1
|
Zhang C, Ji Z, Xu N, Yuan J, Zeng W, Wang Y, He Q, Dong J, Zhang X, Yang D, Jiang W, Yan Y, Shang W, Chu J, Chu Q. Integrating network pharmacology and experimental validation to decipher the pharmacological mechanism of DXXK in treating diabetic kidney injury. Sci Rep 2024; 14:22319. [PMID: 39333622 PMCID: PMC11436795 DOI: 10.1038/s41598-024-73642-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 09/19/2024] [Indexed: 09/29/2024] Open
Abstract
Diabetes mellitus (DM) is a chronic metabolic disease that is highly susceptible to kidney injury. Di'ao XinXueKang capsules (DXXK) is a novel Chinese herbal medicine that has been used in clinical trials for the therapy of DM and kidney disease, but the underlying pharmacological mechanism remains unclear. This study aims to integrate network pharmacology, molecular docking and in vivo experiments to explore the potential mechanisms of DXXK in the treatment of diabetic kidney injury. The chemical constituents of DXXK were extracted from the ETCM and Batman-TCM databases, and then evaluated for their pharmacological activity via the Swiss ADME platform. Multiple disease databases were searched and integrated for DM-related targets. Overlapping targets were then collected to construct a protein-protein interaction (PPI) network. KEGG and GO enrichment analyses were performed based on the Metascape database, and molecular docking was performed using AutoDock Vina software. The main components in DXXK were analyzed by HPLC. The results of network pharmacology and molecular docking were validated in an animal model of DM induced by the combination of a high-fat diet (HFD) and streptozotocin (STZ). We screened and obtained 7 ingredients and identified dioscin, protodioscin, and pseudoprotodioscin as the major components of DXXK by HPLC. A total of 2,216 DM-related pathogenic genes were obtained from DrugBank, GeneCards, OMIM, and DisGeNET databases. KEGG and GO enrichment analyses indicated that the TGF-beta signaling pathway is a critical pathway associated with DM therapy. Molecular docking revealed that the ingredients in DXXK bind to the pivotal targets TGFβ1, Smad2, and Smad3. In diabetic mice, we found that DXXK alleviated diabetic symptoms, lowered blood glucose, improved insulin tolerance, and modulated lipid metabolism. Furthermore, DXXK attenuated renal lesions and fibrosis by downregulating TGFβ1, Smad2, and Smad3. Collectively, our results suggest that DXXK has the potential to regulate glucolipid metabolism in DM, and it may serve as a viable therapeutic option for renoprotection by inhibiting of the TGF-β1/Smad2/3 pathway.
Collapse
Affiliation(s)
- Chenxu Zhang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, People's Republic of China
- School of Graduate, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, People's Republic of China
| | - Zhangxin Ji
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, People's Republic of China
- School of Graduate, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, People's Republic of China
| | - Na Xu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and International Joint Laboratory On Tea Chemistry and Health Effects of Ministry of Education, Anhui Agricultural University, Hefei, 230036, Anhui, People's Republic of China
| | - Jingjing Yuan
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, People's Republic of China
- Research and Technology Center, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, People's Republic of China
| | - Wen Zeng
- Research and Technology Center, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, People's Republic of China
| | - Yadong Wang
- Department of Pathology, School of Integrative Medicine, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, People's Republic of China
| | - Qing He
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, People's Republic of China
- School of Graduate, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, People's Republic of China
| | - Jiaxing Dong
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, People's Republic of China
- School of Graduate, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, People's Republic of China
| | - Xinyu Zhang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, People's Republic of China
- School of Graduate, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, People's Republic of China
| | - Dongmei Yang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, People's Republic of China
- School of Graduate, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, People's Republic of China
| | - Wei Jiang
- School of Nursing, Anhui Medical College, Furong Road Campus, Hefei, 230601, Anhui, People's Republic of China
| | - Yibo Yan
- Second Clinical Medical College, Anhui Medical University, Hefei, 230601, Anhui, People's Republic of China
| | - Wencui Shang
- School of Graduate, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, People's Republic of China
| | - Jun Chu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, People's Republic of China.
- Research and Technology Center, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, People's Republic of China.
- Institute of Surgery, Anhui Academy of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, People's Republic of China.
| | - Quangen Chu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, 230038, Anhui, People's Republic of China.
| |
Collapse
|
2
|
Sztolsztener K, Konstantynowicz-Nowicka K, Pędzińska-Betiuk A, Chabowski A. Concentration-Dependent Attenuation of Pro-Fibrotic Responses after Cannabigerol Exposure in Primary Rat Hepatocytes Cultured in Palmitate and Fructose Media. Cells 2023; 12:2243. [PMID: 37759466 PMCID: PMC10526512 DOI: 10.3390/cells12182243] [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: 07/11/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Hepatic fibrosis is a consequence of liver injuries, in which the overproduction and progressive accumulation of extracellular matrix (ECM) components with the simultaneous failure of matrix turnover mechanisms are observed. The aim of this study was to investigate the concentration-dependent influence of cannabigerol (CBG, Cannabis sativa L. component) on ECM composition with respect to transforming growth factor beta 1 (TGF-β1) changes in primary hepatocytes with fibrotic changes induced by palmitate and fructose media. Cells were isolated from male Wistar rats' livers in accordance with the two-step collagenase perfusion technique. This was followed by hepatocytes incubation with the presence or absence of palmitate with fructose and/or cannabigerol (at concentrations of 1, 5, 10, 15, 25, 30 µM) for 48 h. The expression of ECM mRNA genes and proteins was determined using PCR and Western blot, respectively, whereas media ECM level was evaluated using ELISA. Our results indicated that selected low concentrations of CBG caused a reduction in TGF-β1 mRNA expression and secretion into media. Hepatocyte exposure to cannabigerol at low concentrations attenuated collagen 1 and 3 deposition. The protein and/or mRNA expressions and MMP-2 and MMP-9 secretion were augmented using CBG. Considering the mentioned results, low concentrations of cannabigerol treatment might expedite fibrosis regression and promote regeneration.
Collapse
Affiliation(s)
- Klaudia Sztolsztener
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland; (K.K.-N.); (A.C.)
| | | | - Anna Pędzińska-Betiuk
- Department of Experimental Physiology and Pathophysiology, Medical University of Bialystok, 15-089 Bialystok, Poland;
| | - Adrian Chabowski
- Department of Physiology, Medical University of Bialystok, 15-089 Bialystok, Poland; (K.K.-N.); (A.C.)
| |
Collapse
|
3
|
Nam Y, Kim M, Erdenebileg S, Cha KH, Ryu DH, Kim HY, Lee SH, Jung JH, Nho CW. Sanguisorba officinalis L. Ameliorates Hepatic Steatosis and Fibrosis by Modulating Oxidative Stress, Fatty Acid Oxidation, and Gut Microbiota in CDAHFD-Induced Mice. Nutrients 2023; 15:3779. [PMID: 37686810 PMCID: PMC10490207 DOI: 10.3390/nu15173779] [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: 07/31/2023] [Revised: 08/19/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a leading cause of chronic liver diseases and encompasses non-alcoholic steatosis, steatohepatitis, and fibrosis. Sanguisorba officinalis L. (SO) roots have traditionally been used for their antioxidant properties and have beneficial effects on metabolic disorders, including diabetes and obesity. However, its effects on hepatic steatosis and fibrosis remain unclear. In this study, we explored the effects of a 95% ethanolic SO extract (SOEE) on NAFLD and fibrosis in vivo and in vitro. The SOEE was orally administered to C57BL/6J mice fed a choline-deficient, L-amino-acid-defined, high-fat diet for 10 weeks. The SOEE inhibited hepatic steatosis by modulating hepatic malondialdehyde levels and the expression of oxidative stress-associated genes, regulating fatty-acid-oxidation-related genes, and inhibiting the expression of genes that are responsible for fibrosis. The SOEE suppressed the deposition of extracellular matrix hydroxyproline and mRNA expression of fibrosis-associated genes. The SOEE decreased the expression of fibrosis-related genes in vitro by inhibiting SMAD2/3 phosphorylation. Furthermore, the SOEE restored the gut microbial diversity and modulated specific bacterial genera associated with NAFLD and fibrosis. This study suggests that SOEE might be the potential candidate for inhibiting hepatic steatosis and fibrosis by modulating oxidative stress, fatty acid oxidation, and gut microbiota composition.
Collapse
Affiliation(s)
- Yunseong Nam
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea; (Y.N.); (M.K.); (S.E.); (K.H.C.); (H.Y.K.)
- Smart Farm Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (D.H.R.); (S.H.L.); (J.H.J.)
| | - Myungsuk Kim
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea; (Y.N.); (M.K.); (S.E.); (K.H.C.); (H.Y.K.)
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju 26493, Republic of Korea
| | - Saruul Erdenebileg
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea; (Y.N.); (M.K.); (S.E.); (K.H.C.); (H.Y.K.)
- Smart Farm Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (D.H.R.); (S.H.L.); (J.H.J.)
| | - Kwang Hyun Cha
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea; (Y.N.); (M.K.); (S.E.); (K.H.C.); (H.Y.K.)
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju 26493, Republic of Korea
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
| | - Da Hye Ryu
- Smart Farm Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (D.H.R.); (S.H.L.); (J.H.J.)
| | - Ho Youn Kim
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea; (Y.N.); (M.K.); (S.E.); (K.H.C.); (H.Y.K.)
- Smart Farm Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (D.H.R.); (S.H.L.); (J.H.J.)
| | - Su Hyeon Lee
- Smart Farm Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (D.H.R.); (S.H.L.); (J.H.J.)
| | - Je Hyeong Jung
- Smart Farm Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (D.H.R.); (S.H.L.); (J.H.J.)
| | - Chu Won Nho
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul 02792, Republic of Korea; (Y.N.); (M.K.); (S.E.); (K.H.C.); (H.Y.K.)
- Smart Farm Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea; (D.H.R.); (S.H.L.); (J.H.J.)
| |
Collapse
|
4
|
Sarkar A, Chakraborty D, Kumar V, Malhotra R, Biswas S. Upregulation of leucine-rich alpha-2 glycoprotein: A key regulator of inflammation and joint fibrosis in patients with severe knee osteoarthritis. Front Immunol 2022; 13:1028994. [PMID: 36569927 PMCID: PMC9768428 DOI: 10.3389/fimmu.2022.1028994] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction Osteoarthritis (OA) is a degenerative disease of the joints mainly affecting older individuals. Since the etiology behind the progression of OA is not well understood, several associated consequences, such as synovial joint stiffness and its progression due to joint fibrosis, are still poorly understood. Although a lot of developments have been achieved in the diagnosis and management of OA, synovial fibrosis remains one of the major challenging consequences. The present study was therefore focused on understanding the mechanism of synovial fibrosis, which may further contribute to improving symptomatic treatments, leading to overall improvements in the treatment outcomes of patients with OA. Methods We used advanced proteomic techniques including isobaric tag for relative and absolute quantitation and sequential window acquisition of all theoretical mass spectra for the identification of differentially expressed proteins in the plasma samples of patients with OA. An in silico study was carried out to evaluate the association of the identified proteins with their biological processes related to fibrosis and remodeling of the extracellular matrix (ECM). The most significantly upregulated protein was then validated by Western blot and enzyme-linked immunosorbent assay. The target protein was then further investigated for its role in inflammation and joint fibrosis using an in vitro study model. Results Leucine-rich alpha-2 glycoprotein (LRG1) was found to be the most highly differentially expressed upregulated (9.4-fold) protein in the plasma samples of patients with OA compared to healthy controls. The knockdown of LRG1 followed by in vitro studies revealed that this protein promotes the secretion of the ECM in synovial cells and actively plays a role in wound healing and cell migration. The knockdown of LRG1 further confirmed the reduction of the inflammatory- and fibrosis-related markers in primary cells. Conclusion LRG1 was identified as a highly significant upregulated protein in the plasma samples of patients with OA. It was found to be associated with increased fibrosis and cell migration, leading to enhanced inflammation and joint stiffness in OA pathogenesis.
Collapse
Affiliation(s)
- Ashish Sarkar
- Council of Scientific and Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Delhi University, Delhi, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Debolina Chakraborty
- Council of Scientific and Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Delhi University, Delhi, India,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Vijay Kumar
- All India Institute of Medical Sciences, New Delhi, India
| | | | - Sagarika Biswas
- Council of Scientific and Industrial Research (CSIR)-Institute of Genomics and Integrative Biology, Delhi University, Delhi, India,*Correspondence: Sagarika Biswas,
| |
Collapse
|
5
|
Vijayan N, Perumal MK. A critical review on anti-fibrotic phytochemicals targeting activated hepatic stellate cells. J Food Biochem 2022; 46:e14438. [PMID: 36209494 DOI: 10.1111/jfbc.14438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/29/2022] [Accepted: 09/16/2022] [Indexed: 01/18/2023]
Abstract
Liver fibrosis is a major health concern occurring worldwide. It arises due to prolonged wound healing response of various insults like viral, autoimmune, cholestatic, drug-induced, and metabolic diseases. Currently, there is no clinically approved drug for liver fibrosis treatment. Hepatic stellate cells are the principal liver cells that are activated during liver fibrosis, and targeting these activated cells is an ideal therapeutic strategy. Numerous phytochemicals have been demonstrated in vitro and in vivo treating experimental liver fibrosis; however, none of them have been clinically approved for therapeutic use. This review mainly focuses on such hepatoprotective phytochemicals reported inhibiting major signaling pathways that are dysregulated in activated hepatic stellate cells. PRACTICAL APPLICATIONS: Liver fibrosis is a global health concern and there is no FDA approved drug to treat liver fibrosis. Although notable pharmacological agents like pentoxifylline, gliotoxin, imatinibmesylate, Gleevec, and so on are reported to exhibit anti-fibrotic effect, the major concern is their side effect. Hence, phytochemicals are promising candidates that could be employed against liver fibrosis. In this review, the anti-fibrotic potential of phytochemicals targeting activated HSCs are summarized. Understanding these phytochemicals will further help in the development of agents that are more effective against liver fibrosis.
Collapse
Affiliation(s)
- Nivya Vijayan
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Madan Kumar Perumal
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
6
|
Shi L, Zhang P, Jin R, Chen X, Dong L, Chen W. Dioscin ameliorates inflammatory bowel disease by up-regulating miR-125a-5p to regulate macrophage polarization. J Clin Lab Anal 2022; 36:e24455. [PMID: 35524480 PMCID: PMC9169194 DOI: 10.1002/jcla.24455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/13/2022] [Accepted: 04/16/2022] [Indexed: 11/08/2022] Open
Abstract
PURPOSE Dioscin has been proven to have anti-cancer, anti-inflammatory, and anti-infection roles. However, the role of Dioscin in inflammatory bowel disease (IBD) and its related mechanisms is unclear and needs further study. METHODS The colitis model in mice was established. After Dioscin (20, 40, or 80 mg/kg) treatment, the colon length was measured by a ruler. Histopathology, inflammatory cytokines, gut permeability, tight junction proteins, macrophage infiltration, macrophage polarization, and miR-125a-5p level were detected by hematoxylin-eosin staining, enzyme-linked immunosorbent assay, quantitative real-time polymerase chain reaction (qRT-PCR), FITC-dextran, Western blot, and flow cytometry. In vitro experiments, after RAW264.7 cells induced by lipopolysaccharide (LPS)/interleukin-4 (IL-4), were treated with Dioscin and miR-125a-5p inhibitor, miR-125a-5p level, cell vitality, inflammatory cytokines, and M1/M2 marker genes were measured by qRT-PCR and MTT assay. RESULTS Dioscin (20, 40, or 80 mg/kg) relieved DSS-triggered colitis and restrained the serum and colon of pro-inflammatory cytokines expression. Meanwhile, different concentrations' Dioscin weakened M1 macrophage polarization but facilitated tight junction protein expressions, M2 macrophage polarization, and miR-125a-5p level in colitic mice. Moreover, miR-125a-5p inhibitor reversed the modulation of Dioscin on miR-125a-5p expression, cell vitality, and inflammatory cytokines in lipopolysaccharide (LPS)-induced RAW264.7 cells. We further discovered that Dioscin restrained M1 marker gene (CD16) expression while intensifying M2 marker genes (CD206 and Arginase-1) expressions in vitro, which was reversed by miR-125a-5p inhibitor. CONCLUSION Dioscin modulated macrophage polarization by increasing miR-125a-5p, thereby improving the intestinal epithelial barrier function and reducing IBD.
Collapse
Affiliation(s)
- Lingyan Shi
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
- Department of GastroenterologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Peichen Zhang
- Department of Gastrointestinal SurgeryThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Ruifang Jin
- Department of GastroenterologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Xiaowei Chen
- Department of GastroenterologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Lemei Dong
- Department of GastroenterologyThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Weichang Chen
- Department of GastroenterologyThe First Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
7
|
Shang Q, Zhu L, Shang W, Zeng J, Qi Y. Dioscin exhibits protective effects on in vivo and in vitro asthma models via suppressing TGF-β1/Smad2/3 and AKT pathways. J Biochem Mol Toxicol 2022; 36:e23084. [PMID: 35481609 DOI: 10.1002/jbt.23084] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 03/02/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022]
Abstract
Dioscin is a natural product that possesses protective effects on multiple chronic injuries, but its effects on asthma are not fully understood. Herein, we evaluated its effects on asthmatic mice established by ovalbumin (OVA) sensitization and challenges and further explored the mechanism. Inflammatory cells in bronchoalveolar lavage fluids (BALFs) were analyzed using Diff-Quik staining. OVA-specific immunoglobulin E (IgE)/IgG1 in serum and inflammatory cytokines (interleukin 4[IL-4], IL-5, IL-13, and tumor necrosis factor-α) in BALFs and lung tissues were measured using Enzyme-Linked Immunosorbent Assay Kits. Hematoxylin and eosin, periodic acid-Schiff, and immunohistochemistry staining showed histopathological changes in lung tissues. Epithelial-mesenchymal transition (EMT) in human bronchial epithelial (16HBE) cells was assessed by immunofluorescence staining. Hydroxyproline content was used to evaluate collagen deposition. Polymerase chain reaction and Western blot were performed to measure messenger RNA and protein expression. We found that dioscin treatment (particularly at the dose of 80 mg/kg) significantly inhibited pulmonary inflammation in asthmatic mice, as evidenced by the decreased serum OVA-specific IgE/IgG1 and the reduced inflammatory cells and cytokines in BALFs and lung tissues. Moreover, dioscin effectively ameliorated the goblet cell hyperplasia, mucus hypersecretion, collagen deposition, and smooth muscle hyperplasia in the airways of asthmatic mice. Mechanistically, dioscin restrained the activated TGF-β1/Smad2/3 and protein kinase B (AKT) signal pathways in lung tissues and potently reversed the TGF-β1-induced EMT and phosphorylation of Smad2/3 and AKT in 16HBE cells. Collectively, dioscin displayed protective effects on OVA-induced asthmatic mice via adjusting TGF-β1/Smad2/3 and AKT signal pathways, supporting the fact that dioscin could be a candidate for chronic asthma prevention in the future.
Collapse
Affiliation(s)
- Qian Shang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Li Zhu
- Department of Pulmonary and Critical Care Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China.,Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Weina Shang
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| | - Jia Zeng
- Department of Pulmonary and Critical Care Medicine, People's Hospital of Zhengzhou University, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Yong Qi
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, Henan, China
| |
Collapse
|
8
|
He Y, Luo R, Xia M, Liu J, Yao Y, Min F, Jin R, Wang R, Peng X. Orally Administered Diosgenin Alleviates Colitis in Mice Induced by Dextran Sulfate Sodium through Gut Microbiota Modulation and Short-Chain Fatty Acid Generation. J Med Food 2022; 25:261-271. [PMID: 35320010 DOI: 10.1089/jmf.2021.k.0086] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Diosgenin (DIO) is a kind of steroid sapogenin derived from natural plants. It exerts strong anti-infection, antiallergy, antiviral, and antishock pharmacological properties. In this article, the protective effects of DIO against dextran sulfate sodium (DSS)-induced colitis in mice were researched. Compared with the 2.5% DSS treatment group, 15 mg/kg body weight of diosgenin alleviated colitis disease, evidenced by the increased body weight, the decrease in the disease activity index, and the histological scores. Furthermore, 16S rRNA high-throughput sequencing results demonstrated that DIO improved the colon homeostasis through modulating the gut microbiota, including increases in the relative abundance of several probiotic bacteria, such as Prevotellaceae (from 1.4% to 5.8%), Lactobacillus (from 12.3% to 29.7%), Mucispirillum (from 0.07% to 0.49%), and decreases in the pathogenic bacteria, such as Streptococcus (from 1.6% to 0.6%) and Pseudomonadaceae (from 0.004% to 0%). In addition, the concentration of gut microbial metabolites, total short-chain fatty acids (SCFAs), acetic acid, and propionic acid were significantly increased after DIO supplementation. In conclusion, our findings suggested that DIO attenuates DSS-induced colitis in mice by means of modulating imbalanced gut microbiota and increases in SCFA generation.
Collapse
Affiliation(s)
- Yushu He
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Ruilin Luo
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Manying Xia
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Jiayu Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yanpeng Yao
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Fenyi Min
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Ruyi Jin
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Ruolin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xiaoli Peng
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| |
Collapse
|
9
|
Bandopadhyay S, Anand U, Gadekar VS, Jha NK, Gupta PK, Behl T, Kumar M, Shekhawat MS, Dey A. Dioscin: A review on pharmacological properties and therapeutic values. Biofactors 2022; 48:22-55. [PMID: 34919768 DOI: 10.1002/biof.1815] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 11/27/2021] [Indexed: 02/06/2023]
Abstract
Dioscin has gained immense popularity as a natural, bioactive steroid saponin, which offers numerous medical benefits. The growing global incidence of disease-associated morbidity and mortality continues to compromise human health, facilitating an increasingly urgent need for nontoxic, noninvasive, and efficient treatment alternatives. Natural compounds can contribute vastly to this field. Over recent years, studies have demonstrated the remarkable protective actions of dioscin against a variety of human malignancies, metabolic disorders, organ injuries, and viral/fungal infections. The successful usage of this phytocompound has been widely seen in medical treatment procedures under traditional Chinese medicine, and it is becoming progressively prevalent worldwide. This review provides an insight into the wide spectrum of pharmacological activities of dioscin, as reported and compiled in recent literature. The various novel approaches and applications of dioscin also verify the advantages exhibited by plant extracts against commercially available drugs, highlighting the potential of phytochemical agents like dioscin to be further incorporated into clinical practice.
Collapse
Affiliation(s)
| | - Uttpal Anand
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Vijaykumar Shivaji Gadekar
- Zoology Department, Sangola College (affiliated to Punyashlok Ahilyadevi Holkar Solapur University), Solapur, Maharashtra, India
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering & Technology, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Chandigarh, Punjab, India
| | - Manoj Kumar
- Chemical and Biochemical Processing Division, ICAR - Central Institute for Research on Cotton Technology, Mumbai, Maharashtra, India
| | - Mahipal S Shekhawat
- Plant Biotechnology Unit, Kanchi Mamunivar Government Institute for Postgraduate Studies and Research, Puducherry, India
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, Kolkata, West Bengal, India
| |
Collapse
|
10
|
Siraj MA, Islam MA, Al Fahad MA, Kheya HR, Xiao J, Simal-Gandara J. Cancer Chemopreventive Role of Dietary Terpenoids by Modulating Keap1-Nrf2-ARE Signaling System—A Comprehensive Update. APPLIED SCIENCES 2021; 11:10806. [DOI: 10.3390/app112210806] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
ROS, RNS, and carcinogenic metabolites generate excessive oxidative stress, which changes the basal cellular status and leads to epigenetic modification, genomic instability, and initiation of cancer. Epigenetic modification may inhibit tumor-suppressor genes and activate oncogenes, enabling cells to have cancer promoting properties. The nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that in humans is encoded by the NFE2L2 gene, and is activated in response to cellular stress. It can regulate redox homoeostasis by expressing several cytoprotective enzymes, including NADPH quinine oxidoreductase, heme oxygenase-1, UDP-glucuronosyltransferase, glutathione peroxidase, glutathione-S-transferase, etc. There is accumulating evidence supporting the idea that dietary nutraceuticals derived from commonly used fruits, vegetables, and spices have the ability to produce cancer chemopreventive activity by inducing Nrf2-mediated detoxifying enzymes. In this review, we discuss the importance of these nutraceuticals in cancer chemoprevention and summarize the role of dietary terpenoids in this respect. This approach was taken to accumulate the mechanistic function of these terpenoids to develop a comprehensive understanding of their direct and indirect roles in modulating the Keap1-Nrf2-ARE signaling system.
Collapse
Affiliation(s)
- Md Afjalus Siraj
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawaii at Hilo, Hilo, HI 96720, USA
| | - Md. Arman Islam
- Pharmacy Discipline, Life Science School, Khulna University, Khulna 9208, Bangladesh
| | - Md. Abdullah Al Fahad
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan 31151, Korea
| | - Habiba Rahman Kheya
- Department of Sociology, Faculty of Social Sciences, University of Dhaka, Dhaka 1000, Bangladesh
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo—Ourense Campus, E32004 Ourense, Spain
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Food Science and Technology, University of Vigo—Ourense Campus, E32004 Ourense, Spain
| |
Collapse
|
11
|
Yang L, Li CY, Ouyang JY, Li MZ, Zhan Y, Feng XF, Lu Y, Li MC, Lei JF, Zhao T, Wang L, Zou HY, Zhao H. Trillium tschonoskii rhizomes' saponins induces oligodendrogenesis and axonal reorganization for ischemic stroke recovery in rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114358. [PMID: 34166736 DOI: 10.1016/j.jep.2021.114358] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/18/2021] [Accepted: 06/19/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Trillium tschonoskii Maxim. is one of traditional Chinese medical herbs that has been utilized to treat brain damages and cephalalgia. The neuroprotective effect of total saponins from Trillium tschonoskii rhizome (TSTT) has been demonstrated efficacy in rats following ischemia. However, the axonal remodeling effect of TSTT and the detailed mechanisms after ischemic stroke have not been investigated. AIM OF THE STUDY We aimed to estimate therapeutic role of TSTT in axonal remodeling using magnetic resonance imaging (MRI) technique, and explored possible mechanisms underlying this process followed by histological assays in ischemic rats. METHODS Male Sprague-Dawley (SD) rats underwent permanently focal cerebral ischemia induced by occluding right permanent middle cerebral artery. TSTT was intragastrically administrated 6 h after surgery and once daily for consecutive 15 days. Neurological function was assessed by the motor deficit score and beam walking test. T2 relaxation mapping and diffusion tensor imaging (DTI) were applied for detecting cerebral tissues damages and microstructural integrity of axons. Luxol fast blue (LFB) and transmission electron microscope (TEM) were performed to evaluate histopathology in myelinated axons. Double immunofluorescent staining was conducted to assess oligodendrogenesis. Furthermore, the protein expressions regarding to axonal remodeling related signaling pathways were detected by Western blot assays. RESULTS TSTT treatment (65, 33 mg/kg) markedly improved motor function after ischemic stroke. T2 mapping MRI demonstrated that TSTT decreased lesion volumes, and DTI further confirmed that TSTT preserved axonal microstructure of the sensorimotor cortex and internal capsule. Meanwhile, diffusion tensor tractography (DTT) showed that TSTT elevated correspondent density and length of fiber in the internal capsule. These MRI measurements were confirmed by histological examinations. Notably, TSTT significantly increased Ki67/NG2, Ki67/CNPase double-labeled cells along the boundary zone of ischemic cortex and striatum. Meanwhile, TSTT treatment up-regulated the phosphorylation level of Ser 9 in GSK-3β, and down-regulated phosphorylated β-catenin and CRMP-2 expression. CONCLUSION Taken together, our findings indicated that TSTT (65, 33 mg/kg) enhanced post-stroke functional recovery, amplified endogenous oligodendrogenesis and promoted axonal regeneration. The beneficial role of TSTT might be correlated with GSK-3/β-catenin/CRMP-2 modulating axonal reorganization after ischemic stroke.
Collapse
Affiliation(s)
- Le Yang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| | - Chang-Yi Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| | - Jun-Yao Ouyang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| | - Man-Zhong Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| | - Yu Zhan
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| | - Xue-Feng Feng
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| | - Yun Lu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| | - Ming-Cong Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| | - Jian-Feng Lei
- Medical Imaging laboratory of Core Facility Center, Capital Medical University, Beijing, 100069, China.
| | - Ting Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| | - Lei Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| | - Hai-Yan Zou
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| | - Hui Zhao
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China; Beijing Key Lab of TCM Collateral Disease Theory Research, Beijing, 100069, China.
| |
Collapse
|
12
|
Zhang Y, Li Y, Lin C, Zhang J, Gao H, Chen J. Dioscin-loaded zein nanoparticles alleviate lipopolysaccharide-induced acute kidney injury via the microRNA-let 7i signalling pathways. IET Nanobiotechnol 2021; 15:465-472. [PMID: 34694758 PMCID: PMC8675823 DOI: 10.1049/nbt2.12051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 08/01/2020] [Accepted: 08/17/2020] [Indexed: 11/22/2022] Open
Abstract
The present study investigates the potential role of dioscin (DIO) in the lipopolysaccharide (LPS)-induced kidney injury. For this purpose, DIO-loaded zein nanoparticles (DIO-ZNPs) were formulated and evaluated for physicochemical parameters. The DIO-ZNPs exhibited a controlled release of drug compared with that of the free drug suspension. Results showed that the cell viability of NRK-52E consistently decreased with the increase in LPS from 0.01 µg/ml to 2 µg/ml. When compared with LPS, DIO-induced NPs showed 1.10-, 1.32-, 1.57- and 1.92-fold increase in the cell viability for concentrations of 20 µg/ml, 50 µg/ml, 100 µg/ml and 200 µg/ml, respectively. DIO-ZNPs exhibited the most remarkable recovery in the cell proliferation compared with free DIO as shown by the cellular morphology analysis. Furthermore, Annexin-V staining analysis showed that the LPS-treated cells possess the lowest green fluorescence indicating fewer viable cells, whereas DIO-ZNPs exhibited the maximum green fluorescence comparable with that of the non-treated cells indicating maximum cell viability. Furthermore, the results show that DIO-ZNPs significantly increased the expression of miR-let-7i in the epithelial kidney cells, whereas the expression levels of TLR4 were significantly downregulated compared with that of the LPS-treated cells. In conclusion, miR-let-7i could be an interesting therapeutic target and nanoparticle-based DIO could be a potential candidate in the management of acute kidney injury.
Collapse
Affiliation(s)
- Yun Zhang
- Department of Renal MedicineSecond Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Yuangen Li
- Department of Renal MedicineSecond Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Changda Lin
- Department of Renal MedicineSecond Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Jiequn Zhang
- Department of Renal MedicineSecond Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Hanyuan Gao
- Department of Renal MedicineSecond Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| | - Jinhai Chen
- Department of Renal MedicineSecond Affiliated Hospital of Fujian Medical UniversityQuanzhouFujianChina
| |
Collapse
|
13
|
Guo Q, Chen M, Chen Q, Xiao G, Chen Z, Wang X, Huang Y. Silencing p53 inhibits interleukin 10-induced activated hepatic stellate cell senescence and fibrotic degradation in vivo. Exp Biol Med (Maywood) 2021; 246:447-458. [PMID: 33028080 PMCID: PMC7885051 DOI: 10.1177/1535370220960391] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/28/2020] [Indexed: 12/11/2022] Open
Abstract
Activated hepatic stellate cells are reported to play a significant role in liver fibrogenesis. Beside the phenotype reversion and apoptosis of activated hepatic stellate cells, the senescence of activated hepatic stellate cells limits liver fibrosis. Our previous researches have demonstrated that interleukin-10 could promote hepatic stellate cells senescence via p53 signaling pathway in vitro. However, the relationship between expression of p53 and senescence of activated hepatic stellate cells induced by interleukin-10 in fibrotic liver is unclear. The purpose of present study was to explore whether p53 plays a crucial role in the senescence of activated hepatic stellate cells and degradation of collagen mediated by interleukin-10. Hepatic fibrosis animal model was induced by carbon tetrachloride through intraperitoneal injection and transfection of interleukin-10 gene to liver was performed by hydrodynamic-based transfer system. Depletions of p53 in vivo and in vitro were carried out by adenovirus-based short hairpin RNA against p53. Regression of fibrosis was assessed by liver biopsy and collagen staining. Cellular senescence in the liver was observed by senescence-associated beta-galactosidase (SA-β-Gal) staining. Immunohistochemistry, immunofluorescence double staining, and Western blot analysis were used to evaluate the senescent cell and senescence-related protein expression. Our data showed that interleukin-10 gene treatment could lighten hepatic fibrosis induced by carbon tetrachloride and induce the aging of activated hepatic stellate cells accompanied by up-regulating the expression of aging-related proteins. We further demonstrated that depletion of p53 could abrogate up-regulation of interleukin-10 on the expression of senescence-related protein in vivo and vitro. Moreover, p53 knockout in fibrotic mice could block not only the senescence of activated hepatic stellate cells, but also the degradation of fibrosis induced by interleukin-10 gene intervention. Taken together, our results suggested that interleukin-10 gene treatment could attenuate carbon tetrachloride-induced hepatic fibrosis by inducing senescence of activated hepatic stellate cells in vivo, and this induction was closely related to p53 signaling pathway.
Collapse
Affiliation(s)
- Qilan Guo
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of Geriatrics, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Minghua Chen
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Qingduo Chen
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Guitao Xiao
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Zhixin Chen
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Xiaozhong Wang
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou 350001, China
| | - Yuehong Huang
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou 350001, China
- Department of Geriatrics, Fujian Medical University Union Hospital, Fuzhou 350001, China
| |
Collapse
|
14
|
Parama D, Boruah M, Yachna K, Rana V, Banik K, Harsha C, Thakur KK, Dutta U, Arya A, Mao X, Ahn KS, Kunnumakkara AB. Diosgenin, a steroidal saponin, and its analogs: Effective therapies against different chronic diseases. Life Sci 2020; 260:118182. [PMID: 32781063 DOI: 10.1016/j.lfs.2020.118182] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Chronic diseases are a major cause of mortality worldwide, and despite the recent development in treatment modalities, synthetic drugs have continued to show toxic side effects and development of chemoresistance, thereby limiting their application. The use of phytochemicals has gained attention as they show minimal side effects. Diosgenin is one such phytochemical which has gained importance for its efficacy against the life-threatening diseases, such as cardiovascular diseases, cancer, nervous system disorders, asthma, arthritis, diabetes, and many more. AIM To evaluate the literature available on the potential of diosgenin and its analogs in modulating different molecular targets leading to the prevention and treatment of chronic diseases. METHOD A detailed literature search has been carried out on PubMed for gathering information related to the sources, biosynthesis, physicochemical properties, biological activities, pharmacokinetics, bioavailability and toxicity of diosgenin and its analogs. KEY FINDINGS The literature search resulted in many in vitro, in vivo and clinical trials that reported the efficacy of diosgenin and its analogs in modulating important molecular targets and signaling pathways such as PI3K/AKT/mTOR, JAK/STAT, NF-κB, MAPK, etc., which play a crucial role in the development of most of the diseases. Reports have also revealed the safety of the compound and the adaptation of nanotechnological approaches for enhancing its bioavailability and pharmacokinetic properties. SIGNIFICANCE Thus, the review summarizes the efficacy of diosgenin and its analogs for developing as a potent drug against several chronic diseases.
Collapse
Affiliation(s)
- Dey Parama
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Monikongkona Boruah
- Cell and Molecular Biology Lab, Department of Zoology, Cotton University, Guwahati, Assam 781001, India
| | - Kumari Yachna
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Varsha Rana
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Kishore Banik
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Choudhary Harsha
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Krishan Kumar Thakur
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Uma Dutta
- Cell and Molecular Biology Lab, Department of Zoology, Cotton University, Guwahati, Assam 781001, India
| | - Aditya Arya
- Department of Pharmacology and Therapeutics, School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Xinliang Mao
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, China; Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India.
| |
Collapse
|
15
|
Hu HH, Cao G, Wu XQ, Vaziri ND, Zhao YY. Wnt signaling pathway in aging-related tissue fibrosis and therapies. Ageing Res Rev 2020; 60:101063. [PMID: 32272170 DOI: 10.1016/j.arr.2020.101063] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 02/25/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023]
Abstract
Fibrosis is the final hallmark of pathological remodeling, which is a major contributor to the pathogenesis of various chronic diseases and aging-related organ failure to fully control chronic wound-healing and restoring tissue function. The process of fibrosis is involved in the pathogenesis of the kidney, lung, liver, heart and other tissue disorders. Wnt is a highly conserved signaling in the aberrant wound repair and fibrogenesis, and sustained Wnt activation is correlated with the pathogenesis of fibrosis. In particular, mounting evidence has revealed that Wnt signaling played important roles in cell fate determination, proliferation and cell polarity establishment. The expression and distribution of Wnt signaling in different tissues vary with age, and these changes have key effects on maintaining tissue homeostasis. In this review, we first describe the major constituents of the Wnt signaling and their regulation functions. Subsequently, we summarize the dysregulation of Wnt signaling in aging-related fibrotic tissues such as kidney, liver, lung and cardiac fibrosis, followed by a detailed discussion of its involvement in organ fibrosis. In addition, the crosstalk between Wnt signaling and other pathways has the potential to profoundly add to the complexity of organ fibrosis. Increasing studies have demonstrated that a number of Wnt inhibitors had the potential role against tissue fibrosis, specifically in kidney fibrosis and the implications of Wnt signaling in aging-related diseases. Therefore, targeting Wnt signaling might be a novel and promising therapeutic strategy against aging-related tissue fibrosis.
Collapse
Affiliation(s)
- He-He Hu
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Xia-Qing Wu
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, School of Medicine, University of California Irvine, Irvine, California, 92897, USA
| | - Ying-Yong Zhao
- Faculty of Life Science & Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China.
| |
Collapse
|
16
|
Zhang Q, Chang X, Wang H, Liu Y, Wang X, Wu M, Zhan H, Li S, Sun Y. TGF-β1 mediated Smad signaling pathway and EMT in hepatic fibrosis induced by Nano NiO in vivo and in vitro. ENVIRONMENTAL TOXICOLOGY 2020; 35:419-429. [PMID: 31737983 DOI: 10.1002/tox.22878] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 10/20/2019] [Accepted: 10/26/2019] [Indexed: 06/10/2023]
Abstract
Nickel oxide nanoparticles (Nano NiO) bears hepatotoxicity, while whether it leads to liver fibrosis remains unclear. The aim of this study was to establish the Nano NiO-induced hepatic fibrosis model in vivo and investigate the roles of transforming growth factor β1 (TGF-β1) in Smad pathway activation, epithelial-mesenchymal transition (EMT) occurrence, and extracellular matrix (ECM) deposition in vitro. Male Wistar rats were exposed to 0.015, 0.06, and 0.24 mg/kg Nano NiO by intratracheal instilling twice a week for 9 weeks. HepG2 cells were treated with 100 μg/mL Nano NiO and TGF-β1 inhibitor (SB431542) to explore the mechanism of collagen formation. Results of Masson staining as well as the elevated levels of type I collagen (Col-I) and Col-III suggested that Nano NiO resulted in hepatic fibrosis in rats. Furthermore, Nano NiO increased the protein expression of TGF-β1, p-Smad2, p-Smad3, alpha-smooth muscle actin (α-SMA), matrix metalloproteinase9 (MMP9), and tissue inhibitors of metalloproteinase1 (TIMP1), while decreased the protein content of E-cadherin and Smad7 in rat liver and HepG2 cells. Most importantly, Nano NiO-triggered the abnormal expression of the abovementioned proteins were all alleviated by co-treatment with SB431542, implying that TGF-β1-mediated Smad pathway, EMT and MMP9/TIMP1 imbalance were involved in overproduction of collagen in HepG2 cells. In conclusion, these findings indicated that Nano NiO induced hepatic fibrosis via TGF-β1-mediated Smad pathway activation, EMT occurrence, and ECM deposition.
Collapse
Affiliation(s)
- Qiong Zhang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xuhong Chang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Haibing Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Yunlan Liu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Xiaoxia Wang
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Minmin Wu
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Haibing Zhan
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| | - Sheng Li
- Department occupational disease control, Lanzhou Municipal Center for Disease Control, Lanzhou, China
| | - Yingbiao Sun
- Department of Toxicology, School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
17
|
Zygophyllum album leaves extract prevented hepatic fibrosis in rats, by reducing liver injury and suppressing oxidative stress, inflammation, apoptosis and the TGF-β1/Smads signaling pathways. Exploring of bioactive compounds using HPLC-DAD-ESI-QTOF-MS/MS. Inflammopharmacology 2020; 28:1735-1750. [PMID: 32206981 DOI: 10.1007/s10787-020-00700-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 03/10/2020] [Indexed: 12/12/2022]
Abstract
Zygophyllum album is traditionally used against many illnesses, such as liver disease. The present study investigated the bioactive compounds in methanol extract of Z. album (MEZA) using HPLC-DAD-ESI-QTOF-MS/MS and explored its possible antioxidative, anti-inflammatory, anti-apoptotic, and hepatoprotective effect. Twelve phenolic compounds were identified; isorhamnetin-3-O-rutinoside being the main one was the main composite (144.6 mg/100 g dm). Results showed that MEZA reduced significantly the biochemical markers (AST, ALT, LDH and ALP), and the hepatic oxidative stress indicators (MDA, PC, SOD, CAT, and GPx) in deltamethrin (DLM)-treated rats. Moreover, MEZA limited the inflammatory responses through downregulation of NF-κB gene, which suppressed the production of proinflammatory cytokines (TNF-α, IL-1β, IL-6). Furthermore, Z. album reduced DLM-induced apoptosis by attenuating caspase 3 and p53 mRNA activation. MEZA treatment also alleviated upregulation of α-SMA, type I collagen, and TGF-β1 mRNA in the liver. The possible antifibrotic effect of MEZA was clearly demonstrated by the histopathology examination, using Masson's Trichrome and Sirius Red stainings. Therefore, the current study suggested that the bioactive compounds of Z. album possessed antifibrotic effect against DLM-induced hepatic fibrosis, by protecting liver tissue, and inhibiting oxidative stress, inflammation, apoptosis and the TGF-β1/Smads signaling pathways.
Collapse
|
18
|
Piotrowska-Kempisty H, Nowicki M, Jodynis-Liebert J, Kurpik M, Ewertowska M, Adamska T, Oszmiański J, Kujawska M. Assessment of Hepatoprotective Effect of Chokeberry Juice in Rats Treated Chronically with Carbon Tetrachloride. Molecules 2020; 25:1268. [PMID: 32168847 PMCID: PMC7144002 DOI: 10.3390/molecules25061268] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/19/2022] Open
Abstract
The aim of this study was to compare the protective effects of chokeberry juice and silymarin against chemical-induced liver fibrosis in rats. Liver fibrosis was induced by CCl4 administered two days a week for six weeks. Two groups of rats were co-treated with chokeberry juice, 10 mL/kg/day. or silymarin as a positive control, 100 mg/kg/day for six weeks. Hepatic lipid peroxidation was suppressed by 50% and the activity of hepatic antioxidant enzymes was increased by 19%-173% in rats co-treated with CCl4 and substances tested as compared to rats administered CCl4 alone. Hepatic hydroxyproline was decreased by 24% only in rats treated with silymarin. The messenger RNA (mRNA) expression levels of fibrosis-related molecules, procollagen I, α-SMA, TIMP-1, TGFβ, and TNFα, which were significantly increased in the liver of CCl4-treated rats, were not modulated by substances tested. Histological evaluation revealed a slight protective effect of silymarin against fibrosis. However, in CCl4 + chokeberry-treated rats, the density of vacuolated hepatocytes was significantly lower than that in silymarin administered animals. Chokeberry juice did not demonstrate an antifibrotic effect in the applied experimental model of fibrosis, and the effect of the known antifibrotic agent, silymarin, was very limited.
Collapse
Affiliation(s)
- Hanna Piotrowska-Kempisty
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznań, Poland; (H.P.-K.); (J.J.-L.); (M.K.); (M.E.); (T.A.)
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland;
| | - Jadwiga Jodynis-Liebert
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznań, Poland; (H.P.-K.); (J.J.-L.); (M.K.); (M.E.); (T.A.)
| | - Monika Kurpik
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznań, Poland; (H.P.-K.); (J.J.-L.); (M.K.); (M.E.); (T.A.)
| | - Małgorzata Ewertowska
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznań, Poland; (H.P.-K.); (J.J.-L.); (M.K.); (M.E.); (T.A.)
| | - Teresa Adamska
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznań, Poland; (H.P.-K.); (J.J.-L.); (M.K.); (M.E.); (T.A.)
| | - Jan Oszmiański
- Department of Fruit, Vegetable and Cereal Technology, Environmental and Life Science University, 51-630 Wrocław, Poland;
| | - Małgorzata Kujawska
- Department of Toxicology, Poznan University of Medical Sciences, 60-631 Poznań, Poland; (H.P.-K.); (J.J.-L.); (M.K.); (M.E.); (T.A.)
| |
Collapse
|
19
|
Yin F, Zhou H, Fang Y, Li C, He Y, Yu L, Wan H, Yang J. Astragaloside IV alleviates ischemia reperfusion-induced apoptosis by inhibiting the activation of key factors in death receptor pathway and mitochondrial pathway. JOURNAL OF ETHNOPHARMACOLOGY 2020; 248:112319. [PMID: 31639488 DOI: 10.1016/j.jep.2019.112319] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 10/06/2019] [Accepted: 10/16/2019] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Apoptosis plays an important role in cerebral ischemia-reperfusion injury and triggers a series of pathological changes which may even be life-threatening. Astragaloside-IV (AS-IV), a natural compound extracted from Astragalus (Astragalus membranaceus (Fisch.) Bunge., Leguminosae, Huangqi in Chinese), showed neuroprotective effects in the study of cerebral ischemia-reperfusion injury. In this study we investigate the effects of AS-IV on apoptosis induced by transient cerebral ischemia and reperfusion in rats, as well as the associated regulatory factors. METHODS AS-IV was administrated to male Sprague-Dawley (SD) rats after transient cerebral ischemia and reperfusion surgery (12.5, 25, and 50 mg/kg, once per day, continued for 7 days after surgey). After seven days of continuous administration, neurological function, cerebral infarction volume, and pathological changes of brain tissue were detected. Fas, FasL, Caspase-8, Bax, and Bcl-2 mRNA levels were determined by real-time PCR. Caspase-8, Bid, Cytochrome C (Cyto C), cleaved Caspase-3 proteins were determined by western blot and immunohistochemistry was used to quantify Cyto C. RESULTS AS-IV significantly attenuated the neurological deficit in rats with ischemica-reperfusion injury, and reduced cerebral infarction and neuronal apoptosis. AS-IV inhibited the mRNA upregulation of Fas, FasL, Caspase-8, and Bax/Bcl-2. Furthermore, the protein level of apoptosis cytokines Caspase-8, Bid, cleaved Caspase-3 and Cyto C were also inhibited after ischemia reperfusion, suggesting that AS-IV might alleviate ischemia reperfusion-induced apoptosis by inhibiting the activation of key factors in death receptor pathway and mitochondrial pathway.
Collapse
Affiliation(s)
- Fei Yin
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Huifen Zhou
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yuchen Fang
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chang Li
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yu He
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Li Yu
- Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Haitong Wan
- Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Jiehong Yang
- Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
20
|
Wang C, Li Q, Li T. Dioscin alleviates lipopolysaccharide-induced acute lung injury through suppression of TLR4 signaling pathways. Exp Lung Res 2020; 46:11-22. [PMID: 31931639 DOI: 10.1080/01902148.2020.1711830] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Aim: Acute lung injury (ALI) is a life-threatening inflammatory syndrome that lacks an effective therapy. Dioscin, a natural steroid saponin isolated from a variety of herbs, could serve as an anti-inflammatory agent, as suggested in previous reports. The purpose of this study was to explore the effects of dioscin on lipopolysaccharide (LPS)-induced ALI and validate the potential mechanisms.Materials and Methods: An ALI model was induced by intratracheal administration of LPS. Dioscin (20, 40, and 80 mg/kg) was administered intragastrically once daily for seven consecutive days prior to LPS challenge.Results: Our data revealed that dioscin significantly suppressed LPS-induced lung pathological changes, pulmonary capillary permeability, pulmonary edema, inflammatory cell infiltration, myeloperoxidase (MPO) activity, and cytokine production, including tumor necrosis factor (TNF)-α, interleukin (IL)-6, and keratinocyte chemoattractant (KC). Moreover, dioscin inhibited LPS-induced nuclear factor-kappaB (NF-κB) activation as well as Toll-like receptor 4 (TLR4) expression.Conclusions: In brief, the results indicated that dioscin alleviates LPS-induced ALI through suppression of TLR4 signaling pathways.
Collapse
Affiliation(s)
- Chuntao Wang
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingnian Li
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Li
- Trauma Center/Department of Emergency and Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Choi JH, Kim SM, Lee GH, Jin SW, Lee HS, Chung YC, Jeong HG. Platyconic Acid A, Platycodi Radix-Derived Saponin, Suppresses TGF-1-induced Activation of Hepatic Stellate Cells via Blocking SMAD and Activating the PPAR Signaling Pathway. Cells 2019; 8:cells8121544. [PMID: 31795488 PMCID: PMC6952772 DOI: 10.3390/cells8121544] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/28/2019] [Accepted: 11/28/2019] [Indexed: 02/06/2023] Open
Abstract
Platycodi radix is a widely sold health food worldwide, which contains numerous phytochemicals that are beneficial to health. Previously, we reported that saponin from the roots of Platycodi radix-derived saponin inhibited toxicant-induced liver diseases. Nevertheless, the inhibitory effect of platyconic acid A (PA), the active component of Platycodi radix-derived saponin, on the anti-fibrotic activity involving the SMAD pathway remains unclear. We investigated the inhibitory effects of PA on TGF-β1-induced activation of hepatic stellate cells (HSCs). PA inhibited TGF-β1-enhanced cell proliferation, as well as expression of α-SMA and collagen Iα1 in HSC-T6 cells. PA suppressed TGF-β1-induced smad2/3 phosphorylation and smad binding elements 4 (SBE4) luciferase activity. Reversely, PA restored TGF-β1-reduced expression of smad7 and peroxisome proliferator-activated receptor (PPAR)γ. PA also repressed TGF-β1-induced phosphorylation of Akt and MAPKs. In summary, the results suggest that the inhibitory effect of PA on HSCs occurs through the blocking of SMAD-dependent and SMAD-independent pathways, leading to the suppression of α-SMA and collagen Iα1 expression.
Collapse
Affiliation(s)
- Jae Ho Choi
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (J.H.C.); (S.M.K.); (G.H.L.); (S.W.J.)
| | - Seul Mi Kim
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (J.H.C.); (S.M.K.); (G.H.L.); (S.W.J.)
| | - Gi Ho Lee
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (J.H.C.); (S.M.K.); (G.H.L.); (S.W.J.)
| | - Sun Woo Jin
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (J.H.C.); (S.M.K.); (G.H.L.); (S.W.J.)
| | - Hyun Sun Lee
- Natural Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology, Ochang 28116, Korea;
| | - Young Chul Chung
- Department of Food and Medicine, College of Public Health and Natural Science, International University of Korea, Jinju 52833, Korea;
| | - Hye Gwang Jeong
- Department of Toxicology, College of Pharmacy, Chungnam National University, Daejeon 34134, Korea; (J.H.C.); (S.M.K.); (G.H.L.); (S.W.J.)
- Correspondence: ; Tel.: +82-42-821-5936
| |
Collapse
|
22
|
Wu S, Zhao F, Zhao J, Li H, Chen J, Xia Y, Wang J, Zhao B, Zhao S, Li N. Dioscin improves postmenopausal osteoporosis through inducing bone formation and inhibiting apoptosis in ovariectomized rats. Biosci Trends 2019; 13:394-401. [PMID: 31611520 DOI: 10.5582/bst.2019.01186] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Shan Wu
- The Second Hospital of Jilin University, Changchun, Jilin province, China
| | - Fan Zhao
- China-Japan Union Hospital of Jilin University, Changchun, Jilin province, China
| | - Jing Zhao
- The Second Hospital of Jilin University, Changchun, Jilin province, China
| | - Hui Li
- Qian Wei Hospital of Jilin Province, Changchun, Jilin province, China
| | - Junyu Chen
- The Second Hospital of Jilin University, Changchun, Jilin province, China
| | - Yang Xia
- The Second Hospital of Jilin University, Changchun, Jilin province, China
| | - Junwei Wang
- The Second Hospital of Jilin University, Changchun, Jilin province, China
| | - Benzheng Zhao
- The Second Hospital of Jilin University, Changchun, Jilin province, China
| | - Shuhua Zhao
- The Second Hospital of Jilin University, Changchun, Jilin province, China
| | - Na Li
- Jilin Ginseng Academic, Changchun University of Chinese Medicine, Changchun, Jilin province, China
| |
Collapse
|
23
|
Cao G, Zhu R, Jiang T, Tang D, Kwan HY, Su T. Danshensu, a novel indoleamine 2,3-dioxygenase1 inhibitor, exerts anti-hepatic fibrosis effects via inhibition of JAK2-STAT3 signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 63:153055. [PMID: 31377585 DOI: 10.1016/j.phymed.2019.153055] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 07/03/2019] [Accepted: 07/26/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Indoleamine 2,3-dioxygenase 1 (IDO1), an important intracellular rate-limiting enzyme in the development of Hepatic fibrosis (HF), and has been proposed as a hallmark of HF. Danshensu (DSS) is a major bioactive component that isolated from a edible traditional Chinese medicinal herb Salviae Miltiorrhizae Radix et Rhizoma (Danshen), while, the anti-HF mode and mechanism of action of DSS have not been fully elucidated. METHODS Carbon tetrachloride (CCl4)-induced rat HF model and TGF-β1-induced hepatic stellate cell (HSC) model were employed to assess the in vivo and in vitro anti-HF effects of DSS. HSC-T6 cells stably expressing IDO1, a constitutively active IDO1 mutant, was used to determine the role of JAK2-STAT3 signaling in the DSS's anti-HF effects. RESULTS We found that intragastric administration of DSS potently reduced fibrosis, inhibited IDO1 expression and STAT3 activity both in vitro and in vivo. Using molecular docking and molecular dynamics analysis, DSS was identified as a novel IDO1 inhibitor. Mechanistic studies indicated that DSS inhibited JAK2-STAT3 signaling, it reduced IDO1 expression, STAT3 phosphorylation and STAT3 nuclear localization. More importantly, overexpression of IDO1 diminished DSS's anti-HF effects. CONCLUSION Our findings provide a pharmacological justification for the clinical use of DSS in treating HF, and suggest that DSS has the potential to be developed as a modern alternative and/or complimentary agent for HF treatment and prevention.
Collapse
Affiliation(s)
- Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ruyi Zhu
- School of Pharmacy, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ting Jiang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dongxin Tang
- First Affiliated Hospital of Guiyang College of Traditional Chinese Medicine, Guiyang, China
| | - Hiu Yee Kwan
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Tao Su
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
24
|
Song S, Chu L, Liang H, Chen J, Liang J, Huang Z, Zhang B, Chen X. Protective Effects of Dioscin Against Doxorubicin-Induced Hepatotoxicity Via Regulation of Sirt1/FOXO1/NF-κb Signal. Front Pharmacol 2019; 10:1030. [PMID: 31572199 PMCID: PMC6753638 DOI: 10.3389/fphar.2019.01030] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022] Open
Abstract
Doxorubicin (Dox), an antitumor antibiotic, has therapeutic effects on many kinds of tumors. However, Dox can produce some serious side effects that limit its clinical application. Thus, exploration of effective drug targets or active lead compounds against Dox-induced organ damage is necessary. Dioscin, one natural product, has potent effects against Dox-induced renal injury and cardiotoxicity. However, the effects of dioscin on Dox-induced hepatotoxicity have not been reported. In this study, the results showed that dioscin significantly ameliorated Dox-induced cell injury, reduced reactive oxygen species (ROS) level, and suppressed cell apoptosis in alpha mouse liver 12 (AML-12) cells caused by Dox. In vivo, dioscin evidently decreased the levels of alanine transaminase (ALT), aspartate transaminase (AST), malondialdehyde (MDA); increased the levels of superoxide dismutase (SOD), glutathione (GSH), and glutathione peroxidase (GSH-Px); and alleviated liver injury. Mechanism study showed that dioscin remarkably up-regulated the expression levels of silent information regulator 1 (Sirt1) and heme oxygenase-1 (HO-1) via increase of the nuclear translocation of NF-E2-related factor 2 (Nrf2) and suppressed the expression levels of forkhead box protein O1 (FOXO1) and kelch-like ECH-associated protein-1 (Keap1) to inhibit oxidative stress. Furthermore, dioscin obviously decreased the nuclear translocation of nuclear factor κB (NF-κB) and the mRNA levels of tumor necrosis factor alpha (TNF-α), interleukin 1β (IL-1β), and interleukin 6 (IL-6) to suppress inflammation. Meanwhile, dioscin significantly regulated tumor suppressor P53 (P53) expression level and BCL-2-associated X (BAX)/BCL-2 apoptosis regulator (BCL-2) ratio to inhibit cell apoptosis. These results were further validated by knockdown of Sirt1 using siRNA silencing in AML-12 cells, which confirmed that the target of dioscin against Dox-induced hepatotoxicity was Sirt1/FOXO1/NF-κB signal. In short, our findings showed that dioscin exhibited protective effects against Dox-induced liver damage via suppression of oxidative stress, inflammation, and apoptosis, which should be developed as one new candidate for the prevention of Dox-induced liver injury in the future.
Collapse
Affiliation(s)
- Shasha Song
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Liang Chu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Jin Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Junnan Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Zhao Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, China
| |
Collapse
|
25
|
Yang L, Ren S, Xu F, Ma Z, Liu X, Wang L. Recent Advances in the Pharmacological Activities of Dioscin. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5763602. [PMID: 31511824 PMCID: PMC6710808 DOI: 10.1155/2019/5763602] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/28/2019] [Indexed: 02/07/2023]
Abstract
Dioscin is a typical saponin with multiple pharmacological activities. The past few years have seen an emerging interest in and growing research on this pleiotropic saponin. Here, we review the emerging pharmacological activities reported recently, with foci on its antitumor, antimicrobial, anti-inflammatory, antioxidative, and tissue-protective properties. The potential use of dioscin in therapies of diverse clinical disorders is also discussed.
Collapse
Affiliation(s)
- Longfei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetics, The Second Hospital of Jilin University, Changchun 130041, China
| | - Shengnan Ren
- Department of Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Fei Xu
- Department of Acupuncture and Moxibustion, The Second Hospital of Jilin University, Changchun 130041, China
| | - Zhiming Ma
- Department of Gastrointestinal Nutrition and Hernia Surgery, The Second Hospital of Jilin University, Changchun 130041, China
| | - Xin Liu
- Eye Center, The Second Hospital of Jilin University, Changchun 130024, China
| | - Lufei Wang
- Eye Center, The Second Hospital of Jilin University, Changchun 130024, China
| |
Collapse
|
26
|
Knockout of α-calcitonin gene-related peptide attenuates cholestatic liver injury by differentially regulating cellular senescence of hepatic stellate cells and cholangiocytes. J Transl Med 2019; 99:764-776. [PMID: 30700848 PMCID: PMC6570540 DOI: 10.1038/s41374-018-0178-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/22/2018] [Accepted: 12/06/2018] [Indexed: 12/13/2022] Open
Abstract
α-Calcitonin gene-related peptide (α-CGRP) is a 37-amino acid neuropeptide involved in several pathophysiological processes. α-CGRP is involved in the regulation of cholangiocyte proliferation during cholestasis. In this study, we aimed to evaluate if α-CGRP regulates bile duct ligation (BDL)-induced liver fibrosis by using a α-CGRP knockout (α-CGRP-/-) mouse model. α-CGRP-/- and wild-type (WT) mice were subjected to sham surgery or BDL for 7 days. Then, liver fibrosis and cellular senescence as well as the expression of kinase such as p38 and C-Jun N-terminal protein kinase (JNK) in mitogen-activated protein kinases (MAPK) signaling pathway were evaluated in total liver, together with measurement of cellular senescence in cholangiocytes or hepatic stellate cells (HSCs). There was enhanced hepatic expression of Calca (coding α-CGRP) and the CGRP receptor components (CRLR, RAMP-1 and RCP) in BDL and in both WT α-CGRP-/- and BDL α-CGRP-/- mice, respectively. Moreover, there was increased CGRP serum levels and hepatic mRNA expression of CALCA and CGRP receptor components in late-stage PSC samples compared to healthy control samples. Depletion of α-CGRP reduced liver injury and fibrosis in BDL mice that was associated with enhanced cellular senescence of hepatic stellate cells and reduced senescence of cholangiocytes as well as decreased activation of p38 and JNK MAPK signaling pathway. Cholangiocyte supernatant from BDL α-CGRP-/- mice inhibited the activation and increased cellular senescence of cultured human HSCs (HHSCs) compared to HHSCs stimulated with BDL cholangiocyte supernatant. Taken together, endogenous α-CGRP promoted BDL-induced cholestatic liver fibrosis through differential changes in senescence of HSCs and cholangiocytes and activation of p38 and JNK signaling. Modulation of α-CGRP/CGRP receptor signaling may be key for the management of biliary senescence and liver fibrosis in cholangiopathies.
Collapse
|
27
|
Yu Z, Jv Y, Cai L, Tian X, Huo X, Wang C, Zhang B, Sun C, Ning J, Feng L, Zhang H, Ma X. Gambogic acid attenuates liver fibrosis by inhibiting the PI3K/AKT and MAPK signaling pathways via inhibiting HSP90. Toxicol Appl Pharmacol 2019; 371:63-73. [PMID: 30953615 DOI: 10.1016/j.taap.2019.03.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 03/07/2019] [Accepted: 03/31/2019] [Indexed: 12/17/2022]
Abstract
Gambogic acid (GA), a major ingredient of Garcinia hanburryi, is known to have diverse biological effects. The present study was designed to evaluate the anti-fibrotic effects of GA on hepatic fibrosis and reveal its underlying mechanism. We investigated the anti-fibrotic effect of GA on dimethylnitrosamine and bile duct ligation induced liver fibrosis in rats in vivo. The rat and human hepatic stellate cell lines (HSCs) lines were chose to evaluate the effect of GA in vitro. Our results indicated that GA could significantly ameliorate liver fibrosis associated with improving serum markers, decrease in extracellular matrix accumulation and HSCs activation in vivo. GA significantly inhibited the proliferation of HSC cells and induced the cell cycle arrest at the G1 phase. Moreover, GA triggered autophagy at early time point and subsequent initiates mitochondrial mediated apoptotic pathway resulting in HSC cell death. The mechanism of GA was related to inhibit heat shock protein 90 (HSP90) and degradation of the client proteins inducing PI3K/AKT and MAPK signaling pathways inhibition. This study demonstrated that GA effectively ameliorated liver fibrosis in vitro and in vivo, which provided new insights into the application of GA for liver fibrosis.
Collapse
Affiliation(s)
- Zhenlong Yu
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Yanan Jv
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Lu Cai
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Xiangge Tian
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Xiaokui Huo
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Chao Wang
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Baojing Zhang
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - ChengPeng Sun
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Jing Ning
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Lei Feng
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Houli Zhang
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China.
| | - Xiaochi Ma
- College of Pharmacy, Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
28
|
Mao Z, Han X, Chen D, Xu Y, Xu L, Yin L, Sun H, Qi Y, Fang L, Liu K, Peng J. Potent effects of dioscin against hepatocellular carcinoma through regulating TP53-induced glycolysis and apoptosis regulator (TIGAR)-mediated apoptosis, autophagy, and DNA damage. Br J Pharmacol 2019; 176:919-937. [PMID: 30710454 PMCID: PMC6433650 DOI: 10.1111/bph.14594] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 12/05/2018] [Accepted: 12/18/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE Dioscin shows potent effects against cancers. We aimed to elucidate its pharmacological effects and mechanisms of action on hepatocellular carcinoma (HCC) in vivo and in vitro. EXPERIMENTAL APPROACH Effects of dioscin were investigated in SMMC7721 and HepG2 cells, diethylnitrosamine-induced primary liver cancer in rats, and cell xenografts in nude mice. Isobaric tags for relative and absolution quantitation (iTRAQ)-based proteomics was used to find dioscin's targets and investigate its mechanism. KEY RESULTS In SMMC7721 and HepG2 cells dioscin markedly inhibited cell proliferation and migration, induced apoptosis, autophagy, and DNA damage. It inhibited DEN-induced primary liver cancer in rats, markedly changed body weights and restored levels of α fetoprotein, alanine transaminase, aspartate transaminase, γ-glutamyltransferase, alkaline phosphatase, and Ki67. It also inhibited growth of xenografts in mice. In SMMC7721 cells, 191 differentially expressed proteins were found after dioscin, based on iTRAQ-based assay. TP53-inducible glycolysis and apoptosis regulator (TIGAR) was identified as being significantly down-regulated by dioscin. Dioscin induced cell apoptosis, autophagy, and DNA damage via increasing expression levels of p53, cleaved PARP, Bax, cleaved caspase-3/9, Beclin-1, and LC3 and suppressing those of Bcl-2, p-Akt, p-mammalian target of rapamycin (mTOR), CDK5, p-ataxia telangiectasia-mutated gene (ATM). The transfection of TIGAR siRNA into SMMC7721 cells and xenografts in nude mice further confirmed that the potent activity of dioscin against HCC is evoked by adjusting TIGAR-mediated inhibition of p53, Akt/mTOR, and CDK5/ATM pathways. CONCLUSIONS AND IMPLICATIONS The data suggest that dioscin has potential as a therapeutic, and TIGAR as a drug target for treating HCC.
Collapse
Affiliation(s)
- Zhang Mao
- College of PharmacyDalian Medical UniversityDalianChina
| | - Xu Han
- College of PharmacyDalian Medical UniversityDalianChina
| | - Dahong Chen
- College of PharmacyDalian Medical UniversityDalianChina
| | - Youwei Xu
- College of PharmacyDalian Medical UniversityDalianChina
| | - Lina Xu
- College of PharmacyDalian Medical UniversityDalianChina
| | - Lianhong Yin
- College of PharmacyDalian Medical UniversityDalianChina
| | - Huijun Sun
- College of PharmacyDalian Medical UniversityDalianChina
| | - Yan Qi
- College of PharmacyDalian Medical UniversityDalianChina
| | - Lingling Fang
- College of PharmacyDalian Medical UniversityDalianChina
| | - Kexin Liu
- College of PharmacyDalian Medical UniversityDalianChina
| | - Jinyong Peng
- College of PharmacyDalian Medical UniversityDalianChina
- Key Laboratory for Basic and Applied Research on Pharmacodynamic Substances of Traditional Chinese Medicine of Liaoning ProvinceDalian Medical UniversityDalianChina
- National‐Local Joint Engineering Research Center for Drug Development (R&D) of Neurodegenerative DiseasesDalian Medical UniversityDalianChina
| |
Collapse
|
29
|
Cao YJ, Xu Y, Liu B, Zheng X, Wu J, Zhang Y, Li XS, Qi Y, Sun YM, Wen WB, Hou L, Wan CP. Dioscin, a Steroidal Saponin Isolated from Dioscorea nipponica, Attenuates Collagen-Induced Arthritis by Inhibiting Th17 Cell Response. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:423-437. [PMID: 30827153 DOI: 10.1142/s0192415x19500216] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Dioscin, a steroidal saponin isolated from Dioscorea nipponica Makino, has previously been shown to possess antiarthritic effects. However, the underlying mechanism is still elusive. Herein, we investigated the therapeutic effects of dioscin on collagen-induced arthritis (CIA) in DBA/1 mice and related mechanism. Cytokine production in CII-specific immune responses were measured by enzyme-linked immunosorbent assay (ELISA); Th17 cell-related gene expression, including IL-17A, ROR[Formula: see text] and IL-23p19, were detected by qPCR analysis; Surface marker, T regulatory (Treg) cells and intracellular cytokines (IL-17A and IFN-[Formula: see text]) were evaluated by flow cytometry. We performed Th17 cell differentiation assay in vitro. Results showed that, in vivo, dioscin treatment significantly reduced the severity of CIA, which was accompanied by decreased Th17 response, but not Th1 and Treg response; dioscin-treated mice also showed lower percentage of CD11b[Formula: see text] Gr-1[Formula: see text] neutrophils; In vitro, dioscin treatment suppressed the differentiation of naive CD4[Formula: see text] T cells into Th17 cell and decreased IL-17A production. Collectively, our results indicate that dioscin exerts antiarthritic effects by inhibiting Th17 cell immune response.
Collapse
Affiliation(s)
- Yong-Jun Cao
- Department of Rheumatology, Nantong Hospital Affiliated to Nanjing, University of Traditional Chinese Medicine, Nantong, Jiangsu 226001, P. R. China
| | - Ying Xu
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Bei Liu
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Xi Zheng
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Jian Wu
- Department of Rheumatology, Nantong Hospital Affiliated to Nanjing, University of Traditional Chinese Medicine, Nantong, Jiangsu 226001, P. R. China
| | - Ying Zhang
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Xiao-Si Li
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Yan Qi
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Yu-Meng Sun
- Department of Rheumatology, Nantong Hospital Affiliated to Nanjing, University of Traditional Chinese Medicine, Nantong, Jiangsu 226001, P. R. China
| | - Wei-Bo Wen
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| | - Lifei Hou
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Chun-Ping Wan
- The No. 1 Affiliated Hospital of Yunnan, University of Traditional Chinese Medicine, Kunming 650021, P. R. China
| |
Collapse
|
30
|
Tao X, Yin L, Xu L, Peng J. Dioscin: A diverse acting natural compound with therapeutic potential in metabolic diseases, cancer, inflammation and infections. Pharmacol Res 2018; 137:259-269. [PMID: 30315966 DOI: 10.1016/j.phrs.2018.09.022] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 09/19/2018] [Accepted: 09/20/2018] [Indexed: 01/11/2023]
Abstract
Currently, the numbers of patients with cancer, fibrosis, diabetes, chronic kidney disease, stroke and osteoporosis are increasing fast and fast. It's critical necessary to discovery lead compounds for new drug development. Dioscin, one active compound in some medicinal plants, has anti-inflammation, immunoregulation, hypolipidemic, anti-viral, anti-fungal and anti-allergic effects. In recent years, dioscin has reached more and more attention with its potent effects to treat liver, kidney, brain, stomach and intestine damages, and metabolic diseases including diabetes, osteoporosis, obesity, hyperuricemia as well as its anti-cancer activities through adjusting multiple targets and multiple signals. Therefore, dioscin is a promising multi-target candidate to treat various diseases. This review paper summarized the progress on pharmacological activities and mechanisms of dioscin, which may provide useful data for development and exploration of this natural product in the further.
Collapse
Affiliation(s)
- Xufeng Tao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian, 116044, China.
| |
Collapse
|
31
|
Molecular mechanism and inhibitory targets of dioscin in HepG2 cells. Food Chem Toxicol 2018; 120:143-154. [DOI: 10.1016/j.fct.2018.07.016] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/22/2018] [Accepted: 07/06/2018] [Indexed: 12/16/2022]
|
32
|
Karimi J, Mohammadalipour A, Sheikh N, Khodadadi I, Hashemnia M, Goudarzi F, Khanjarsim V, Solgi G, Hajilooi M, Bahabadi M, Kheiripour N, Hedayatyanfard K. Protective effects of combined Losartan and Nilotinib on carbon tetrachloride (CCl 4)-induced liver fibrosis in rats. Drug Chem Toxicol 2018; 43:468-478. [PMID: 30207194 DOI: 10.1080/01480545.2018.1504960] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tyrosine kinase inhibitors (TKIs) have been developed as therapeutic compounds for inhibiting the progression of liver fibrosis. In the present study, the simultaneous treatment of Nilotinib (TKIs) and Losartan was studied. Forty rats were divided into eight groups of fibrosis induced by carbon tetrachloride (CCl4) and therapeutics (Nilotinib, Losartan, and combination therapy). In the end, serum parameters of the liver and gene expression analysis of transforming growth factor-β1, its receptors (TβRII), platelet-derived growth factor, its receptors (PDGFRβ), matrix metalloproteinases (MMP-2 and MMP-9), tumor necrosis factor-α, cytochrome P450 2E1, and collagen1 type 1 were performed. The oxidant/antioxidant factors were also analyzed. Histopathology analysis along with α-SMA immunohistochemistry and hydroxyproline evaluation was also conducted for a more in-depth study. The overall results indicated a better therapeutic effect of co-treatment of Nilotinib-Losartan in comparison with the treatment of each of them alone. Interestingly, some gene and protein factors and fibrotic indices were reduced even to the normal levels of the control group. The results of this study suggest that co-administration of these two combinations, strengthens their anti-fibrotic properties and, due to the routine use of these compounds against AML and blood pressure, these compounds can be used with caution against human liver fibrosis.
Collapse
Affiliation(s)
- Jamshid Karimi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Adel Mohammadalipour
- Faculty of Pharmacy and Pharmaceutical Sciences, Department of Clinical Biochemistry, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Nasrin Sheikh
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Khodadadi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mohammad Hashemnia
- Veterinary Medicine Faculty, Departments of Pathobiology, Razi University, Kermanshah, Iran
| | - Farjam Goudarzi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Vahid Khanjarsim
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ghasem Solgi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehrdad Hajilooi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Majid Bahabadi
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nejat Kheiripour
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran
| | - Keshvad Hedayatyanfard
- Faculty of Medicine, Department of Pharmacology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
33
|
Lou D, Han J, Zhou L, Ma H, Xv J, Shou J, Xu Z, Jiang L, Qian Y. Fibroblast growth factor receptor 1 antagonism attenuates lipopolysaccharide-induced activation of hepatic stellate cells via suppressing inflammation. Exp Ther Med 2018; 16:2909-2916. [PMID: 30250515 PMCID: PMC6143916 DOI: 10.3892/etm.2018.6586] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 07/27/2018] [Indexed: 12/13/2022] Open
Abstract
Activated hepatic stellate cells (HSCs) serve key roles in hepatic fibrosis by producing excessive extracellular matrix (ECM) components. Lipopolysaccharide (LPS) has been found to be associated with hepatic fibrogenesis through direct interactions with HSCs. Recently, the fibroblast growth factor receptor 1 (FGFR1) signalling system was identified as a key player in the process of liver fibrosis. In the present study it was evaluated whether FGFR1 mediated LPS-induced HSCs activation. In cultured cells, FGFR1 was inhibited by either siRNA silencing or by a small-molecule inhibitor in LPS-stimulated HSCs. The blockade of FGFR1 decreased LPS-induced nuclear factor-κB (NF-κB) activation, inflammatory cytokine release, fibrosis, and cell proliferation in HSCs. It was further indicated that LPS triggered FGFR1 phosphorylation via TLR4/c-Src. These findings confirmed the detrimental effect of FGFR1 activation in the pathogenesis of LPS-related HSC activation and revealed that FGFR1 may be an ideal therapeutic target for LPS-induced liver fibrosis.
Collapse
Affiliation(s)
- Dayong Lou
- Medication Department, Zhuji People's Hospital of Zhejiang Province, Zhuji, Shaoxing, Zhejiang 311800, P.R. China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Liqin Zhou
- Medication Department, Zhuji People's Hospital of Zhejiang Province, Zhuji, Shaoxing, Zhejiang 311800, P.R. China
| | - Huanjie Ma
- Medication Department, Zhuji People's Hospital of Zhejiang Province, Zhuji, Shaoxing, Zhejiang 311800, P.R. China
| | - Jianjiang Xv
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Junwei Shou
- Medication Department, Zhuji People's Hospital of Zhejiang Province, Zhuji, Shaoxing, Zhejiang 311800, P.R. China
| | - Zhixiu Xu
- Medication Department, Zhuji People's Hospital of Zhejiang Province, Zhuji, Shaoxing, Zhejiang 311800, P.R. China
| | - Liqin Jiang
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang 314000, P.R. China
| | - Yuanyuan Qian
- Medication Department, Zhuji People's Hospital of Zhejiang Province, Zhuji, Shaoxing, Zhejiang 311800, P.R. China
| |
Collapse
|
34
|
Yao H, Tao X, Xu L, Qi Y, Yin L, Han X, Xu Y, Zheng L, Peng J. Dioscin alleviates non-alcoholic fatty liver disease through adjusting lipid metabolism via SIRT1/AMPK signaling pathway. Pharmacol Res 2018; 131:51-60. [PMID: 29574225 DOI: 10.1016/j.phrs.2018.03.017] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 02/09/2023]
Abstract
Dioscin, one natural product, has active effect against non-alcoholic fatty liver disease (NAFLD) in our previous work. However, the pharmacological data are insufficient and the mechanisms have not been reported. Thus, this study aims to comprehensively investigate the effects and molecular mechanisms of dioscin against NAFLD. The primary cultured hepatocytes, AML-12 and HepG-2 cells were treated with palmic acid (PA) after dioscin treatment. The mice and rats were induced by high fat diet to establish the in vivo models of NAFLD. Dioscin obviously alleviated liver lipid accumulation symptoms and improved the levels of serum and hepatic biochemical parameters in vitro and in vivo. Further investigations revealed that dioscin significantly attenuated lipid metabolism via adjusting SIRT1/AMPK signal pathway to regulate the expression levels of SREBP-1c, CPT, FAS, SCD, FoxO1 and ATGL. In addition, suppression of SIRT1 by Nicotinamide or abrogation of AMPK by Compound C eliminated the inhibitory effects of dioscin on lipid metabolism. Therefore, our findings further demonstrated that dioscin markedly prevented NAFLD through adjusting lipid metabolism via SIRT1/AMPK signal pathway, which should be developed as a new candidate for NAFLD.
Collapse
Affiliation(s)
- Hong Yao
- College of Pharmacy, Dalian Medical University, Western 9, Lvshunnan Road, Dalian, 116044, China
| | - Xufeng Tao
- College of Pharmacy, Dalian Medical University, Western 9, Lvshunnan Road, Dalian, 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9, Lvshunnan Road, Dalian, 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Western 9, Lvshunnan Road, Dalian, 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9, Lvshunnan Road, Dalian, 116044, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, Western 9, Lvshunnan Road, Dalian, 116044, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, Western 9, Lvshunnan Road, Dalian, 116044, China
| | - Lingli Zheng
- College of Pharmacy, Dalian Medical University, Western 9, Lvshunnan Road, Dalian, 116044, China; The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9, Lvshunnan Road, Dalian, 116044, China.
| |
Collapse
|
35
|
Ogaly HA, Eltablawy NA, Abd-Elsalam RM. Antifibrogenic Influence of Mentha piperita L. Essential Oil against CCl 4-Induced Liver Fibrosis in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4039753. [PMID: 29849890 PMCID: PMC5933010 DOI: 10.1155/2018/4039753] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 01/10/2018] [Accepted: 01/18/2018] [Indexed: 12/22/2022]
Abstract
Essential oils of some aromatic plants provide an effective nonmedicinal option to control liver fibrosis. Mentha piperita L. essential oil (MPEO) have been reported to possess protective effects against hepatotoxicity. However, its effect against liver fibrosis remains unknown. The present study investigated the antifibrogenic potential of MPEO and its underlying mechanisms. Forty male rats divided into 4 groups were used: group 1 served as normal control, group 2 (liver fibrosis) received CCl4 (2.5 mL/kg, IP, twice weekly) for 8 weeks, group 3 concurrently received CCl4 plus MPEO (50 mg/kg, IP, daily, from the 3rd week), and group 4 received MPEO only. MPOE significantly improved the liver injury markers, lipid peroxidation (LPO), antioxidant capacity, CYP2E1 gene expressionand liver histology. Furthermore, MPOE ameliorated liver fibrosis as evidenced by the reduced expression of desmin, α-smooth muscle actin (α-SMA), transforming growth factor-β1 (TGF-β1), and SMAD3 proteins. In addition, MPOE counteracted the p53 upregulation induced by CCl4 at both mRNA and protein levels. In conclusion, MPOE could effectively attenuate hepatic fibrosis mainly through improving the redox status, suppressing p53 and subsequently modulating TGF-β1 and SMAD3 protein expression. These data promote the use of MPOE as a promising approach in antifibrotic therapy.
Collapse
Affiliation(s)
- Hanan A. Ogaly
- Department of Chemistry, College of Sciences, King Khalid University, Abha, Saudi Arabia
| | - Nadia A. Eltablawy
- Biochemistry Division, National Organization for Drug Control and Research (NODCAR), Giza, Egypt
| | - Reham M. Abd-Elsalam
- Department of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
36
|
Oliveira DRDE, Nepomuceno DD, Castro RN, Braz R, Carvalho MGDE. Special metabolites isolated from Urochloa humidicola (Poaceae). AN ACAD BRAS CIENC 2018. [PMID: 28640339 DOI: 10.1590/0001-3765201720160126] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
This study aims to identify special metabolites in polar extracts from Urochloa humidicola (synonym Brachiaria humidicola) that have allelopathic effects and induce secondary photosensitization in ruminants. The compounds were isolated and identified via chromatographic and spectroscopic techniques. The compounds 4-hydroxy-3-methoxy-benzoic acid, trans-4-hydroxycinnamic acid, and p-hydroxy-benzoic acid; the flavonols isorhamnetin-3-O-β-d-glucopyranoside and methyl quercetin-3-O-β-d-glucuronate; and kaempferitrin, quercetin-3-O-α-l-rhamnopyranoside, and tricin were identified in the extract from the leaves of Urochloa humidicola. Two furostanic saponins, namely, dioscin and 3-O-α-l-rhamnopyranosyl-(1-4)-[α-l-rhamnopyranosyl-(1-2)]-β-d-glucopyranosyl-penogenin, as well as catechin-7-O-β-d-glucopyranoside were identified in the methanolic extract obtained from the roots of this plant. This species features a range of metabolites that may be toxic for animals if used in food and may interfere with the growth medium, thereby inhibiting the development of other species.
Collapse
Affiliation(s)
- Débora R DE Oliveira
- Universidade Federal Rural do Rio de Janeiro, Instituto de Ciências Exatas, Departamento de Química, Rodovia BR 465, Km 07, 23890-000 Seropédica, RJ, Brazil
| | - Delci D Nepomuceno
- Universidade Federal Rural do Rio de Janeiro, Instituto de Zootecnia, Departamento de Nutrição Animal e Pastagens, Rodovia BR 465, Km 07, 23890-000 Seropédica, RJ, Brazil
| | - Rosane N Castro
- Universidade Federal Rural do Rio de Janeiro, Instituto de Ciências Exatas, Departamento de Química, Rodovia BR 465, Km 07, 23890-000 Seropédica, RJ, Brazil
| | - Raimundo Braz
- Universidade Federal Rural do Rio de Janeiro, Instituto de Ciências Exatas, Departamento de Química, Rodovia BR 465, Km 07, 23890-000 Seropédica, RJ, Brazil.,Setor de Química de Produtos Naturais, Universidade Estadual do Norte Fluminense Darcy Ribeiro, 28013-600 Campos dos Goytacazes, RJ, Brazil
| | - Mário G DE Carvalho
- Universidade Federal Rural do Rio de Janeiro, Instituto de Ciências Exatas, Departamento de Química, Rodovia BR 465, Km 07, 23890-000 Seropédica, RJ, Brazil
| |
Collapse
|
37
|
Wang YC, Wu DW, Wu TC, Wang L, Chen CY, Lee H. Dioscin overcome TKI resistance in EGFR-mutated lung adenocarcinoma cells via down-regulation of tyrosine phosphatase SHP2 expression. Int J Biol Sci 2018; 14:47-56. [PMID: 29483824 PMCID: PMC5821048 DOI: 10.7150/ijbs.22209] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 12/14/2017] [Indexed: 11/23/2022] Open
Abstract
Resistance to tyrosine kinase inhibitors (TKIs) results in tumor relapse and poor prognosis in patients with lung adenocarcinoma. TKI resistance caused by epidermal growth factor receptor (EGFR) mutations at T790M and c-Met amplification occurs through persistent activation of the MEK/ERK and PI3K/AKT signaling pathways. We therefore expected that dual inhibitors of both signaling pathways could overcome TKI resistance in lung adenocarcinoma. Here, dioscin was selected from a product library of Chinese naturally occurring compounds and overcame TKI resistance in EGFR-mutated lung adenocarcinoma cells. Mechanistically, dioscin may down-regulate the expression of SH2 domain-containing phosphatase-2 (SHP2) at the transcription level by increasing p53 binding to the SHP2 promoter due to reactive oxygen species (ROS). Simultaneous inhibition of MEK/ERK and PI3K/AKT activation via decreased SHP2 expression and its interaction with GAB1 may be responsible for dioscin-mediated TKI sensitivity. A higher unfavorable response to TKI therapy occurred more commonly in patients with high SHP2 mRNA expression than in patients with low SHP2 mRNA expression. Therefore, we suggest that dioscin may act as a dual inhibitor of the MEK/ERK and PI3K/AKT signaling pathways to overcome TKI resistance via dysregulation of SHP2 expression in lung adenocarcinoma.
Collapse
Affiliation(s)
- Yao-Chen Wang
- Department of Internal Medicine, Chung Shan Medical University and Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University and Hospital, Taichung, Taiwan
| | - De-Wei Wu
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Chin Wu
- Department of Internal Medicine, Chung Shan Medical University and Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University and Hospital, Taichung, Taiwan
| | - Lee Wang
- Department of Public Health, Chung Shan Medical University, Taichung, Taiwan
| | - Chih-Yi Chen
- School of Medicine, Chung Shan Medical University and Hospital, Taichung, Taiwan.,Department of Surgery, Chung Shan Medical University and Hospital, Taichung, Taiwan
| | - Huei Lee
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
38
|
Zheng L, Yin L, Xu L, Qi Y, Li H, Xu Y, Han X, Liu K, Peng J. Protective effect of dioscin against thioacetamide-induced acute liver injury via FXR/AMPK signaling pathway in vivo. Biomed Pharmacother 2018; 97:481-488. [PMID: 29091898 DOI: 10.1016/j.biopha.2017.10.153] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 10/25/2017] [Accepted: 10/25/2017] [Indexed: 12/30/2022] Open
Abstract
Our previous works showed that dioscin, a natural product, could protect liver from acute liver damages induced by dimethylnitrosamine, ethanol, carbon tetrachloride and acetaminophen. However, the effect of dioscin on thioacetamide (TAA)-induced acute liver injury still remained unknown. The purpose of this study was to investigate whether dioscin confers a protective effect against TAA-induced acute liver injury in rats and mice. The results showed that dioscin decreased the serum levels of ALT, AST, and rehabilitated histopathological changes compared with the model groups. In addition, dioscin obviously increased the levels of GSH, GSH-Px, SOD, and significantly reduced MDA levels compared with the model groups. Mechanistic study showed that dioscin significantly up-regulated the expression levels of FXR, p-AMPKα, and then increased the expression levels of Nrf2, HO-1, NQO-1, GCLM and GST. Furthermore, dioscin obviously down-regulated the expression levels of NF-κB (p65), ICAM-1, HMGB1, COX-2, TNF-α, IL-1β and IL-6. Taken together, dioscin showed protective effect against TAA-induced acute liver injuries in rats and mice and the effects might be obtained through inhibiting oxidative stress and inflammation via FXR/AMPK signal pathway. These findings provided a new insight on the role of doscin in the treatment of acute liver injury.
Collapse
Affiliation(s)
- Lingli Zheng
- Department of Pharmaceuticals, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China; College of Pharmacy, Dalian Medical University, 9 Western Lvshun South Road, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, 9 Western Lvshun South Road, Dalian 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, 9 Western Lvshun South Road, Dalian 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, 9 Western Lvshun South Road, Dalian 116044, China
| | - Hua Li
- College of Pharmacy, Dalian Medical University, 9 Western Lvshun South Road, Dalian 116044, China.
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, 9 Western Lvshun South Road, Dalian 116044, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, 9 Western Lvshun South Road, Dalian 116044, China
| | - Kexin Liu
- College of Pharmacy, Dalian Medical University, 9 Western Lvshun South Road, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, 9 Western Lvshun South Road, Dalian 116044, China.
| |
Collapse
|
39
|
Xu L, Gu L, Tao X, Xu Y, Qi Y, Yin L, Han X, Peng J. Effect of dioscin on promoting liver regeneration via activating Notch1/Jagged1 signal pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 38:107-117. [PMID: 29425642 DOI: 10.1016/j.phymed.2017.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 10/20/2017] [Accepted: 11/12/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND Development of novel candidates to promote liver regeneration is critical important after partial hepatectomy (PH). Dioscin, a natural product, shows potent effect on liver protection in our previous works. PURPOSE This work aimed to investigate the effect and underlying mechanisms of dioscin on liver regeneration. METHODS The promoting proliferation effects of dioscin on mouse hepatocytem AML12 cells, rat primary hepatocytes, rats and mice after 70% PH were evaluated. RESULTS Dioscin significantly promoted proliferation of rat primary hepatocytes and AML12 cells through MTT, BrdU and PCNA staining assays. Meanwhile, dioscin rapidly recovered the liver to body weight ratios, declined ALT and AST levels, and relieved hepatocytes necrosis compared with 70% PH operation groups in rats and mice. Mechanistic test showed that dioscin significantly increased Notch1 and Jagged1 levels, and accelerated γ-secretase activity by up-regulating PS1 expression, leading to nuclear translocation of Notch1 intracellular domain (NICD1). Subsequently, the significant activation of Notch-dependent target genes (Hey1, Hes1, EGFR, VEGF), and cell-cycle regulatory proteins (CyclinD1, CyclinE1, CDK4 and CDK2) were all recognized. In addition, these results were further confirmed by Notch1 siRNA silencing and inhibition of γ-secretase by DAPT (a well-characterized γ-secretase inhibitor) in vitro. CONCLUSIONS Dioscin, as a novel efficient γ-secretase activator, NICD1 nucleus translocation promoter and cell cycle regulator, markedly activated Notch1/Jagged1 pathway to promote hepato-proliferation. Our findings provide novel insights into dioscin as a natural product with facilitating liver regeneration after PH.
Collapse
Affiliation(s)
- Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lina Gu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xufeng Tao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| |
Collapse
|
40
|
Yang F, Luo L, Zhu ZD, Zhou X, Wang Y, Xue J, Zhang J, Cai X, Chen ZL, Ma Q, Chen YF, Wang YJ, Luo YY, Liu P, Zhao L. Chlorogenic Acid Inhibits Liver Fibrosis by Blocking the miR-21-Regulated TGF-β1/Smad7 Signaling Pathway in Vitro and in Vivo. Front Pharmacol 2017; 8:929. [PMID: 29311932 PMCID: PMC5742161 DOI: 10.3389/fphar.2017.00929] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 12/08/2017] [Indexed: 12/30/2022] Open
Abstract
Aims: Chlorogenic acid (CGA) is a phenolic acid that has a wide range of pharmacological effects. However, the protective effects and mechanisms of CGA on liver fibrosis are not clear. This study explored the effects of CGA on miR-21-regulated TGF-β1/Smad7 liver fibrosis in the hepatic stellate LX2 cell line and in CCl4-induced liver fibrosis in Sprague-Dawley rats. Methods: The mRNA expression of miR-21, Smad7, connective tissue growth factor (CTGF), α-smooth muscle actin (α-SMA), tissue inhibitor of metalloproteinase 1 (TIMP-1), matrix metalloproteinase-9 (MMP-9), and transforming growth factor-β1 (TGF-β1) and the protein levels of Smad2, p-Smad2, Smad3, p-Smad3, Smad2/3, p-Smad2/3, Smad7, CTGF, α-SMA, TIMP-1, MMP-9 and TGF-β1 were assayed in LX2 cells and liver tissue. The effects of CGA after miR-21 knockdown or overexpression were analyzed in LX2 cells. The liver tissue and serum were collected for histopathological examination, immunohistochemistry (IHC) and ELISA. Results: The mRNA expression of miR-21, CTGF, α-SMA, TIMP-1, and TGF-β1 and the protein expression of p-Smad2, p-Smad3, p-Smad2/3, CTGF, α-SMA, TIMP-1, and TGF-β1 were inhibited by CGA both in vitro and in vivo. Meanwhile, CGA elevated the mRNA and protein expression of Smad7 and MMP-9. After miR-21 knockdown and overexpression, the downstream molecules also changed accordingly. CGA also lessened the degree of liver fibrosis in the pathological manifestation and reduced α-SMA and collagen I expression in liver tissue and TGF-β1 in serum. Conclusion: CGA might relieve liver fibrosis through the miR-21-regulated TGF-β1/Smad7 signaling pathway, which suggests that CGA might be a new anti-fibrosis agent that improves liver fibrosis.
Collapse
Affiliation(s)
- Fan Yang
- Department of Hepatology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Lei Luo
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan, China
| | - Zhi-De Zhu
- Guangxi University of Chinese Medicine, Nanning, China
| | - Xuan Zhou
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Wang
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan, China
| | - Juan Xue
- Department of Gastroenterology, Hubei Provincial Hospital of Traditional Chinese and Western Medicine, Wuhan, China
| | - Juan Zhang
- Department of Pulmonary Diseases, Jingmen City Hospital of Traditional Chinese Medicine, Jingmen, China
| | - Xin Cai
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan, China
| | - Zhi-Lin Chen
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Ma
- School of Life Sciences, Hubei University, Wuhan, China
| | - Yun-Fei Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu-Jie Wang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying-Ying Luo
- Department of Integrated Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pan Liu
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan, China
| | - Lei Zhao
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
41
|
Peng M, Yang XF. Relationship between mTOR signaling pathway and hepatic stellate cells function. Shijie Huaren Xiaohua Zazhi 2017; 25:3141-3148. [DOI: 10.11569/wcjd.v25.i35.3141] [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] [Indexed: 02/06/2023] Open
Abstract
The activation of hepatic stellate cells (HSCs) is generally considered to be the central link in the formation of hepatic fibrosis. Various factors can regulate the function of HSCs through multiple signaling pathways, of which the mammalian target of rapamycin (mTOR) signaling pathway is especially important. Elucidating the relationship between the mTOR signaling pathway and the proliferation, apoptosis, autophagy, and senescence of HSCs can provide new therapeutic targets and methods for the clinical treatment of hepatic fibrosis. This paper discusses the relationship between the mTOR signaling pathway and the function of HSCs.
Collapse
Affiliation(s)
- Min Peng
- Department of Gastroenterology, Affiliated Nanhua Hospital, University of South China, Hengyang 421002, Hunan Province, China
| | - Xue-Feng Yang
- Department of Gastroenterology, Affiliated Nanhua Hospital, University of South China, Hengyang 421002, Hunan Province, China
| |
Collapse
|
42
|
Wan Y, McDaniel K, Wu N, Ramos-Lorenzo S, Glaser T, Venter J, Francis H, Kennedy L, Sato K, Zhou T, Kyritsi K, Huang Q, Annable T, Wu C, Glaser S, Alpini G, Meng F. Regulation of Cellular Senescence by miR-34a in Alcoholic Liver Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2788-2798. [PMID: 29128099 PMCID: PMC5718103 DOI: 10.1016/j.ajpath.2017.08.027] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 08/16/2017] [Accepted: 08/22/2017] [Indexed: 12/12/2022]
Abstract
Alcoholic liver disease remains a major cause of liver-related morbidity and mortality, which ranges from alcoholic steatohepatitis to fibrosis/cirrhosis and hepatocellular carcinoma, and the related mechanisms are understood poorly. In this study, we aimed to investigate the role of miR-34a in alcohol-induced cellular senescence and liver fibrosis. We found that hepatic miR-34a expression was upregulated in ethanol-fed mice and heavy drinkers with steatohepatitis compared with respective controls. Mice treated with miR-34a Vivo-Morpholino developed less severe liver fibrosis than wild-type mice after 5 weeks of ethanol feeding. Further mechanism exploration showed that inhibition of miR-34a increased cellular senescence of hepatic stellate cells (HSCs) in ethanol-fed mice, although it decreased senescence in total liver and hepatocytes, which was verified by the changes of senescence-associated β-galactosidase and gene expression. Furthermore, enhanced cellular senescence was observed in liver tissues from steatohepatitis patients compared with healthy controls. In addition, the expression of transforming growth factor-β1, drosophila mothers against decapentaplegic protein 2 (Smad2), and Smad3 was decreased after inhibition of miR-34a in ethanol-fed mice. Our in vitro experiments showed that silencing of miR-34a partially blocked activation of HSCs by lipopolysaccharide and enhanced senescence of HSCs. Furthermore, inhibition of miR-34a decreased lipopolysaccharide-induced fibrotic gene expression in cultured hepatocytes. In conclusion, our data suggest that miR-34a functions as a profibrotic factor that promotes alcohol-induced liver fibrosis by reducing HSC senescence and increasing the senescence of hepatocytes.
Collapse
Affiliation(s)
- Ying Wan
- Division of Research, Central Texas Veterans Healthcare System, Temple, Texas; Baylor Scott & White Health Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, Texas; Department of Internal Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas; Department of Pathophysiology, Southwest Medical University, Luzhou, China
| | - Kelly McDaniel
- Division of Research, Central Texas Veterans Healthcare System, Temple, Texas; Baylor Scott & White Health Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, Texas; Department of Internal Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
| | - Nan Wu
- Department of Internal Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
| | - Sugeily Ramos-Lorenzo
- Baylor Scott & White Health Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, Texas
| | - Trenton Glaser
- Baylor Scott & White Health Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, Texas
| | - Julie Venter
- Department of Internal Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
| | - Heather Francis
- Division of Research, Central Texas Veterans Healthcare System, Temple, Texas; Baylor Scott & White Health Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, Texas; Department of Internal Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
| | - Lindsey Kennedy
- Division of Research, Central Texas Veterans Healthcare System, Temple, Texas; Department of Internal Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
| | - Keisaku Sato
- Department of Internal Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
| | - Tianhao Zhou
- Division of Research, Central Texas Veterans Healthcare System, Temple, Texas; Department of Internal Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
| | - Konstantina Kyritsi
- Department of Internal Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
| | - Qiaobing Huang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, China
| | - Tami Annable
- Baylor Scott & White Health Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, Texas; Temple Health and Bioscience District, Temple, Texas
| | - Chaodong Wu
- Department of Nutrition and Food Science, Texas A&M University, College Station, Texas
| | - Shannon Glaser
- Division of Research, Central Texas Veterans Healthcare System, Temple, Texas; Baylor Scott & White Health Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, Texas; Department of Internal Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas
| | - Gianfranco Alpini
- Division of Research, Central Texas Veterans Healthcare System, Temple, Texas; Baylor Scott & White Health Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, Texas; Department of Internal Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas.
| | - Fanyin Meng
- Division of Research, Central Texas Veterans Healthcare System, Temple, Texas; Baylor Scott & White Health Digestive Disease Research Center, Baylor Scott & White Healthcare, Temple, Texas; Department of Internal Medicine, Texas A&M University Health Science Center College of Medicine, Temple, Texas.
| |
Collapse
|
43
|
Li X, Zhu L, Wang B, Yuan M, Zhu R. Drugs and Targets in Fibrosis. Front Pharmacol 2017; 8:855. [PMID: 29218009 PMCID: PMC5703866 DOI: 10.3389/fphar.2017.00855] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 11/08/2017] [Indexed: 01/18/2023] Open
Abstract
Fibrosis contributes to the development of many diseases and many target molecules are involved in fibrosis. Currently, the majority of fibrosis treatment strategies are limited to specific diseases or organs. However, accumulating evidence demonstrates great similarities among fibroproliferative diseases, and more and more drugs are proved to be effective anti-fibrotic therapies across different diseases and organs. Here we comprehensively review the current knowledge on the pathological mechanisms of fibrosis, and divide factors mediating fibrosis progression into extracellular and intracellular groups. Furthermore, we systematically summarize both single and multiple component drugs that target fibrosis. Future directions of fibrosis drug discovery are also proposed.
Collapse
Affiliation(s)
- Xiaoyi Li
- Department of Gastroenterology, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Lixin Zhu
- Department of Pediatrics, Digestive Diseases and Nutrition Center, State University of New York at Buffalo, Buffalo, NY, United States
- Genome, Environment and Microbiome Community of Excellence, State University of New York at Buffalo, Buffalo, NY, United States
| | - Beibei Wang
- Department of Gastroenterology, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Meifei Yuan
- Center for Drug Discovery, SINO High Goal Chemical Technology Co., Ltd., Shanghai, China
| | - Ruixin Zhu
- Department of Gastroenterology, School of Life Sciences and Technology, Shanghai East Hospital, Tongji University, Shanghai, China
| |
Collapse
|
44
|
El-Mezayen NS, El-Hadidy WF, El-Refaie WM, Shalaby T, Khattab MM, El-Khatib AS. Hepatic stellate cell-targeted imatinib nanomedicine versus conventional imatinib: A novel strategy with potent efficacy in experimental liver fibrosis. J Control Release 2017; 266:226-237. [DOI: 10.1016/j.jconrel.2017.09.035] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/24/2017] [Accepted: 09/26/2017] [Indexed: 02/07/2023]
|
45
|
Zhu M, Hao S, Liu T, Yang L, Zheng P, Zhang L, Ji G. Lingguizhugan decoction improves non-alcoholic fatty liver disease by altering insulin resistance and lipid metabolism related genes: a whole trancriptome study by RNA-Seq. Oncotarget 2017; 8:82621-82631. [PMID: 29137289 PMCID: PMC5669915 DOI: 10.18632/oncotarget.19734] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 06/29/2017] [Indexed: 12/23/2022] Open
Abstract
Lingguizhugan decoction, a classic traditional Chinese medicine formula, has been used to treat non-alcoholic fatty liver disease (NAFLD), however, the underlying mechanisms remains unclear. In the present study, we compared the phenotype of the normal rats (fed with chow diet), high-fat-diet (HFD) induced NAFLD rats and Lingguizhugan decoction (LGZG, comprises four Chinese herbs: Poria, Ramulus Cinnamomi, Rhizoma Atractylodis Macrocephalae, and Radix Glycyrrhizae.) intervened rats, and detected whole genome gene expression by RNA-Seq. Our results demonstrated that LGZG decoction attenuated phenotypic characteristics of NAFLD rats. RNA-Seq data analysis revealed that gene expression profiles exerted differential patterns between different groups. 2690 (1445 up-regulated, 1245 down-regulated) genes in NAFLD versus (vs) normal group, 69 (16 up-regulated, 53 down-regulated) genes in LGZG vs NAFLD group, and 42 overlapped (12 up- regulated, 30 down-regulated) genes between NAFLDvs normal group and LGZG vs NAFLD group were identified as differentially expressed. GO, pathway enrichment and PPI networks analysis of the overlapped genes revealed that LGZG decoction might attenuate NAFLD possibly by affecting insulin resistance and lipid metabolism related pathways (e.g., PI3K-Akt, AMPK). Differentially expressed genes involved in these pathways such as Pik3r1, Foxo1, Foxo3, Scd1, Col3a1 and Fn1 might be candidate targets for treating NAFLD.
Collapse
Affiliation(s)
- Mingzhe Zhu
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Public Health College, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Shijun Hao
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Liu
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lili Yang
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Peiyong Zheng
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Zhang
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Guang Ji
- Institute of Digestive Diseases, China-Canada Center of Research for Digestive Diseases, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
46
|
Li C, Lu Y, Du S, Li S, Zhang Y, Liu F, Chen Y, Weng D, Chen J. Dioscin Exerts Protective Effects Against Crystalline Silica-induced Pulmonary Fibrosis in Mice. Theranostics 2017; 7:4255-4275. [PMID: 29158824 PMCID: PMC5695011 DOI: 10.7150/thno.20270] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Accepted: 08/29/2017] [Indexed: 12/15/2022] Open
Abstract
Inhalation of crystalline silica particles leads to pulmonary fibrosis, eventually resulting in respiratory failure and death. There are few effective drugs that can delay the progression of this disease; thus, patients with silicosis are usually only offered supportive care. Dioscin, a steroidal saponin, exhibits many biological activities and health benefits including its protective effects against hepatic fibrosis. However, the effect of dioscin on silicosis is unknown. Methods: We employed experimental mouse mode of silicosis. Different doses of dioscin were gavaged to the animals 1 day after crystalline silica instillation to see the effect of dioscin on crystalline silica induced pulmonary fibrosis. Also, we used RAW264.7 and NIH-3T3 cell lines to explore dioscin effects on macrophages and fibroblasts. Dioscin was also oral treatment but 10 days after crystalline silica instillation to see its effect on established pulmonary fibrosis. Results: Dioscin treatment reduced pro-inflammation and pro-fibrotic cytokine secretion by modulating innate and adaptive immune responses. It also reduced the recruitment of fibrocytes, protected epithelial cells from crystalline silica injury, inhibited transforming growth factor beta/Smad3 signaling and fibroblast activation. Together, these effects delayed the progression of crystalline silica-induced pulmonary fibrosis. The mechanism by which dioscin treatment alleviated CS-induced inflammation appeared to be via the reduction of macrophage, B lymphocyte, and T lymphocte infiltration into lung. Dioscin inhibits macrophages and fibroblasts from secreting pro-inflammatory cytokines and may also function as a modulator of T helper cells responses, concurrent with attenuated phosphorylation of the apoptosis signal-regulating kinase 1-p38/c-Jun N-terminal kinase pathway. Also, dioscin could block the phosphorylation of Smad3 in fibroblast. Oral treatment of dioscin could also effectively postpone the progression of established silicosis. Conclusion: Oral treatment dioscin delays crystalline silica-induced pulmonary fibrosis and exerts pulmonary protective effects in mice. Dioscin may be a novel and potent candidate for protection against crystalline silica-induced pulmonary fibrosis.
Collapse
|
47
|
Yin L, Qi Y, Xu Y, Xu L, Han X, Tao X, Song S, Peng J. Dioscin Inhibits HSC-T6 Cell Migration via Adjusting SDC-4 Expression: Insights from iTRAQ-Based Quantitative Proteomics. Front Pharmacol 2017; 8:665. [PMID: 29033837 PMCID: PMC5627034 DOI: 10.3389/fphar.2017.00665] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 09/06/2017] [Indexed: 12/25/2022] Open
Abstract
Hepatic stellate cells (HSCs) migration, an important bioprocess, contributes to the development of liver fibrosis. Our previous studies have found the potent activity of dioscin against liver fibrosis by inhibiting HSCs proliferation, triggering the senescence and inducing apoptosis of activated HSCs, but the molecular mechanisms associated with cell migration were not clarified. In this work, iTRAQ (isobaric tags for relative and absolution quantitation)-based quantitative proteomics study was carried out, and a total of 1566 differentially expressed proteins with fold change ≥2.0 and p < 0.05 were identified in HSC-T6 cells treated by dioscin (5.0 μg/mL). Based on Gene Ontology classification, String and KEGG pathway assays, the effects of dioscin to inhibit cell migration via regulating SDC-4 were carried out. The results of wound-healing, cell migration and western blotting assays indicated that dioscin significantly inhibit HSC-T6 cell migration through SDC-4-dependent signal pathway by affecting the expression levels of Fn, PKCα, Src, FAK, and ERK1/2. Specific SDC-4 knockdown by shRNA also blocked HSC-T6 cell migration, and dioscin slightly enhanced the inhibiting effect. Taken together, the present work showed that SDC-4 played a crucial role on HSC-T6 cell adhesion and migration of dioscin against liver fibrosis, which may be one potent therapeutic target for fibrotic diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jinyong Peng
- College of Pharmacy, Dalian Medical UniversityDalian, China
| |
Collapse
|
48
|
Guo Y, Liang X, Meng M, Chen H, Wei X, Li M, Li J, Huang R, Wei J. Hepatoprotective effects of Yulangsan flavone against carbon tetrachloride (CCl 4)-induced hepatic fibrosis in rats. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 33:28-35. [PMID: 28887917 DOI: 10.1016/j.phymed.2017.07.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 05/22/2017] [Accepted: 07/02/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND Yulangsan flavone (YLSF) was extracted from the root of Millettia pulchra Kurz var-laxior (Dunn) Z. Wei, which has been widely used for liver disease treatment in the Guangxi province of China. HYPOTHESIS/PURPOSE The study was conducted to demonstrate the hepatoprotective effects of YLSF against CCl4-induced hepatic fibrosis in rats, meanwhile revealing the potential mechanism. STUDY DESIGN Sprague-Dawley (SD) rats of both sexes were randomly divided into two groups: hepatic fibrosis group and normal control (NC) group. The rats in the hepatic fibrosis group were given 1 ml/kg 50% CCl4 (1:1 mixed with peanut oil), while those in the NC group were given 1 ml/kg normal saline (NS), both via intragastric administration. The established experimental rat model from the hepatic fibrosis group was confirmed by pathological inspection and randomly divided into five groups: three YLSF groups (20 mg/kg, 40 mg/kg and 80 mg/kg), a colchicine group (0.20 mg/kg) and a model group (10 ml/kg NS). All rats were treated with corresponding drugs or NS once a day for four consecutive weeks. Twenty-four hours after the last administration, blood serum and hepatic tissue were collected. METHODS The activities of ALT and AST in the serum and the levels of SOD, MDA, GSH and GSH-Px in hepatic tissue were analysed, the indexes of liver, spleen and thymus were counted, the degree of hepatic injury was examined using HE and Masson staining, and the mRNA expression of Col-1, TIMP-1 and TGF-β1 in hepatic tissues was detected. RESULTS Compared with the model group, experimental results showed that YLSF and colchicine could reduce the levels of AST, ALT and MDA, increase the levels of SOD, GSH and GSH-Px, enhance rat survivability, decrease the liver, spleen and thymus index, significantly lessen collagen deposition and tissue damage and down-regulate the mRNA expression of Col-1, TIMP-1 and TGF-β1. CONCLUSIONS Our findings confirm that YLSF has a certain curative effect on rats with liver fibrosis induced by CCl4, and its mechanism may include attenuating free radicals, inhibiting lipid peroxidation and accelerating extracellular matrix degradation by down-regulating expression of related genes.
Collapse
Affiliation(s)
- Youjia Guo
- Department of Pharmacology, Guangxi Medical University, 22, Shuangyong Road, Nanning 530021, Guangxi, P.R. China
| | - Xingmei Liang
- Department of Pharmacology, Guangxi Medical University, 22, Shuangyong Road, Nanning 530021, Guangxi, P.R. China
| | - Mingyu Meng
- Department of Pharmacology, Guangxi Medical University, 22, Shuangyong Road, Nanning 530021, Guangxi, P.R. China
| | - Hongxia Chen
- Department of Pharmacology, Guangxi Medical University, 22, Shuangyong Road, Nanning 530021, Guangxi, P.R. China
| | - Xiaojie Wei
- Department of Pharmacology, Guangxi Medical University, 22, Shuangyong Road, Nanning 530021, Guangxi, P.R. China
| | - Mingyan Li
- Department of Pharmacology, Guangxi Medical University, 22, Shuangyong Road, Nanning 530021, Guangxi, P.R. China
| | - Juman Li
- Department of Pharmacology, Guangxi Medical University, 22, Shuangyong Road, Nanning 530021, Guangxi, P.R. China
| | - Renbin Huang
- Department of Pharmacology, Guangxi Medical University, 22, Shuangyong Road, Nanning 530021, Guangxi, P.R. China.
| | - Jinbin Wei
- Department of Pharmacology, Guangxi Medical University, 22, Shuangyong Road, Nanning 530021, Guangxi, P.R. China
| |
Collapse
|
49
|
Xu L, Yin L, Tao X, Qi Y, Han X, Xu Y, Song S, Li L, Sun P, Peng J. Dioscin, a potent ITGA5 inhibitor, reduces the synthesis of collagen against liver fibrosis: Insights from SILAC-based proteomics analysis. Food Chem Toxicol 2017; 107:318-328. [PMID: 28689917 DOI: 10.1016/j.fct.2017.07.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 11/26/2022]
Abstract
Inhibiting collagen generation is one effective method to treat liver fibrosis. Dioscin showed protective effect against liver fibrosis in our previous studies, and in the present work, SILAC-based proteomics was employed to test the underlying mechanism. A total of 121 differentially expressed proteins caused by dioscin in LX-2 cells were found, and dioscin significantly decreased the expression levels of FN, FAK1, ITGA5, p-PI3K/PI3K, p-Akt/Akt, p-mTOR/mTOR, Col1a1, Col1a2, Col2a1, Col5a1, Col6a1, and increased 2ABB level in vivo and in vitro. Thus, we elucidated that dioscin specifically suppressed collagen synthesis through modulating PI3K/Akt pathway. In addition, we found that dioscin directly targeted with ITGA5 by molecular docking assay. SiRNA and overexpression transfection tests showed that ITGA5 siRNA plus dioscin slightly altered the effect of ITGA5 siRNA, and ITGA5 DNA transfection reversed the inhibitory effect of dioscin on collagen expressions via PI3K/Akt pathway. Our data explicated that dioscin should be considered as a novel and potent ITGA5 inhibitor to suppress collagen synthesis, which can also be developed as an effective food and healthcare product against hepatic fibrosis.
Collapse
Affiliation(s)
- Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xufeng Tao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Shasha Song
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lei Li
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Pengyuan Sun
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| |
Collapse
|
50
|
Wan Y, Meng F, Wu N, Zhou T, Venter J, Francis H, Kennedy L, Glaser T, Bernuzzi F, Invernizzi P, Glaser S, Huang Q, Alpini G. Substance P increases liver fibrosis by differential changes in senescence of cholangiocytes and hepatic stellate cells. Hepatology 2017; 66:528-541. [PMID: 28256736 PMCID: PMC5519428 DOI: 10.1002/hep.29138] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 02/01/2017] [Accepted: 02/25/2017] [Indexed: 12/16/2022]
Abstract
UNLABELLED Substance P (SP) is involved in the proliferation of cholangiocytes in bile duct-ligated (BDL) mice and human cholangiocarcinoma growth by interacting with the neurokinin-1 receptor (NK-1R). To identify whether SP regulates liver fibrosis during cholestasis, wild-type or NK-1R knockout (NK-1R-/- ) mice that received BDL or sham surgery and multidrug resistance protein 2 knockout (Mdr2-/- ) mice treated with either an NK-1R antagonist (L-733,060) or saline were used. Additionally, wild-type mice were treated with SP or saline intraperitoneally. In vivo, there was increased expression of tachykinin precursor 1 (coding SP) and NK-1R in both BDL and Mdr2-/- mice compared to wild-type mice. Expression of tachykinin precursor 1 and NK-1R was significantly higher in liver samples from primary sclerosing cholangitis patients compared to healthy controls. Knockout of NK-1R decreased BDL-induced liver fibrosis, and treatment with L-733,060 resulted in decreased liver fibrosis in Mdr2-/- mice, which was shown by decreased sirius red staining, fibrosis gene and protein expression, and reduced transforming growth factor-β1 levels in serum and cholangiocyte supernatants. Furthermore, we observed that reduced liver fibrosis in NK-1R-/- mice with BDL surgery or Mdr2-/- mice treated with L-733,060 was associated with enhanced cellular senescence of hepatic stellate cells and decreased senescence of cholangiocytes. In vitro, L-733,060 inhibited SP-induced expression of fibrotic genes in hepatic stellate cells and cholangiocytes; treatment with L-733,060 partially reversed the SP-induced decrease of senescence gene expression in cultured hepatic stellate cells and the SP-induced increase of senescence-related gene expression in cultured cholangiocytes. CONCLUSION Collectively, our results demonstrate the regulatory effects of the SP/NK-1R axis on liver fibrosis through changes in cellular senescence during cholestatic liver injury. (Hepatology 2017;66:528-541).
Collapse
Affiliation(s)
- Ying Wan
- Research, Central Texas Veterans Health Care System, Temple, TX,Baylor Scott & White Digestive Disease Research Center, Scott & White, Temple, TX,Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, China
| | - Fanyin Meng
- Research, Central Texas Veterans Health Care System, Temple, TX,Baylor Scott & White Digestive Disease Research Center, Scott & White, Temple, TX,Operational Funds, Baylor Scott & White, Temple, TX,Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White, Temple, TX
| | - Nan Wu
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White, Temple, TX
| | - Tianhao Zhou
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White, Temple, TX
| | - Julie Venter
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White, Temple, TX
| | - Heather Francis
- Research, Central Texas Veterans Health Care System, Temple, TX,Baylor Scott & White Digestive Disease Research Center, Scott & White, Temple, TX,Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White, Temple, TX
| | - Lindsey Kennedy
- Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White, Temple, TX
| | - Trenton Glaser
- Baylor Scott & White Digestive Disease Research Center, Scott & White, Temple, TX
| | | | | | - Shannon Glaser
- Research, Central Texas Veterans Health Care System, Temple, TX,Baylor Scott & White Digestive Disease Research Center, Scott & White, Temple, TX,Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White, Temple, TX
| | - Qiaobing Huang
- Department of Pathophysiology, Key Lab for Shock and Microcirculation Research of Guangdong Province, Southern Medical University, Guangzhou, China
| | - Gianfranco Alpini
- Research, Central Texas Veterans Health Care System, Temple, TX,Baylor Scott & White Digestive Disease Research Center, Scott & White, Temple, TX,Department of Medicine, Division Gastroenterology, Texas A&M University Health Science Center and Baylor Scott & White, Temple, TX
| |
Collapse
|