagy-related genes, and protein expression indicated the adverse effects of DQ on the liver. The results of our experimental trial showed that DQ induces non-significant toxic effects on pro-apoptotic factors like BAX, BAK1, TNF-α, caspase series, and p53. The results revealed that anti-apoptotic gene Parkin was significantly upregulated, while an upward trend was also observed for Bcl2, suggesting that involvement of the anti-apoptotic factors in ducklings plays an important role in DQ poisoning. Results showed that DQ significantly increased the protein expression level of the autophagy factor Beclin 1 in the liver. Results on key autophagy factors like LC3A, LC3B, and p62 showed an upward trend at gene level, while the protein expression level of both LC3B and p62 reduced that might be associated with process of translation affected by the pro-apoptotic components such as apoptotic protease that inhibits the occurrence of autophagy while initiating cell apoptosis. The above results indicate that DQ can induce cell autophagy and apoptosis and the exposed organism may resist the toxic effects of DQ by increasing anti-apoptotic factors.
Collapse
Affiliation(s)
- Jiaxin Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yalin Su
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Renzhao Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Fei Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Peng Shang
- College of Animal Science, Tibet Agriculture and Animal Husbandry College, Linzhi, China
| | - Riaz Hussain
- Department of Pathology, Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Dayou Shi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
37
|
Nalimu F, Oloro J, Kahwa I, Ogwang PE. Review on the phytochemistry and toxicological profiles of Aloe vera and Aloe ferox. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021; 7:145. [PMID: 34307697 PMCID: PMC8294304 DOI: 10.1186/s43094-021-00296-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/05/2021] [Indexed: 12/11/2022] Open
Abstract
Background Aloe vera and Aloe ferox have over the years been among the most sought-after Aloe species in the treatment of ailments worldwide. This review provides categorized literature on the phytochemical and scientifically proven toxicological profiles of A. vera and A. ferox to facilitate their exploitation in therapy. Main body of the abstract Original full-text research articles were searched in PubMed, ScienceDirect, Research gate, Google Scholar, and Wiley Online Library using specific phrases. Phenolic acids, flavonoids, tannins, and anthraquinones were the main phytochemical classes present in all the two Aloe species. Most of the phytochemical investigations and toxicity studies have been done on the leaves. Aloe vera and Aloe ferox contain unique phytoconstituents including anthraquinones, flavonoids, tannins, sterols, alkaloids, and volatile oils. Aloe vera hydroalcoholic leaf extract showed a toxic effect on Kabir chicks at the highest doses. The methanolic, aqueous, and supercritical carbon dioxide extracts of A. vera leaf gel were associated with no toxic effects. The aqueous leaf extract of A. ferox is well tolerated for short-term management of ailments but long-term administration may be associated with organ toxicity. Long-term administration of the preparations from A. vera leaves and roots was associated with toxic effects. Short conclusion This review provides beneficial information about the phytochemistry and toxicity of A. vera and A. ferox and their potential in the treatment of COVID-19 which up to date has no definite cure. Clinical trials need to be carried out to clearly understand the toxic effects of these species.
Collapse
Affiliation(s)
- Florence Nalimu
- Pharm-Bio Technology and Traditional Medicine Centre of Excellence, Mbarara University of Science and Technology, Mbarara, Uganda.,Department of Pharmaceutical Sciences, Faculty of Medicine, Mbarara University of Science and Technology, P.O. Box 1410, Mbarara, Uganda
| | - Joseph Oloro
- Pharm-Bio Technology and Traditional Medicine Centre of Excellence, Mbarara University of Science and Technology, Mbarara, Uganda.,Department of Pharmacology and Therapeutics, Faculty of Medicine, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Ivan Kahwa
- Pharm-Bio Technology and Traditional Medicine Centre of Excellence, Mbarara University of Science and Technology, Mbarara, Uganda.,Department of Pharmacy, Faculty of Medicine, Mbarara University of Science and Technology, Mbarara, Uganda
| | - Patrick Engeu Ogwang
- Pharm-Bio Technology and Traditional Medicine Centre of Excellence, Mbarara University of Science and Technology, Mbarara, Uganda.,Department of Pharmacy, Faculty of Medicine, Mbarara University of Science and Technology, Mbarara, Uganda
| |
Collapse
|
38
|
Lin FJ, Li H, Wu DT, Zhuang QG, Li HB, Geng F, Gan RY. Recent development in zebrafish model for bioactivity and safety evaluation of natural products. Crit Rev Food Sci Nutr 2021; 62:8646-8674. [PMID: 34058920 DOI: 10.1080/10408398.2021.1931023] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The zebrafish is a species of freshwater fish, popular in aquariums and laboratories. Several advantageous features have facilitated zebrafish to be extensively utilized as a valuable vertebrate model in the lab. It has been well-recognized that natural products possess multiple health benefits for humans. With the increasing demand for natural products in the development of functional foods, nutraceuticals, and natural cosmetics, the zebrafish has emerged as an unprecedented tool for rapidly and economically screening and identifying safe and effective substances from natural products. This review first summarized the key factors for the management of zebrafish in the laboratory, followed by highlighting the current progress on the establishment and applications of zebrafish models in the bioactivity evaluation of natural products. In addition, the zebrafish models used for assessing the potential toxicity or health risks of natural products were involved as well. Overall, this review indicates that zebrafish are promising animal models for the bioactivity and safety evaluation of natural products, and zebrafish models can accelerate the discovery of novel natural products with potential health functions.
Collapse
Affiliation(s)
- Fang-Jun Lin
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China.,Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, USA
| | - Hang Li
- Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
| | - Ding-Tao Wu
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China
| | - Qi-Guo Zhuang
- China-New Zealand Belt and Road Joint Laboratory on Kiwifruit, Sichuan Provincial Academy of Natural Resource Sciences, Chengdu, China
| | - Hua-Bin Li
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Fang Geng
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China
| | - Ren-You Gan
- Key Laboratory of Coarse Cereal Processing (Ministry of Agriculture and Rural Affairs), Sichuan Engineering & Technology Research Center of Coarse Cereal Industralization, Chengdu University, Chengdu, China.,Research Center for Plants and Human Health, Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu, China
| |
Collapse
|
39
|
Fu Q, Pan H, Tang Y, Rong J, Zheng Z. MiR-200a-3p Aggravates DOX-Induced Cardiotoxicity by Targeting PEG3 Through SIRT1/NF-κB Signal Pathway. Cardiovasc Toxicol 2021; 21:302-313. [PMID: 33638775 DOI: 10.1007/s12012-020-09620-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
Doxorubicin (DOX) is a widely used cytotoxic drug whose application is limited by its severe side effects. Little was known regarding how to offset its side effects. Therefore this study aims to explore the role of miR-200a-3p in DOX-induced cardiotoxicity and its possible mechanism. DOX-induced myocardial injury rat models were established, which were then injected with miR-200a-3p inhibitor (miR-200a-3p suppression) to observe the effects of miR-200a-3p on cell proliferation, and apoptosis. Heart function and weights of rat models were also measured. Cardiomyocytes were induced by DOX, in which PEG3 knockdown or corresponding plasmids were transfected to assess the possible effect of PEG3 on cell activity. Dual luciferase reporter assay was applied to verify the binding of PEG3 with miR-200a-3p. Elevated levels of lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB) and left ventricular end-diastolic pressure (LVEDP), as well as suppressed left ventricular systolic pressure (LVSP) and ± dp/dt max were showed in myocardial injury rat models. DOX induced myocardial injury and increased miR-200a-3p expression levels. miR-200a-3p inhibitor could partially attenuate DOX-induced cardiotoxicity in rat models, while PEG3 could regulate myocardial injury in DOX-treated cell models. miR-200a-3p, by targeting PEG3 through SIRT1/NF-κB signal pathway, regulated cell proliferation, inflammation and apoptosis of myocardiocytes. The results in current study demonstrated that miR-200a-3p regulates cell proliferation and apoptosis of cardiomyocytes by targeting PEG3 through SIRT1/NF-κB signal pathway. This result may provide a potential clue for the treatment of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Qinghua Fu
- Department of Cardiovasology, Hunan Provincial People's Hospital, Changsha, Hunan, 410000, People's Republic of China.
| | - Hongwei Pan
- Department of Cardiovasology, Hunan Provincial People's Hospital, Changsha, Hunan, 410000, People's Republic of China
| | - Yi Tang
- Department of Cardiovasology, Hunan Provincial People's Hospital, Changsha, Hunan, 410000, People's Republic of China
| | - Jingjing Rong
- Department of Cardiovasology, Hunan Provincial People's Hospital, Changsha, Hunan, 410000, People's Republic of China
| | - Zhaofen Zheng
- Department of Cardiovasology, Hunan Provincial People's Hospital, Changsha, Hunan, 410000, People's Republic of China
| |
Collapse
|
40
|
Liu Y, Wang Y, Ling X, Yan Z, Wu D, Liu J, Lu G. Effects of Nanoplastics and Butyl Methoxydibenzoylmethane on Early Zebrafish Embryos Identified by Single-Cell RNA Sequencing. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2021; 55:1885-1896. [PMID: 33445878 DOI: 10.1021/acs.est.0c06479] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Nanoplastics with small particle sizes and high surface area/volume ratios easily absorb environmental pollutants and affect their bioavailability. In this study, polystyrene nanoplastic beads (PS-NPBs) with a particle size of 100 nm and butyl methoxydibenzoylmethane (BMDBM) sunscreen in personal-care products were chosen as target pollutants to study their developmental toxicity and interactive effects on zebrafish embryos. The exposure period was set from 2 to 12 h postfertilization (hpf). BMDBM and PS-NPBs significantly upregulated genes related to antioxidant enzymes and downregulated the gene expression of aromatase and DNA methyltransferases, but the influenced genes were not exactly the same. The combined exposure reduced the adverse effects on the expression of all genes. With the help of the single-cell RNA sequencing technology, neural mid cells were identified as the target cells of both pollutants, and brain development, head development, and the notch signaling pathway were the functions they commonly altered. The key genes and functions that are specifically affected by BMDBM and/or PS-NPBs were identified. BMDBM mainly affects the differentiation and fate of neurons in the central nervous system through the regulation of her5, her6, her11, lfng, pax2a, and fgfr4. The PS-NPBs regulate the expression of olig2, foxg1a, fzd8b, six3a, rx1, lhx2b, nkx2.1a, and sfrp5 to alter nervous system development, retinal development, and stem cell differentiation. The phenotypic responses of zebrafish larvae at 120 hpf were tested, and significant inhibition of locomotor activity was found, indicating that early effects on the central nervous system would have a sustained impact on the behavior of zebrafish.
Collapse
Affiliation(s)
- Yuxuan Liu
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China
| | - Yonghua Wang
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China
| | - Xin Ling
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China
| | - Zhenhua Yan
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China
| | - Donghai Wu
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China
| | - Jianchao Liu
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China
| | - Guanghua Lu
- Key Laboratory of Integrated Regulation and Resource Development on Shallow Lake of Ministry of Education, College of Environment, Hohai University, Nanjing 210098, China
| |
Collapse
|
41
|
Rao T, Liu YT, Zeng XC, Li CP, Ou-Yang DS. The hepatotoxicity of Polygonum multiflorum: The emerging role of the immune-mediated liver injury. Acta Pharmacol Sin 2021; 42:27-35. [PMID: 32123300 PMCID: PMC7921551 DOI: 10.1038/s41401-020-0360-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023]
Abstract
Herbal and dietary supplements (HDS)-induced liver injury has been a great concern all over the world. Polygonum multiflorum Thunb., a well-known Chinese herbal medicine, is recently drawn increasing attention because of its hepatotoxicity. According to the clinical and experimental studies, P. multiflorum-induced liver injury (PM-DILI) is considered to be immune-mediated idiosyncratic liver injury, but the role of immune response and the underlying mechanisms are not completely elucidated. Previous studies focused on the direct toxicity of PM-DILI by using animal models with intrinsic drug-induced liver injury (DILI). However, most epidemiological and clinical evidence demonstrate that PM-DILI is immune-mediated idiosyncratic liver injury. The aim of this review is to assess current epidemiological, clinical and experimental evidence about the possible role of innate and adaptive immunity in the idiosyncratic hepatotoxicity of P. multiflorum. The potential effects of factors associated with immune tolerance, including immune checkpoint molecules and regulatory immune cells on the individual's susceptibility to PM-DILI are also discussed. We conclude by giving our hypothesis of possible immune mechanisms of PM-DILI and providing suggestions for future studies on valuable biomarkers identification and proper immune models establishment.
Collapse
Affiliation(s)
- Tai Rao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410008, China.
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China.
| | - Ya-Ting Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410008, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Xiang-Chang Zeng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410008, China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China
| | - Chao-Peng Li
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, 410205, China
| | - Dong-Sheng Ou-Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Pharmacogenetics, Institute of Clinical Pharmacology, Central South University, Changsha, 410008, China.
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Changsha, 410008, China.
- Hunan Key Laboratory for Bioanalysis of Complex Matrix Samples, Changsha, 410205, China.
| |
Collapse
|
42
|
Qiang L, Cheng J. Exposure to polystyrene microplastics impairs gonads of zebrafish (Danio rerio). CHEMOSPHERE 2021; 263:128161. [PMID: 33297137 DOI: 10.1016/j.chemosphere.2020.128161] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 05/23/2023]
Abstract
Microplastic contamination poses a great threat to the health of aquatic species, which may affect their reproduction and result in ecological consequences. There is a need to further elucidate the potential impact microplastics can impose on aquatic species. In this study, the effects of exposure to polystyrene microplastics on reproductive organs, and the underlying response mechanisms, were investigated using zebrafish Danio rerio. After 21 days of continuous waterborne exposure, no significant difference was observed at the lower concentration of 10 μg/L. At concentrations above 100 μg/L, significantly enhanced reactive oxygen species (ROS) level was found in both male and female liver and gonads. At the concentration of 1000 μg/L, significantly increased apoptosis levels were observed in male testes, triggering increased expression of p53-mediated apoptotic pathways; histological alteration in the form of a significant decrease in testis basement membrane thickness was also observed. This study demonstrated that exposure to microplastics can induce molecular responses and histological alterations in fish gonads, implying potential adverse impact on fish reproductive organs. This work provided new insights on the reproductive damage microplastics can cause in fish and have implications in fields of freshwater ecology and environmental toxicology.
Collapse
Affiliation(s)
- Liyuan Qiang
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai, 200062, China
| | - Jinping Cheng
- State Key Laboratory of Estuarine and Coastal Research, East China Normal University, Shanghai, 200062, China; Hong Kong Branch of Southern Marine Science and Engineering Guangdong Lab (Guangzhou) & Department of Ocean Science, School of Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China.
| |
Collapse
|
43
|
Li HY, Yang JB, Li WF, Qiu CX, Hu G, Wang ST, Song YF, Gao HY, Liu Y, Wang Q, Wang Y, Cheng XL, Wei F, Jin HT, Ma SC. In vivo hepatotoxicity screening of different extracts, components, and constituents of Polygoni Multiflori Thunb. in zebrafish (Danio rerio) larvae. Biomed Pharmacother 2020; 131:110524. [PMID: 33152900 DOI: 10.1016/j.biopha.2020.110524] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/07/2020] [Accepted: 07/11/2020] [Indexed: 12/18/2022] Open
Abstract
Polygonum multiflorum Thunb. (PM) is a traditional Chinese medicine, commonly used to treat a variety of diseases. However, the hepatotoxicity associated with PM hampers its clinical application and development. In this study, we refined the zebrafish hepatotoxicity model with regard to the following endpoints: liver size, liver gray value, and the area of yolk sac. The levels of alanine aminotransferase, aspartate transaminase, albumin, and microRNAs-122 were evaluated to verify the model. Subsequently, this model was used to screen different extracts, components, and constituents of PM, including 70 % EtOH extracts of PM, four fractions from macroporous resin (components A, B, C, and D), and 19 compounds from component D. We found that emodin, chrysophanol, emodin-8-O-β-D-glucopyranoside, (cis)-emodin-emodin dianthrones, and (trans)-emodin-emodin dianthrones showed higher hepatotoxicity compared to other components in PM, whereas polyphenols showed lower hepatotoxicity. To the best of our knowledge, this study is the first to identify that dianthrones may account for the hepatotoxicity of PM. We believe that these findings will be helpful in regulating the hepatotoxicity of PM.
Collapse
Affiliation(s)
- Hong-Ying Li
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jian-Bo Yang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Wan-Fang Li
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Cai-Xia Qiu
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Guang Hu
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Shu-Ting Wang
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yun-Fei Song
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Hui-Yu Gao
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Yue Liu
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Qi Wang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Ying Wang
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Xian-Long Cheng
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Feng Wei
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Hong-Tao Jin
- New Drug Safety Evaluation Center, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China; Beijing Union-Genius Pharmaceutical Technology Co. Ltd, Beijing, 100176, China.
| | - Shuang-Cheng Ma
- Institute for Control of Chinese Traditional Medicine and Ethnic Medicine, National Institutes for Food and Drug Control, Beijing, 100050, China.
| |
Collapse
|
44
|
Zhuang T, Gu X, Zhou N, Ding L, Yang L, Zhou M. Hepatoprotection and hepatotoxicity of Chinese herb Rhubarb (Dahuang): How to properly control the "General (Jiang Jun)" in Chinese medical herb. Biomed Pharmacother 2020; 127:110224. [PMID: 32559851 DOI: 10.1016/j.biopha.2020.110224] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 02/06/2023] Open
Abstract
Chinese herb Rhubarb (Dahuang), one of the most widely used traditional Chinese medicine in clinical application for over a thousand years and known as the "General (Jiang Jun)" in Chinese medical herb, currently used clinically for long-term treatment of gastrointestinal diseases and chronic liver diseases. Through previous researches, it has been identified that Rhubarb possessed a good hepatoprotective effect, which primarily protected liver from oxidation, fibrosis and cirrhosis, liver failure, hepatocellular carcinoma and various types of hepatitis. Meanwhile, it has been recently reported that long-term administration of Rhubarb preparation may undertake the risk of liver damage, which has aroused worldwide doubts about the safety of Rhubarb. Therefore, how to correctly understand the "two-way" effect of Rhubarb on liver protection and liver toxicity provides a basis for scientific evaluation of Rhubarb's efficacy on liver and side effects, as well as guiding clinical rational drug use. In this review, the mechanisms of Rhubarb how to play a role in hepatoprotection and why it causes hepatotoxic potential will be elaborated in detail and critically. In addition, some positive clinical guidances are also advised on how to reduce its hepatotoxicity in medical treatment.
Collapse
Affiliation(s)
- Tongxi Zhuang
- Center for Chinese Medicine Therapy and Systems Biology, Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xinyi Gu
- Center for Chinese Medicine Therapy and Systems Biology, Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Nian Zhou
- Center for Chinese Medicine Therapy and Systems Biology, Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lili Ding
- Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li Yang
- Center for Chinese Medicine Therapy and Systems Biology, Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key Laboratory of Complex Prescriptions and MOE Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Mingmei Zhou
- Center for Chinese Medicine Therapy and Systems Biology, Institute for Interdisciplinary Medicine Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
45
|
Yan Y, Shi N, Han X, Li G, Wen B, Gao J. UPLC/MS/MS-Based Metabolomics Study of the Hepatotoxicity and Nephrotoxicity in Rats Induced by Polygonum multiflorum Thunb. ACS OMEGA 2020; 5:10489-10500. [PMID: 32426606 PMCID: PMC7227050 DOI: 10.1021/acsomega.0c00647] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/03/2020] [Indexed: 05/21/2023]
Abstract
Polygonum multiflorum Thunb. (PM) is one of the most frequently used natural products in China. Its hepatotoxicity has been proven and reported. However, chronic PM toxicity is a dynamic process, and a few studies have reported the long-term hepatotoxic mechanism of PM or its nephrotoxicity. To elucidate the mechanism of hepatotoxicity and nephrotoxicity induced by PM after different administration times, different samples from rats were systematically investigated by traditional biochemical analysis, histopathological observation, and nontargeted metabolomics. The concentrations of direct bilirubin (DBIL) at 4 weeks and total bile acid, DBIL, uric acid, and blood urea nitrogen at 8 weeks were significantly increased in the treatment group compared with those in the control group. Approximately, 12 metabolites and 24 proteins were considered as unique toxic biomarkers and targets. Metabolic pathway analysis showed that the primary pathways disrupted by PM were phenylalanine and tyrosine metabolism, which resulted in liver injury, accompanied by chronic kidney injury. As the administration time increased, the toxicity of PM gradually affected vitamin B6, bile acid, and bilirubin metabolism, leading to aggravated liver injury, abnormal biochemical indicators, and marked nephrotoxicity. Our results suggest that the hepatotoxicity and nephrotoxicity caused by PM are both dynamic processes that affect different metabolic pathways at different administration times, which indicated that PM-induced liver and kidney injury should be treated differently in the clinic according to the degree of injury.
Collapse
Affiliation(s)
- Yan Yan
- Dongfang
Hospital, Beijing University of Chinese
Medicine, No. 6 Fangxingyuan 1st Block, Fengtai District, Beijing 100078, P. R.
China
| | - Ning Shi
- Pharmaceutical
Department of Characteristic Medical Center, Strategic Support Force, Beijing 100101, P. R. China
| | - Xuyang Han
- Beijing
Institute of Traditional Chinese Medicine, Beijing Hospital of Traditional
Chinese Medicine, Capital Medical University, Beijing 100010, P. R. China
| | - Guodong Li
- Beijing
University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing 100078, P. R.
China
| | - Binyu Wen
- Dongfang
Hospital, Beijing University of Chinese
Medicine, No. 6 Fangxingyuan 1st Block, Fengtai District, Beijing 100078, P. R.
China
| | - Jian Gao
- Beijing
University of Chinese Medicine, No. 11, Bei San Huan Dong Lu, Chaoyang District, Beijing 100078, P. R.
China
| |
Collapse
|
46
|
Liu DM, Yang D, Zhou CY, Wu JS, Zhang GL, Wang P, Wang F, Meng XL. Aloe-emodin induces hepatotoxicity by the inhibition of multidrug resistance protein 2. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 68:153148. [PMID: 32028185 DOI: 10.1016/j.phymed.2019.153148] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/05/2019] [Accepted: 12/07/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Aloe-emodin (AE) is among the primary bioactive anthraquinones present in traditional Chinese medicinal plants such as Rheum palmatum L. Multidrug resistance protein 2 (ABCC2/ MRP2) is an important efflux transporter of substances associated with cellular oxidative stress. However, the effects of traditional Chinese medicine on this protein remain unclear. PURPOSE The aim of this research is to study the role of ABCC2 in AE-induced hepatotoxicity. METHODS The expression of ABCC2 protein and mRNA levels were analyzed by Western-Blotting and qRT-PCR, respectively. The intracellular oxidative stress caused by AE was evaluated by quantifying the levels of intracellular reactive oxygen species, malondialdehyde, glutathione reduced and oxidized glutathione. The levels of adenosine triphosphate, mitochondrial membrane potential and mitochondrial DNA were explored to evaluate the effects of AE on mitochondrial function. The effects of AE on cell apoptosis and cell cycle were detected by flow cytometry. To further clarify the key role of ABCC2 in AE induced cytotoxicity, we used pCI-neo-ABCC2 plasmid to over express ABCC2 protein, and small interfering RNA was used to knockdown ABCC2 in HepG2 cells. Additionally, we investigated the impact of AE on ABCC2 degradation pathway and the hepatotoxic effects of AE in mice. RESULTS AE was found to inhibit ABCC2 transport activity, downregulate ABCC2 expression and altered intracellular redox balance. Induction of oxidative stress resulted in depletion of intracellular glutathione reduced, mitochondria dysfunction and activation of apoptosis. ABCC2 overexpression significantly reduced AE-induced intracellular oxidative stress and cell death, which was enhanced by ABCC2 knockdown. Furthermore, AE was observed to promote ABCC2 degradation through induction of autophagy and hepatotoxicity was induced in mice by promoting ABCC2 degradation. CONCLUSIONS The inhibition of ABCC2 is a novel effect of AE that triggers oxidative stress and apoptosis. These findings are helpful in understanding the toxicological effects of AE-containing medicinal plants.
Collapse
Affiliation(s)
- De-Ming Liu
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610037, China; Department of Dermatology, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400011, China
| | - Dong Yang
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610037, China
| | - Chun-Yan Zhou
- Department of Dermatology, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400011, China
| | - Jia-Si Wu
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610037, China
| | - Guo-Lin Zhang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
| | - Ping Wang
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610037, China
| | - Fei Wang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China.
| | - Xian-Li Meng
- College Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 610037, China.
| |
Collapse
|
47
|
Orozco-Hernández JM, Gómez Oliván LM, Heredia-García G, Luja-Mondragón M, Islas-Flores H, SanJuan-Reyes N, Galar-Martínez M, García-Medina S, Dublán-García O. Genotoxic and cytotoxic alterations induced by environmentally-relevant concentrations of amoxicillin in blood cells of Cyprinus carpio. CHEMOSPHERE 2019; 236:124323. [PMID: 31319313 DOI: 10.1016/j.chemosphere.2019.07.054] [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: 04/24/2019] [Revised: 06/27/2019] [Accepted: 07/07/2019] [Indexed: 06/10/2023]
Abstract
Amoxicillin (AMX) is a pharmaceutical widely employed in human and veterinary medicine worldwide. Its wide production and use has led to this pharmaceutical being released into the environment in concentrations that range from ng L-1 to μg L-1. Previous studies have demonstrated that this antibiotic generates toxic effects, amongst which alterations to embryonic development and oxidative stress in aquatic organisms, is noteworthy. Nonetheless, it is necessary to characterize the risks that this pharmaceutical represents for species of economic interest such as Cyprinus carpio, in a more precise manner. The aim of this work was to demonstrate if AMX, at environmentally-relevant concentrations, is capable of inducing genotoxic/cytotoxic alterations in C. carpio. In order to evaluate genotoxicity, the comet assay and micronucleus test were used; in order to determine cytotoxic effects, caspase-3 activity and the TUNEL assay were carried out. The results showed that the effects of the biomarkers had their maximum at 72 h; considering the DNA damage in the comet assay, 0.039 μg L-1 resulted in a 29% increase compared to control, and 1.67 μg L-1 caused a 40% increase; micronucleus frequency increased by 205% in C1 and by 311% in C2 when compared to control; compared to control, caspase-3 activity increased 262% in C1 and 787% in C2; for the TUNEL assay, DNA fragmentation increased by 86% in C1 and 120% in C2 compared to control. The results showed that environmentally-relevant concentrations, AMX was capable of generating DNA damage and cytotoxic effects in blood cells of the common carp.
Collapse
Affiliation(s)
- José Manuel Orozco-Hernández
- Laboratorio de Toxicología Ambiental de la Facultad de Química de la Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Colonia Universidad, CP, 50120, Toluca, Estado de México, Mexico
| | - Leobardo Manuel Gómez Oliván
- Laboratorio de Toxicología Ambiental de la Facultad de Química de la Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Colonia Universidad, CP, 50120, Toluca, Estado de México, Mexico.
| | - Gerardo Heredia-García
- Laboratorio de Toxicología Ambiental de la Facultad de Química de la Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Colonia Universidad, CP, 50120, Toluca, Estado de México, Mexico
| | - Marlenee Luja-Mondragón
- Laboratorio de Toxicología Ambiental de la Facultad de Química de la Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Colonia Universidad, CP, 50120, Toluca, Estado de México, Mexico
| | - Hariz Islas-Flores
- Laboratorio de Toxicología Ambiental de la Facultad de Química de la Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Colonia Universidad, CP, 50120, Toluca, Estado de México, Mexico
| | - Nely SanJuan-Reyes
- Laboratorio de Toxicología Acuática del Departamento de Farmacia de la Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Avenida Wilfrido Massieu y Manuel Stampa, Colonia Industrial Vallejo, CDMX, CP, 07700, Mexico
| | - Marcela Galar-Martínez
- Laboratorio de Toxicología Acuática del Departamento de Farmacia de la Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Avenida Wilfrido Massieu y Manuel Stampa, Colonia Industrial Vallejo, CDMX, CP, 07700, Mexico
| | - Sandra García-Medina
- Laboratorio de Toxicología Acuática del Departamento de Farmacia de la Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Avenida Wilfrido Massieu y Manuel Stampa, Colonia Industrial Vallejo, CDMX, CP, 07700, Mexico
| | - Octavio Dublán-García
- Laboratorio de Toxicología Ambiental de la Facultad de Química de la Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan s/n, Colonia Universidad, CP, 50120, Toluca, Estado de México, Mexico
| |
Collapse
|
48
|
Fei X, He Y, Chen J, Man W, Chen C, Sun K, Ding B, Wang C, Xu R. The role of Toll-like receptor 4 in apoptosis of brain tissue after induction of intracerebral hemorrhage. J Neuroinflammation 2019; 16:234. [PMID: 31771613 PMCID: PMC6880548 DOI: 10.1186/s12974-019-1634-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 11/06/2019] [Indexed: 02/08/2023] Open
Abstract
Background Inflammation and apoptosis caused by intracerebral hemorrhage (ICH) are two important factors that affect patient prognosis and survival. Toll-like receptor 4 (TLR4) triggers activation of the inflammatory pathway, causing synthesis and release of inflammatory factors. The inflammatory environment also causes neuronal apoptosis. However, no studies have reported the role of TLR4 in inflammation and apoptosis. Methods We performed survival curve analysis and behavioral scores on TLR4 knockout mice and wild-type mice after inducing ICH. We used TLR4 knockout mice and wild-type mice to make ICH models with type VII collagenase and explored the link between TLR4 in inflammation and apoptosis. We used Western blot to detect the expression of apoptosis-related proteins, inflammatory factors, and their receptors at different time points after ICH induction. The effects of TLR4 on apoptosis were observed by TUNEL, Hoechst, and HE staining techniques. The association with TLR4 in inflammation and apoptosis was explored using IL-1β and TNF-α antagonists. Data conforming to a normal distribution are expressed as mean ± standard deviation. Grade and quantitative data were compared with rank sum test and t test between two groups. P < 0.05 was considered statistically significant. Results TLR4 knockout significantly increased the survival rate of ICH mice. The scores of TLR4 knockout mice were significantly lower than those of wild-type mice. We found that TLR4 knockout mice significantly inhibited apoptosis and the expression of inflammatory factors after the induction of ICH. The apoptosis of ICH-induced mice was significantly improved after injecting IL-1β and TNF-α antagonists. Moreover, the anti-apoptotic effect of the antagonist in wild-type mice is more pronounced. A single injection of the antagonist failed to improve apoptosis in TLR4 knockout mice. Conclusions We conclude that TLR4-induced inflammation after ICH promotes neuronal apoptosis. IL-1β and TNF-α antagonists attenuate this apoptotic effect. Therefore, targeting TLR4 in patients with clinical ICH may attenuate inflammatory response, thereby attenuating apoptosis and improving prognosis.
Collapse
Affiliation(s)
- Xiaowei Fei
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China.,Affiliated Bayi Brain Hospital, General Army Hospital, Beijing, 10000, China.,Dapartment of Physiology, Dalian Medical University, Dalian, 116044, China
| | - Yeting He
- Department of Neurosurgery, Second Affiliated Hospital of Dalian Medical University, Dalian, 116044, China
| | - Jia Chen
- Affiliated BaYi Children's Hospital, Clinical Medical College in The Seventh Medical Center of PLA General Hospital, Southern Medical University, Beijing, China
| | - Weitao Man
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Chen Chen
- Affiliated Bayi Brain Hospital, General Army Hospital, Beijing, 10000, China
| | - Kai Sun
- Affiliated Bayi Brain Hospital, General Army Hospital, Beijing, 10000, China
| | - Boyun Ding
- Affiliated Bayi Brain Hospital, General Army Hospital, Beijing, 10000, China
| | - Chongwu Wang
- Affiliated Bayi Brain Hospital, General Army Hospital, Beijing, 10000, China.
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China. .,Affiliated Bayi Brain Hospital, General Army Hospital, Beijing, 10000, China.
| |
Collapse
|
49
|
Heredia-García G, Gómez-Oliván LM, Orozco-Hernández JM, Luja-Mondragón M, Islas-Flores H, SanJuan-Reyes N, Galar-Martínez M, García-Medina S, Dublán-García O. Alterations to DNA, apoptosis and oxidative damage induced by sucralose in blood cells of Cyprinus carpio. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 692:411-421. [PMID: 31351285 DOI: 10.1016/j.scitotenv.2019.07.165] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/03/2019] [Accepted: 07/11/2019] [Indexed: 06/10/2023]
Abstract
Sucralose (SUC) is an organochlorine that is used as a common sweetener in different dietary products around the world. Its extended use and production have led to this product is released into the environment in concentrations ranging from ng L-1 to μg L-1 in surface waters, groundwaters, wastewater treatment plants and ocean waters. A previous study carried out by our research team demonstrated that SUC is capable of inducing oxidative stress in Cyprinus carpio at environmentally-relevant concentrations. The aim of this study was to evaluate if SUC was capable of inducing alterations to DNA, apoptosis, and oxidative damage in the blood cells of C. carpio. Carps were exposed to two environmentally-relevant concentrations (0.05 and 155 μg L-1) of SUC, and the following biomarkers were determined: comet assay, micronucleus test (MN), caspase-3 activity, TUNEL assay, hydroperoxide content, lipid peroxidation level, protein carbonyl content and superoxide dismutase and catalase activities. Results obtained showed that SUC is capable of inducing DNA damage. A maximum increase of 35% and 23% were observed for c1 and c2, respectively in the comet assay; increases of 586% and 507.7% for c1 and c2, respectively, were found at 72 h through the MN test. The activity of caspase-3 showed a greater response for c1 and c2 at 96 h, with 271% and 493.5%, respectively. TUNEL assay also showed the highest response at 96 h, with 51.8 for c1 and 72.9 for c2; c1 y c2 were able to induce oxidative stress with the highest expression at 72 h. A correlation between DNA damage biomarkers, apoptosis and plasma levels of SUC in both concentrations were observed. With the data obtained, we can conclude that SUC, at environmentally-relevant concentrations, was capable of generating DNA alterations, apoptosis and oxidative stress in blood cells in common carp.
Collapse
Affiliation(s)
- Gerardo Heredia-García
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Leobardo Manuel Gómez-Oliván
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico.
| | - José Manuel Orozco-Hernández
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Marlenee Luja-Mondragón
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Hariz Islas-Flores
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| | - Nely SanJuan-Reyes
- Laboratorio de Toxicología Acuática, Departamento de Farmacia, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Av. Wilfrido Massieu s/n y cerrada Manuel Stampa, Col. Industrial Vallejo, Ciudad de México CP 07700, Mexico
| | - Marcela Galar-Martínez
- Laboratorio de Toxicología Acuática, Departamento de Farmacia, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Av. Wilfrido Massieu s/n y cerrada Manuel Stampa, Col. Industrial Vallejo, Ciudad de México CP 07700, Mexico
| | - Sandra García-Medina
- Laboratorio de Toxicología Acuática, Departamento de Farmacia, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Unidad Profesional Adolfo López Mateos, Av. Wilfrido Massieu s/n y cerrada Manuel Stampa, Col. Industrial Vallejo, Ciudad de México CP 07700, Mexico
| | - Octavio Dublán-García
- Laboratorio de Toxicología Ambiental, Facultad de Química, Universidad Autónoma del Estado de México, Paseo Colón intersección Paseo Tollocan, Colonia Residencial Colón, CP 50120 Toluca, Estado de México, Mexico
| |
Collapse
|
50
|
Cassar S, Adatto I, Freeman JL, Gamse JT, Iturria I, Lawrence C, Muriana A, Peterson RT, Van Cruchten S, Zon LI. Use of Zebrafish in Drug Discovery Toxicology. Chem Res Toxicol 2019; 33:95-118. [PMID: 31625720 DOI: 10.1021/acs.chemrestox.9b00335] [Citation(s) in RCA: 330] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Unpredicted human safety events in clinical trials for new drugs are costly in terms of human health and money. The drug discovery industry attempts to minimize those events with diligent preclinical safety testing. Current standard practices are good at preventing toxic compounds from being tested in the clinic; however, false negative preclinical toxicity results are still a reality. Continual improvement must be pursued in the preclinical realm. Higher-quality therapies can be brought forward with more information about potential toxicities and associated mechanisms. The zebrafish model is a bridge between in vitro assays and mammalian in vivo studies. This model is powerful in its breadth of application and tractability for research. In the past two decades, our understanding of disease biology and drug toxicity has grown significantly owing to thousands of studies on this tiny vertebrate. This Review summarizes challenges and strengths of the model, discusses the 3Rs value that it can deliver, highlights translatable and untranslatable biology, and brings together reports from recent studies with zebrafish focusing on new drug discovery toxicology.
Collapse
Affiliation(s)
- Steven Cassar
- Preclinical Safety , AbbVie , North Chicago , Illinois 60064 , United States
| | - Isaac Adatto
- Stem Cell and Regenerative Biology , Harvard University , Cambridge , Massachusetts 02138 , United States
| | - Jennifer L Freeman
- School of Health Sciences , Purdue University , West Lafayette , Indiana 47907 , United States
| | - Joshua T Gamse
- Drug Safety Evaluation , Bristol-Myers Squibb , New Brunswick , New Jersey 08901 , United States
| | | | - Christian Lawrence
- Aquatic Resources Program , Boston Children's Hospital , Boston , Massachusetts 02115 , United States
| | | | - Randall T Peterson
- Pharmacology and Toxicology, College of Pharmacy , University of Utah , Salt Lake City , Utah 84112 , United States
| | | | - Leonard I Zon
- Stem Cell Program and Division of Hematology/Oncology, Children's Hospital and Dana Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Medical School, Harvard Stem Cell Institute, Stem Cell and Regenerative Biology Department , Harvard University , Boston , Massachusetts 02138 , United States
| |
Collapse
|