1
|
Lou Y, Luan YT, Rong WQ, Gai Y. Corilagin alleviates podocyte injury in diabetic nephropathy by regulating autophagy via the SIRT1-AMPK pathway. World J Diabetes 2024; 15:1916-1931. [PMID: 39280180 PMCID: PMC11372637 DOI: 10.4239/wjd.v15.i9.1916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 06/04/2024] [Accepted: 07/02/2024] [Indexed: 08/27/2024] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is the most frequent chronic microvascular consequence of diabetes, and podocyte injury and malfunction are closely related to the development of DN. Studies have shown that corilagin (Cor) has hepatoprotective, anti-inflammatory, antibacterial, antioxidant, anti-hypertensive, anti-diabetic, and anti-tumor activities. AIM To explore the protective effect of Cor against podocyte injury in DN mice and the underlying mechanisms. METHODS Streptozotocin and a high-fat diet were combined to generate DN mice models, which were then divided into either a Cor group or a DN group (n = 8 in each group). Mice in the Cor group were intraperitoneally injected with Cor (30 mg/kg/d) for 12 wk, and mice in the DN group were treated with saline. Biochemical analysis was used to measure the blood lipid profiles. Hematoxylin and eosin staining was used to detect pathological changes in kidney tissue. Immunohistochemistry and Western blotting were used to assess the protein expression of nephrin and podocin. Mouse podocyte cells (MPC5) were cultured and treated with glucose (5 mmol/L), Cor (50 μM), high glucose (HG) (30 mmol/L), and HG (30 mmol/L) plus Cor (50 μM). Real-time quantitative PCR and Western blotting were performed to examine the effects of Cor on podocyte autophagy. RESULTS Compared with the control group, the DN mice models had increased fasting blood glucose, glycosylated hemoglobin, triglycerides, and total cholesterol, decreased nephrin and podocin expression, increased apoptosis rate, elevated inflammatory cytokines, and enhanced oxidative stress. All of the conditions mentioned above were alleviated after intervention with Cor. In addition, Cor therapy improved SIRT1 and AMPK expression (P < 0.001), inhibited reactive oxygen species and oxidative stress, and elevated autophagy in HG-induced podocytes (P < 0.01). CONCLUSION Cor alleviates podocyte injury by regulating autophagy via the SIRT1-AMPK pathway, thereby exerting its protective impact on renal function in DN mice.
Collapse
Affiliation(s)
- Yu Lou
- Department of Preventive Treatment of Disease, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China
| | - Yu-Ting Luan
- Department of Infectious Diseases, Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China
| | - Wen-Qing Rong
- Department of General Practice (Including Medical Oncology), Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China
| | - Yun Gai
- Department of General Practice (Including Medical Oncology), Seventh People's Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200137, China
| |
Collapse
|
2
|
Zhang X, Tian B, Cong X, Ning Z. Corilagin Alleviates Ang II-Induced Cardiac Fibrosis by Regulating the PTEN/AKT/mTOR Pathway. Dose Response 2024; 22:15593258241261198. [PMID: 39301185 PMCID: PMC11412214 DOI: 10.1177/15593258241261198] [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: 02/08/2024] [Accepted: 05/27/2024] [Indexed: 09/22/2024] Open
Abstract
This research aimed to evaluate the therapeutic effect of corilagin (Cor) against angiotensin II (Ang II)-induced cardiac fibrosis and its underlying mechanisms. C57BL/6 mice (male, 8-10 weeks) received saline or Ang II (2.0 mg/kg/day) via subcutaneous infusion and intraperitoneal injection of Cor (30 mg/kg) for 28 days. Ang II induction increased the fibrotic area, whereas Cor treatment inhibited the fibrotic area significantly. Cor markedly reduced the Ang II-induced cardiac fibroblasts. Cor significantly inhibited Ang II-induced increase in expressions of smooth muscle alpha-actin (α-SMA), collagen I, collagen III, transforming growth factor beta 1 (TGF-β1), fibronectin, and connective tissue growth factor (CTGF). Cor suppressed the intracellular reactive oxygen species (ROS) production. Cor therapy reduced Ang II-induced malondialdehyde (MDA) content, whereas superoxide dismutase (SOD) and catalase (CAT) activities were increased (all, P < .001). Moreover, Ang II induction elevated the expression of phosphorylated phosphatase and tensin homolog (p-PTEN), phosphorylated protein kinase B (p-AKT) (Ser473) and phosphorylated mammalian target of rapamycin (p-mTOR) (Ser 2448), whereas Cor reduced their expressions. Cor treatment inhibited the migration ability of the cardiac fibroblast, whereas a PTEN inhibitor, VO-ohpic, increased the migration capability. Cor could have a protective effect against Ang II-induced cardiac fibrosis via inhibition of the PTEN/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Xiaogang Zhang
- Department of Cardiology, Shanghai Pudong New Area Zhoupu Hospital (Shanghai Health Medical College Affiliated Zhoupu Hospital), Shanghai, China
| | - Bei Tian
- Department of Nursing, Shanghai Pudong New Area Zhoupu Hospital (Shanghai Health Medical College Affiliated Zhoupu Hospital), Shanghai, China
| | - Xinpeng Cong
- Department of Cardiology, Shanghai Pudong New Area Zhoupu Hospital (Shanghai Health Medical College Affiliated Zhoupu Hospital), Shanghai, China
| | - Zhongping Ning
- Department of Cardiology, Shanghai Pudong New Area Zhoupu Hospital (Shanghai Health Medical College Affiliated Zhoupu Hospital), Shanghai, China
| |
Collapse
|
3
|
Xue X, Li Y, Yao Y, Zhang S, Peng C, Li Y. A comprehensive review of miR-21 in liver disease: Big impact of little things. Int Immunopharmacol 2024; 134:112116. [PMID: 38696909 DOI: 10.1016/j.intimp.2024.112116] [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: 02/21/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 05/04/2024]
Abstract
microRNAs (miRNAs), a class of non-coding RNA with 20-24 nucleotides, are defined as the powerful regulators for gene expression. miR-21 is a multifunctional miRNA enriched in the circulatory system and multiple organs, which not only serves as a non-invasive biomarker in disease diagnosis, but also participates in many cellular activities. In various chronic liver diseases, the increase of miR-21 affects glycolipid metabolism, viral infection, inflammatory and immune cell activation, hepatic stellate cells activation and tissue fibrosis, and autophagy. Moreover, miR-21 is also a liaison in the deterioration of chronic liver disease to hepatocellular carcinoma (HCC), and it impacts on cell proliferation, apoptosis, migration, invasion, angiogenesis, immune escape, and epithelial-mesenchymal transformation by regulating target genes expression in different signaling pathways. In current research on miRNA therapy, some natural products can exert the hepatoprotective effects depending on the inhibition of miR-21 expression. In addition, miR-21-based therapeutic also play a role in regulating intracellular miR-21 levels and enhancing the efficacy of chemotherapy drugs. Herein, we systemically summarized the recent progress of miR-21 on biosynthesis, biomarker function, molecular mechanism and miRNA therapy in chronic liver disease and HCC, and looked forward to outputting some information to enable it from bench to bedside.
Collapse
Affiliation(s)
- Xinyan Xue
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yanzhi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuxin Yao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shenglin Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
4
|
Wang Y, Li Y, Chen Y, Mao J, Ji J, Zhang S, Liu P, Pronyuk K, Fisher D, Dang Y, Zhao L. Corilagin relieves atherosclerosis via the toll-like receptor 4 signaling pathway in vascular smooth muscle cells. Int J Immunopathol Pharmacol 2024; 38:3946320241254083. [PMID: 38869980 PMCID: PMC11179462 DOI: 10.1177/03946320241254083] [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: 09/13/2023] [Accepted: 04/24/2024] [Indexed: 06/15/2024] Open
Abstract
INTRODUCTION Corilagin possesses a diverse range of pharmacologic bioactivities. However, the specific protective effects and mechanisms of action of corilagin in the context of atherosclerosis remain unclear. In this study, we investigated the impact of corilagin on the toll-like receptor (TLR)4 signaling pathway in a mouse vascular smooth muscle cell line (MOVAS) stimulated by oxidized low-density lipoprotein (ox-LDL). Additionally, we examined the effects of corilagin in Sprague-Dawley rats experiencing atherosclerosis. METHODS The cytotoxicity of corilagin was assessed using the CCK8 assay. MOVAS cells, pre-incubated with ox-LDL, underwent treatment with varying concentrations of corilagin. TLR4 expression was modulated by either downregulation through small interfering (si)RNA or upregulation via lentivirus transfection. Molecular expression within the TLR4 signaling pathway was analyzed using real-time polymerase chain reaction (PCR) and Western blotting. The proliferation capacity of MOVAS cells was determined through cell counting. In a rat model, atherosclerosis was induced in femoral arteries using an improved guidewire injury method, and TLR4 expression in plaque areas was assessed using immunofluorescence. Pathological changes were examined through hematoxylin and eosin staining, as well as Oil-Red-O staining. RESULTS Corilagin demonstrated inhibitory effects on the TLR4 signaling pathway in MOVAS cells pre-stimulated with ox-LDL, consequently impeding the proliferative impact of ox-LDL. The modulation of TLR4 expression, either through downregulation or upregulation, similarly influenced the expression of downstream molecules. In an in vivo context, corilagin exhibited the ability to suppress TLR4 and MyD88 expression in the plaque lesion areas of rat femoral arteries, thereby alleviating the formation of atherosclerotic plaques. CONCLUSION Corilagin can inhibit the TLR4 signaling pathway in VSMCs, possibly by downregulating TLR4 expression and, consequently, relieving atherosclerosis.
Collapse
MESH Headings
- Animals
- Toll-Like Receptor 4/metabolism
- Hydrolyzable Tannins/pharmacology
- Rats, Sprague-Dawley
- Signal Transduction/drug effects
- Atherosclerosis/drug therapy
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Lipoproteins, LDL/metabolism
- Male
- Glucosides/pharmacology
- Glucosides/therapeutic use
- Mice
- Cell Line
- Rats
- Cell Proliferation/drug effects
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Disease Models, Animal
- Myeloid Differentiation Factor 88/metabolism
Collapse
Affiliation(s)
- Yujie Wang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunfei Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinqian Mao
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyu Ji
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shaojun Zhang
- National & Local Joint Engineering Research Centre for High-Throughput Drug Screening Technology, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, China
| | - Pan Liu
- Department of Pediatrics, Wuchang Hospital, Wuhan, China
| | - Khrystyna Pronyuk
- Department of Infectious Diseases, Bogomolets National Medical University, Kyiv, Ukraine
| | - David Fisher
- Department of Medical Biosciences, Faculty of Natural Sciences, University of the Western Cape, Cape Town, South Africa
- School of Health Professions, University of Missouri, Columbia, MO, USA
| | - Yiping Dang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Sabir U, Gu HM, Zhang DW. Extracellular matrix turnover: phytochemicals target and modulate the dual role of matrix metalloproteinases (MMPs) in liver fibrosis. Phytother Res 2023; 37:4932-4962. [PMID: 37461256 DOI: 10.1002/ptr.7959] [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: 03/31/2023] [Revised: 06/05/2023] [Accepted: 07/02/2023] [Indexed: 11/10/2023]
Abstract
Extracellular matrix (ECM) resolution by matrix metalloproteinases (MMPs) is a well-documented mechanism. MMPs play a dual and complex role in modulating ECM degradation at different stages of liver fibrosis, depending on the timing and levels of their expression. Increased MMP-1 combats disease progression by cleaving the fibrillar ECM. Activated hepatic stellate cells (HSCs) increase expression of MMP-2, -9, and -13 in different chemicals-induced animal models, which may alleviate or worsen disease progression based on animal models and the stage of liver fibrosis. In the early stage, elevated expression of certain MMPs may damage surrounding tissue and activate HSCs, promoting fibrosis progression. At the later stage, downregulation of MMPs can facilitate ECM accumulation and disease progression. A number of phytochemicals modulate MMP activity and ECM turnover, alleviating disease progression. However, the effects of phytochemicals on the expression of different MMPs are variable and may depend on the disease models and stage, and the dosage, timing and duration of phytochemicals used in each study. Here, we review the most recent advances in the role of MMPs in the effects of phytochemicals on liver fibrogenesis, which indicates that further studies are warranted to confirm and define the potential clinical efficacy of these phytochemicals.
Collapse
Affiliation(s)
- Usman Sabir
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Hong-Mei Gu
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Da-Wei Zhang
- Department of Pediatrics and Group on the Molecular and Cell Biology of Lipids, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
6
|
Sato H, Hara T, Meng S, Tsuji Y, Arao Y, Saito Y, Sasaki K, Kobayashi S, Doki Y, Eguchi H, Ishii H. Multifaced roles of desmoplastic reaction and fibrosis in pancreatic cancer progression: Current understanding and future directions. Cancer Sci 2023; 114:3487-3495. [PMID: 37480223 PMCID: PMC10475783 DOI: 10.1111/cas.15890] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/28/2023] [Accepted: 06/06/2023] [Indexed: 07/23/2023] Open
Abstract
Desmoplastic reaction is a fibrosis reaction that is characterized by a large amount of dense extracellular matrix (ECM) and dense fibrous stroma. Fibrotic stroma around the tumor has several different components, including myofibroblasts, collagen, and other ECM molecules. This stromal reaction is a natural response to the tissue injury process, and fibrosis formation is a key factor in pancreatic cancer development. The fibrotic stroma of pancreatic cancer is associated with tumor progression, metastasis, and poor prognosis. Reportedly, multiple processes are involved in fibrosis, which is largely associated with the upregulation of various cytokines, chemokines, matrix metalloproteinases, and other growth factors that promote tumor growth and metastasis. Fibrosis is also associated with immunosuppressive cell recruitment, such as regulatory T cells (Tregs) with suppressing function to antitumor immunity. Further, dense fibrosis restricts the flow of nutrients and oxygen to the tumor cells, which can contribute to drug resistance. Furthermore, the dense collagen matrix can act as a physical barrier to block the entry of drugs into the tumor, thereby further contributing to drug resistance. Thus, understanding the mechanism of desmoplastic reaction and fibrosis in pancreatic cancer will open an avenue to innovative medicine and improve the prognosis of patients suffering from this disease.
Collapse
Grants
- 17cm0106414h0002 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology
- JP21lm0203007 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology
- 18KK0251 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology
- 19K2265 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology
- 20H00541 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology
- 21K19526 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology
- 22H03146 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology
- 22K19559 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology
- 16H06279 Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology
- Mitsubishi Foundation
- Mitsubishi Foundation
Collapse
Affiliation(s)
- Hiromichi Sato
- Department of Medical Data ScienceCenter of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineOsakaJapan
- Department of Gastrointestinal SurgeryOsaka University Graduate School of MedicineOsakaJapan
| | - Tomoaki Hara
- Department of Medical Data ScienceCenter of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineOsakaJapan
| | - Sikun Meng
- Department of Medical Data ScienceCenter of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineOsakaJapan
| | - Yoshiko Tsuji
- Department of Medical Data ScienceCenter of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineOsakaJapan
| | - Yasuko Arao
- Department of Medical Data ScienceCenter of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineOsakaJapan
| | - Yoshiko Saito
- Department of Medical Data ScienceCenter of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineOsakaJapan
| | - Kazuki Sasaki
- Department of Medical Data ScienceCenter of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineOsakaJapan
- Department of Gastrointestinal SurgeryOsaka University Graduate School of MedicineOsakaJapan
| | - Shogo Kobayashi
- Department of Gastrointestinal SurgeryOsaka University Graduate School of MedicineOsakaJapan
| | - Yuichiro Doki
- Department of Gastrointestinal SurgeryOsaka University Graduate School of MedicineOsakaJapan
| | - Hidetoshi Eguchi
- Department of Gastrointestinal SurgeryOsaka University Graduate School of MedicineOsakaJapan
| | - Hideshi Ishii
- Department of Medical Data ScienceCenter of Medical Innovation and Translational ResearchOsaka University Graduate School of MedicineOsakaJapan
| |
Collapse
|
7
|
Wang M, Cao L. Hydrolysable tannins as a potential therapeutic drug for the human fibrosis-associated disease. Drug Dev Res 2023; 84:1096-1113. [PMID: 37386756 DOI: 10.1002/ddr.22089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 05/22/2023] [Accepted: 06/06/2023] [Indexed: 07/01/2023]
Abstract
Fibrosis is a pathological change with abnormal tissue regeneration due to a response to persistent injury, which is extensively related to organ damage and failure, leading to high morbidity and mortality worldwide. Although the pathogenesis of fibrosis has been comprehensively elucidated, there are few effective therapies for treating fibrotic diseases. Natural products are increasingly regarded as an effective strategy for fibrosis with numerous favorable functions. Hydrolysable tannins (HT) are a type of natural products that have the potential to treat the fibrotic disease. In this review, we describe some biological activities and the therapeutic prospects of HT in organ fibrosis. Furthermore, the underlying mechanisms of inhibition of HT on fibrotic organs in relation to inflammation, oxidative stress, epithelial-mesenchymal transition, fibroblast activation and proliferation, and extracellular matrix accumulation are discussed. Understanding the mechanism of HT against fibrotic diseases will provide a new strategy for the prevention and attenuation of fibrosis progression.
Collapse
Affiliation(s)
- Meiwei Wang
- The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Linghui Cao
- The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| |
Collapse
|
8
|
Shan L, Wang F, Zhai D, Meng X, Liu J, Lv X. Matrix metalloproteinases induce extracellular matrix degradation through various pathways to alleviate hepatic fibrosis. Biomed Pharmacother 2023; 161:114472. [PMID: 37002573 DOI: 10.1016/j.biopha.2023.114472] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/20/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Liver fibrosis is the common consequence of various chronic liver injuries and is mainly characterized by the imbalance between the production and degradation of extracellular matrix, which leads to the accumulation of interstitial collagen and other matrix components. Matrix metalloproteinases (MMPs) and their specific inhibitors, that is, tissue inhibitors of metalloproteinases (TIMPs), play a crucial role in collagen synthesis and lysis. Previous in vivo and in vitro studies of our laboratory found repressing extracellular matrix (ECM) accumulation by restoring the balance between MMPs and TIMPs can alleviate liver fibrosis. We conducted a review of articles published in PubMed and Science Direct in the last decade until February 1, 2023, which were searched for using these words "MMPs/TIMPs" and "Hepatic Fibrosis." Through a literature review, this article reviews the experimental studies of liver fibrosis based on MMPs/TIMPs, summarizes the components that may exert an anti-liver fibrosis effect by affecting the expression or activity of MMPs/TIMPs, and attempts to clarify the mechanism of MMPs/TIMPs in regulating collagen homeostasis, so as to provide support for the development of anti-liver fibrosis drugs. We found the MMP-TIMP-ECM interaction can result in better understanding of the pathogenesis and progression of hepatic fibrosis from a different angle, and targeting this interaction may be a promising therapeutic strategy for hepatic fibrosis. Additionally, we summarized and analyzed the drugs that have been found to reduce liver fibrosis by changing the ratio of MMPs/TIMPs, including medicine natural products.
Collapse
Affiliation(s)
- Liang Shan
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China; The Key Laboratory of Major Autoimmune Diseases, Hefei 230032, Anhui, China
| | - Fengling Wang
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China
| | - Dandan Zhai
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China
| | - Xiangyun Meng
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China
| | - Jianjun Liu
- Department of Pharmacy, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui 230011, China.
| | - Xiongwen Lv
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Medical University, Hefei 230032, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei 230032, China; The Key Laboratory of Major Autoimmune Diseases, Hefei 230032, Anhui, China.
| |
Collapse
|
9
|
Dong BS, Liu FQ, Yang WN, Li XD, Shi MJ, Li MR, Yan XL, Zhang H. Huangqi Decoction, a compound Chinese herbal medicine, inhibits the proliferation and activation of hepatic stellate cells by regulating the long noncoding RNA-C18orf26-1/microRNA-663a/transforming growth factor-β axis. JOURNAL OF INTEGRATIVE MEDICINE 2023; 21:47-61. [PMID: 36456413 DOI: 10.1016/j.joim.2022.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 10/31/2022] [Indexed: 11/17/2022]
Abstract
OBJECTIVE Huangqi Decoction (HQD), a classical traditional Chinese medicine formula, has been used as a valid treatment for alleviating liver fibrosis; however, the underlying molecular mechanism is still unknown. Although our previous studies showed that microRNA-663a (miR-663a) suppresses the proliferation and activation of hepatic stellate cells (HSCs) and the transforming growth factor-β/small mothers against decapentaplegic (TGF-β/Smad) pathway, whether long noncoding RNAs (lncRNAs) are involved in HSC activation via the miR-663a/TGF-β/Smad signaling pathway has not yet reported. The present study aimed to investigate the roles of lncRNA lnc-C18orf26-1 in the activation of HSCs and the mechanism by which HQD inhibits hepatic fibrosis. METHODS The expression levels of lnc-C18orf26-1, miR-663a and related genes were measured by quantitative reverse transcription-polymerase chain reaction. HSCs were transfected with the miR-663a mimic or inhibitor and lnc-C18orf26-1 small interfering RNAs. The water-soluble tetrazolium salt-1 assay was used to assess the proliferation rate of HSCs. Changes in lncRNA expression were evaluated in miR-663a-overexpressing HSCs by using microarray to identify miR-663a-regulated lncRNAs. RNA hybrid was used to predict the potential miR-663a binding sites on lncRNAs. Luciferase reporter assays further confirmed the interaction between miR-663a and the lncRNA. The expression levels of collagen α-2(I) chain (COL1A2), α-smooth muscle actin (α-SMA) and TGF-β/Smad signaling pathway-related proteins were determined using Western blotting. RESULTS Lnc-C18orf26-1 was upregulated in TGF-β1-activated HSCs and competitively bound to miR-663a. Knockdown of lnc-C18orf26-1 inhibited HSC proliferation and activation, downregulated TGF-β1-stimulated α-SMA and COL1A2 expression, and inhibited the TGF-β1/Smad signaling pathway. HQD suppressed the proliferation and activation of HSCs. HQD increased miR-663a expression and decreased lnc-C18orf26-1 expression in HSCs. Further studies showed that HQD inhibited the expression of COL1A2, α-SMA, TGF-β1, TGF-β type I receptor (TGF-βRI) and phosphorylated Smad2 (p-Smad2) in HSCs, and these effects were reversed by miR-663a inhibitor treatment. CONCLUSION Our study identified lnc-C18orf26-1 and miR-663a as promising therapeutic targets for hepatic fibrosis. HQD inhibits HSC proliferation and activation at least partially by regulating the lnc-C18orf26-1/miR-663a/TGF-β1/TGF-βRI/p-Smad2 axis.
Collapse
Affiliation(s)
- Ben-Sheng Dong
- Traditional Chinese Medicine Epigenomics Research Center, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fu-Qun Liu
- Department of Rheumatology and Immunology, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing 211299, Jiangsu Province, China; Department of Rheumatology and Immunology, Yangzhou University Medical College, Yangzhou 225000, Jiangsu Province, China
| | - Wen-Na Yang
- Department of Rheumatology and Immunology, Nanjing Lishui District Hospital of Traditional Chinese Medicine, Nanjing 211299, Jiangsu Province, China; Department of Rheumatology and Immunology, Yangzhou University Medical College, Yangzhou 225000, Jiangsu Province, China
| | - Xiao-Dong Li
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University, Kita-gun, Kagawa 761-0793, Japan
| | - Miao-Juan Shi
- Traditional Chinese Medicine Epigenomics Research Center, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mao-Rong Li
- Traditional Chinese Medicine Epigenomics Research Center, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiu-Li Yan
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200437, China.
| | - Hui Zhang
- Traditional Chinese Medicine Epigenomics Research Center, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
10
|
Shams F, Azeem A, Shams A, Tawab A, Rehman S, Tariq S, Latief N, Ijaz B. Flavonoid rich extract of Trigonella foenum-graecum leaves ameliorate liver fibrosis. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
11
|
Liao M, Zhang R, Wang Y, Mao Z, Wu J, Guo H, Zhang K, Jing Y, Zhang C, Song H, Chen X, Wei G. Corilagin prevents non-alcoholic fatty liver disease via improving lipid metabolism and glucose homeostasis in high fat diet-fed mice. Front Nutr 2022; 9:983450. [PMID: 36071929 PMCID: PMC9443665 DOI: 10.3389/fnut.2022.983450] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 07/27/2022] [Indexed: 12/19/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has been considered to be one of the most common chronic liver diseases. However, no validated pharmacological therapies have been officially proved in clinic due to its complex pathogenesis. The purpose of this study was to examine the protective effects of Corilagin (referred to Cori) against NAFLD in mice under a high fat diet (HFD) condition. Mice were fed either a normal control diet (NCD) or HFD with or without Cori (5 or 10 mg/kg body weight) for 15 weeks. In our results, Cori treatment significantly attenuated HFD-induced hepatic steatosis, high NAFLD activity score (NAD) and liver injury. Consistently, Cori treatment remarkably alleviated HFD-induced hepatic lipid accumulation (e.g., triglycerides (TG) and total cholesterol (TC) contents in liver), and improved plasma lipid concentrations (e.g., plasma TG, TC, low-density lipoprotein cholesterol (LDL-c), high-density lipoprotein cholesterol (HDL-c)). Moreover, Cori treatment ameliorated NAFLD associated metabolic disorders such as glucose intolerance and insulin resistance in HFD-fed mice. Additionally, Cori treatment dramatically changed HFD-induced liver gene expression profiles, and identified overlapped differentially expressed genes (DEGs) between NCD vs. HFD group and HFD vs. HCR (high fat diet plus treatment with Cori) group. With these DEGs, we observed a marked enrichment of Gene Ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, which were closely associated with the metabolic balance in liver. Particularly, we found several potential hub proteins against NAFLD development with analyses of protein-protein interaction (PPI) network and qPCR assays. Collectively, our results revealed the important protective effects of Cori against the progress of NAFLD, which was probably mediated through improving dysregulated lipid metabolism and insulin resistance in HFD-fed mice. Additionally, Cori-dependent overlapped DEGs might serve as a featured NAFLD-associated gene expression signature for the diagnosis, treatment, as well as drug discovery and development of NAFLD in the near future.
Collapse
Affiliation(s)
- Mingjuan Liao
- Department of Traditional Chinese Medicine, The Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rong Zhang
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongling Wang
- Department of Traditional Chinese Medicine, The Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Yongling Wang,
| | - Ziming Mao
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Wu
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huaqi Guo
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kaiwen Zhang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Jing
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Caoxu Zhang
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huaidong Song
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Chen
- Department of Endocrinology, Shanghai Gongli Hospital, Shanghai, China
- Xia Chen,
| | - Gang Wei
- Department of Traditional Chinese Medicine, The Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The Core Laboratory in Medical Center of Clinical Research, Department of Molecular Diagnostics & Endocrinology, Shanghai Ninth People’s Hospital, State Key Laboratory of Medical Genomics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Key Laboratory of Pollution Exposure and Health Intervention of Zhejiang Province, Hangzhou, China
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- *Correspondence: Gang Wei,
| |
Collapse
|
12
|
Yan F, Cheng D, Wang H, Gao M, Zhang J, Cheng H, Wang C, Zhang H, Xiong H. Corilagin Ameliorates Con A-Induced Hepatic Injury by Restricting M1 Macrophage Polarization. Front Immunol 2022; 12:807509. [PMID: 35095894 PMCID: PMC8792905 DOI: 10.3389/fimmu.2021.807509] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 12/22/2021] [Indexed: 12/24/2022] Open
Abstract
Immune-mediated hepatic injury plays a key role in the initiation and pathogenesis of diverse liver diseases. However, treatment choice for immune-mediated hepatic injury remains limited. Corilagin, a natural ellagitannin extracted from various traditional Chinese medicines, has been demonstrated to exhibit multiple pharmacological activities, such as anti-inflammatory, anti-tumor, and hepatoprotective properties. The present study aimed to investigate the effects of corilagin on immune-mediated hepatic injury using a murine model of concanavalin A (Con A)-induced hepatitis, which is well-characterized to study acute immune-mediated hepatitis. Herein, mice were administered corilagin (25 mg/kg) intraperitoneally twice at 12 h intervals, and 1 h later, the mice were challenged with Con A (20 mg/kg body weight); serum and liver samples were collected after 12 h. The results showed that corilagin significantly increased the survival of mice and reduced serum alanine transaminase (ALT) and aspartate aminotransferase (AST) levels. In addition, corilagin markedly improved histopathological damage, hepatocyte apoptosis, and oxidative stress in the liver. The activation of M1 macrophages in the hepatic mononuclear cells was also significantly reduced compared with that in the control group. The expression of M1 macrophage-associated proinflammatory cytokines and genes, including interleukin (IL)-6, IL-12, and inducible nitric oxide synthase (iNOS), was also decreased after corilagin treatment. Finally, the results demonstrated that corilagin regulated macrophage polarization by modulating the mitogen-activated protein kinases (MAPK), nuclear factor (NF)-κB, and interferon regulatory factor (IRF) signaling pathways. Thus, the findings indicate that corilagin protects mice from Con A-induced immune-mediated hepatic injury by limiting M1 macrophage activation via the MAPK, NF-κB, and IRF signaling pathways, suggesting corilagin as a possible treatment choice for immune-mediated hepatic injury.
Collapse
Affiliation(s)
- Fenglian Yan
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China.,Jining Key Laboratory of Immunology, Jining Medical University, Jining, China
| | - Dalei Cheng
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China.,Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haiyan Wang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China.,Jining Key Laboratory of Immunology, Jining Medical University, Jining, China
| | - Min Gao
- Clinical Laboratory, Jining First People's Hospital, Jining, China
| | - Junfeng Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China.,Jining Key Laboratory of Immunology, Jining Medical University, Jining, China
| | - Hongyan Cheng
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China.,Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Changying Wang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China.,Jining Key Laboratory of Immunology, Jining Medical University, Jining, China
| | - Hui Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China.,Jining Key Laboratory of Immunology, Jining Medical University, Jining, China
| | - Huabao Xiong
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China.,Jining Key Laboratory of Immunology, Jining Medical University, Jining, China
| |
Collapse
|
13
|
Jia S, He D, Liang X, Cheng P, Liu J, Chen M, Wang C, Zhang H, Meng C. Corilagin induces apoptosis and inhibits autophagy of HL‑60 cells by regulating miR‑451/HMGB1 axis. Mol Med Rep 2021; 25:34. [PMID: 34850958 PMCID: PMC8669704 DOI: 10.3892/mmr.2021.12550] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 08/12/2021] [Indexed: 11/17/2022] Open
Abstract
Corilagin is the primary active component of the Euphorbia phyllanthus plant and has significant anti-cancer properties. However, the biological effects and mechanisms of corilagin on acute myeloid leukemia (AML) have not been clarified. The Cell Counting Kit-8 and Carboxyfluorescein Diacetate Succinimidyl Ester assay results showed that corilagin significantly inhibited proliferation of the AML cell line HL-60 in a time- and dose-dependent manner. Western blotting and flow cytometry analysis were performed to determine the levels of apoptosis in HL-60 cells. The protein levels of cleaved caspase-3 and Bak were upregulated, while Bcl-xl was downregulated in cells treated with corilagin. The percentage of early- and late-stage apoptotic cells increased following corilagin treatment in a dose-dependent manner, indicating that the intrinsic mitochondrial apoptosis pathway was activated by corilagin. Simultaneously, western blotting and immunofluorescence results revealed that autophagy was suppressed; this was accompanied by a decrease in light chain 3-II (LC3-II) conversion and autophagosomes. MicroRNA (miRNA/miR) profile analysis showed that corilagin elevated the expression of the tumor suppressor miR-451, while the mRNA and protein levels of high mobility group protein B1 (HMGB1), the target of miR-451, decreased following exposure to corilagin. Knockdown of miR-451 decreased the downregulation of HMGB1 caused by corilagin, indicating negative regulation of HMGB1 by miR-451 during corilagin treatment. Furthermore, knockdown of miR-451 also attenuated corilagin-induced proliferation inhibition of HL-60 cells, implying that miR-451 was essential for the proliferation inhibitory effect of corilagin. In conclusion, these results indicated that corilagin induced apoptosis and inhibited autophagy in HL-60 cells by regulating the miR-451/HMGB1 axis, and corilagin may be a novel therapeutic drug for the treatment of AML.
Collapse
Affiliation(s)
- Shu Jia
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Dongye He
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Xiao Liang
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Panpan Cheng
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Jilan Liu
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Mingtai Chen
- Medical Research Center, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Cuiling Wang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Hao Zhang
- Department of Hematology, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Chunyang Meng
- Department of Spine Surgery, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| |
Collapse
|
14
|
Su Z, Guo Y, Huang X, Feng B, Tang L, Zheng G, Zhu Y. Phytochemicals: Targeting Mitophagy to Treat Metabolic Disorders. Front Cell Dev Biol 2021; 9:686820. [PMID: 34414181 PMCID: PMC8369426 DOI: 10.3389/fcell.2021.686820] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/02/2021] [Indexed: 12/21/2022] Open
Abstract
Metabolic disorders include metabolic syndrome, obesity, type 2 diabetes mellitus, non-alcoholic fatty liver disease and cardiovascular diseases. Due to unhealthy lifestyles such as high-calorie diet, sedentary and physical inactivity, the prevalence of metabolic disorders poses a huge challenge to global human health, which is the leading cause of global human death. Mitochondrion is the major site of adenosine triphosphate synthesis, fatty acid β-oxidation and ROS production. Accumulating evidence suggests that mitochondrial dysfunction-related oxidative stress and inflammation is involved in the development of metabolic disorders. Mitophagy, a catabolic process, selectively degrades damaged or superfluous mitochondria to reverse mitochondrial dysfunction and preserve mitochondrial function. It is considered to be one of the major mechanisms responsible for mitochondrial quality control. Growing evidence shows that mitophagy can prevent and treat metabolic disorders through suppressing mitochondrial dysfunction-induced oxidative stress and inflammation. In the past decade, in order to expand the range of pharmaceutical options, more and more phytochemicals have been proven to have therapeutic effects on metabolic disorders. Many of these phytochemicals have been proved to activate mitophagy to ameliorate metabolic disorders. Given the ongoing epidemic of metabolic disorders, it is of great significance to explore the contribution and underlying mechanisms of mitophagy in metabolic disorders, and to understand the effects and molecular mechanisms of phytochemicals on the treatment of metabolic disorders. Here, we investigate the mechanism of mitochondrial dysfunction in metabolic disorders and discuss the potential of targeting mitophagy with phytochemicals for the treatment of metabolic disorders, with a view to providing a direction for finding phytochemicals that target mitophagy to prevent or treat metabolic disorders.
Collapse
Affiliation(s)
- Zuqing Su
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yanru Guo
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xiufang Huang
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bing Feng
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lipeng Tang
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangjuan Zheng
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Zhu
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
15
|
Liu J, Qin X, Ma W, Jia S, Zhang X, Yang X, Pan D, Jin F. Corilagin induces apoptosis and autophagy in NRF2‑addicted U251 glioma cell line. Mol Med Rep 2021; 23:320. [PMID: 33760110 PMCID: PMC7974271 DOI: 10.3892/mmr.2021.11959] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
Corilagin, extracted from the Euphorbiaceae and Phyllanthus plants, inhibits the growth of a number of types of tumors. Compared with temozolomide, the traditional chemotherapy drug, corilagin has demonstrated stronger antitumor activity. However, the pharmaceutical mechanism of corilagin in glioma remains unclear. Nuclear factor erythroid 2 like 2 (NFE2L2 or NRF2) is positively associated with several types of tumor including glioma. In the present study, NRF2 expression was higher in glioma tissues compared with non-glioma specimens. Therefore, it was hypothesized that corilagin targets NRF2 regulation of U251 cell apoptosis. The present study used Hoechst 33258 staining to demonstrate that corilagin induced glioma cell apoptosis and observed that the expression of the apoptosis-related gene Bcl-2 was reduced. In addition, corilagin induced autophagy and promoted the conversion of light chain 3 (LC3) protein from LC3I to LC3II. NRF2 expression was downregulated by corilagin stimulation. Furthermore, the gene expression pattern following knockdown of NRF2 in U251 cells using siRNA was consistent with corilagin stimulation. Therefore, it was preliminarily concluded that corilagin induces apoptosis and autophagy by reducing NRF2 expression.
Collapse
Affiliation(s)
- Jilan Liu
- Department of Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Xianyun Qin
- Department of Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Wenyuan Ma
- Department of Neurosurgery, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Shu Jia
- Department of Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Xiaobei Zhang
- Department of Central Laboratory, Affiliated Hospital of Jining Medical University, Jining, Shandong 272029, P.R. China
| | - Xinlin Yang
- Department of Orthopaedic Surgery, Orthopaedic Research Labs, University of Virginia, Charlottesville, VA 22908, USA
| | - Dongfeng Pan
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22908, USA
| | - Feng Jin
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
16
|
Liang Z, Li J, Zhao L, Deng Y. miR‑375 affects the hedgehog signaling pathway by downregulating RAC1 to inhibit hepatic stellate cell viability and epithelial‑mesenchymal transition. Mol Med Rep 2021; 23:182. [PMID: 33398380 PMCID: PMC7809903 DOI: 10.3892/mmr.2020.11821] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 10/13/2020] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs/miRs) are a class of non-coding RNAs that serve crucial roles in liver cancer and other liver injury diseases. However, the expression profile and mechanisms underlying miRNAs in liver fibrosis are not completely understood. The present study identified the novel miR-375/Rac family small GTPase 1 (RAC1) regulatory axis in liver fibrosis. Reverse transcription-quantitative PCR was performed to detect miR-375 expression levels. MTT, flow cytometry and western blotting were performed to explore the in vitro roles of miR-375. The dual-luciferase reporter gene assay was performed to determine the potential mechanism underlying miR-375 in liver fibrosis. miR-375 expression was significantly downregulated in liver fibrosis tissues and cells compared with healthy control tissues and hepatocytes, respectively. Compared with the pre-negative control group, miR-375 overexpression inhibited mouse hepatic stellate cell (HSC) viability and epithelial-mesenchymal transition, and alleviated liver fibrosis. The dual-luciferase reporter assay results demonstrated that miR-375 bound to RAC1. Moreover, the results indicated that miR-375 regulated the hedgehog signaling pathway via RAC1 to restrain HSC viability and EMT, thus exerting its anti-liver fibrosis function. The present study identified the miR-375/RAC1 axis as a novel regulatory axis associated with the development of liver fibrosis.
Collapse
Affiliation(s)
- Zhiwei Liang
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Jian Li
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Longshuan Zhao
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yilei Deng
- Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
17
|
Lu F, Zhang WJ, Zhai S, Sun YW, Chen QJ, Yang XL, Zhang CF, Wang CZ, Yuan CS. Anti-migraine effect of wine-processed Radix scutellariae: Pharmacodynamic verification in nitroglycerin-induced rats and correlation study between compounds dissolution and the fractal dimension. JOURNAL OF ETHNOPHARMACOLOGY 2020; 263:113131. [PMID: 32730879 DOI: 10.1016/j.jep.2020.113131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 05/11/2020] [Accepted: 06/23/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Wine-processed Radix scutellariae (RS) is the processed product of RS, which is the dried root of Scutellaria baicalensis Georgi. It is recorded in Chinese traditional formula that wine-processed RS has the effect of anti-migraine, while the effect has not been confirmed and the possible mechanism remains unclear. AIM OF THE STUDY To verify the anti-migraine effect of wine-processed RS in nitroglycerin (NTG)-induced rats and explore the correlation between compounds dissolution and the pore structure based on fractal theory. MATERIALS AND METHODS In the validation of pharmacodynamics, the effects of wine-processed RS on migraines were firstly evaluated by observing the number of head-scratching of rats, then investigated by determining the levels of nitric oxide (NO), calcitonin gene-related peptide (CGRP) and the expression of c-Fos in the brain of NTG-induced rat models using ELISA and immunohistochemical assessments. In the correlation study, the stir-frying time of RS was set to 5 min, 10 min and 15 min. The scanning electron microscope (SEM) and mercury intrusion method were used to explore the pore structure and main parameters of the pore structure including pore size distribution, pore volume, porosity, surface area and fractal dimension. The compounds dissolution of total flavonoids and five major components containing baicalein, baicalin, scutellarin, wogonin and wogonoside was determined by UV-Vis spectrophotometry and HPLC separately. RESULTS The animal experiments had shown that wine-processed RS could significantly reduce the head-scratching times of NTG-induced rat models (p < 0.01) and markedly decrease the levels of NO (p < 0.01), CGRP (p < 0.05) and the expression of c-Fos (p < 0.01) compared with model group. The data indicated that wine-processing would affect the dissolution of compounds by changing the pore structure of RS. The order of positive correlation between pore structure parameters and compounds' dissolution was total surface area > fractal dimension (r > 0) and the order of negative correlation was average pore size > total porosity > total volume (r < 0). Compared with the other sample groups (p < 0.05), the wine-processed RS stir-fried for 10 min had a pore structure which was more favorable for compounds dissolution. CONCLUSIONS Wine-processing could strengthen the anti-migraine effect of RS by changing the pore structure of RS, which is linked to the dissolution of compounds. The RS stir-fried for 10 min may be more effective in treating migraine.
Collapse
Affiliation(s)
- Fang Lu
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, JS, 210009, China
| | - Wen-Jun Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, JS, 210009, China
| | - Shuo Zhai
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, JS, 210009, China
| | - Yue-Wen Sun
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, JS, 210009, China
| | - Qiu-Jing Chen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, JS, 210009, China
| | - Xiao-Lin Yang
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Chun-Feng Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, JS, 210009, China; Tang Center of Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL, 60637, USA.
| | - Chong-Zhi Wang
- Tang Center of Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL, 60637, USA
| | - Chun-Su Yuan
- Tang Center of Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
18
|
Jin X, Fu Q, Gu Z, Zhang Z, Lv H. Injectable corilagin/low molecular weight chitosan/PLGA-PEG-PLGA thermosensitive hydrogels for localized cancer therapy and promoting drug infiltration by modulation of tumor microenvironment. Int J Pharm 2020; 589:119772. [DOI: 10.1016/j.ijpharm.2020.119772] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/26/2020] [Accepted: 08/11/2020] [Indexed: 02/09/2023]
|
19
|
Yuan H, Zhang J, Yin X, Liu T, Yue X, Li C, Wang Y, Li D, Wang Q. The protective role of corilagin on renal calcium oxalate crystal-induced oxidative stress, inflammatory response, and apoptosis via PPAR-γ and PI3K/Akt pathway in rats. Biotechnol Appl Biochem 2020; 68:1323-1331. [PMID: 33080078 DOI: 10.1002/bab.2054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/15/2020] [Indexed: 11/07/2022]
Abstract
Kidney stones, also known as calcium oxalate (CaOx) nephrolithiasis, are often asymptomatic, leading to kidney injury and renal failure complications. Corilagin is a gallotannin found in various plants and is known to elicit various biological activities. The present study aimed to elucidate the renoprotective effect of corilagin against the rats' renal stones deposition. The rats were induced for nephrolithiasis (CaOx deposition) using 0.75% ethylene glycol in their drinking water. Then, they were treated with corilagin at 50 and 100 mg/kg/day for 4 weeks. At the end of the experimental period, the rats were killed; blood and renal tissues were collected for various histological, biochemical, and gene expression analyses. The results demonstrated that the rats had renal calculi displaying a significant increase in serum creatinine (59.39 μmol/L) and blood urea nitrogen (19.03 mmol/L) levels compared with controls. Moreover, the malondialdehyde (13.29 nmol/mg) level was found to increase with a profound reduction in antioxidants' activities with upregulated inflammatory cytokines. In contrast, the RT-PCR and immunohistochemistry analysis demonstrated a substantial reduction in cell survival markers PPAR-γ and PI3K/Akt with an apparent increase in apoptosis markers genes expressions in rats suffering from renal stones. Thus, the present study results suggest that corilagin could suppress renal CaOx crystal-induced oxidative stress, inflammatory response, and apoptosis via PPAR-γ and PI3K/Akt-mediated pathway.
Collapse
Affiliation(s)
- Haibo Yuan
- Department of Urology, Baoding No. 1 Central Hospital, Baoding, Hebei, 071000, People's Republic of China
| | - Jinghong Zhang
- Department of Urology, Baoding No. 1 Central Hospital, Baoding, Hebei, 071000, People's Republic of China
| | - Xiaosong Yin
- Department of Urology, Baoding No. 1 Central Hospital, Baoding, Hebei, 071000, People's Republic of China
| | - Tongwei Liu
- Department of Urology, Baoding No. 1 Central Hospital, Baoding, Hebei, 071000, People's Republic of China
| | - Xiao Yue
- Department of Urology, Baoding No. 1 Central Hospital, Baoding, Hebei, 071000, People's Republic of China
| | - Chuangui Li
- Department of Urology, Baoding No. 1 Central Hospital, Baoding, Hebei, 071000, People's Republic of China
| | - Yuanyuan Wang
- Department of Urology, Baoding No. 1 Central Hospital, Baoding, Hebei, 071000, People's Republic of China
| | - Ding Li
- Department of Urology, Baoding No. 1 Central Hospital, Baoding, Hebei, 071000, People's Republic of China
| | - Qiang Wang
- Department of Urology, Baoding No. 1 Central Hospital, Baoding, Hebei, 071000, People's Republic of China
| |
Collapse
|
20
|
Lu J, Ye C, Huang Y, Huang D, Tang L, Hou W, Kuang Z, Chen Y, Xiao S, Yishake M, He R. Corilagin suppresses RANKL-induced osteoclastogenesis and inhibits oestrogen deficiency-induced bone loss via the NF-κB and PI3K/AKT signalling pathways. J Cell Mol Med 2020; 24:10444-10457. [PMID: 32681612 PMCID: PMC7521306 DOI: 10.1111/jcmm.15657] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 06/24/2020] [Accepted: 06/29/2020] [Indexed: 12/15/2022] Open
Abstract
Over‐activated osteoclastogenesis, which is initiated by inflammation, has been implicated in osteoporosis. Corilagin, a natural compound extracted from various medicinal herbaceous plants, such as Cinnamomum cassia, has antioxidant and anti‐inflammatory activities. We found that Corilagin suppressed osteoclast differentiation in a dose‐dependent manner, significantly decreased osteoclast‐related gene expression and impaired bone resorption by osteoclasts. Moreover, phosphorylation of members of the nuclear factor‐kappaB (NF‐κB) and PI3K/AKT signalling pathways was reduced by Corilagin. In a murine model of osteoporosis, Corilagin inhibited osteoclast functions in vivo and restored oestrogen deficiency‐induced bone loss. In conclusion, our findings suggested that Corilagin inhibited osteoclastogenesis by down‐regulating the NF‐κB and PI3K/AKT signalling pathways, thus showing its potential possibility for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Jinwei Lu
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Chenyi Ye
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Yanyong Huang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China.,Department of Orthopedic Surgery, The First People's Hospital of Xiaoshan District, Hangzhou, China
| | - Donghui Huang
- Department of Orthopedic Surgery, Hangzhou Third Hospital, Hangzhou, China
| | - Lan Tang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Weiduo Hou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Zhihui Kuang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Yazhou Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Shining Xiao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| | - Mumingjiang Yishake
- Orthopedics Department, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Rongxin He
- Department of Orthopedic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Orthopedics Research Institute of Zhejiang University, Hangzhou, China
| |
Collapse
|