1
|
Wang C, Jiao S, Zhou R, Huang P, Zeng B, Yang Z, Wang J. Momordin Ic ameliorates psoriasis skin damage in mice via the IL-23/IL-17 axis. Arch Dermatol Res 2024; 316:474. [PMID: 39007937 DOI: 10.1007/s00403-024-03023-y] [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: 04/02/2024] [Revised: 04/02/2024] [Accepted: 04/26/2024] [Indexed: 07/16/2024]
Abstract
Psoriasis, a chronic and easily recurring inflammatory skin disease, causes a great economic burden to the patient's family because the etiology and mechanism are still unclear and the treatment cycle is long. In this study, the function and related mechanisms of Momordin Ic in psoriasis were investigated. The IMQ-induced mouse psoriasis model was constructed. The protective effects of different doses of Momordin Ic on psoriasis skin damage in mice were detected by PASI score, HE staining and Ki-67 staining. A psoriasis-like keratinocyte model was established at the cellular level using M5 (IL-17A, IL-22, oncostatin M, IL-1α, and TNF-α) triggered HaCaT. The effects of Momordin Ic upon HaCaT cell biological behavior were examined using MTT and CCK-8 assays. In terms of mechanism, the expression level of each inflammatory factor was assessed using IHC staining and/or ELISA, qRT-PCR, the expression of oxidative stress-related indicators was detected biochemically, and western blot was performed to detect the levels of key proteins of the Wnt signaling and VEGF. As the results shown, at the in vivo level, Momordin Ic significantly alleviated skin damage, reduced PASI score and inhibited hyperproliferation of keratinized cells in psoriasis mice. At the cellular level, Momordin Ic also significantly reversed M5-induced hyperproliferation of HaCaT keratinocytes. In terms of mechanism, Momordin Ic significantly inhibited the IL-23/IL-17 axis, dramatically elevated the levels of intracellular antioxidants including SOD, GSH-Px, and CAT, and significantly down-regulated the levels of the indicator of oxidative damage, malondialdehyde (MDA). In addition, Momordin Ic also significantly inhibited the level of β-catenin, a pivotal protein of the Wnt signaling, C-Myc, a target gene of the Wnt signaling, and VEGF, a critical protein of angiogenesis. In conclusion, Momordin Ic can be involved in the skin-protective effects of psoriasis by multiple mechanisms, including inhibition of the Wnt signaling pathway and the IL-23/IL-17 axis, and suppression of oxidative damageand VEGF expression. Momordin Ic has been proven to be an underlying therapeutic drug for the treatment of psoriasis.
Collapse
Affiliation(s)
- Chang Wang
- Department of Dermatology, the Second Affiliated Hospital, The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410005, Hunan, China
| | - Simin Jiao
- Department of Dermatology, the Second Affiliated Hospital, The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410005, Hunan, China
| | - Rong Zhou
- Department of Dermatology, the Second Affiliated Hospital, The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410005, Hunan, China
| | - Pan Huang
- Department of Dermatology, the Second Affiliated Hospital, The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410005, Hunan, China
| | - Bijun Zeng
- Department of Dermatology, the Second Affiliated Hospital, The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410005, Hunan, China
| | - Zhibo Yang
- Department of Dermatology, the Second Affiliated Hospital, The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410005, Hunan, China
| | - Junwen Wang
- Department of Dermatology, the Second Affiliated Hospital, The Domestic First-Class Discipline Construction Project of Chinese Medicine, Hunan University of Chinese Medicine, Changsha, 410005, Hunan, China.
| |
Collapse
|
2
|
Pocasap P, Prawan A, Kongpetch S, Senggunprai L. Network pharmacology- and cell-based assessments identify the FAK/Src pathway as a molecular target for the antimetastatic effect of momordin Ic against cholangiocarcinoma. Heliyon 2024; 10:e32352. [PMID: 38961933 PMCID: PMC11219314 DOI: 10.1016/j.heliyon.2024.e32352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Previous studies have indicated the efficacy of momordin Ic (MIc), a plant-derived triterpenoid, against several types of cancers, implying its potential for further development. However, comprehensive insights into the molecular mechanisms and targets of MIc in cholangiocarcinoma (CCA) are lacking. This study aimed to investigate the actions of MIc against CCA at the molecular level. Network pharmacology analysis was first employed to predict the mechanisms and targets of MIc. The results unveiled the potential involvement of MIc in apoptosis and cell migration, pinpointing Src and FAK as key targets. Subsequently, cell-based assays, in accordance with FAK/Src-associated metastasis, were conducted, demonstrating the ability of MIc to attenuate the metastatic behaviours of KKU-452 cells. The in vitro results further indicated the capability of MIc to suppress the epithelial-mesenchymal transition (EMT) process, notably by downregulating EMT regulators, including N-cadherin, vimentin, ZEB2 and FOXC1/2 expression. Furthermore, MIc suppressed the activation of the FAK/Src signalling pathway, influencing critical downstream factors such as MMP-9, VEGF, ICAM-1, and c-Myc. Molecular docking simulations also suggested that MIc could interact with FAK and Src domains and restrain kinases from being activated by hindering ATP binding. In conclusion, this study employs a comprehensive approach encompassing network pharmacology analysis, in vitro assays, and molecular docking to unveil the mechanisms and targets of MIc in CCA. MIc mitigates metastatic behaviours and suppresses key pathways, offering a promising avenue for future therapeutic strategies against this aggressive cancer.
Collapse
Affiliation(s)
- Piman Pocasap
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Auemduan Prawan
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Sarinya Kongpetch
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Laddawan Senggunprai
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, 40002, Thailand
| |
Collapse
|
3
|
Sun J, Hu JR, Liu CF, Li Y, Wang W, Fu R, Guo M, Wang HL, Pang M. ANKRD49 promotes the metastasis of NSCLC via activating JNK-ATF2/c-Jun-MMP-2/9 axis. BMC Cancer 2023; 23:1108. [PMID: 37964204 PMCID: PMC10644579 DOI: 10.1186/s12885-023-11612-9] [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: 04/07/2023] [Accepted: 11/04/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Ankyrin repeat domain 49 (ANKRD49) has been found to be highly expressed in multiple cancer including lung adenocarcinoma (LUAD) and lung squamous carcinoma (LUSC). However, the function of ANKRD49 in the pathogenesis of NSCLC still remains elusive. Previously, ANKRD49 has been demonstrated to promote the invasion and metastasis of A549 cells, a LUAD cell line, via activating the p38-ATF-2-MMP2/MMP9 pathways. Considering the heterogeneity of tumor cells, the function and mechanism of ANKRD49 in NSCLC need more NSCLC-originated cells to clarify. METHODS Real-time qPCR was employed to test ANKRD49 expression levels in nine pairs of fresh NSCLC tissues and the corresponding adjacent normal tissues. The function of ANKRD49 was investigated using overexpression and RNA interference assays in lung adenocarcinoma cell line (NCI-H1299) and lung squamous carcinoma cell line (NCI-H1703) through gelatin zymography, cell counting kit-8, colony formation, wound healing, migration and invasion assays mmunoprecipitation was performed to in vitro. Immunoprecipitation was performed to test the interaction of c-Jun and ATF2. Chromatin immunoprecipitation was conducted to assess the transcriptional regulation of ATF2/c-Jun on MMP-2/9. Moreover, the tumorigenicity of ANKRD49 was evaluated in nude mice models and the involved signal molecular was also measured by immunohistochemical method. RESULTS We found that the levels of ANKRD49 in cancerous tissues were higher than those in adjacent normal tissues. in vitro assay showed that ANKRD49 promoted the migration and invasion of NCI-H1299 and NCI-H1703 cells via enhancing the levels of MMP-2 and MMP-9. Furthermore, ANKRD49 elevated phosphorylation of JNK and then activated c-Jun and ATF2 which interact in nucleus to promote the binding of ATF2:c-Jun with the promoter MMP-2 or MMP-9. In vivo assay showed that ANKRD49 promoted lung metastasis of injected-NSCLC cells and the high metastatic rate was positively correlated with the high expression of ANKRD49, MMP-2, MMP-9, p-JNK, p-c-Jun and p-ATF2. CONCLUSION The present study indicated that ANKRD49 accelerated the invasion and metastasis of NSCLC cells via JNK-mediated transcription activation of c-Jun and ATF2 which regulated the expression of MMP-2/MMP-9. The molecular mechanisms of ANKRD49's function is different from those found in A549 cells. The current study is a supplement and improvement to the previous research.
Collapse
Affiliation(s)
- Jia Sun
- Department of Pulmonary and Critical Care Medicine, Shanxi Province Key Laboratory of Respiratory Disease, the First Hospital, Shanxi Medical University, NHC Key Laboratory of Pneumoconiosis, Taiyuan, Shanxi, 030001, China
- Department of Laboratorial Medicine, Changzhi Traditional Chinese Medicine Hospital, Changzhi, 046000, China
| | - Jin-Rui Hu
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, No. 55 Wenhua Street, Jinzhong, Shanxi, 030600, China
| | - Chao-Feng Liu
- Department of Pulmonary and Critical Care Medicine, Shanxi Province Key Laboratory of Respiratory Disease, the First Hospital, Shanxi Medical University, NHC Key Laboratory of Pneumoconiosis, Taiyuan, Shanxi, 030001, China
| | - Yuan Li
- Department of Respiratory Medicine 1, Shanxi Hospital Affiliated to Cancer Hospital, Shanxi Province Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, 030013, China
| | - Wei Wang
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, No. 55 Wenhua Street, Jinzhong, Shanxi, 030600, China
| | - Rong Fu
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, No. 55 Wenhua Street, Jinzhong, Shanxi, 030600, China
| | - Min Guo
- Laboratory of Animal Center, Shanxi Medical University, Taiyuan, 030001, China
| | - Hai-Long Wang
- School of Basic Medicine, Basic Medical Sciences Center, Shanxi Medical University, No. 55 Wenhua Street, Jinzhong, Shanxi, 030600, China.
| | - Min Pang
- Department of Pulmonary and Critical Care Medicine, Shanxi Province Key Laboratory of Respiratory Disease, the First Hospital, Shanxi Medical University, NHC Key Laboratory of Pneumoconiosis, Taiyuan, Shanxi, 030001, China.
- Department of Pulmonary and Critical Care Medicine, the First Hospital, Shanxi Medical University, No. 85 Jiefang South Road, Taiyuan, Shanxi, 030001, China.
| |
Collapse
|
4
|
Tan M, Zhao Q, Wang X, Zhao B. Study on extraction, isolation and biological activity of saponins from quinoa bran. J FOOD PROCESS PRES 2022. [DOI: 10.1111/jfpp.17155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Minghui Tan
- College of Food Science and Technology Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Province Engineering Laboratory for Marine Biological Products, Key Laboratory of Advanced Processing of Aquatic Product of Guangdong Higher Education Institution, Zhanjiang 524088 China
- Collaborative Innovation Center of Seafood Deep Processing Dalian Polytechnic University,Dalian 116034
| | - Qingsheng Zhao
- Division of Green Biochemical Process, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering Chinese Academy of Sciences 100190 Beijing China
- University of Chinese Academy of Sciences 100049 Beijing China
| | - Xiaodong Wang
- Division of Green Biochemical Process, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering Chinese Academy of Sciences 100190 Beijing China
| | - Bing Zhao
- Division of Green Biochemical Process, State Key Laboratory of Biochemical Engineering, Institute of Process Engineering Chinese Academy of Sciences 100190 Beijing China
| |
Collapse
|
5
|
Yang K, Li C, Wang Y, Hao J. Micro-Vibration Environment Promotes Bone Marrow Mesenchymal Stem Cells (BMSCs) Healing of Fracture Ends and Matrix Metalloproteinase-9 (MMP-9) Expression. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
When the muscle is stimulated by micro-vibration environment, it will produce a mechanical effect on skeletal muscle, thereby promoting growth of skeletal muscle cells. Bone marrow mesenchymal stem cells (BMSCs), as mechanically sensitive cells, have ability to multipolarize and multiple
tropisms. This experiment explores the effect of BMSC cells on fracture end healing in fracture rats in a micro-vibration environment, and further explores whether it promotes the healing of fracture end to provide biological treatment ideas for the clinical repair of fracture patients. After
modeling, SD rats were assigned into blank group, control group, and experimental group (treated with BMSCs) followed by analysis of bone volume fraction and bone trabecular thickness and number by Micro-CT, callus growth by H&E staining, and expression of p38 and MMP-9 by immunohistochemical
staining. The BV/TV value of experimental group was (0.41±0.06), Tb.Th value (0.08±0.01), Tb.N value (3.96±0.48) and was higher than other two groups (P < 0.05). The growth of capillaries, trabecular bone, fibrous callus and cartilaginous callus in experimental
group showed increased growth and the calcification was observed at the edge of cartilage callus. In addition, experimental group showed increased distribution area of MMP-9 and elevated expression of MMP-9 and p38MAPK. In conclusion, the micro-vibration environment can effectively promote
the chemotaxis of BMSC cells to the fracture site to activate ossification, thereby promoting the proliferation and ossification and differentiation of BMSC, and further promoting the repair of fracture ends.
Collapse
Affiliation(s)
- Kai Yang
- Department of Orthopedics, Lanzhou University Second Hospital, Chengguan District, Lanzhou City, Gansu Province, 730030, China
| | - Chenmei Li
- Department of Orthopedics, Lanzhou University Second Hospital, Chengguan District, Lanzhou City, Gansu Province, 730030, China
| | - Yapeng Wang
- Department of Orthopedics, Lanzhou University Second Hospital, Chengguan District, Lanzhou City, Gansu Province, 730030, China
| | - Junlong Hao
- Department of Orthopedics, Lanzhou University Second Hospital, Chengguan District, Lanzhou City, Gansu Province, 730030, China
| |
Collapse
|
6
|
Kong L, Ji H, Gan X, Cao S, Li Z, Jin Y. Knockdown of CD44 inhibits proliferation, migration and invasion of osteosarcoma cells accompanied by downregulation of cathepsin S. J Orthop Surg Res 2022; 17:154. [PMID: 35264209 PMCID: PMC8905747 DOI: 10.1186/s13018-022-03048-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 03/02/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Osteosarcoma (OS) is a malignant bone tumour of mesenchymal origin. These tumours are characterised by rich vascularisation, therefore promoting rapid proliferation and facilitating metastasis. CD44 has been reported to be involved in OS, but its role and molecular mechanisms in the pathogenesis of the disease are not fully determined. METHODS In this study, we investigated the antitumor effect of CD44 on the development of OS and further explored the molecular mechanisms. The expression of CD44, cathepsin S and MMP-9 was detected by Western blot (WB) and reverse transcription-polymerase chain reaction (RT-qPCR) in different cell lines (MG63, U2OS OS and hFOB 1.19). To elucidate the role of CD44 in OS, MG63 and U2OS cells were treated with small interference RNA (siRNA) to knock down CD44, and the knockdown efficiency was validated with GFP and RT-qPCR. Furthermore, cell proliferation was assayed using Cell Counting Kit‑8 (CCK-8) and colony formation assays, and cell migration and invasion were assayed by transwell and wound-healing assays. RESULTS We found that CD44 expression in the MG63 and U2OS OS cell lines was markedly increased compared to that of the human osteoblast hFOB 1.19 cell line. Knockdown of CD44 inhibited proliferation, migration and invasion of MG63 and U2OS cells. Cathepsin S expression in the MG63 and U2OS OS cell lines was increased compared to that of the human osteoblast hFOB 1.19 cell line. When CD44 was knocked down, its expression level went down. CONCLUSION Taken together, our data reinforced the evidence that CD44 knockdown inhibited cell proliferation, migration and invasion of OS cells accompanied by altered expression of cathepsin S. These findings offer new clues for OS development and progression, suggesting CD44 as a potential therapeutic target for OS.
Collapse
Affiliation(s)
- Lingwei Kong
- Department of Orthopaedics, The Affiliated Hospital of Chengde Medical College, No. 1 Nanyingzi Street, Chengde, 067000, Hebei, China
| | - Hairu Ji
- Pathology Teaching and Research Section, Chengde Medical College, Chengde, 067000, Hebei, China
| | - Xintian Gan
- Department of Orthopaedics, The Affiliated Hospital of Chengde Medical College, No. 1 Nanyingzi Street, Chengde, 067000, Hebei, China
| | - Sheng Cao
- Department of Orthopaedics, The Affiliated Hospital of Chengde Medical College, No. 1 Nanyingzi Street, Chengde, 067000, Hebei, China
| | - Zhehong Li
- Department of Orthopaedics, The Affiliated Hospital of Chengde Medical College, No. 1 Nanyingzi Street, Chengde, 067000, Hebei, China
| | - Yu Jin
- Department of Orthopaedics, The Affiliated Hospital of Chengde Medical College, No. 1 Nanyingzi Street, Chengde, 067000, Hebei, China.
| |
Collapse
|
7
|
Chen Z, Ye X, Yuan K, Liu W, Liu K, Li Y, Huang C, Yu Z, Wu D. Lycorine nanoparticles induce apoptosis through mitochondrial intrinsic pathway and inhibit migration and invasion in HepG2 cells. IEEE Trans Nanobioscience 2021; 21:549-559. [PMID: 34851831 DOI: 10.1109/tnb.2021.3132104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Lycorine-nanoparticles (LYC-NPs) were successfully synthesized using anti-solvent precipitation-freeze drying method, and characterized using transmission electron microscopy (TEM), particle size analysis and Fourier transform infrared spectroscopy (FTIR). Then, the antitumor effects of LYC-NPs against HepG2 cells were investigated, and the underlying molecular mechanisms were explored. Our results showed that LYC-NPs displayed potent antiproliferative against HepG2 cells concentration dependently. Flow cytometry analysis exhibited that LYC-NPs triggered apoptosis and impeded cell cycle in G0/G1 phase. Moreover, the up-regulated expression of cleaved caspases-3 and Bax, and decrease of mitochondrial membrane potential and the Bcl-2 expression were involved in LYC-NPs apoptosis, implying that LYC-NPs induced apoptosis via the mitochondrial-mediated apoptosis pathway. Furthermore, LYC-NPs distinctly impaired HepG2 cells migration and invasion with down-regulation of matrix metalloproteinase-2 (MMP-2) and MMP-9 expression. These results indicated that LYC-NPs could be an favorable agent for restraining the growth and metastasis of HepG2 cells.
Collapse
|
8
|
Serala K, Steenkamp P, Mampuru L, Prince S, Poopedi K, Mbazima V. In vitro antimetastatic activity of Momordica balsamina crude acetone extract in HT-29 human colon cancer cells. ENVIRONMENTAL TOXICOLOGY 2021; 36:2196-2205. [PMID: 34272816 DOI: 10.1002/tox.23333] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/19/2021] [Accepted: 07/07/2021] [Indexed: 06/13/2023]
Abstract
Plant-derived compounds and/or extracts have proven to be beneficial for the treatment of a broad spectrum of cancers with minimal side effects. In this study, we investigated whether a crude acetone extract of Momordica balsamina (MBE) can interfere with the metastatic ability of HT-29 colorectal cancer (CRC) cells. The phytochemical composition of MBE was determined by ultra-performance liquid chromatography and cytotoxic effects by the MTT and acridine orange/ethidium bromide staining assays. The effect of MBE on the formation of reactive oxygen species was assessed using the DCFH2 -DA assay. Wound healing assay, transwell cell invasion assay, cell adhesion assay, and the extracellular matrix-cell adhesion array were used to assess the antimetastatic effects of MBE. The effect of MBE on the expression of TNF-α, NF-κB, TIMP-3, MMP-2, and MMP-9 was assessed by western blot analysis. Our results showed that MBE consists of a mixture of compounds without a known anticancer activity in CRC and exhibits cytotoxicity against HT-29 cells. MBE also suppressed reactive oxygen species formation, cell invasion, cell migration, and cell adhesion. The reduction of cell invasion was associated with the downregulation of TNF-α, NF-κB, MMP2, and MMP9 and upregulation of TIMP-3 proteins. We concluded that MBE inhibits the metastatic ability of HT-29 CRC cells in vitro.
Collapse
Affiliation(s)
- Karabo Serala
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Sovenga, South Africa
| | - Paul Steenkamp
- Centre for Plant Metabolomics Research, Department of Biochemistry, Faculty of Science, University of Johannesburg, Auckland Park, South Africa
| | - Leseilane Mampuru
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Sovenga, South Africa
| | - Sharon Prince
- Division of Cell Biology, Department of Human Biology, University of Cape Town, Cape Town, South Africa
| | - Kgomotso Poopedi
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Sovenga, South Africa
| | - Vusi Mbazima
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Sovenga, South Africa
| |
Collapse
|
9
|
Zhang HF, Gao X, Wang X, Chen X, Huang Y, Wang L, Xu ZW. The mechanisms of renin-angiotensin system in hepatocellular carcinoma: From the perspective of liver fibrosis, HCC cell proliferation, metastasis and angiogenesis, and corresponding protection measures. Biomed Pharmacother 2021; 141:111868. [PMID: 34328104 DOI: 10.1016/j.biopha.2021.111868] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/04/2021] [Accepted: 06/28/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common cancers worldwide, of which the occurrence and development involve a variety of pathophysiological processes, such as liver fibrosis, hepatocellular malignant proliferation, metastasis, and tumor angiogenesis. Some important cytokines, such as TGF-β, PI3K, protein kinase B (Akt), VEGF and NF-κB, can regulate the growth, proliferation, diffusion, metastasis, and apoptosis of HCC cells by acting on the corresponding signaling pathways. Besides, many studies have shown that the formation of HCC is closely related to the main components of renin-angiotensin system (RAS), such as Ang II, ACE, ACE2, MasR, AT1R, and AT2R. Therefore, this review focused on liver fibrosis, HCC cell proliferation, metastasis, tumor angiogenesis, and corresponding protective measures. ACE-Ang II-AT1 axis and ACE2-Ang-(1-7)-MasR axis were taken as the main lines to introduce the mechanism of RAS in the occurrence and development of HCC, so as to provide references for future clinical work and scientific research.
Collapse
Affiliation(s)
- Hai-Feng Zhang
- Department of Clinical Medical, the First Clinical Medical College of Anhui Medical University, Hefei, Anhui 230032, China
| | - Xiang Gao
- Department of Clinical Medical, the First Clinical Medical College of Anhui Medical University, Hefei, Anhui 230032, China
| | - Xuan Wang
- Department of Clinical Medical, the Second Clinical Medical College of Anhui Medical University, Hefei, Anhui 230032, China
| | - Xin Chen
- Department of Clinical Medical, the Second Clinical Medical College of Anhui Medical University, Hefei, Anhui 230032, China
| | - Yu Huang
- Department of Clinical Medical, the First Clinical Medical College of Anhui Medical University, Hefei, Anhui 230032, China
| | - Lang Wang
- Department of Clinical Medical, the First Clinical Medical College of Anhui Medical University, Hefei, Anhui 230032, China
| | - Zhou-Wei Xu
- Department of Emergency Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China; Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, China.
| |
Collapse
|
10
|
Xianjun F, Xirui X, Jie T, Huiwen M, Shaojun Z, Qiaoyun L, Yunxin L, Xuqun S. Momordin Ic induces G0/1 phase arrest and apoptosis in colon cancer cells by suppressing SENP1/c-MYC signaling pathway. J Pharmacol Sci 2021; 146:249-258. [PMID: 34049792 DOI: 10.1016/j.jphs.2021.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 04/06/2021] [Accepted: 04/30/2021] [Indexed: 01/02/2023] Open
Abstract
Momordin Ic (MI) is a natural pentacyclic triterpenoid enriched in various Chinese natural medicines such as the fruit of Kochia scoparia (L.) Schrad. Studies have shown that MI presents antitumor properties in liver and prostate cancers. However, the activity and potential mechanisms of MI against colorectal cancer remain elusive. Here, we showed that MI inhibited cell proliferation with G0/1 phase cell cycle arrest in colon cancer cells. Moreover, it was observed that MI increased apoptosis compared to untreated cells. Further investigation showed that the SUMOylation of c-Myc was enhanced by MI and led to the down-regulated protein level of c-Myc, which is involved in regulating cell proliferation and apoptosis. SENP1 has been demonstrated to be critical for the SUMOylation of c-Myc. Meanwhile, knockdown of SENP1 by siRNA abolished the effects of MI on c-Myc level and cell viability in colon cancer cells. Together, these results revealed that MI exerted an anti-tumor activity in colon cancer cells via SENP1/c-Myc signaling pathway. These finding provide an insight into the potential of MI for colon cancer therapy.
Collapse
Affiliation(s)
- Fang Xianjun
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China
| | - Xian Xirui
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Tang Jie
- Controlled Release Pharmaceutical Preparation Laboratory of Hefei University of Technology, Anhui, Hefei, 230000, PR China
| | - Mu Huiwen
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Zheng Shaojun
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China
| | - Ling Qiaoyun
- School of Pharmacy, Anhui Medical University, Anhui, Hefei, 230032, PR China
| | - Liu Yunxin
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, PR China; Nanjing First Hospital, Nanjing Medical University, Nanjing, 210029, PR China.
| | - Sun Xuqun
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, PR China.
| |
Collapse
|
11
|
Li Q, Hu L, Li J, Yu P, Hu F, Wan B, Xu M, Cheng H, Yu W, Jiang L, Shi Y, Li J, Duan M, Long Y, Liu WT. Hydrogen Attenuates Endotoxin-Induced Lung Injury by Activating Thioredoxin 1 and Decreasing Tissue Factor Expression. Front Immunol 2021; 12:625957. [PMID: 33767697 PMCID: PMC7985449 DOI: 10.3389/fimmu.2021.625957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/18/2021] [Indexed: 01/19/2023] Open
Abstract
Endotoxin-induced lung injury is one of the major causes of death induced by endotoxemia, however, few effective therapeutic options exist. Hydrogen inhalation has recently been shown to be an effective treatment for inflammatory lung injury, but the underlying mechanism is unknown. In the current study we aim to investigate how hydrogen attenuates endotoxin-induced lung injury and provide reference values for the clinical application of hydrogen. LPS was used to establish an endotoxin-induced lung injury mouse model. The survival rate and pulmonary pathologic changes were evaluated. THP-1 and HUVECC cells were cultured in vitro. The thioredoxin 1 (Trx1) inhibitor was used to evaluate the anti-inflammatory effects of hydrogen. Hydrogen significantly improved the survival rate of mice, reduced pulmonary edema and hemorrhage, infiltration of neutrophils, and IL-6 secretion. Inhalation of hydrogen decreased tissue factor (TF) expression and MMP-9 activity, while Trx1 expression was increased in the lungs and serum of endotoxemia mice. LPS-stimulated THP-1 and HUVEC-C cells in vitro and showed that hydrogen decreases TF expression and MMP-9 activity, which were abolished by the Trx1 inhibitor, PX12. Hydrogen attenuates endotoxin-induced lung injury by decreasing TF expression and MMP-9 activity via activating Trx1. Targeting Trx1 by hydrogen may be a potential treatment for endotoxin-induced lung injury.
Collapse
Affiliation(s)
- Qian Li
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
- Department of Anesthesiology, Jinling College Affiliated to Nanjing Medical University, Nanjing, China
| | - Liang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Juan Li
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Pan Yu
- Department of Burn and Plastic Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Fan Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Bing Wan
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Miaomiao Xu
- Department of Anesthesiology, Jinling College Affiliated to Nanjing Medical University, Nanjing, China
| | - Huixian Cheng
- Department of Anesthesiology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China
| | - Wanyou Yu
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Liping Jiang
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Yadan Shi
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Jincan Li
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Manlin Duan
- Department of Anesthesiology, Jinling College Affiliated to Nanjing Medical University, Nanjing, China
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
- Department of Anesthesiology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China
- Department of Anesthesiology, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Yun Long
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Wen-Tao Liu
- Department of Anesthesiology, Jiangning Hospital Affiliated to Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
12
|
Kochiae Fructus, the Fruit of Common Potherb Kochia scoparia (L.) Schrad: A Review on Phytochemistry, Pharmacology, Toxicology, Quality Control, and Pharmacokinetics. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:5382684. [PMID: 33603816 PMCID: PMC7868135 DOI: 10.1155/2021/5382684] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 12/08/2020] [Accepted: 01/19/2021] [Indexed: 11/17/2022]
Abstract
Kochiae Fructus (KF) is the fruit of an annual potherb Kochia scoparia (Linn.) Schrad and has been traditionally used for the treatment of diseases in the skin, eyes, and urinary tract for thousands of years in China. Recent studies have showed its anti-inflammatory, antifungal, antiallergic, and antipruritogenic effects to clarify the mechanisms of these actions. Meanwhile, its other effects, such as anticancer, hypoglycemic, and hepatoprotective effects, also have been reported. The achievement of these therapeutic effects is contributed by its chemical constituents. A total of 153 compounds have been identified in KF, mainly including triterpenoids, flavonoids, carbohydrates, amino acids, organic acids, and essential oils. Momordin Ic is the representative triterpene glycoside compound, which is used as a phytochemical marker for the quality control of Kochiae Fructus. The research on toxicity is insufficient, and only one article reported that the LD50 was 7.15 ± 0.03 g/kg for water extract of KF after oral administration in KM mice. In addition, the pharmacokinetic study was carried out on momordin Ic with linear pharmacokinetic characteristics. Above all, this review provides comprehensive information about Kochiae Fructus and may provide the theoretic foundation of its clinical application and further development.
Collapse
|
13
|
Miethe C, Torres L, Beristain J, Zamora M, Price RS. The role of visfatin and resistin in an in vitro model of obesity-induced invasive liver cancer. Can J Physiol Pharmacol 2020; 99:839-846. [PMID: 33356858 DOI: 10.1139/cjpp-2020-0377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Obesity is associated with the development of liver disease and its progression to hepatocellular carcinoma. This link may be attributed to adipocytokines such as visfatin and resistin which have been shown to promote liver cancer incidence and progression. Studies have yet to determine the role of visfatin and resistin in liver cancer, specifically in the context of obesity. The objective of this study was to investigate the effect of neutralizing visfatin and resistin in obese (OB) or normal weight (NW) sera to determine the contribution of these proteins in obesity-induced invasive liver cancer. Sera from OB or NW males was used to determine the efficacy of neutralizing visfatin and resistin to reduce the obesity-induced liver cancer phenotype. HepG2 and SNU-449 cells were exposed to OB and NW sera ± antibodies for visfatin or resistin. The neutralizing antibodies differentially suppressed invasion, reactive oxygen species production, and matrix metalloproteinase-9 secretion. These changes corresponded with a decrease in phosphorylated extracellular signal-regulated kinases and protein kinase B in HepG2 cells, but differences were not observed in CAP1 or β-catenin. In conclusion, visfatin and resistin have differential roles in obesity-associated liver cancer and may be potential targets to reverse the impact of obesity on liver cancer progression.
Collapse
Affiliation(s)
- Candace Miethe
- Nutrition and Foods, Texas State University, San Marcos, TX, USA
| | - Linda Torres
- Department of Psychology, Texas State University, San Marcos, TX, USA
| | | | - Megan Zamora
- Nutrition and Foods, Texas State University, San Marcos, TX, USA
| | - Ramona S Price
- Nutrition and Foods, Texas State University, San Marcos, TX, USA
| |
Collapse
|
14
|
Wang J, Han Y, Wang M, Zhao Q, Chen X, Liu X. Natural triterpenoid saponin Momordin Ic suppresses HepG2 cell invasion via COX-2 inhibition and PPARγ activation. Toxicol In Vitro 2020; 65:104784. [DOI: 10.1016/j.tiv.2020.104784] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 01/03/2020] [Accepted: 01/22/2020] [Indexed: 12/12/2022]
|
15
|
Gong F, Chen MF, Chen J, Li C, Zhou C, Hong P, Sun S, Qian ZJ. Boiled Abalone Byproduct Peptide Exhibits Anti-Tumor Activity in HT1080 Cells and HUVECs by Suppressing the Metastasis and Angiogenesis in Vitro. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:8855-8867. [PMID: 31343893 DOI: 10.1021/acs.jafc.9b03005] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Abalone (Haliotis discus hannai) is a precious seafood in the market. It has been reported that biological active substances derived from abalone have anti-oxidative, anti-inflammatory, anti-bacterial, and anti-thrombosis potential. However, there were few studies to assess whether they have anti-cancer potential. In this study, we evaluated the anti-metastasis and anti-pro-angiogenic factors and mechanism of action of boiled abalone byproduct peptide (BABP, EMDEAQDPSEW) in human fibrosarcoma (HT1080) cells and human umbilical vein endothelial cells (HUVECs). The results demonstrated that BABP treatment significantly lowers migration and the invasion of HT1080 cells and HUVECs. BABP inhibits phorbol 12-myristate 13-acetate (PMA)-induced matrix metalloproteinase (MMP) expression and activity by blocking mitogen-activated protein kinases (MAPKs) and NF-κB signaling and hypoxia-induced vascular endothelial growth factor (VEGF) secretion and hypoxia inducible factor (HIF)-1α accumulation through suppressing the AKT/mTOR signal pathway. BABP treatment inhibits VEGF-induced VEGFR-2 expression and tube formation in HUVECs. The effect of BABP on anti-metastatic and anti-vascular activity in HT1080 cells and HUVECs revealed that BABP may be a potential pharmacophore for tumor therapy in the future.
Collapse
Affiliation(s)
- Fang Gong
- College of Food Science and Technology , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
| | - Mei-Fang Chen
- College of Food Science and Technology , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
| | - Jiali Chen
- College of Food Science and Technology , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
| | - ChengYong Li
- School of Chemistry and Environment , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
- Shenzhen Institute of Guangdong Ocean University , Shenzhen , Guangdong 518114 , China
| | - ChunXia Zhou
- College of Food Science and Technology , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
| | - PengZhi Hong
- College of Food Science and Technology , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
| | - ShengLi Sun
- School of Chemistry and Environment , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
| | - Zhong-Ji Qian
- School of Chemistry and Environment , Guangdong Ocean University , Zhanjiang , Guangdong 524088 , China
- Shenzhen Institute of Guangdong Ocean University , Shenzhen , Guangdong 518114 , China
| |
Collapse
|