1
|
Chien HJ, Liu CJ, Ying TH, Wu PJ, Wang JW, Ting YH, Hsieh YH, Wang SC. Timosaponin AIII Inhibits Migration and Invasion Abilities in Human Cervical Cancer Cells through Inactivation of p38 MAPK-Mediated uPA Expression In Vitro and In Vivo. Cancers (Basel) 2022; 15:cancers15010037. [PMID: 36612038 PMCID: PMC9817900 DOI: 10.3390/cancers15010037] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/15/2022] [Accepted: 12/18/2022] [Indexed: 12/24/2022] Open
Abstract
Cervical cancer is one of the most common gynecologic cancers globally that require novel approaches. Timosaponin AIII (TSAIII) is a steroidal saponin that displays beneficial effects in antitumor activities. However, the effect of TSAIII on human cervical cancer remains unknown. In this study, we found that TSAIII showed no influence on cell viability, cytotoxicity, cell cycle distribution and apoptosis induction in human cervical cancer cells. TSAIII was revealed to have a significant inhibitory effect on cell migration and invasion through the downregulation of invasion-related uPA expression and p38 MAPK activation in both human cervical cancer cells and cervical cancer stem cells (CCSCs), indicating that the p38 MAPK-uPA axis mediated the TSAIII-inhibited capacity of cellular migration and invasion. In a synergistic inhibition assay, a TSAIII plus p38 siRNA cotreatment revealed a greater inhibition of uPA expression, migration and invasion in human cervical cancer cells. In an immunodeficient mouse model, TSAIII significantly inhibited lung metastases from human cervical cancer SiHa cells without TSAIII-induced toxicity. These findings first revealed the inhibitory effects of TSAIII on the progression of human cervical cancer through its downregulation of p38 MAPK-uPA axis activation. Therefore, TSAIII might provide a potential strategy for auxiliary therapy in human cervical cancer.
Collapse
Affiliation(s)
- Hung-Ju Chien
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Obstetrics and Gynecology, Changhua Christian Hospital, Changhua 50006, Taiwan
| | - Chung-Jung Liu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807378, Taiwan
- Regenetative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Tsung-Ho Ying
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Department of Obstetrics and Gynecology, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Pei-Ju Wu
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Jiunn-Wei Wang
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807378, Taiwan
- Regenetative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Department of Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
| | - Yi-Hsuan Ting
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Correspondence: (Y.-H.H.); (S.-C.W.)
| | - Shih-Chiang Wang
- Department of Obstetrics and Gynecology, Chung-Kang Branch, Cheng Ching Hospital, Taichung 40764, Taiwan
- Correspondence: (Y.-H.H.); (S.-C.W.)
| |
Collapse
|
2
|
Mao Z, Gao M, Zhao X, Li L, Peng J. Neuroprotective Effect of Dioscin against Parkinson's Disease via Adjusting Dual-Specificity Phosphatase 6 (DUSP6)-Mediated Oxidative Stress. Molecules 2022; 27:3151. [PMID: 35630630 PMCID: PMC9146847 DOI: 10.3390/molecules27103151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/06/2022] [Accepted: 05/10/2022] [Indexed: 12/10/2022] Open
Abstract
Exploration of lead compounds against Parkinson's disease (PD), a neurodegenerative disease, is of great important. Dioscin, a bioactive natural product, shows various pharmacological effects. However, the activities and mechanisms of dioscin against PD have not been well investigated. In this study, the tests on 6-hydroxydopamine (6-OHDA)-induced PC12 cells and rats were carried out. The results showed that dioscin dramatically improved cell viability, decreased reactive oxygen species (ROS) levels, improved motor behavior and tyrosine hydroxylase(TH) levels and restored the levels of glutathione (GSH) and malondialdehyde (MDA) in rats. Mechanism investigation showed that dioscin not only markedly increased the expression level of dual- specificity phosphatase 6 (DUSP6) by 1.87-fold in cells and 2.56-fold in rats, and decreased phospho-extracellular regulated protein kinases (p-ERK) level by 2.12-fold in cells and 2.34-fold in rats, but also increased the levels of nuclear factor erythroid2-related factor 2 (Nrf2), hemeoxygenase-1 (HO-1), superoxide dismutase (SOD) and decreased the levels of kelch-1ike ECH-associated protein l (Keap1) in vitro and in vivo. Furthermore, DUSP6 siRNA transfection experiment in PC12 cells validated the protective effects of dioscin against PD via regulating DUSP6 to adjust the Keap1/Nrf2 pathway. Our data supported that dioscin has protection against PD in regulating oxidative stress via DUSP6 signal, which should be considered as an efficient candidate for the treatment of PD in the future.
Collapse
Affiliation(s)
- Zhang Mao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China; (Z.M.); (M.G.); (X.Z.)
- College of Intergrative Medicine, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Meng Gao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China; (Z.M.); (M.G.); (X.Z.)
| | - Xuerong Zhao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China; (Z.M.); (M.G.); (X.Z.)
| | - Lili Li
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China; (Z.M.); (M.G.); (X.Z.)
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| |
Collapse
|
3
|
Orientin Alleviates Liver Inflammation via Downregulation of ZEB-2/PTEN Markers—Hepatic Stellate Cells Approach. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12052725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Liver inflammation is associated with an increased risk of liver fibrosis that substantially progresses to cirrhosis. Recently, usage of the herbal supplement has been increased because of its emerging role to dominate oxidative stress in hepatic injury. Orientin is one of the bioactive flavonoids that possesses a diversity of curative activities. Therefore, the present study was conducted to evaluate the anti-inflammatory role of orientin (1 mg/kg) in vitro in lipopolysaccharide (LPS)-induced inflammation in hepatic stellate cells (HSCs) and in vivo in carbon tetrachloride (CCl4)-induced liver fibrosis in mice. Moreover, the current study was supported by in silico investigation. Orientin demonstrated protection against LPS-induced HSC inflammation as evidenced by a decrease in iNOS, NO, and TNF-α and inhibition of the fibrotic markers ZEB-2 and PTEN. In addition, orientin afforded protection against CCl4-induced liver fibrosis in mice as shown from decreased AST/ALT ratio, inhibition of the pro-inflammatory mediators TNF-α, IL-6, IL-8, and IFN-γ, reduction of fibrotic markers ZEB-2 and PTEN, and improvement of the histopathological changes. Furthermore, the docking study demonstrated virtual interactions of orientin with ZEB-2 and PTEN. Taken together, the current study suggested that the protective effects of orientin against LPS- and CCl4-induced liver inflammation are via inhibition of fibrotic markers and reduction of pro-inflammatory mediators.
Collapse
|
4
|
Keller-Pinter A, Gyulai-Nagy S, Becsky D, Dux L, Rovo L. Syndecan-4 in Tumor Cell Motility. Cancers (Basel) 2021; 13:cancers13133322. [PMID: 34282767 PMCID: PMC8268284 DOI: 10.3390/cancers13133322] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Cell migration is crucial fReaor metastasis formation and a hallmark of malignancy. The primary cause of high mortality among oncology patients is the ability of cancer cells to metastasize. To form metastasis, primary tumor cells must be intrinsically able to move. The transmembrane, heparan sulfate proteoglycan syndecan-4 (SDC4) exhibits multiple functions in signal transduction by regulating Rac1 GTPase activity and consequently actin remodeling, as well as regulating focal adhesion kinase, protein kinase C-alpha and the level of intracellular calcium. By affecting several signaling pathways and biological processes, SDC4 is involved in cell migration under physiological and pathological conditions as well. In this review, we discuss the SDC4-mediated cell migration focusing on the role of SDC4 in tumor cell movement. Abstract Syndecan-4 (SDC4) is a ubiquitously expressed, transmembrane proteoglycan bearing heparan sulfate chains. SDC4 is involved in numerous inside-out and outside-in signaling processes, such as binding and sequestration of growth factors and extracellular matrix components, regulation of the activity of the small GTPase Rac1, protein kinase C-alpha, the level of intracellular calcium, or the phosphorylation of focal adhesion kinase. The ability of this proteoglycan to link the extracellular matrix and actin cytoskeleton enables SDC4 to contribute to biological functions like cell adhesion and migration, cell proliferation, cytokinesis, cellular polarity, or mechanotransduction. The multiple roles of SDC4 in tumor pathogenesis and progression has already been demonstrated; therefore, the expression and signaling of SDC4 was investigated in several tumor types. SDC4 influences tumor progression by regulating cell proliferation as well as cell migration by affecting cell-matrix adhesion and several signaling pathways. Here, we summarize the general role of SDC4 in cell migration and tumor cell motility.
Collapse
Affiliation(s)
- Aniko Keller-Pinter
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (S.G.-N.); (D.B.); (L.D.)
- Correspondence:
| | - Szuzina Gyulai-Nagy
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (S.G.-N.); (D.B.); (L.D.)
| | - Daniel Becsky
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (S.G.-N.); (D.B.); (L.D.)
| | - Laszlo Dux
- Department of Biochemistry, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (S.G.-N.); (D.B.); (L.D.)
| | - Laszlo Rovo
- Department of Oto-Rhino-Laryngology and Head-Neck Surgery, University of Szeged, H-6725 Szeged, Hungary;
| |
Collapse
|
5
|
Pessolano E, Belvedere R, Novizio N, Filippelli A, Perretti M, Whiteford J, Petrella A. Mesoglycan connects Syndecan-4 and VEGFR2 through Annexin A1 and formyl peptide receptors to promote angiogenesis in vitro. FEBS J 2021; 288:6428-6446. [PMID: 34058069 PMCID: PMC9290969 DOI: 10.1111/febs.16043] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/06/2021] [Accepted: 05/28/2021] [Indexed: 12/11/2022]
Abstract
Mesoglycan is a mixture of glycosaminoglycans (GAG) with fibrinolytic effects and the potential to enhance skin wound repair. Here, we have used endothelial cells isolated from wild‐type (WT) and Syndecan‐4 null (Sdc4‐/‐) C57BL/6 mice to demonstrate that mesoglycan promotes cell motility and in vitro angiogenesis acting on the co‐receptor Syndecan‐4 (SDC4). This latter is known to participate in the formation and release of extracellular vesicles (EVs). We characterized EVs released by HUVECs and assessed their effect on angiogenesis. Particularly, we focused on Annexin A1 (ANXA1) containing EVs, since they may contribute to tube formation via interactions with Formyl peptide receptors (FPRs). In our model, the bond ANXA1‐FPRs stimulates the release of vascular endothelial growth factor (VEGF‐A) that interacts with vascular endothelial receptor‐2 (VEGFR2) and activates the pathway enhancing cell motility in an autocrine manner, as shown by wound healing/invasion assays, and the induction of endothelial to mesenchymal transition (EndMT). Thus, we have shown for the first time that mesoglycan exerts its pro‐angiogenic effects in the healing process triggering the activation of the three interconnected molecular axis: mesoglycan‐SDC4, EVs‐ANXA1‐FPRs, and VEGF‐A‐VEGFR2.
Collapse
Affiliation(s)
- Emanuela Pessolano
- Department of Pharmacy, University of Salerno, Fisciano, Italy.,The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | | | - Nunzia Novizio
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Amelia Filippelli
- Department of Medicine, Surgery, and Dentistry, University of Salerno, Baronissi, Italy
| | - Mauro Perretti
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | - James Whiteford
- The William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, UK
| | | |
Collapse
|
6
|
Yang H, Liu Y, Zhao MM, Guo Q, Zheng XK, Liu D, Zeng KW, Tu PF. Therapeutic potential of targeting membrane-spanning proteoglycan SDC4 in hepatocellular carcinoma. Cell Death Dis 2021; 12:492. [PMID: 33990545 PMCID: PMC8121893 DOI: 10.1038/s41419-021-03780-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 12/15/2022]
Abstract
Syndecan-4 (SDC4) functions as a major endogenous membrane-associated receptor and widely regulates cytoskeleton, cell adhesion, and cell migration in human tumorigenesis and development, which represents a charming anti-cancer therapeutic target. Here, SDC4 was identified as a direct cellular target of small-molecule bufalin with anti-hepatocellular carcinoma (HCC) activity. Mechanism studies revealed that bufalin directly bond to SDC4 and selectively increased SDC4 interaction with substrate protein DEAD-box helicase 23 (DDX23) to induce HCC genomic instability. Meanwhile, pharmacological promotion of SDC4/DDX23 complex formation also inactivated matrix metalloproteinases (MMPs) and augmented p38/JNK MAPKs phosphorylation, which are highly associated with HCC proliferation and migration. Notably, specific knockdown of SDC4 or DDX23 markedly abolished bufalin-dependent inhibition of HCC proliferation and migration, indicating SDC4/DDX23 signaling axis is highly involved in the HCC process. Our results indicate that membrane-spanning proteoglycan SDC4 is a promising druggable target for HCC, and pharmacological regulation of SDC4/DDX23 signaling axis with small-molecule holds great potential to benefit HCC patients.
Collapse
Affiliation(s)
- Heng Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yang Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Mei-Mei Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Qiang Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xi-Kang Zheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Dan Liu
- Proteomics Laboratory, Medical and Healthy Analytical Center, Peking University Health Science Center, Beijing, 100191, China
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| | - Peng-Fei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
7
|
Hsieh YH, Hsu WH, Yang SF, Liu CJ, Lu KH, Wang PH, Lin RC. Potential Antimetastatic Effect of Timosaponin AIII against Human Osteosarcoma Cells through Regulating the Integrin/FAK/Cofilin Axis. Pharmaceuticals (Basel) 2021; 14:ph14030260. [PMID: 33799345 PMCID: PMC8000016 DOI: 10.3390/ph14030260] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 03/12/2021] [Indexed: 12/13/2022] Open
Abstract
Timosaponin AIII (TSAIII) is a steroidal saponin which demonstrates anti-tumour activities. However, the effect of TSAIII on human osteosarcoma cells remains largely unknown. In this study, we demonstrated that TSAIII exerted a significant inhibitory effect on the distribution of cytoskeletal F-actin and cytoskeletal-related proteins, which contributed to the suppression of cell migration and invasion, without inhibiting cell growth or apoptosis. In the synergistic inhibitory analysis, cotreatment of TSAIII with αVβ3 integrin inhibitor [Cyclo(RGDyK)] or focal adhesion kinase (FAK) inhibitor (PF-573228) exerted greater synergistic inhibitory effects on the expression of Intergin αVβ3/FAK/cofilin axis, thus inhibiting the migration and invasion capacities of human osteosarcoma cells. TSAIII was demonstrated to significantly inhibit the pulmonary metastasis formation of human osteosarcoma cells in vivo in metastasis animal models. These findings reveal the inhibitory effects of TSAIII on the metastasis progression of human osteosarcoma cells and the regulation of integrin-αVβ3-FAK-Src and TESK1/p-cofilin mediated cytoskeletal F-actin pathway. Therefore, TSAIII might represent a novel strategy for the auxiliary treatment of human osteosarcoma cells.
Collapse
Affiliation(s)
- Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (Y.-H.H.); (S.-F.Y.); (P.-H.W.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Wen-Hung Hsu
- Division of Gastroenterology, Department of Internal Medicine, Kaoshiung Medical University Hospital, Kaoshiung, Medical University, Kaoshiung 80756, Taiwan; (W.-H.H.); (C.-J.L.)
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (Y.-H.H.); (S.-F.Y.); (P.-H.W.)
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Chung-Jung Liu
- Division of Gastroenterology, Department of Internal Medicine, Kaoshiung Medical University Hospital, Kaoshiung, Medical University, Kaoshiung 80756, Taiwan; (W.-H.H.); (C.-J.L.)
| | - Ko-Hsiu Lu
- Department of Orthopedics, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 40201, Taiwan;
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Pei-Han Wang
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; (Y.-H.H.); (S.-F.Y.); (P.-H.W.)
| | - Renn-Chia Lin
- Department of Orthopedics, Chung Shan Medical University Hospital, Chung Shan Medical University, Taichung 40201, Taiwan;
- School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Correspondence:
| |
Collapse
|
8
|
Qiao YL, Zhou JJ, Liang JH, Deng XP, Zhang ZJ, Huang HL, Li S, Dai SF, Liu CQ, Luan ZL, Yu ZL, Sun CP, Ma XC. Uncaria rhynchophylla ameliorates unpredictable chronic mild stress-induced depression in mice via activating 5-HT 1A receptor: Insights from transcriptomics. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 81:153436. [PMID: 33360346 DOI: 10.1016/j.phymed.2020.153436] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/29/2020] [Accepted: 12/03/2020] [Indexed: 06/12/2023]
Abstract
BACKGROUND Depression is a pervasive or persistent mental disorder that causes mood, cognitive and memory deficits. Uncaria rhynchophylla has been widely used to treat central nervous system diseases for a long history, although its efficacy and potential mechanism are still uncertain. PURPOSE The present study aimed to investigate anti-depression effect and potential mechanism of U. rhynchophylla extract (URE). STUDY DESIGN AND METHODS A mouse depression model was established using unpredictable chronic mild stress (UCMS). Effects of URE on depression-like behaviours, neurotransmitters, and neuroendocrine hormones were investigated in UCMS-induced mice. The potential target of URE was analyzed by transcriptomics and bioinformatics methods and validated by RT-PCR and Western blot. The agonistic effect on 5-HT1A receptor was assayed by dual-luciferase reporter system. RESULTS URE ameliorated depression-like behaviours, and modulated levels of neurotransmitters and neuroendocrine hormones, including 5-hydroxytryptamine (5-HT), 5-hydroxyindole acetic acid (5-HIAA), dopamine (DA), 3,4-dihydroxyphenylacetic acid (DOPAC), homovanillic acid (HVA), corticosterone (CORT), corticotropin-releasing hormone (CRH), and adrenocorticotropic hormone (ACTH), in UCMS-induced mice. Transcriptomics and bioinformatics results indicated that URE could regulate glutamatergic, cholinergic, serotonergic, and GABAergic systems, especially neuroactive ligand-receptor and cAMP signaling pathways, revealing that Htr1a encoding 5-HT1A receptor was a potential target of URE. The expression levels of downstream proteins of 5-HT1A signaling pathway 5-HT1A, CREB, BDNF, and PKA were increased in UCMS-induced mice after URE administration, and URE also displayed an agonistic effect against 5-HT1A receptor with an EC50 value of 17.42 μg/ml. CONCLUSION U. rhynchophylla ameliorated depression-like behaviours in UCMS-induced mice through activating 5-HT1A receptor.
Collapse
Affiliation(s)
- Yan-Ling Qiao
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Jun-Jun Zhou
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Jia-Hao Liang
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Xiao-Peng Deng
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Zhan-Jun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Hui-Lian Huang
- Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Song Li
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Shu-Fang Dai
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Chun-Qing Liu
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Zhi-Lin Luan
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Zhen-Long Yu
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China
| | - Cheng-Peng Sun
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China.
| | - Xiao-Chi Ma
- Dalian Key Laboratory of Metabolic Target Characterization and Traditional Chinese Medicine, College of Pharmacy, College of Integrative Medicine, Department of Neurosurgery, The First and Second Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian, China; State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China.
| |
Collapse
|
9
|
Zhang T, Li Y, Song Y, Chen X, Li J, Peng Q, He J, Fei X. Curcumin- and Cyclopamine-Loaded Liposomes to Enhance Therapeutic Efficacy Against Hepatic Fibrosis. Drug Des Devel Ther 2020; 14:5667-5678. [PMID: 33380787 PMCID: PMC7767702 DOI: 10.2147/dddt.s287442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/11/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND AND PURPOSE Hepatic fibrosis is a public health problem characterized by activation of hepatic stellate cells (HSCs), which triggers excessive production of extracellular matrix (ECM). Inhibition of HSC activation may be an effective treatment. Since various pathways control HSC activation, a combination of drugs with different mechanisms may be more effective than monotherapy. METHODS Here, we prepared liposomes loaded with curcumin and cyclopamine to inhibit HSC activation. We systematically analyzed the physicochemical characteristics of liposomes loaded with the two drugs, as well as their effects on HSC proliferation, activation and collagen production on gene, protein and cellular levels. RESULTS The prepared liposomes helped solubilize both drugs, contributing to their uptake by cells. Liposomes loaded with both drugs inhibited cell proliferation, migration and invasion, as well as induced more apoptosis and perturbed the cell cycle more than the free combination of both drugs in solution or liposomes loaded with either drug alone. Liposomes loaded with both drugs strongly suppressed HSC activation and collagen secretion. CONCLUSION Our results suggest that liposome encapsulation can increase the uptake of curcumin and cyclopamine as well as the synergism between them in anti-fibrosis. This approach shows potential for treating hepatic fibrosis.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu610041, People’s Republic of China
| | - Yanping Li
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, People’s Republic of China
| | - Yi Song
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu610041, People’s Republic of China
| | - Xiaoshuang Chen
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu610041, People’s Republic of China
| | - Jing Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu610041, People’s Republic of China
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu610041, People’s Republic of China
| | - Jinhan He
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu610041, People’s Republic of China
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital of Sichuan University, Chengdu, People’s Republic of China
| | - Xiaofan Fei
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu610041, People’s Republic of China
| |
Collapse
|
10
|
Wang P, Wang C, Liu C. Antitumor effects of dioscin in A431 cells via adjusting ATM/p53-mediated cell apoptosis, DNA damage and migration. Oncol Lett 2020; 21:59. [PMID: 33281970 PMCID: PMC7709553 DOI: 10.3892/ol.2020.12321] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 10/26/2020] [Indexed: 11/27/2022] Open
Abstract
Skin cancer is the deadliest type of malignant disease and causes primary mortality worldwide. Dioscin, which exists in medicinal plants, has potent anticancer effects. However, its effects on skin cancer remain unknown. In the present study, the activity and mechanism of dioscin on the human skin cancer A431 cell line were investigated, MTT, colony formation, Transwell, wound-healing, TUNEL, Comet, immunofluorescence and western blot assays were used to assess the effects of dioscin on A431 cells. The results of MTT, colony formation, Transwell and wound-healing assays revealed that dioscin suppressed proliferation, colony formation and invasion of the cancer cells. TUNEL and comet assays demonstrated that dioscin exhibited significant effects on cell apoptosis and DNA damage. Investigations into the mechanism revealed that the expression levels of phosphorylated Ataxia telangiectasia-mutated (ATM) were considerably activated by dioscin, which significantly upregulated the expression levels of p53 to activate mitochondrial apoptosis signaling. Furthermore, the expression levels of BAX, cleaved caspase-3/9 and cleaved poly (ADP-ribose) polymerase were upregulated, and the expression levels of BCL-2 were downregulated by dioscin. Additionally, dioscin markedly downregulated the expression levels of matrix metalloproteinase 2 (MMP2), MMP9, RHO and cdc42, which are all associated with tumor invasion. In addition, p53-small interfering RNA transfection experiments indicated that dioscin exhibited excellent activity against skin cancer in vitro by decreasing p53 expression. Overall, the present results suggested that dioscin inhibited skin cancer cell proliferation via adjusting ATM/p53-mediated cell apoptosis, migration and DNA damage, which should be considered as a potential option for future treatments of skin cancer.
Collapse
Affiliation(s)
- Peng Wang
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, P.R. China
| | - Chun Wang
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, P.R. China
| | - Chunying Liu
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning 110847, P.R. China
| |
Collapse
|
11
|
Becsky D, Szabo K, Gyulai-Nagy S, Gajdos T, Bartos Z, Balind A, Dux L, Horvath P, Erdelyi M, Homolya L, Keller-Pinter A. Syndecan-4 Modulates Cell Polarity and Migration by Influencing Centrosome Positioning and Intracellular Calcium Distribution. Front Cell Dev Biol 2020; 8:575227. [PMID: 33178691 PMCID: PMC7593626 DOI: 10.3389/fcell.2020.575227] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/17/2020] [Indexed: 12/24/2022] Open
Abstract
Efficient cell migration requires cellular polarization, which is characterized by the formation of leading and trailing edges, appropriate positioning of the nucleus and reorientation of the Golgi apparatus and centrosomes toward the leading edge. Migration also requires the development of an asymmetrical front-to-rear calcium (Ca2+) gradient to regulate focal adhesion assembly and actomyosin contractility. Here we demonstrate that silencing of syndecan-4, a transmembrane heparan sulfate proteoglycan, interferes with the correct polarization of migrating mammalian myoblasts (i.e., activated satellite stem cells). In particular, syndecan-4 knockdown completely abolished the intracellular Ca2+ gradient, abrogated centrosome reorientation and thus decreased cell motility, demonstrating the role of syndecan-4 in cell polarity. Additionally, syndecan-4 exhibited a polarized distribution during migration. Syndecan-4 knockdown cells exhibited decreases in the total movement distance during directional migration, maximum and vectorial distances from the starting point, as well as average and maximum cell speeds. Super-resolution direct stochastic optical reconstruction microscopy images of syndecan-4 knockdown cells revealed nanoscale changes in the actin cytoskeletal architecture, such as decreases in the numbers of branches and individual branch lengths in the lamellipodia of the migrating cells. Given the crucial importance of myoblast migration during embryonic development and postnatal muscle regeneration, we conclude that our results could facilitate an understanding of these processes and the general role of syndecan-4 during cell migration.
Collapse
Affiliation(s)
- Daniel Becsky
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Kitti Szabo
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Szuzina Gyulai-Nagy
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Tamas Gajdos
- Department of Optics and Quantum Electronics, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Zsuzsa Bartos
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Center of Excellence, Budapest, Hungary
| | - Arpad Balind
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Laszlo Dux
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Peter Horvath
- Institute of Biochemistry, Biological Research Centre, Hungarian Academy of Sciences, Szeged, Hungary
| | - Miklos Erdelyi
- Department of Optics and Quantum Electronics, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Laszlo Homolya
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences Center of Excellence, Budapest, Hungary
| | - Aniko Keller-Pinter
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| |
Collapse
|
12
|
Yin L, Zhang Y, Shi H, Feng Y, Zhang Z, Zhang L. Proteomic profiling of hepatic stellate cells in alcohol liver fibrosis reveals proteins involved in collagen production. Alcohol 2020; 86:81-91. [PMID: 32171770 DOI: 10.1016/j.alcohol.2020.02.167] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 01/17/2020] [Accepted: 02/28/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Hepatic stellate cell (HSC) activation has central functions in alcohol-induced liver fibrosis. Proteins of HSCs in alcoholic liver fibrosis (ALF) are still not completely understood. Here, we performed a proteomic study to discover proteins related to ALF using HSCs isolated from a rat model. METHODS Sprague-Dawley rats were fed with ethanol for 2 or 6 weeks. Liver histology was assessed using Sirius red and Oil red O staining. HSCs were enriched by using Percoll density gradient centrifugation, and analyzed using flow cytometry. Proteins extracted from HSCs were separated using two-dimensional electrophoresis (2DE). Differentially expressed proteins were identified using liquid chromatography-mass spectrometry (LC-MS). The characteristics of the differentially expressed proteins were analyzed using the UniProtKB database and STRING software. The mRNA levels of two differentially expressed proteins were analyzed using real-time RT-PCR, of which NADH dehydrogenase (ubiquinone) flavoprotein 2, mitochondrial (Ndufv2) was further investigated using Western blot (WB) and immunohistochemical analysis in the ALF model and human liver tissues. The relationship between Ndufv2 and alcohol stimulation was evaluated using WB. Next, Ndufv2 was knocked-down by shRNA in the HSC-T6 cell line. Three genes (encoding collagen, metalloproteinase inhibitor 1 [TIMP-1], and α-smooth muscle actin [a-SMA]) related to HSC activation were detected. RESULTS An ALF model was successfully established, with a liver fibrosis score of 1-2 (S1-2), and some big fat vacuoles development. Twenty-one non-abundant proteins with more than a 2-fold difference were identified using mass spectrometry, including 7 upregulated and 14 downregulated proteins. These differential proteins are a response to antigen presentation, mitochondrial metabolism, ethanol, and collagen degradation. Among them, two upregulated proteins (Ndufv2 and ATP synthase subunit alpha, mitochondrial [ATP5a1]) were involved in mitochondrial metabolism in ALF, and showed concurrent changes in mRNA and protein levels. Ndufv2 was upregulated in HSCs, as shown by WB, in non-parenchymal cells (NPCs) in the rat model and human liver tissues, and detected using immunohistochemistry. Ndufv2 was also upregulated after alcohol stimulation. Following Ndufv2 knockdown, collagen, TIMP-1, and α-SMA were downregulated compared with that in the controls. CONCLUSIONS A proteomic study was performed to discover proteins related to ALF in HSCs isolated from a rat model. Twenty-one differentially expressed proteins were identified, including proteins involved in mitochondrial metabolism and antigen presentation. Ndufv2, an upregulated protein in ALF, might be involved in ALF through regulating the production of fibrosis factors.
Collapse
|
13
|
Yang Y, Zhao J, Song X, Li L, Li F, Shang J, Wang WW. Amygdalin reduces lipopolysaccharide-induced chronic liver injury in rats by down-regulating PI3K/AKT, JAK2/STAT3 and NF-κB signalling pathways. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2688-2697. [PMID: 31257932 DOI: 10.1080/21691401.2019.1634084] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This study was aimed to evaluate the anti-inflammatory potential of AG on lipopolysaccharide (LPS) -induced liver injury and investigate the underlying mechanism. Administration of LPSs in the rat produced rat liver injury model which was ascertained at histological and molecular levels. Those models were treated with a range of doses of LPSs (0.5, 1.0 and 1.5 mg/kg body weight), followed by measurement physical parameter and function of the liver. Within the max treatment doses, no toxicity was shown but protective effects of AG were evidenced by regulation of physical parameters and functions of the liver. Interestingly, nuclear factor kappa B (NF-κB) levels and inflammatory factors were down-regulated by AG. Furthermore, the histopathological analysis demonstrated that AG promoted recovery from dysfunction of liver tissue in the rats, which was further confirmed by observing expression changes of inflammation-associated proteins. Particularly, alteration in the PI3K/AKT and JAK2/STAT3 signalling pathway protein expression were regulated by AG in a dose-dependent manner, indicating the mechanism underlying the relief effect of AG in liver injury. Our study demonstrated the potential of AG in the management of complications related to liver injury.
Collapse
Affiliation(s)
- Yang Yang
- a Department of Nosocomial Infection Management, the First Affiliated Hospital of Zhengzhou University , Zhengzhou , PR China
| | - Jie Zhao
- b Center of Telemedicine, the First Affiliated Hospital of Zhengzhou University , Zhengzhou , PR China
| | - Xiaoqin Song
- b Center of Telemedicine, the First Affiliated Hospital of Zhengzhou University , Zhengzhou , PR China
| | - Lifeng Li
- b Center of Telemedicine, the First Affiliated Hospital of Zhengzhou University , Zhengzhou , PR China
| | - Fuqin Li
- a Department of Nosocomial Infection Management, the First Affiliated Hospital of Zhengzhou University , Zhengzhou , PR China
| | - Jia Shang
- c Department of Infectious Diseases, Henan Provincial People's Hospital , Zhengzhou , Henan , PR China
| | - Wei-Wei Wang
- d Department of Pathology, The Fourth Affiliated Hospital Of Nantong University & The Sixth People's Hospital of Yancheng City , Yancheng , PR China
| |
Collapse
|
14
|
Design, Preparation, and Characterization of Dioscin Nanosuspensions and Evaluation of Their Protective Effect against Carbon Tetrachloride-Induced Acute Liver Injury in Mice. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:3907915. [PMID: 31814841 PMCID: PMC6878791 DOI: 10.1155/2019/3907915] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/20/2018] [Accepted: 07/17/2019] [Indexed: 02/06/2023]
Abstract
The purpose of this study was to prepare a dioscin nanosuspension (Dio-NS) that has a better distance and high solubility for oral administration and to evaluate its hepatoprotective effects. Optimal primary manufacture parameters, including shear time, shear speed, emulation temperature, pressure, and cycles of homogenization, were determined by single-factor experiments. The concentrations of dioscin, SDS, and soybean lecithin were optimized using the central composite design-response surface method, and their effects on the mean particle size (MPS) and particle size distribution of Dio-NS were investigated. Characterization of the Dio-NS formulations included examinations of the surface morphology and physical status of dioscin in Dio-NS, the stability of Dio-NS at different temperatures, in vitro solubility, and liver protective effect in vivo. Under optimal conditions, Dio-NS had an MPS of 106.72 nm, polydispersity index of 0.221, and zeta potential of −34.27 mV. Furthermore, the proportion of dioscin in Dio-NS was approximately 21.26%. The observation of particles with a spherical shape and the disappearance of crystalline peaks indicated that the physical and chemical properties of Dio-NS were altered. Furthermore, we observed that the dissolution of Dio-NS was superior to that of a physical mixture and Dio-GZF. Moreover, Dio-NS was demonstrated to have a protective effect against CCl4-induced acute liver damage in mice that was equivalent to that of silymarin (a positive control drug) at the same dose. The good hepatoprotective effect of our Dio-NS preparation can provide a theoretical basis for investigating its absorption mechanisms in the body.
Collapse
|
15
|
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
|
16
|
Chiang KC, Lai CY, Chiou HL, Lin CL, Chen YS, Kao SH, Hsieh YH. Timosaponin AIII inhibits metastasis of renal carcinoma cells through suppressing cathepsin C expression by AKT/miR-129-5p axis. J Cell Physiol 2019; 234:13332-13341. [PMID: 30604866 DOI: 10.1002/jcp.28010] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 12/10/2018] [Indexed: 12/14/2022]
Abstract
Timosaponin AIII (TSAIII) is a steroidal saponin that exerts anticancer activity on various cancer cells. In this study, we explore the effects of TSAIII on renal cell carcinoma (RCC) cells. Our findings show that TSAIII treatment (<8 μM) insignificantly influenced cell viability and cell cycle distribution of human RCC cell lines 786-O, A-498, and ACHN. Further observations revealed that TSAIII inhibited migration and invasion of 786-O and A-498 cells, as well as significantly decreased the production and expression of cathepsin C (CTSC) in both the cell types. Kinase cascade analysis exhibited that PI3K/AKT activation was inhibited, but PTEN expression was increased, in response to TSAIII treatments. Combining TSAIII and PI3K inhibitors, LY294002 synergically reduced the migration and invasion of 786-O and A-498 cells, as well as decreased the CTSC expression in both the cell types. We also observed that miR-129-5p bound to CTSC gene and suppressed the expression of CTSC and demonstrated that the miR-129-5p expression was synergically enhanced by TSAIII and LY294002. In addition, pretreatment with antago-miR-129-5p significantly restored the CTSC expression and the migration and invasion of TSAIII-treated 786-O cells. In conclusion, our findings reveal that TSAIII inhibits the metastatic properties of RCC cells, contributing to the inhibition of PI3K/AKT and the increase of miR-129-5p and the subsequent downregulation of CTSC. This suggests that TSAIII has significant antimetastatic activity against RCC cells and may be beneficial to RCC treatments.
Collapse
Affiliation(s)
- Kuang-Chung Chiang
- Deprtment of Urology, Chung-Kang Branch, Cheng-Ching General Hospital, Taichung, Taiwan
| | - Chung-Yu Lai
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Thoracic Surgery, Chung-Kang Branch, Cheng-Ching General Hospital, Taichung, Taiwan
| | - Hui-Ling Chiou
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Liang Lin
- Institute of Biochemistry, Microbiology, and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Yong-Syuan Chen
- Institute of Biochemistry, Microbiology, and Immunology, Chung Shan Medical University, Taichung, Taiwan
| | - Shao-Hsuan Kao
- Institute of Biochemistry, Microbiology, and Immunology, Chung Shan Medical University, Taichung, Taiwan.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Biochemistry, Microbiology, and Immunology, Chung Shan Medical University, Taichung, Taiwan.,Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan.,Department of Biochemistry, of School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
17
|
Piceatannol attenuates D-GalN/LPS-induced hepatoxicity in mice: Involvement of ER stress, inflammation and oxidative stress. Int Immunopharmacol 2018; 64:131-139. [DOI: 10.1016/j.intimp.2018.08.037] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 08/20/2018] [Accepted: 08/26/2018] [Indexed: 12/12/2022]
|
18
|
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
|
19
|
Yao Z, Han J, Lou S, Jiang L, Xu J, Zhou Y, Fang W, Li X, Zhou L, Qian Y, Lou D. Schisandrin B attenuates lipopolysaccharide-induced activation of hepatic stellate cells through Nrf-2-activating anti-oxidative activity. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:4917-4925. [PMID: 31949567 PMCID: PMC6962939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 08/25/2018] [Indexed: 06/10/2023]
Abstract
Activated hepatic stellate cells (HSCs) are known to have a potential role in increasing the deposition of ECM and elevating proliferation in liver fibrosis, which can be driven by lipopolysaccharide (LPS). Schisandrin B (SB) is a dibenzocyclooctadiene derivative of Schisandra chinensis with anti-oxidative stress activity, but its effective target is unknown. Here, we have evaluated whether SB is protective against the LPS-induced activation of HSCs and have explored the underlying anti-oxidative stress mechanisms of SB. HSCs were treated with SB 1 h prior to LPS, and then incubated for indicated time. Nrf-2 in HSCs was inhibited genetically. The simultaneous effects on Nrf-2 activity, oxidative stress, cell proliferation, and ECM deposition were examined. SB decreased LPS-induced cell proliferation, fibrosis, and oxidative stress in HSCs. We further demonstrated that the protective effects of SB in LPS-induced HSCs activation involve the modulation of Nrf-2. SB, specifically targeting Nrf-2, attenuates the oxidative stress in HSCs. SB also reduces LPS-induced fibrosis and cell viability in HSCs. In addition, Nrf-2 may serve as a therapeutic target for infections or periods of chronic oxidative stress and may help with future drug discovery.
Collapse
Affiliation(s)
- Zhongcai Yao
- Zhuji People’s Hospital of Zhejiang ProvinceZhuji, Shaoxing, Zhejiang, China
| | - Jibo Han
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing UniversityJiaxing, Zhejiang, China
| | - Shuyi Lou
- National Computer Network Emergency Response Technical Team/Coordination Center of ChinaBeijing, China
| | - Liqin Jiang
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing UniversityJiaxing, Zhejiang, China
| | - Jianjiang Xu
- Department of Cardiology, The Second Affiliated Hospital of Jiaxing UniversityJiaxing, Zhejiang, China
| | - Yaxi Zhou
- Zhuji People’s Hospital of Zhejiang ProvinceZhuji, Shaoxing, Zhejiang, China
| | - Weiqin Fang
- Zhuji Sixth People’s Hospital of Zhejiang ProvinceZhuji, Shaoxing, Zhejiang, China
| | - Xiaolin Li
- Zhuji People’s Hospital of Zhejiang ProvinceZhuji, Shaoxing, Zhejiang, China
| | - Liqin Zhou
- Zhuji People’s Hospital of Zhejiang ProvinceZhuji, Shaoxing, Zhejiang, China
| | - Yuanyuan Qian
- Zhuji People’s Hospital of Zhejiang ProvinceZhuji, Shaoxing, Zhejiang, China
| | - Dayong Lou
- Zhuji People’s Hospital of Zhejiang ProvinceZhuji, Shaoxing, Zhejiang, China
| |
Collapse
|
20
|
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
|
21
|
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
|