1
|
Yang B, Jue XY, Luo SF, Tan ZB, Yang LN, Feng YT, Tan YZ, Liu B, Zhang JZ, Deng B, Wu WW, Zhang SW. Ilexgenin A Alleviates Myocardial Ferroptosis in Response to Ischemia Reperfusion Injury via the SIRT1 Pathway. Phytother Res 2025; 39:938-956. [PMID: 39698933 DOI: 10.1002/ptr.8414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024]
Abstract
Myocardial ischemia-reperfusion (I/R) injury has emerged as an increasingly serious cardiovascular health concern worldwide, with ferroptosis playing a pivotal role as the underlying pathogenic process. This study aimed to investigate the pharmacological effect and mechanism of Ilexgenin A on cardiomyocyte ferroptosis induced by myocardial I/R injury. In vivo, we established a murine anterior descending artery ligation/recanalization model to evaluate the cardioprotective effect of Ilexgenin A. Bioinformatics analysis, molecular docking, and Surface Plasmon Resonance imaging were conducted to predict the pharmacological targets of Ilexgenin A. In vitro experiments, the neonatal rat cardiomyocytes (NRCMs) were utilized to further explore the mechanism of Ilexgenin A in inhibiting ferroptosis using chemiluminescence and immunofluorescence staining, electron microscopy, biochemical assay, RT-qPCR, western blotting, and so on. The results showed that Ilexgenin A protected against cardiac dysfunction, ameliorated myocardial ferroptosis and mitochondrial damage induced by murine myocardial I/R injury via the silence information regulator 1 (SIRT1) pathway, the trend was consistently observed in NRCMs. Additionally, the SIRT1 knockdown by siRNA delivery partially abrogated the beneficial effects of Ilexgenin A on ameliorating mitochondrial damage, and then aggravated erastin-induced ferroptosis in NRCMs. Overall, Our research demonstrated that the inhibition of ferroptosis via the SIRT1 pathway was one of the mechanisms by which Ilexgenin A exerted cardioprotective effect.
Collapse
Affiliation(s)
- Bo Yang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiao-Yu Jue
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shang-Fei Luo
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhang-Bin Tan
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Li-Ning Yang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yun-Ting Feng
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yong-Zhen Tan
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bin Liu
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jing-Zhi Zhang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bo Deng
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wei-Wei Wu
- Department of Rehabilitation, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Shuang-Wei Zhang
- Department of Traditional Chinese Medicine, Guangzhou Institute of Cardiovascular Disease, State Key Laboratory of Respiratory Disease, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
2
|
Wen S, Cai X, Zhou K, Min Y, Shang C, Shen L, Deng L, Liu D, Qiao G, Shen X. Metabolome and comparative genome provide insights into secondary metabolites generation of a rare karst-growing Rhododendron in vitro culture. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2025; 121:e17235. [PMID: 39935165 DOI: 10.1111/tpj.17235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/18/2024] [Indexed: 02/13/2025]
Abstract
Rhododendron species have the potential to be rich in secondary metabolites with pharmaceutical or industrial value. However, there is a lack of comprehensive metabolome studies at the genome level, particularly for unique and rare species like Rhododendron bailiense, which exclusively grows in karst environments in Guizhou, southwest China. Recently, genome assembly data for this species was available. In this study, nontargeted metabolomics was employed to investigate the secondary metabolites profile of R. bailiense callus. The callus of R. bailiense was induced using 0.2 mg L-1 TDZ (Thidiazuron) + 0.1 mg L-1 IBA (3-Indole butyric acid). A comparison between light-treated calli and dark-cultured calli revealed differential accumulation of metabolites, particularly in flavonoids, terpenoids, coumarins, and hydroxycinnamic acids, known for their beneficial effects such as antioxidant, anticancer, and anti-inflammatory properties. Proanthocyanidins, with various health-promoting effects, were found to accumulate significantly in dark-cultured calli. Light conditions promoted diterpene and triterpene products, whereas darkness favored sesquiterpene products. Additionally, the study demonstrated the potential of utilizing Agrobacterium transformation technology on callus suspension cells to enhance secondary metabolite production. Comparison with the genome of Rhododendron molle revealed that the R. bailiense genome exhibited active 'glycosyltransferase activity,' possessed a higher number of copies of monoterpene and sesquiterpene terpene synthases, and contained high copies of specific cytochrome P450 members (CYP71, CYP76, CYP79, CYP82, CYP736). This study offers valuable insights and potential strategies for the biosynthesis and production of Rhododendron secondary metabolites with pharmaceutical or industrial significance.
Collapse
Affiliation(s)
- Sulin Wen
- Department of Plant Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
- Key Laboratory of Mountain Plant Resources Protection and Germplasm Innovation (Ministry of Education), Institute of Agro-bioengineering/College of Life Sciences, Guizhou University, Guiyang, 550025, Guizhou Province, China
- School of Design, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaowei Cai
- Key Laboratory of Mountain Plant Resources Protection and Germplasm Innovation (Ministry of Education), Institute of Agro-bioengineering/College of Life Sciences, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Kui Zhou
- Key Laboratory of Mountain Plant Resources Protection and Germplasm Innovation (Ministry of Education), Institute of Agro-bioengineering/College of Life Sciences, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Yi Min
- Key Laboratory of Mountain Plant Resources Protection and Germplasm Innovation (Ministry of Education), Institute of Agro-bioengineering/College of Life Sciences, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Chunqiong Shang
- Key Laboratory of Mountain Plant Resources Protection and Germplasm Innovation (Ministry of Education), Institute of Agro-bioengineering/College of Life Sciences, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Luonan Shen
- Key Laboratory of Mountain Plant Resources Protection and Germplasm Innovation (Ministry of Education), Institute of Agro-bioengineering/College of Life Sciences, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Lin Deng
- Key Laboratory of Mountain Plant Resources Protection and Germplasm Innovation (Ministry of Education), Institute of Agro-bioengineering/College of Life Sciences, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Di Liu
- Majorbio Bio-Pharm Technology Co., Ltd, Shanghai, 201203, China
| | - Guang Qiao
- Key Laboratory of Mountain Plant Resources Protection and Germplasm Innovation (Ministry of Education), Institute of Agro-bioengineering/College of Life Sciences, Guizhou University, Guiyang, 550025, Guizhou Province, China
| | - Xiaohui Shen
- Department of Plant Science, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
- School of Design, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
3
|
Zhou Q, Wang Y, Cheng Y, Zhou J, Liu W, Ma X, Tang S, Tang S, Tang C. Ilexgenin A inhibits lipid accumulation in macrophages and reduces the progression of atherosclerosis through PTPN2/ERK1/2/ABCA1 signalling pathway. Biochim Biophys Acta Mol Cell Biol Lipids 2024; 1869:159533. [PMID: 39009241 DOI: 10.1016/j.bbalip.2024.159533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
Macrophage lipid accumulation indicates a pathological change in atherosclerosis. Ilexgenin A (IA), a pentacyclic triterpenoid compound, plays a role in preventing inflammation, bacterial infection, and fatty liver and induces a potential anti-atherogenic effect. However, the anti-atherosclerotic mechanism remains unclear. The present study investigated the effects of IA on lipid accumulation in macrophage-derived foam cells and atherogenesis in apoE-/- mice. Our results indicated that the expression of adenosine triphosphate-binding cassette transporter A1 (ABCA1) was up-regulated by IA, promoting cholesterol efflux and reducing lipid accumulation in macrophages, which may be regulated by the protein tyrosine phosphatase non-receptor type 2 (PTPN2)/ERK1/2 signalling pathway. IA attenuated the progression of atherosclerosis in high-fat diet-fed apoE-/- mice. PTPN2 knockdown with siRNA or treatment with an ERK1/2 agonist (Ro 67-7476) impeded the effects of IA on ABCA1 upregulation and cholesterol efflux in macrophages. These results suggest that IA inhibits macrophage lipid accumulation and alleviates atherosclerosis progression via the PTPN2/ERK1/2 signalling pathway.
Collapse
Affiliation(s)
- Qinyi Zhou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China; Department of Cardiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yang Wang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yaqiong Cheng
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jing Zhou
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Wang Liu
- Department of Gastrointestinal Surgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan, China
| | - Xiaofeng Ma
- Department of Cardiology, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Shilin Tang
- Department of Critical Care Medicine, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Shangshu Tang
- The Seventh Affiliated Hospital University of South China / Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China.
| | - Chaoke Tang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China.
| |
Collapse
|
4
|
Kanwal A, Zhang Z. Exploring common pathogenic association between Epstein Barr virus infection and long-COVID by integrating RNA-Seq and molecular dynamics simulations. Front Immunol 2024; 15:1435170. [PMID: 39391317 PMCID: PMC11464307 DOI: 10.3389/fimmu.2024.1435170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/27/2024] [Indexed: 10/12/2024] Open
Abstract
The term "Long-COVID" (LC) is characterized by the aftereffects of COVID-19 infection. Various studies have suggested that Epstein-Barr virus (EBV) reactivation is among the significant reported causes of LC. However, there is a lack of in-depth research that could largely explore the pathogenic mechanism and pinpoint the key genes in the EBV and LC context. This study mainly aimed to predict the potential disease-associated common genes between EBV reactivation and LC condition using next-generation sequencing (NGS) data and reported naturally occurring biomolecules as inhibitors. We applied the bulk RNA-Seq from LC and EBV-infected peripheral blood mononuclear cells (PBMCs), identified the differentially expressed genes (DEGs) and the Protein-Protein interaction (PPI) network using the STRING database, identified hub genes using the cytoscape plugins CytoHubba and MCODE, and performed enrichment analysis using ClueGO. The interaction analysis of a hub gene was performed against naturally occurring bioflavonoid molecules using molecular docking and the molecular dynamics (MD) simulation method. Out of 357 common genes, 22 genes (CCL2, CCL20, CDCA2, CEP55, CHI3L1, CKAP2L, DEPDC1, DIAPH3, DLGAP5, E2F8, FGF1, NEK2, PBK, TOP2A, CCL3, CXCL8, DEPDC1, IL6, RETN, MMP2, LCN2, and OLR1) were classified as hub genes, and the remaining ones were classified as neighboring genes. Enrichment analysis showed the role of hub genes in various pathways such as immune-signaling pathways, including JAK-STAT signaling, interleukin signaling, protein kinase signaling, and toll-like receptor pathways associated with the symptoms reported in the LC condition. ZNF and MYBL TF-family were predicted as abundant TFs controlling hub genes' transcriptional machinery. Furthermore, OLR1 (PDB: 7XMP) showed stable interactions with the five shortlisted refined naturally occurring bioflavonoids, i.e., apigenin, amentoflavone, ilexgenin A, myricetin, and orientin compounds. The total binding energy pattern was observed, with amentoflavone being the top docked molecule (with a binding affinity of -8.3 kcal/mol) with the lowest total binding energy of -18.48 kcal/mol. In conclusion, our research has predicted the hub genes, their molecular pathways, and the potential inhibitors between EBV and LC potential pathogenic association. The in vivo or in vitro experimental methods could be utilized to functionally validate our findings, which would be helpful to cure LC or to prevent EBV reactivation.
Collapse
Affiliation(s)
- Ayesha Kanwal
- MOE Key Laboratory for Cellular Dynamics and Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhiyong Zhang
- MOE Key Laboratory for Cellular Dynamics and Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Department of Physics, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
5
|
Xie Y, Jin Y, Wen J, Li G, Huai X, Duan Y, Ni F, Fu J, Li M, Li L, Yan M, Cao L, Xiao W, Yang H, Wang ZZ. A novel Alisma orientale extract alleviates non-alcoholic steatohepatitis in mice via modulation of PPARα signaling pathway. Biomed Pharmacother 2024; 176:116908. [PMID: 38850668 DOI: 10.1016/j.biopha.2024.116908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/10/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD), particularly advanced non-alcoholic steatohepatitis (NASH), leads to irreversible liver damage. This study investigated the therapeutic effects and potential mechanism of a novel extract from traditional Chinese medicine Alisma orientale (Sam.) Juzep (AE) on free fatty acid (FFA)-induced HepG2 cell model and high-fat diet (HFD) + carbon tetrachloride (CCl4)-induced mouse model of NASH. C57BL/6 J mice were fed a HFD for 10 weeks. Subsequently, the mice were injected with CCl4 to induce NASH and simultaneously treated with AE at daily doses of 50, 100, and 200 mg/kg for 4 weeks. At the end of the treatment, animals were fasted for 12 h and then sacrificed. Blood samples and liver tissues were collected for analysis. Lipid profiles, oxidative stress, and histopathology were examined. Additionally, a polymerase chain reaction (PCR) array was used to predict the molecular targets and potential mechanisms involved, which were further validated in vivo and in vitro. The results demonstrated that AE reversed liver damage (plasma levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), hepatocyte ballooning, hepatic steatosis, and NAS score), the accumulation of hepatic lipids (TG and TC), and oxidative stress (MDA and GSH). PCR array analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that AE protects against NASH by regulating the adipocytokine signaling pathway and influencing nuclear receptors such as PPARα. Furthermore, AE increased the expression of peroxisome proliferator-activated receptor gamma coactivator-1α (PPARGC1α) and reversed the decreased expression of PPARα in NASH mice. Moreover, in HepG2 cells, AE reduced FFA-induced lipid accumulation and oxidative stress, which was dependent on PPARα up-regulation. Overall, our findings suggest that AE may serve as a potential therapeutic approach for NASH by inhibiting lipid accumulation and reducing oxidative stress specifically through the PPARα pathway.
Collapse
Affiliation(s)
- Yan Xie
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Kanion School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Yimin Jin
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Kanion School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, PR China
| | - Jianhui Wen
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Guiping Li
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Xue Huai
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Yueyang Duan
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Fuyong Ni
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Juan Fu
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Ming Li
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Liang Li
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Ming Yan
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Liang Cao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Wei Xiao
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 200120, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China
| | - Hao Yang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 200120, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China.
| | - Zhen-Zhong Wang
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Lianyungang, Jiangsu 222001, PR China; Kanion School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210046, PR China; Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, Jiangsu 222001, PR China.
| |
Collapse
|
6
|
Yang WQ, Lu QP, Chen CX, Zhu LP, Zhang X, Xu W, Hu LS, Chen J, Zhao ZX. Six undescribed 23-norursane triterpenoids from the biotransformation of ilexgenin a by endophytic fungi and their vascular protective activity. Fitoterapia 2024; 176:106053. [PMID: 38838828 DOI: 10.1016/j.fitote.2024.106053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/01/2024] [Accepted: 06/01/2024] [Indexed: 06/07/2024]
Abstract
Biotransformation of ursane-type triterpenoid ilexgenin A by endophytic fungi Lasiodiplodia sp. MQD-4 and Pestalotiopsis sp. ZZ-1, isolated from Ilex pubescences and Callicarpa kwangtungensis respectively, was investigated for the first time. Six previously undescribed metabolites (1-6) with 23-norursane triterpenoids skeleton were isolated and their structures were unambiguously established by the analysis of spectroscopic data and single-crystal X-ray crystallographic experiments. Decarboxylation, oxidation, and hydroxylation reactions were observed on the triterpenoid skeleton. Especially, the decarboxylation of C-23 provided definite evidence to understand the biogenetic process of 23-norursane triterpenoids. Moreover, the qualitative analysis of the extract of I. pubescences showed metabolites 1, 3, 4, and 6 could be detected in the originated plant, indicating biotransformation by endophytic fungi is a practical strategy for the isolation of novel natural products. Finally, all isolates were evaluated for the protective activities against H2O2-induced HUVECs dysfunction in vitro. Compound 5 could improve the viability of endothelial cells and decrease the level of intracellular ROS.
Collapse
Affiliation(s)
- Wei-Qun Yang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Qi-Ping Lu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Cai-Xin Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Li-Ping Zhu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xiao Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Wei Xu
- College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Le-Shi Hu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jie Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| | - Zhong-Xiang Zhao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China.
| |
Collapse
|
7
|
Mao J, Tian Y, Luo N. An ion channel-based prognostic model identified TRPV2 and GJB3 as immunotherapy determinants in pancreatic cancer. Heliyon 2024; 10:e27301. [PMID: 38560261 PMCID: PMC10979059 DOI: 10.1016/j.heliyon.2024.e27301] [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: 12/21/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 04/04/2024] Open
Abstract
Background Less than 10% of people who have pancreatic ductal adenocarcinoma (PDAC) will survive the malignancy for five years. The ion channel genes-related biomarker and predictive model were needed for exploitation. Methods Differentially expressed ion channel genes (DEICGs) were detected in PDAC patients. GO and KEGG enrichment analysis was conducted on DEICGs. The prognostic genes were found using Cox regression analysis. After that, a risk model was created and examined. A nomogram was created based on independent predictive analysis. The molecular functions of two risk groups were explored. Immune checkpoint molecule expression was compared in two risk groups. We evaluated the possible cancer immunotherapy response in two risk groups using the TIDE method. We further examined how TRPV2 functions in PDAC as a potent oncogene and regulates the activity of macrophages by in vitro validation, including CCK8, EdU, and Transwell assays. Results A total of twenty-four DEICGs were found. Next, we discovered that two DEICGs (TRPV2 and GJB3) were connected to PDAC patients' overall survival (OS). The risk model was created and validated, and a nomogram was used to forecast the overall survival of PDAC patients. The high-risk group considerably accumulated oncogenic pathways. Furthermore, we discovered a correlation between the expression of critical immunological checkpoints and the risk score. Furthermore, patients in the high-risk category had a lower chance of benefiting from immune therapy. The HPA database confirmed that TRPV2 is expressed as a protein. Lastly, TRPV2 controls macrophage activity and acts as a potent oncogene in PDAC. Conclusion Altogether, this study suggested that two ion channel genes, TRPV2 and GJB3, were potential biomarkers for the prognosis of PDAC and immunotherapy targets, and the research will be crucial for creating novel PDAC treatment targets and predictive molecular indicators.
Collapse
Affiliation(s)
- Jiakai Mao
- Department of Hepatobiliary and Pancreatic Surgery, Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yu Tian
- Department of Vascular Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Nan Luo
- Department of Infection, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
8
|
Zhu Y, Tang H, Zhao H, Lu J, Lin K, Ni J, Zhao B, Wu G, Tan C. Vinpocetine represses the progression of nonalcoholic steatohepatitis in mice by mediating inflammasome components via NF-κB signaling. GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:366-376. [PMID: 37562770 DOI: 10.1016/j.gastrohep.2023.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 06/15/2023] [Accepted: 07/18/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND Inflammasome activation is known to be involved in nonalcoholic steatohepatitis (NASH). Vinpocetine is a derivative of vincamine and is reported to suppress the activation of inflammasome. METHODS This study explored the therapeutical potential of Vinpocetine on NASH. Mice were fed with a choline-deficient (MCD) or chow diet in the presence or absence of Vinpocetine for 8 weeks. H&E staining and biochemical assays were determined to evaluate the hepatic steatosis and fibrosis symptoms. In addition, primary hepatocytes and Kupffer cells were isolated and induced by MCD or lipopolysaccharides/cholesterol crystals with or without Vinpocetine. ELISAs, qPCR, and Western blotting were applied to determine the levels of NASH-related biomarkers in both in vivo mouse model and in vitro cell models. RESULTS Treatment of Vinpocetine did not cause observable side effects against and MCD-induced cells and mouse NASH model. However, treatment of Vinpocetine ameliorated hepatic steatosis and fibrosis and suppressed the levels of alanine transaminase and aspartate transferase in the mouse NASH model. In addition, treatment of Vinpocetine suppressed the mRNA and protein levels of inflammasome components both in vitro and in vivo. CONCLUSION Vinpocetine suppressed NASH in mice by mediating inflammasome components via nuclear factor κB signaling.
Collapse
Affiliation(s)
- Yingwei Zhu
- Department of Gastroenterology, Jiangnan University Medical Center (JUMC), No. 68 Zhongshan Road, Wuxi 214002, Jiangsu, China; Department of Gastroenterology, Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, No. 68 Zhongshan Road, Wuxi 214002, Jiangsu, China
| | - Hong Tang
- Department of Pathology, Jiangnan University Medical Center (JUMC), No. 68 Zhongshan Road, Wuxi 214002, Jiangsu, China
| | - Han Zhao
- Department of Gastroenterology, Jiangnan University Medical Center (JUMC), No. 68 Zhongshan Road, Wuxi 214002, Jiangsu, China
| | - Jian Lu
- Department of Gastroenterology, Jiangnan University Medical Center (JUMC), No. 68 Zhongshan Road, Wuxi 214002, Jiangsu, China
| | - Kai Lin
- Department of Gastroenterology, Jiangnan University Medical Center (JUMC), No. 68 Zhongshan Road, Wuxi 214002, Jiangsu, China
| | - Jingbin Ni
- Department of Gastroenterology, Jiangnan University Medical Center (JUMC), No. 68 Zhongshan Road, Wuxi 214002, Jiangsu, China
| | - Bo Zhao
- Department of Gastroenterology, Jiangnan University Medical Center (JUMC), No. 68 Zhongshan Road, Wuxi 214002, Jiangsu, China
| | - Gaojue Wu
- Department of Gastroenterology, Jiangnan University Medical Center (JUMC), No. 68 Zhongshan Road, Wuxi 214002, Jiangsu, China; Department of Gastroenterology, Wuxi No. 2 People's Hospital, Affiliated Wuxi Clinical College of Nantong University, No. 68 Zhongshan Road, Wuxi 214002, Jiangsu, China.
| | - Chunxiao Tan
- Department of Gastroenterology, Jiangnan University Medical Center (JUMC), No. 68 Zhongshan Road, Wuxi 214002, Jiangsu, China.
| |
Collapse
|
9
|
Manoharan S, Saha S, Murugesan K, Santhakumar A, Perumal E. Natural bioactive compounds and STAT3 against hepatocellular carcinoma: An update. Life Sci 2024; 337:122351. [PMID: 38103726 DOI: 10.1016/j.lfs.2023.122351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/23/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Hepatocellular carcinoma (HCC) is a challenging and very fatal liver cancer. The signal transducer and activator of transcription 3 (STAT3) pathway is a crucial regulator of tumor development and are ubiquitously active in HCC. Therefore, targeting STAT3 has emerged as a promising approach for preventing and treating HCC. Various natural bioactive compounds (NBCs) have been proven to target STAT3 and have the potential to prevent and treat HCC as STAT3 inhibitors. Numerous kinds of STAT3 inhibitors have been identified, including small molecule inhibitors, peptide inhibitors, and oligonucleotide inhibitors. Due to the undesirable side effects of the conventional therapeutic drugs against HCC, the focus is shifted to NBCs derived from plants and other natural sources. NBCs can be broadly classified into the categories of terpenes, alkaloids, carotenoids, and phenols. Most of the compounds belong to the family of terpenes, which prevent tumorigenesis by inhibiting STAT3 nuclear translocation. Further, through STAT3 inhibition, terpenes downregulate matrix metalloprotease 2 (MMP2), matrix metalloprotease 9 (MMP9) and vascular endothelial growth factor (VEGF), modulating metastasis. Terpenes also suppress the anti-apoptotic proteins and cell cycle markers. This review provides comprehensive information related to STAT3 abrogation by NBCs in HCC with in vitro and in vivo evidences.
Collapse
Affiliation(s)
- Suryaa Manoharan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Shreejit Saha
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Krishnasanthiya Murugesan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Aksayakeerthana Santhakumar
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India.
| |
Collapse
|
10
|
Zhang Z, Zhang Q, Yu Y, Su S. Epigallocatechin gallate inhibits ovarian cancer cell growth and induces cell apoptosis via activation of FOXO3A. In Vitro Cell Dev Biol Anim 2023; 59:739-746. [PMID: 38038884 DOI: 10.1007/s11626-023-00830-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/12/2023] [Indexed: 12/02/2023]
Abstract
Epigallocatechin gallate (EGCG), a bioactive component in tea, displays broad anti-cancer effects. Our study was designed to evaluate the anti-cancer effects of EGCG on ovarian cancer and explored the underlying molecular mechanisms. To evaluate the in vitro inhibitory effects of EGCG against ovarian cancer, MTT assay, colony formation assay, apoptosis assay, and wound healing assay, were performed. Besides, the inhibitory effects of EGCG on tumor growth in the xenograft animal model were evaluated by measuring tumor volume and tumor weight. Moreover, Western blotting and qPCR were used to evaluate the levels of target genes and proteins. Treatment with EGCG inhibited cell migration and cell survival, and promoted cell apoptosis in A2780 and SKOV3 cells. Interestingly, treatment with EGCG inhibited the tumor growth in the xenograft animal model. The mechanistic study revealed that treatment with EGCG induced the activation of FOXO3A and suppressed the expression of c-Myc both in vitro and in vivo. Our findings demonstrate that EGCG suppress ovarian cancer cell growth, which may be due to its regulation on FOXO3A and c-Myc.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Gynecology, Zibo Central Hospital, No. 54 Gongqingtuan West Road, Zibo, 255036, Shandong, China
| | - Qinghua Zhang
- Department of Gynecology, Zibo Central Hospital, No. 54 Gongqingtuan West Road, Zibo, 255036, Shandong, China
| | - Yani Yu
- Department of Gynecology, Zibo Central Hospital, No. 54 Gongqingtuan West Road, Zibo, 255036, Shandong, China
| | - Shan Su
- Department of Gynecology, Zibo Central Hospital, No. 54 Gongqingtuan West Road, Zibo, 255036, Shandong, China.
| |
Collapse
|
11
|
Su Y, Liu J, Zheng Z, Shi L, Huang W, Huang X, Ye C, Qi J, Wang W, Zhuang H. NSUN5-FTH1 Axis Inhibits Ferroptosis to Promote the Growth of Gastric Cancer Cells. Cell Biochem Biophys 2023; 81:553-560. [PMID: 37528314 DOI: 10.1007/s12013-023-01152-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 07/23/2023] [Indexed: 08/03/2023]
Abstract
Recent studies revealed that NOP2/Sun RNA methyltransferase 5 (NSUN5) - ferritin heavy chain (FTH1) pathway is associated with ferroptosis in stem cells, whereas its roles in gastric cancer are still unclear. Our study aims to investigate the roles of the NSUN5-FTH1 axis in gastric cancer (GC) and its molecular mechanisms. Stable cell lines were constructed on SGC7901 cells by using shRNAs and pcDNA3.1 expression vectors, respectively. CCK-8 kits were used to determine cell viability. Biochemicals assays were used to detect lipid reactive oxygen species (ROS) and intracellular Fe2+ levels. RNA immunoprecipitation assay, qPCR, and Western blotting were used to determine the changes in biomarkers. GC xenograft mouse model was established to confirm the observation in vivo. An elevation of NSUN5 was observed in GC tumor tissues. NSUN5 inhibited ferroptosis including decreasing cell viability and increasing levels of lipid ROS and Fe2+ in GC cells. Besides, a positive correlation was also observed between NSUN5 and FTH1. Interestingly, NSUN5 regulated the levels of FTH1, instead of FTH1 regulating NSUN5 in GC cells. NSUN5-FTH1 axis regulated erastin-induced ferroptosis in SGC7901 cells. Consistently, silencing NSUN5 or FTH1 inhibited the growth of the SGC7901 tumor in vivo. NSUN5-FTH1 axis promoted the growth of GC cells in part by the regulation of ferroptosis.
Collapse
Affiliation(s)
- Yibin Su
- Department of Gastrointestinal Surgery, Quanzhou First Hospital affiliated to Fujian Medical University, No. 248 East Street, Licheng District, Quanzhou, 362000, Fujian, China.
| | - Jiangrui Liu
- Department of Gastrointestinal Surgery, Quanzhou First Hospital affiliated to Fujian Medical University, No. 248 East Street, Licheng District, Quanzhou, 362000, Fujian, China
| | - Zhihua Zheng
- Department of Gastrointestinal Surgery, Quanzhou First Hospital affiliated to Fujian Medical University, No. 248 East Street, Licheng District, Quanzhou, 362000, Fujian, China
| | - Liangpan Shi
- Department of Gastrointestinal Surgery, Quanzhou First Hospital affiliated to Fujian Medical University, No. 248 East Street, Licheng District, Quanzhou, 362000, Fujian, China
| | - Wenchang Huang
- Department of Gastrointestinal Surgery, Quanzhou First Hospital affiliated to Fujian Medical University, No. 248 East Street, Licheng District, Quanzhou, 362000, Fujian, China
| | - Xiaohui Huang
- Department of Gastrointestinal Surgery, Quanzhou First Hospital affiliated to Fujian Medical University, No. 248 East Street, Licheng District, Quanzhou, 362000, Fujian, China
| | - Chengwei Ye
- Department of Gastrointestinal Surgery, Quanzhou First Hospital affiliated to Fujian Medical University, No. 248 East Street, Licheng District, Quanzhou, 362000, Fujian, China
| | - Jinyu Qi
- Department of Gastrointestinal Surgery, Quanzhou First Hospital affiliated to Fujian Medical University, No. 248 East Street, Licheng District, Quanzhou, 362000, Fujian, China
| | - Weidong Wang
- Department of Gastrointestinal Surgery, Quanzhou First Hospital affiliated to Fujian Medical University, No. 248 East Street, Licheng District, Quanzhou, 362000, Fujian, China
| | - Haibin Zhuang
- Department of Gastrointestinal Surgery, Quanzhou First Hospital affiliated to Fujian Medical University, No. 248 East Street, Licheng District, Quanzhou, 362000, Fujian, China
| |
Collapse
|
12
|
Pinho JO, Matias M, Godinho-Santos A, Amaral JD, Mendes E, Jesus Perry M, Paula Francisco A, Rodrigues CMP, Manuela Gaspar M. A step forward on the in vitro and in vivo assessment of a novel nanomedicine against melanoma. Int J Pharm 2023; 640:123011. [PMID: 37146952 DOI: 10.1016/j.ijpharm.2023.123011] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 04/11/2023] [Accepted: 04/27/2023] [Indexed: 05/07/2023]
Abstract
Melanoma is the most aggressive form of skin cancer, with increasing incidence and mortality rates. To overcome current treatment limitations, a hybrid molecule (HM) combining a triazene and a ʟ-tyrosine analogue, was recently synthesized, incorporated in long blood circulating liposomes (LIP HM) and validated in an immunocompetent melanoma model. The present work constitutes a step forward in the therapeutic assessment of HM formulations. Here, human melanoma cells, A375 and MNT-1, were used and dacarbazine (DTIC), a triazene drug clinically available as first-line treatment for melanoma, constituted the positive control. In cell cycle analysis, A375 cells, after 24-h incubation with HM (60 μM) and DTIC (70 μM), resulted in a 1.2 fold increase (related to control) in the percentage of cells in G0/G1 phase. The therapeutic activity was evaluated in a human murine melanoma model (subcutaneously injected with A375 cells) to most closely resemble the human pathology. Animals treated with LIP HM exhibited the highest antimelanoma effect resulting in a 6-, 5- and 4-fold reduction on tumor volume compared to negative control, Free HM and DTIC groups, respectively. No toxic side effects were detected. Overall, these results constitute another step forward in the validation of the antimelanoma activity of LIP HM, using a murine model that more accurately simulates the pathology that occurs in human patients.
Collapse
Affiliation(s)
- Jacinta O Pinho
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Mariana Matias
- CICS-UBI-Health Sciences Research Centre, University of Beira Interior, 6200-506 Covilhã, Portugal.
| | - Ana Godinho-Santos
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Joana D Amaral
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Eduarda Mendes
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Maria Jesus Perry
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Ana Paula Francisco
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - Cecília M P Rodrigues
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| | - M Manuela Gaspar
- Research Institute for Medicines, iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
13
|
Paskeh MDA, Ghadyani F, Hashemi M, Abbaspour A, Zabolian A, Javanshir S, Razzazan M, Mirzaei S, Entezari M, Goharrizi MASB, Salimimoghadam S, Aref AR, Kalbasi A, Rajabi R, Rashidi M, Taheriazam A, Sethi G. Biological impact and therapeutic perspective of targeting PI3K/Akt signaling in hepatocellular carcinoma: Promises and Challenges. Pharmacol Res 2023; 187:106553. [PMID: 36400343 DOI: 10.1016/j.phrs.2022.106553] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/09/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022]
Abstract
Cancer progression results from activation of various signaling networks. Among these, PI3K/Akt signaling contributes to proliferation, invasion, and inhibition of apoptosis. Hepatocellular carcinoma (HCC) is a primary liver cancer with high incidence rate, especially in regions with high prevalence of viral hepatitis infection. Autoimmune disorders, diabetes mellitus, obesity, alcohol consumption, and inflammation can also lead to initiation and development of HCC. The treatment of HCC depends on the identification of oncogenic factors that lead tumor cells to develop resistance to therapy. The present review article focuses on the role of PI3K/Akt signaling in HCC progression. Activation of PI3K/Akt signaling promotes glucose uptake, favors glycolysis and increases tumor cell proliferation. It inhibits both apoptosis and autophagy while promoting HCC cell survival. PI3K/Akt stimulates epithelial-to-mesenchymal transition (EMT) and increases matrix-metalloproteinase (MMP) expression during HCC metastasis. In addition to increasing colony formation capacity and facilitating the spread of tumor cells, PI3K/Akt signaling stimulates angiogenesis. Therefore, silencing PI3K/Akt signaling prevents aggressive HCC cell behavior. Activation of PI3K/Akt signaling can confer drug resistance, particularly to sorafenib, and decreases the radio-sensitivity of HCC cells. Anti-cancer agents, like phytochemicals and small molecules can suppress PI3K/Akt signaling by limiting HCC progression. Being upregulated in tumor tissues and clinical samples, PI3K/Akt can also be used as a biomarker to predict patients' response to therapy.
Collapse
Affiliation(s)
- Mahshid Deldar Abad Paskeh
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Ghadyani
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Alireza Abbaspour
- Cellular and Molecular Research Center,Qazvin University of Medical Sciences, Qazvin, Iran
| | - Amirhossein Zabolian
- Resident of department of Orthopedics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Salar Javanshir
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrnaz Razzazan
- Medical Student, Student Research Committee, Golestan University of Medical Sciences, Gorgan, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Translational Sciences, Xsphera Biosciences Inc. 6, Tide Street, Boston, MA 02210, USA
| | - Alireza Kalbasi
- Department of Pharmacy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| |
Collapse
|
14
|
Wang F, Shan H, Song G, Chen S, Zhang C, Liu Y, Wu T. 17β-Estradiol attenuates inflammation and tendon degeneration in a rat model of Achilles tendinitis. Immunopharmacol Immunotoxicol 2022; 44:556-564. [PMID: 35404181 DOI: 10.1080/08923973.2022.2065639] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 04/09/2022] [Indexed: 11/05/2022]
Abstract
INTRODUCTION 17β-Estradiol (E2) is an immune-regulatory agent with anti-inflammatory effects. However, it is still unknown whether E2 exerts pharmacological properties against Achilles tendinitis (AT). This study aims to investigate the effects of E2 on AT and its underlying mechanisms. MATERIALS AND METHODS The established model of Achilles tendinitis was intraperitoneally injected with E2 (10, 20, or 30 μg/kg/d). After 8 weeks, biomechanical properties of the Achilles tendon were determined. Hydroxyproline content and tendon degeneration-related biomarkers were determined. The levels of inflammatory cytokines and apoptotic-related biomarkers in tendon tissues were determined. Furthermore, western blotting was determined to detect the expressions of ER-α and the PI3K/Akt pathway in tendon tissues. RESULTS E2 relieved AT-related symptoms in a dose-dependent manner. E2 ameliorated tendon degeneration by regulating tendon degeneration-related biomarkers (e.g. collagen types I and III, Decorin (DCN), and tenascin-C). Besides, treatment with E2 suppressed inflammatory cytokines and increased anti-inflammatory cytokines. Treatment with E2 also regulated cell apoptosis in tendon tissues. The underlying mechanism study revealed that treatment with E2 activated ER-α and upregulated the PI3K/Akt pathway. CONCLUSION The regulatory effects of E2 on inflammation and tendon degeneration in a rat model of AT were associated with the ER-α and the PI3K/Akt signaling pathways.
Collapse
Affiliation(s)
- Feng Wang
- Department of Orthopaedical Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Haojie Shan
- Department of Orthopaedical Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Guoxun Song
- Department of Orthopaedical Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Song Chen
- Department of Orthopaedical Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chengyuan Zhang
- Department of Orthopaedical Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yingjie Liu
- Department of Orthopaedical Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Tianyi Wu
- Department of Orthopaedical Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
15
|
Ding W, Chen X, Yang L, Chen Y, Song J, Bu W, Feng B, Zhang M, Luo Y, Jia X, Feng L. Combination of ShuangDan Capsule and Sorafenib Inhibits Tumor Growth and Angiogenesis in Hepatocellular Carcinoma Via PI3K/Akt/mTORC1 Pathway. Integr Cancer Ther 2022; 21:15347354221078888. [PMID: 35234063 PMCID: PMC8894619 DOI: 10.1177/15347354221078888] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a high mortality liver cancer. The existing treatments (transplantation, chemotherapy, and individualized treatment) with limitations. However, drug combination provides a viable option for hepatocellular carcinoma treatment. A Chinese patent medicine, ShuangDan Capsules (SDC), has been clinically prescribed to hepatocellular carcinoma patients as adjuvant therapy and has shown good antitumor activity. The purpose of this study was to investigate whether SDC could improve the anti-cancer effect and mitigate adverse reactions of sorafenib on HCC in vivo. Magnetic resonance imaging (MRI), immunohistochemistry, and western blot were executed to reveal the potential mechanisms of the combination of SDC and sorafenib on HCC. Tumors appeared hyperintense on T2 sequence images relative to the adjacent normal liver in MRI. Combination of SDC and sorafenib inhibited the progression of DEN (Diethylnitrosamine)-induced HCC. In the HepG2 xenografts model, sorafenib plus SDC exhibited greater suppression on tumor growth than individual treatment accompanied with decreased expression of VEGF, VEGFA, Ki67, CD31 and increased expression of caspase-3. Furthermore, PI3K/Akt/mTORC1 pathway was inhibited by co-administration. Sorafenib monotherapy elicited hepatotoxicity for specific expression in the up-regulated level of aspartate transaminase (AST) and AST/glutamic-pyruvic transaminase (ALT) ratio, but the co-administration could remedy this adverse effect. These dates indicated that the combination of SDC and sorafenib might offer a potential therapy for HCC.
Collapse
Affiliation(s)
- Wenbo Ding
- Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xiuwei Chen
- Yuhuatai District Maternity and Child Care Clinic, Nanjing, P.R. China
| | - Licheng Yang
- China Pharmaceutical University, Nanjing, P.R. China
| | - Yaping Chen
- China Pharmaceutical University, Nanjing, P.R. China
| | - Jie Song
- Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Weiquan Bu
- Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Bin Feng
- Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Meng Zhang
- China Pharmaceutical University, Nanjing, P.R. China
| | - Yi Luo
- Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xiaobin Jia
- China Pharmaceutical University, Nanjing, P.R. China
| | - Liang Feng
- China Pharmaceutical University, Nanjing, P.R. China
| |
Collapse
|
16
|
Zhou A, Cai Q, Hong Y, Lv Y. Down-Regulation of Casein Kinase 1α Contributes to Endometriosis through Phosphatase and Tensin Homolog/Autophagy-Related 7-Mediated Autophagy. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:2195-2202. [PMID: 34809787 DOI: 10.1016/j.ajpath.2021.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/09/2021] [Accepted: 08/18/2021] [Indexed: 06/13/2023]
Abstract
The present study aimed to explore the roles of casein kinase 1α (CK1α) in endometriosis and its underlying mechanisms. Endometrial specimen were collected from the patients and healthy volunteers. The expression patterns of CK1α, phosphatase and tensin homolog (PTEN), and autophagy-related proteins were determined using immunohistochemistry staining, Western blot analysis, and quantitative RT-PCR. Besides, the CK1α-overexpressing cells and PTEN knockdown cells were constructed in the endometrial stromal cells isolated from endometriosis patients. In addition, the cells were transfected with pcDNA3.1-CK1α or pcDNA3.1-CK1α plus siRNA- PTEN. The expressions of CK1α, PTEN, and autophagy-related proteins were determined using Western blot and quantitative RT-PCR. The expressions of CK1α and autophagy-related 7 (Atg7) were significantly decreased in the ectopic endometrium compared with the eutopic endometrium. Spearman rank correlation analysis revealed positive correlations between CK1α and PTEN, CK1α and Atg7, and PTEN and Atg7. In addition, CK1α, PTEN, and autophagy-related proteins were down-regulated in ectopic endometrium. Interestingly, overexpression of CK1α significantly increased the expressions of autophagy-related proteins, whereas the protein expression of autophagy-related proteins was decreased with PTEN knock-down. CK1α regulated PTEN/Atg7-mediated autophagy in endometriosis.
Collapse
Affiliation(s)
- Aixiu Zhou
- Department of Gynaecology and Obstetrics, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Qiongyi Cai
- Department of Gynaecology and Obstetrics, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Yiting Hong
- Department of Gynaecology and Obstetrics, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Yuchun Lv
- Department of Gynaecology and Obstetrics, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China.
| |
Collapse
|
17
|
Anti-Hepatocellular Carcinoma Biomolecules: Molecular Targets Insights. Int J Mol Sci 2021; 22:ijms221910774. [PMID: 34639131 PMCID: PMC8509806 DOI: 10.3390/ijms221910774] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/25/2021] [Accepted: 09/26/2021] [Indexed: 12/15/2022] Open
Abstract
This report explores the available curative molecules directed against hepatocellular carcinoma (HCC). Limited efficiency as well as other drawbacks of existing molecules led to the search for promising potential alternatives. Understanding of the cell signaling mechanisms propelling carcinogenesis and driven by cell proliferation, invasion, and angiogenesis can offer valuable information for the investigation of efficient treatment strategies. The complexity of the mechanisms behind carcinogenesis inspires researchers to explore the ability of various biomolecules to target specific pathways. Natural components occurring mainly in food and medicinal plants, are considered an essential resource for discovering new and promising therapeutic molecules. Novel biomolecules normally have an advantage in terms of biosafety. They are also widely diverse and often possess potent antioxidant, anti-inflammatory, and anti-cancer properties. Based on quantitative structure-activity relationship studies, biomolecules can be used as templates for chemical modifications that improve efficiency, safety, and bioavailability. In this review, we focus on anti-HCC biomolecules that have their molecular targets partially or completely characterized as well as having anti-cancer molecular mechanisms that are fairly described.
Collapse
|
18
|
Yun BD, Son SW, Choi SY, Kuh HJ, Oh TJ, Park JK. Anti-Cancer Activity of Phytochemicals Targeting Hypoxia-Inducible Factor-1 Alpha. Int J Mol Sci 2021; 22:ijms22189819. [PMID: 34575983 PMCID: PMC8467787 DOI: 10.3390/ijms22189819] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/02/2021] [Accepted: 09/03/2021] [Indexed: 12/15/2022] Open
Abstract
Hypoxia-inducible factor-1 alpha (HIF-1α) is overexpressed in cancer, leading to a poor prognosis in patients. Diverse cellular factors are able to regulate HIF-1α expression in hypoxia and even in non-hypoxic conditions, affecting its progression and malignant characteristics by regulating the expression of the HIF-1α target genes that are involved in cell survival, angiogenesis, metabolism, therapeutic resistance, et cetera. Numerous studies have exhibited the anti-cancer effect of HIF-1α inhibition itself and the augmentation of anti-cancer treatment efficacy by interfering with HIF-1α-mediated signaling. The anti-cancer effect of plant-derived phytochemicals has been evaluated, and they have been found to possess significant therapeutic potentials against numerous cancer types. A better understanding of phytochemicals is indispensable for establishing advanced strategies for cancer therapy. This article reviews the anti-cancer effect of phytochemicals in connection with HIF-1α regulation.
Collapse
Affiliation(s)
- Ba Da Yun
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (B.D.Y.); (S.W.S.); (S.Y.C.)
| | - Seung Wan Son
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (B.D.Y.); (S.W.S.); (S.Y.C.)
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (B.D.Y.); (S.W.S.); (S.Y.C.)
| | - Hyo Jeong Kuh
- Department of Medical Life Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Tae-Jin Oh
- Department of Pharmaceutical Engineering and Biotechnology, SunMoon University, 70 Sunmoon-ro 221, Tangjeong-myeon, Asan-si 31460, Korea;
| | - Jong Kook Park
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 24252, Korea; (B.D.Y.); (S.W.S.); (S.Y.C.)
- Correspondence: ; Tel.: +82-33-248-2114
| |
Collapse
|
19
|
Wang Y, Jiang L. Role of vitamin D-vitamin D receptor signaling on hyperoxia-induced bronchopulmonary dysplasia in neonatal rats. Pediatr Pulmonol 2021; 56:2335-2344. [PMID: 33878208 DOI: 10.1002/ppul.25418] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/25/2021] [Accepted: 04/03/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Vitamin D exerts therapeutic effects on bronchopulmonary dysplasia (BPD), but its underlying mechanisms remain unclear. The present study was designed to investigate the effects of vitamin D on hyperoxia-induced BPD and elucidate the underlying mechanisms. METHODS Neonatal rats were exposed to either room air (control) or 75% O2 (hyperoxia) and intraperitoneally injected with vitamin D3. After 14 days, a histopathological examination was performed in the lungs of rats. Serum 25-hydroxyvitamin D (25OHD) was measured by liquid chromatography-tandom mass spectrometry (LC-MS)/MS. Interleukin 1 beta (IL-1β) and interferon gamma (IFN-γ) were measured by specific enzyme-linked immunosorbent assays. The messenger RNA and protein levels of vitamin D receptor (VDR), vascular endothelial growth factor (VEGF), VEGF receptor 2 (VEGFR2), and hypoxia-inducible factor 1α (HIF-1α) were determined by real-time quantitative reverse transcription polymerase chain reaction and immunoblot analysis, respectively. RESULTS Treatment with vitamin D3 increased serum 25OHD and upregulated VDR in lung tissues with or without hyperoxia. In addition, treatment with vitamin D3 attenuated alveolar simplification, increased VEGF and VEGFR2, and protected alveolar simplification induced by hyperoxia. Furthermore, treatment with vitamin D3 resulted in a decrease of IL-1β and IFN-γ and an increase of HIF-1α in lung tissues under hyperoxia conditions. CONCLUSION Vitamin D exerts protective effects on hyperoxia-induced BPD in neonatal rats by regulating vitamin D-VDR signaling pathways.
Collapse
Affiliation(s)
- Yuchun Wang
- Department of Pediatrics, Hebei Medical University, Shijiazhuang, Hebei, China.,Department of Neonatology, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Lian Jiang
- Department of Pediatrics, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
20
|
Yin W, Xu J, Mao Y. Synergistic effects of autophagy inhibitors combined with cisplatin against cisplatin-resistant nasopharyngeal cancer cells. Biochem Cell Biol 2021; 99:322-329. [PMID: 34038188 DOI: 10.1139/bcb-2020-0283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
This study explored the synergistic effects of autophagy inhibitors combined with cisplatin against cisplatin-resistant nasopharyngeal cancer cells by treating HNE-1 and cisplatin (diamminedichloroplatinum; DDP)-resistant HNE1/DDP nasopharyngeal cancer cell lines with DDP, autophagy inhibitors, or a combination of autophagy inhibitors and DDP. Cell viability was determined via MTT (colorimetric) and colony-forming assays, and the rate of apoptosis was determined using propidium iodide (PI) and annexin V double-staining. The expressions of proteins were determined by Western blotting. For our in-vivo studies, a murine xenograft model was established to evaluate the anti-tumor effects of the combination of autophagy inhibitor and DDP. The results showed that treatment with DDP increased the expressions of ATP-binding cassette sub-family B member 1 (ABCB1), ATP Binding Cassette Subfamily C Member 1 (ABCC1), and P-glycoprotein 1 (P-gp) in the HNE1/DDP cell lines. Treatment with chloroquine decreased the expression levels of ABCB1, ABCC1, and P-gp, and increased the formation of LC3-II and the expression levels of p62 in the HNE1/DDP cells. Additionally, the combination of autophagy inhibitors and DDP produced a synergistic effect on DDP-induced cell death and apoptosis. Furthermore, the combination of the autophagy inhibitor and DDP showed significant anti-tumor effects in the xenograft mouse model. In summary, autophagy inhibitors show synergistic anti-tumor effects with DDP in vitro against DDP-resistant nasopharyngeal cancer cells and in vivo in our xenograft murine model.
Collapse
Affiliation(s)
- Wei Yin
- Hangzhou Cancer Hospital, No. 34 Yanguanxiang, Hangzhou Zhejiang 310002, China
| | - Jianfeng Xu
- Department of Otolaryngology Head and Neck Surgery, Jiande Second People's Hospital, No. 223 Zongfu Street, Meicheng Town, Jiande Zheijang 311064, China
| | - Yanjiao Mao
- Hangzhou Cancer Hospital, No. 34 Yanguanxiang, Hangzhou Zhejiang 310002, China
| |
Collapse
|
21
|
Wang H, Wu C, Kong D. miR-140-5p Overexpression Protects Against Lipopolysaccharide-Induced Necrotizing Pneumonia via Targeting Toll-Like Receptor 4. Cell Mol Bioeng 2021; 14:339-348. [PMID: 34295443 DOI: 10.1007/s12195-021-00673-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 04/27/2021] [Indexed: 01/04/2023] Open
Abstract
Objective This study is to identify the effects of miRNA-140-5p on necrotizing pneumonia (NP) and its underlying mechanism. Methods The mRNA levels of miRNA-140-5p and TLR4 and secretion of IL-1β, IL-6, and TNF-α in peripheral blood from children with NP and healthy volunteers were determined using qRT-PCR and specific ELISAs. The interactions between miRNA-140-5p and TLR4 were investigated using a dual-luciferase reporter system. Cell viabilities were determined using a CCK-8 assay. qRT-PCR, western blotting, and specific ELISAs were applied to determine the expressions of genes in the cells. Peripheral blood from children with NP had significantly elevated levels of TLR4 but significantly lower levels of miR-140-5p compared to the control. Results Spearman's rank correlation analysis showed a negative correlation between TLR4 and miR-140-5p. miR-140-5p regulated the expressions of TLR4 in A549 cells. Additionally, LPS induced a significant enhancement in the levels of TLR4 but significant reduction in the levels of miR-140-5p. The overexpression of miR-140-5p suppressed cell apoptosis and induced the release of inflammatory cytokines in the LPS-induced A549 cells. Conclusion The underlying mechanisms of miR-140-5p on the regulation of TLR4 are in part by the regulation of p65. The miR-140-5p inhibits necrotizing pneumonia by regulating TLR-4 via TNF-p65 signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1007/s12195-021-00673-0.
Collapse
Affiliation(s)
- Haichao Wang
- School of Biology and Food Engineering, Suzhou University, No. 49 Bianhe Middle Road, Suzhou, 234000 Anhui China
| | - Changhao Wu
- School of Biology and Food Engineering, Suzhou University, No. 49 Bianhe Middle Road, Suzhou, 234000 Anhui China
| | - Dehui Kong
- School of Biology and Food Engineering, Suzhou University, No. 49 Bianhe Middle Road, Suzhou, 234000 Anhui China
| |
Collapse
|
22
|
Yang H, Li GP, Liu Q, Zong SB, Li L, Xu ZL, Zhou J, Cao L, Wang ZZ, Zhang QC, Li M, Fan QR, Hu HF, Xiao W. Neuroprotective effects of Ginkgolide B in focal cerebral ischemia through selective activation of prostaglandin E2 receptor EP4 and the downstream transactivation of epidermal growth factor receptor. Phytother Res 2021; 35:2727-2744. [PMID: 33452698 DOI: 10.1002/ptr.7018] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 12/28/2020] [Accepted: 12/31/2020] [Indexed: 11/07/2022]
Abstract
The present study was undertaken to identify whether prostaglandin E2 receptor is the potential receptor/binding site for Ginkgolide A, Ginkgolide B, Ginkgolide K, and Bilobalide, the four main ingredients of the Ginkgo biloba L., leaves. Using functional assays, we identified EP4, coupled with Gs protein, as a target of Ginkgolide B. In human neuroblastoma SH-SY5Y cells suffered from oxygen-glucose deprivation/reperfusion, Ginkgolide B-activated PKA, Akt, and ERK1/2 as well as Src-mediated transactivation of epidermal growth factor receptor. These resulted in downstream signaling pathways, which enhanced cell survival and inhibited apoptosis. Knockdown of EP4 prevented Ginkgolide B-mediated Src, epidermal growth factor receptor (EGFR), Akt, and ERK1/2 phosphorylation and neuroprotective effects. Moreover, Src inhibitor prevented Ginkgolide B-mediated EGFR transactivation and the downstream Akt and ERK1/2 activation, while the phosphorylation of PKA induced by Ginkgolide B was not affected, indicating Ginkgolide B might transactivate EGFR in a ligand-independent manner. EP4 knockdown in a rat middle cerebral artery occlusion (MCAO) model prevented Ginkgolide B-mediated infarct size reduction and neurological assessment improvement. At the same time, the increased expressions of p-Akt, p-ERK1/2, p-PKA, p-Src, and p-EGFR and the deceased expression of cleaved capases-3 induced by Ginkgolide B in cerebral cortex were blocked due to EP4 knockdown. In conclusion, Ginkgolide B exerts neuroprotective effects in rat MCAO model through the activation of EP4 and the downstream transactivation of EGFR.
Collapse
Affiliation(s)
- Hao Yang
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Gui-Ping Li
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Qiu Liu
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Shao-Bo Zong
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Liang Li
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Zhi-Liang Xu
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Jun Zhou
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Liang Cao
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Zhen-Zhong Wang
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Quan-Chang Zhang
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Ming Li
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Qi-Ru Fan
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Han-Fei Hu
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| | - Wei Xiao
- State Key Laboratory of New-Tech for Chinese Medicine Pharmaceutic Process, Jiangsu Kanion Pharmaceutical Co., Ltd., Lianyungang, China
| |
Collapse
|
23
|
Yan Y, Qian H, Cao Y, Zhu T. Nuclear factor-κB inhibitor Bay11-7082 inhibits gastric cancer cell proliferation by inhibiting Gli1 expression. Oncol Lett 2021; 21:301. [PMID: 33732377 PMCID: PMC7905653 DOI: 10.3892/ol.2021.12562] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 01/28/2021] [Indexed: 02/07/2023] Open
Abstract
Dysregulated nuclear factor (NF)-κB signaling pathway is involved in gastric carcinogenesis. The present study aimed to investigate the antitumor effects of the NF-κB inhibitor, Bay11-7082, on gastric cancer (GC) and elucidate its underlying molecular mechanisms. The MTT assay was performed to assess the effects of Bay11-7082 on the proliferation of HGC27 and MKN45 gastric cancer cells. In addition, the Transwell and wound healing assays were performed to determine cell migration and invasion, respectively. Reverse transcription-quantitative PCR and western blot analyses were performed to detect the mRNA and protein expression levels of the target genes. The results demonstrated that the half-maximal inhibitory concentration (IC50) of Bay11-7082 in HGC27 cells was 24.88, 6.72 and 4.23 nM at 24, 48 and 72 h, respectively. Furthermore, the IC50 of Bay11-7082 in MKN45 cells was 29.11, 11.22 and 5.88 nM at 24, 48 and 72 h, respectively. Treatment with Bay11-7082 significantly suppressed the cell migratory and invasive abilities compared with the control group. Notably, Bay11-7082 suppressed GLI Family Zinc Finger 1 (Gli1) mRNA and protein expression levels. Taken together, the results of the present study demonstrated that Bay11-7082 inhibited GC cell proliferation, at least in part through inhibition of Gli1.
Collapse
Affiliation(s)
- Yan Yan
- Department of Pharmacology, The Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, Jiangsu 215600, P.R. China
| | - Heya Qian
- Department of Oncology, The Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, Jiangsu 215600, P.R. China
| | - Ying Cao
- Department of Pharmacology, The Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, Jiangsu 215600, P.R. China
| | - Tao Zhu
- Department of Laboratory, The Affiliated Zhangjiagang Hospital of Soochow University, Zhangjiagang, Jiangsu 215600, P.R. China
| |
Collapse
|
24
|
Cao H, Wang D, Gao R, Chen L, Feng Y. Down regulation of U2AF1 promotes ARV7 splicing and prostate cancer progression. Biochem Biophys Res Commun 2021; 541:56-62. [PMID: 33477033 DOI: 10.1016/j.bbrc.2020.12.111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 12/23/2020] [Indexed: 12/17/2022]
Abstract
The present study aims to investigate the roles of U2 Small Nuclear RNA Auxiliary Factor 1 (U2AF1) in the resistance to anti-androgen treatment in prostate cancer and its underlying mechanism. U2AF1 and androgen receptor variant 7 (ARV7) knockdown and overexpression were introduced in PC3 and DU145 cells. In addition, a bicalutamide-resistant PC3 (PC3 BR) cell line was also constructed. Cell count, MTT and soft agar colony formation assays were performed to evaluate cell proliferation. qRT-PCR was applied to determine the mRNA levels of U2AF1, ARV7 and Mitogen-Activated Protein Kinase 1 (MAPK1). Western blot was used to determine the MAPK1 protein expression. A negative correlation between ARV7 and U2AF1 in prostate tumor tissues was observed. U2AF1 downregulation was correlated with poor prognosis in prostate cancer patients. U2AF1 exhibited a negative correlation with ARV7 and its downregulation promoted prostate cancer cell proliferation and bicalutamide resistance. The regulatory effects of U2AF1 on ARV7 splicing were associated with MAPK1. U2AF1 affected prostate cancer proliferation and anti-androgen resistance by regulating ARV7 splicing.
Collapse
Affiliation(s)
- Hongwen Cao
- Surgical Department I (Urology Department), Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725 Wanping Road South, Xuhui District, Shanghai, 200032, China
| | - Dan Wang
- Surgical Department I (Urology Department), Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725 Wanping Road South, Xuhui District, Shanghai, 200032, China
| | - Renjie Gao
- Surgical Department I (Urology Department), Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725 Wanping Road South, Xuhui District, Shanghai, 200032, China
| | - Lei Chen
- Surgical Department I (Urology Department), Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725 Wanping Road South, Xuhui District, Shanghai, 200032, China.
| | - Yigeng Feng
- Surgical Department I (Urology Department), Longhua Hospital Shanghai University of Traditional Chinese Medicine, No. 725 Wanping Road South, Xuhui District, Shanghai, 200032, China.
| |
Collapse
|
25
|
Yousefi H, Mashouri L, Okpechi SC, Alahari N, Alahari SK. Repurposing existing drugs for the treatment of COVID-19/SARS-CoV-2 infection: A review describing drug mechanisms of action. Biochem Pharmacol 2021; 183:114296. [PMID: 33191206 PMCID: PMC7581400 DOI: 10.1016/j.bcp.2020.114296] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 02/06/2023]
Abstract
The outbreak of a novel coronavirus (SARS-CoV-2) has caused a major public health concern across the globe. SARS-CoV-2 is the seventh coronavirus that is known to cause human disease. As of September 2020, SARS-CoV-2 has been reported in 213 countries and more than 31 million cases have been confirmed, with an estimated mortality rate of ∼3%. Unfortunately, a drug or vaccine is yet to be discovered to treat COVID-19. Thus, repurposing of existing cancer drugs will be a novel approach in treating COVID-19 patients. These drugs target viral replication cycle, viral entry and translocation to the nucleus. Some can enhance innate antiviral immune response as well. Hence this review focuses on comprehensive list of 22 drugs that work against COVID-19 infection. These drugs include fingolimod, colchicine, N4-hydroxycytidine, remdesivir, methylprednisone, oseltamivir, icatibant, perphanizine, viracept, emetine, homoharringtonine, aloxistatin, ribavirin, valrubicin, famotidine, almitrine, amprenavir, hesperidin, biorobin, cromolyn sodium, and antibodies- tocilzumab and sarilumab. Also, we provide a list of 31 drugs that are predicted to function against SARS-CoV-2 infection. In summary, we provide succinct overview of various therapeutic modalities. Among these 53 drugs, based on various clinical trials and literature, remdesivir, nelfinavir, methylpredinosolone, colchicine, famotidine and emetine may be used for COVID-19. SIGNIFICANCE: It is of utmost important priority to develop novel therapies for COVID-19. Since the effect of SARS-CoV-2 is so severe, slowing the spread of diseases will help the health care system, especially the number of visits to Intensive Care Unit (ICU) of any country. Several clinical trials are in works around the globe. Moreover, NCI developed a recent and robust response to COVID-19 pandemic. One of the NCI's goals is to screen cancer related drugs for identification of new therapies for COVID-19. https://www.cancer.gov/news-events/cancer-currents-blog/2020/covid-19-cancer-nci-response?cid=eb_govdel.
Collapse
Affiliation(s)
- Hassan Yousefi
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA, USA
| | - Ladan Mashouri
- Department of Medical Sciences, University of Arkansas, Little Rock, AK, USA
| | - Samuel C Okpechi
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA, USA
| | - Nikhilesh Alahari
- Department of Biological Engineering, Louisiana State University, Baton Rouge, LA, USA
| | - Suresh K Alahari
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center School of Medicine, New Orleans, LA, USA; Stanley Scott Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| |
Collapse
|
26
|
Qi X, Xing Y, Wang X. Blockade of CCL2/CCR2 Signaling Pathway Exerts Anti-Inflammatory Effects and Attenuates Gestational Diabetes Mellitus in a Genetic Mice Model. Horm Metab Res 2021; 53:56-62. [PMID: 33022739 DOI: 10.1055/a-1250-8221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The chemokine (C-C motif) ligand 2 (CCL2) and its receptor CCR2 are involved in gestational diabetes mellitus (GDM). The present study aims to explore the effects of CCL2 blocking on GDM. Serum CCL2, interleukin (IL)-6, and tumor necrosis factor (TNF)-α were determined in GDM patients and healthy volunteers. C57BL/KsJdb/+mouse was used as the GDM model and CCL2 antibody (αCCL2) was applied. Flow cytometry was applied to determine the frequency of macrophages. Quantitative reverse transcription PCR (RT-qPCR) and western blot were determined to detect the mRNA and protein expressions, respectively. Enzyme-linked immunosorbent assay (ELISA) was applied to determine the levels of inflammatory cytokines and serum insulin. Serum CCL2 was correlated with inflammatory cytokines (IL-6 and TNF-α) in the GDM patients. Besides, the results showed high expressions of CCL2 in the visceral adipose tissue (VAT) and placenta tissue in the GDM mice. Flow cytometry and immunohistochemistry (IHC) staining showed the accumulations of macrophages in these tissues. Treatment of αCCL2 attenuated the GDM symptoms and ameliorated the inflammation. Furthermore, the treatment of αCCL2 improved reproductive outcomes in the GDM mice. Blockade of CCL2 attenuated GDM symptoms and reduced inflammatory cytokines in a genetic mice model.
Collapse
Affiliation(s)
- Xinying Qi
- The Second Department of Obstetrics, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Yanping Xing
- The Second Department of Obstetrics, Cangzhou Central Hospital, Cangzhou, Hebei, China
| | - Xuezhen Wang
- The Second Department of Obstetrics, Cangzhou Central Hospital, Cangzhou, Hebei, China
| |
Collapse
|
27
|
Zhu H, Zhou W, Wan Y, Ge K, Lu J, Jia C. Nanomedicine-mediated induction of immunogenic cell death and prevention of PD-L1 overexpression for enhanced hepatocellular carcinoma therapy. Cancer Nanotechnol 2020. [DOI: 10.1186/s12645-020-00072-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Abstract
Background
The present study aims to develop a nanoparticle encapsulating doxorubicin (DOX) and programmed death-ligand 1 (PD-L1) siRNA and evaluate its anti-tumor effects on hepatoma carcinoma (HCC).
Methods
Nanoparticle encapsulating DOX and PD-L1 siRNA (NPDOX/siPD-L1) was characterized by dynamic light scattering and transmission electron microscopy. Flow cytometry was applied to analyze cell populations, NPDOX/siPD-L1 internalization, and cell apoptosis. Real-Time (RT)-quantitative reverse transcription (qPCR) and western blotting were used to determine the mRNA and protein levels, respectively. Released ATP was determined using ATP determination kit and cytokines were determined using specific ELISAs. A tumor-bearing animal model was established to evaluate the anti-tumor effects of NPDOX/siPD-L1.
Results
Treatment of NPDOX/siPD-L1 induced immunogenic cell death (ICD) and PD-L1 overexpression in HCC. In vivo study demonstrated that intravenously injection of NPDOX/siPD-L1 significantly inhibited the tumor volume and PD-L1 expressions of tumor tissue in the H22 tumor-bearing animal model. Besides, the treatment of NPDOX/siPD-L1 also regulated the populations of matured dendritic cells and cytotoxic T cells and the productions of cytokines in the tumor tissues.
Conclusion
Taken together, NPDOX/siPD-L1 showed significant anti-tumor effects on HCC by the induction of ICD and inhibition of PD-L1 overexpression.
Collapse
|
28
|
Liu J, Yang T, Zhang Y, Wang S. Promotion of BZW2 by LINC00174 through miR-4500 inhibition enhances proliferation and apoptosis evasion in laryngeal papilloma. Cancer Cell Int 2020; 20:471. [PMID: 33005104 PMCID: PMC7525952 DOI: 10.1186/s12935-020-01559-3] [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: 01/05/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Background We aimed to explore the roles of basic leucine zipper and W2 domains (BZW) 2 in the human papillomavirus-infected laryngeal papillomatosis. Methods In the present study, BZW 2 knockdown and overexpressed cell lines were constructed. CCK-8 and colony formation assays were used to determine cell proliferation. Caspase-3 activity and nucleosomes fragmentation assays were used to determine cell apoptosis. qRT-PCR and Western blot were employed to evaluate the mRNA and protein levels of target genes, respectively. Luciferase and biotin-coupled miRNA pulldown assays were used to examine the interactions between mRNA and mRNA. Results We observed the levels of BZW2 were up-regulated in the laryngeal papilloma (LP) tissues as compared with adjacent tissues. The knockdown of BZW2 significantly inhibited cell proliferation and promoted cell apoptosis in the LP cells. Additionally, we identified the expressions of BZW2 negatively regulated by miR-4500. Luciferase and biotin-coupled miRNA pulldown assays demonstrated that LINC00174 competed with the BZW2 for binding with miR-4500. Moreover, the results showed that LINC00174/miR-4500/BZW2 axis regulated cell proliferation and apoptosis. Conclusion Our results demonstrated that the regulation of LINC00174/miR-4500/BZW2 axis might be used as an effective strategy for treatment of human papillomavirus-infected laryngeal papillomatosis.
Collapse
Affiliation(s)
- Jiajia Liu
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| | - Tao Yang
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| | - Ying Zhang
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| | - Shuhui Wang
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011 Hunan China
| |
Collapse
|
29
|
Lesovaya EA, Savinkova AV, Morozova OV, Lylova ES, Zhidkova EM, Kulikov EP, Kirsanov KI, Klopot A, Baida G, Yakubovskaya MG, Gordon LI, Readhead B, Dudley JT, Budunova I. A Novel Approach to Safer Glucocorticoid Receptor-Targeted Anti-lymphoma Therapy via REDD1 (Regulated in Development and DNA Damage 1) Inhibition. Mol Cancer Ther 2020; 19:1898-1908. [PMID: 32546661 PMCID: PMC7875139 DOI: 10.1158/1535-7163.mct-19-1111] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/31/2020] [Accepted: 06/09/2020] [Indexed: 11/16/2022]
Abstract
Glucocorticoids are widely used for therapy of hematologic malignancies. Unfortunately, chronic treatment with glucocorticoids commonly leads to adverse effects including skin and muscle atrophy and osteoporosis. We found recently that REDD1 (regulated in development and DNA damage 1) plays central role in steroid atrophy. Here, we tested whether REDD1 suppression makes glucocorticoid-based therapy of blood cancer safer. Unexpectedly, approximately 50% of top putative REDD1 inhibitors selected by bioinformatics screening of Library of Integrated Network-Based Cellular Signatures database (LINCS) were PI3K/Akt/mTOR inhibitors. We selected Wortmannin, LY294002, and AZD8055 for our studies and showed that they blocked basal and glucocorticoid-induced REDD1 expression. Moreover, all PI3K/mTOR/Akt inhibitors modified glucocorticoid receptor function shifting it toward therapeutically important transrepression. PI3K/Akt/mTOR inhibitors enhanced anti-lymphoma effects of Dexamethasone in vitro and in vivo, in lymphoma xenograft model. The therapeutic effects of PI3K inhibitor+Dexamethasone combinations ranged from cooperative to synergistic, especially in case of LY294002 and Rapamycin, used as a previously characterized reference REDD1 inhibitor. We found that coadministration of LY294002 or Rapamycin with Dexamethasone protected skin against Dexamethasone-induced atrophy, and normalized RANKL/OPG ratio indicating a reduction of Dexamethasone-induced osteoporosis. Together, our results provide foundation for further development of safer and more effective glucocorticoid-based combination therapy of hematologic malignancies using PI3K/Akt/mTOR inhibitors.
Collapse
Affiliation(s)
- Ekaterina A Lesovaya
- N.N. Blokhin NMRCO, Moscow, Russia
- I.P. Pavlov Ryazan State Medical University, Ryazan, Russia
| | | | | | | | | | | | | | - Anna Klopot
- Department of Dermatology, Northwestern University, Chicago, Illinois
| | - Gleb Baida
- Department of Dermatology, Northwestern University, Chicago, Illinois
| | | | - Leo I Gordon
- Division of Hematology Oncology; Northwestern University; Chicago, Illinois
| | - Ben Readhead
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Joel T Dudley
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Irina Budunova
- Department of Dermatology, Northwestern University, Chicago, Illinois.
| |
Collapse
|
30
|
Nanoparticle-facilitated delivery of BAFF-R siRNA for B cell intervention and rheumatoid arthritis therapy. Int Immunopharmacol 2020; 88:106933. [PMID: 32866781 DOI: 10.1016/j.intimp.2020.106933] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/06/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023]
Abstract
The present study was designed to explore the effects of B-cell activating factor receptor (BAFF-R) siRNA encapsulated nanoparticles on collagen-induced arthritis (CIA). BAFF-R siRNA encapsulated nanoparticles (NP-siBAFF-R) were constructed using a double emulsion method and was characterized by dynamic light scattering and transmission electron microscopy. Cellular uptake of nanoparticles was determined using flow cytometry. The CIA mouse model was established and the mice were intravenously injected with nanoparticles. NP-siBAFF-R effectively decreased the expression of BAFF-R in B cells and facilitated the delivery of siRNA into B cells. Treatment of NPsiBAFF-R ameliorated rheumatoid arthritis (RA) symptoms in the CIA mouse model via decreasing the arthritis score, mean ankle diameter, the levels of anti-collagen IgG in serum and increasing the expression of collagen type II and osteocalcin in dissected joint tissues. Additionally, treatment of NPsiBAFF-R decreased the percentage and number of B cells and inhibited the production of pro-inflammatory cytokines in RA mice. These results demonstrate that NP-siBAFF-R may provide an effective strategy for RA treatment.
Collapse
|
31
|
Liu J, Li X, Ke A. High-mobility group box-1 induces mechanical pain hypersensitivity through astrocytic connexin 43 via the toll-like receptor-4/JNK signaling pathway. Synapse 2020; 75:e22184. [PMID: 32761652 DOI: 10.1002/syn.22184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/26/2020] [Accepted: 07/27/2020] [Indexed: 12/28/2022]
Abstract
The present study aimed to investigate the effects of high-mobility group box-1 (HMGB-1) on mechanical pain hypersensitivity and the underlying mechanism. Mouse primary astrocytes were isolated and treated as specified. A CCK-8 assay was used to determine cytotoxicity and a gap junctional communication assay was performed. Ethidium bromide (EtBr) uptake was used to evaluate the hemichannel activity of primary astrocytes. A mouse model of neuropathic pain was developed and paw withdrawal threshold was used to evaluate hind paw sensitivity. RT-qPCR and Western blot were used to determine mRNA and protein expression of genes, respectively. ELISA was used to measure the release of inflammatory cytokines. Treatment with HMGB-1 increased the expression of both toll-like receptor-4 (TLR-4) and connexin 43 (Cx43) in mouse primary astrocytes. HMGB-1 also promoted gap junctional intercellular communication and hemichannel function. Our results also demonstrated that HMGB-1-regulated Cx43 through the JNK signaling pathway, and Cx43 was involved in HMGB-1-mediated inflammation in astrocytes. In vivo analysis supported the idea that HMGB-1-induced mechanical hypersensitivity was associated with Cx43. We therefore conclude that HMGB-1-induced mechanical pain hypersensitivity occurs through modulating astrocytic Cx43 via the TLR-4/JNK signaling pathway.
Collapse
Affiliation(s)
- Jiang Liu
- Department of Anesthesiology, Tangshan Gongren Hospital, Tangshan, China
| | - Xiuhua Li
- Department of Anesthesiology, Tangshan Gongren Hospital, Tangshan, China
| | - Ana Ke
- Department of Anesthesiology, Tangshan Gongren Hospital, Tangshan, China
| |
Collapse
|
32
|
Ci R, Zhang K, Zhu A, Zang W. Dendrobine attenuates isoniazid- and rifampicin-induced liver injury by inhibiting miR-295-5p. Hum Exp Toxicol 2020; 39:1671-1680. [PMID: 32633153 DOI: 10.1177/0960327120937047] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The present study aims to investigate the protective effects of Dendrobine and its underlying mechanisms on liver injury induced by isoniazid (INH) and rifampicin (RIF). A mouse model of liver injury was induced by intragastrically administration of 100 mg/kg INH and 100 mg/kg RIF for 14 days. The mice were intragastrically administrated with Dendrobine (50, 100, and 200 mg/kg) before the administration of INH and RIF. Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were determined. Oxidative stress markers including glutathione, superoxide dismutase, and malondialdehyde in the liver were measured and liver histopathological examinations were performed. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blot were applied to determine the mRNA and protein expressions, respectively. Luciferase reporter assay was used to evaluate the interactions between miR-295-5p and CYP1A2. Dendrobine significantly decreased serum ALT and AST and inhibited the liver index and ameliorated the liver histological changes induced by INH and RIF. Besides, Dendrobine also regulated oxidative stress status in the liver by the regulation of CYP1A2. Moreover, mmu-miR-295-5p was identified to target CYP1A2 and to regulate the expression of CYP1A2. In summary, Dendrobine ameliorated INH and RIF induced mouse liver injury by miR-295-5p-mediated CYP1A2 expression.
Collapse
Affiliation(s)
- R Ci
- Department of Tuberculosis, Shijiazhuang No. 5 City Hospital, Shijiazhuang, Hebei, China
| | - K Zhang
- Department of Tuberculosis, Shijiazhuang No. 5 City Hospital, Shijiazhuang, Hebei, China
| | - A Zhu
- Department of Tuberculosis, Shijiazhuang No. 5 City Hospital, Shijiazhuang, Hebei, China
| | - W Zang
- Department of Tuberculosis, Shijiazhuang No. 5 City Hospital, Shijiazhuang, Hebei, China
| |
Collapse
|
33
|
Fan G, Wei X, Xu X. Is the era of sorafenib over? A review of the literature. Ther Adv Med Oncol 2020; 12:1758835920927602. [PMID: 32518599 PMCID: PMC7252361 DOI: 10.1177/1758835920927602] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 04/27/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most severe diseases worldwide. For the different stages of HCC, there are different clinical treatment strategies, such as surgical therapy for the early stage, and transarterial chemoembolization (TACE) and selective internal radiation therapy (SIRT) for intermediate-stage disease. Systemic treatment, which uses mainly targeted drugs, is the standard therapy against advanced HCC. Sorafenib is an important first-line therapy for advanced HCC. As a classically effective drug, sorafenib can increase overall survival markedly. However, it still has room for improvement because of the heterogeneity of HCC and acquired resistance. Scientists have reported the acquired sorafenib resistance is associated with the anomalous expression of certain genes, most of which are also related with HCC onset and development. Combining sorafenib with inhibitors targeting these genes may be an effective treatment. Combined treatment may not only overcome drug resistance, but also inhibit the expression of carcinoma-related genes. This review focuses on the current status of sorafenib in advanced HCC, summarizes the inhibitors that can combine with sorafenib in the treatment against HCC, and provides the rationale for clinical trials of sorafenib in combination with other inhibitors in HCC. The era of sorafenib in the treatment of HCC is far from over, as long as we find better methods of medication.
Collapse
Affiliation(s)
- Guanghan Fan
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine; NHC Key Laboratory of Combined Multi-organ Transplantation; Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS; Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
| | - Xuyong Wei
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine; NHC Key Laboratory of Combined Multi-organ Transplantation; Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS; Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, First Affiliated Hospital, Zhejiang University School of Medicine; NHC Key Laboratory of Combined Multi-organ Transplantation; Key Laboratory of the diagnosis and treatment of organ Transplantation, CAMS; Key Laboratory of Organ Transplantation, Zhejiang Province, 79 QingChun Road, Hangzhou, 310003, China
| |
Collapse
|
34
|
Sorafenib-Loaded Long-Circulating Nanoliposomes for Liver Cancer Therapy. BIOMED RESEARCH INTERNATIONAL 2020; 2020:1351046. [PMID: 32461962 PMCID: PMC7229545 DOI: 10.1155/2020/1351046] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/14/2020] [Accepted: 03/13/2020] [Indexed: 12/24/2022]
Abstract
A type of sorafenib- (SOR-) loaded long-circulating nanoliposome was constructed, and the targeting performance and antitumor effects of the prepared liposome were evaluated in the present study. Polyethylene glycol- (PEG-) modified long-circulating nanoliposomes (LC-NPs) were designed and prepared using reverse evaporation, and the LC-NPs were used for delivering sorafenib (LC-PEG-SOR-NPs). Then, the anti-VEGFR antibody as a targeting moiety was chemically coupled with LC-PEG-SOR-NPs to form liver cancer-targeted nanoliposomes (anti-VEGFR-LC-PEG-SOR-NPs). The drug entrapment and loading efficiency were measured. And the cancer-targeting performance and therapeutic efficiency were evaluated both in vitro and in vivo. The anti-VEGFR-LC-PEG-SOR-NPs with an average of 119.8 ± 4.2 nm showed a uniform spherical structure. The drug entrapment and loading efficiency were 92.5% and 18.5%, respectively. The killing efficiency of anti-VEGFR-LC-PEG-SOR-NPs was up to 18% after incubating with liver cancer cells for 72 h. Furthermore, the anti-VEGFR-LC-PEG-SOR-NPs could actively target at the tumor region and could efficiently inhibit tumor growth with negligible side effects. This newly designed nanoliposomes had desirable dispersibility, high drug entrapment efficiency, tumor targeting and therapeutic efficiency, and good safety. As a biocompatible nanocomposite, it was promising to become a novel and useful tumor-targeting nanodrug for liver cancer therapy.
Collapse
|
35
|
Zhang L, Wang Z. Circular RNA hsa_circ_0004812 impairs IFN-induced immune response by sponging miR-1287-5p to regulate FSTL1 in chronic hepatitis B. Virol J 2020; 17:40. [PMID: 32188476 PMCID: PMC7079541 DOI: 10.1186/s12985-020-01314-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/06/2020] [Indexed: 12/18/2022] Open
Abstract
Background The present study aims to explore the functions of circular RNA hsa_circ_0004812 in chronic hepatitis B (CHB) and its underlying molecular mechanisms. Methods The expression of circular RNA (circRNA)_0004812 was examined using qRT-PCR and Western blot in blood and liver tissues from CHB patients and healthy volunteers. In the in vitro study, the expression levels of circular RNA hsa_circ_0004812, miR-1287-5p, interferon (IFN)-α, IFN-β were determined using qRT-PCR and Western blotting in HBV-infected hepatoma cells, respectively. Luciferase and biotin pull-down assays were used to investigate the interactions between miR-1287-5p and circ_0004812. Results Levels of circ_0004812 were upregulated in CHB patients and HBV-infected hepatoma cells. Knockdown of circ_0004812 increased the expression of IFN-α and IFN-β in HBV-infected Huh7 cells. MiR-1287-5p was identified as a target of circ_0004812 whose overexpression inhibited the expression of miR-1287-5p. Additionally, circ_0004812 promoted the expression of Follistatin-related protein (FSTL) 1 through inhibiting miR-1287-5p. Circ_0004812/miR-1287-5p/FSTL1 axis regulated HBV-induced immune suppression. Conclusion Circ_0004812 was identified as a potential target for CHB infection. Circ_0004812 promoted the expression of FSTL1 by inhibiting miR-1287-5p.
Collapse
Affiliation(s)
- Liangdong Zhang
- Second Department of Infectious Diseases, Cangzhou Central Hospital, Xinhua West Road, Cangzhou, 061001, Hebei Province, China.
| | - Zichao Wang
- Second Department of Infectious Diseases, Cangzhou Central Hospital, Xinhua West Road, Cangzhou, 061001, Hebei Province, China
| |
Collapse
|
36
|
Su L, Zhang X, Zheng L, Wang M, Zhu Z, Li P. Mutation of Isocitrate Dehydrogenase 1 in Cholangiocarcinoma Impairs Tumor Progression by Inhibiting Isocitrate Metabolism. Front Endocrinol (Lausanne) 2020; 11:189. [PMID: 32373065 PMCID: PMC7187788 DOI: 10.3389/fendo.2020.00189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/17/2020] [Indexed: 12/17/2022] Open
Abstract
Aim: Isocitrate dehydrogenase 1 (IDH1) is key enzyme involved in cellular metabolism and DNA repair. Mutations in IDH1 occur in up to 25% of cholangiocarcinomas. The present study aimed to explore the features of cellosaurus REB cells with mutant and wide-type IDH1. Methods: To compare the features of IDH1 knockout and mutation in cholangiocarcinoma, we firstly constructed the IDH1 knockout and IDH1 mutation cell lines. We then evaluated the viability of these cell lines using the cell count assay and MTT assay. Next, we determined cell migration and invasion using the Transwell assay. Additionally, to evaluate the effects of IDH1 on cellular metabolism, the levels of α-ketoglutarate (α-KG) and nicotinamide adenine dinucleotide phosphate (NADPH) were determined using enzyme-linked immunosorbent assay. We then applied ChIPbase dataset to explore the genes that were regulated by IDH1. Results: High frequency of mutated IDH1 was observed in the cholangiocarcinoma and IDH1 R132C was presented in more than 80% of mutations. The results showed that IDH1 knockout decreased cell proliferation, migration and invasion, whereas the overexpression of IDH1 in IDH1 knockout cell line recovered its proliferation, migration and invasion capacities. Additionally, IDH1 mutation reduced the levels of NADPH and α-KG. Furthermore, investigation into the underlying mechanisms revealed that IDH1 overexpression induced the expression of aldehyde dehydrogenase 1 thereby promoting cell proliferation, migration and invasion. Conclusion:IDH1 plays an important role in cholangiocarcinoma and its mutation impairs tumor progression in part by inhibition of isocitrate metabolism.
Collapse
Affiliation(s)
- Li Su
- Department of Integrated Traditional and Western Medicine in Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xinglong Zhang
- Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Lei Zheng
- Department of Integrated Traditional and Western Medicine in Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Miaomiao Wang
- Department of Integrated Traditional and Western Medicine in Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhifa Zhu
- Department of Integrated Traditional and Western Medicine in Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ping Li
- Department of Integrated Traditional and Western Medicine in Oncology, First Affiliated Hospital of Anhui Medical University, Hefei, China
- *Correspondence: Ping Li
| |
Collapse
|
37
|
Loss of Tyrosine Phosphatase Delta Promotes Gastric Cancer Progression via Signal Transducer and Activator of Transcription 3 Pathways. Dig Dis Sci 2019; 64:3164-3172. [PMID: 31041642 DOI: 10.1007/s10620-019-05637-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/20/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND The protein tyrosine phosphatase delta (PTPRD) is a tumor suppressor, and its role in gastric cancer (GC) remains poorly understood. METHODS The expressions of PTPRD were determined based on public data. In addition, the mRNA expressions of PTPRD in the GC samples, and the expressions of PTPRD in the GC cell lines including HGC27, SGC790, and BGC823, and gastric epithelial cell line GES-1 were determined by western blotting and quantitative real-time PCR. Furthermore, PTPRD siRNA was transfected into the HGC27 cell line, and then, cell proliferation, migration, and invasion were evaluated. The activity of signal transducer and activator of transcription 3 (STAT3) pathways in HGC27 cells transfected with PTPRD siRNA was determined by western blotting. RESULTS PTPRD deletion was found in the GC patients, and this deletion was found to be correlated with poor prognosis in the GC patients. Expression of PTPRD was significantly downregulated in gastric carcinoma specimens and tumor cell lines when compared with those in normal controls. PTPRD also plays a key role in the GC cells proliferation, invasion, and migration. Silencing PTPRD expression by siRNA dramatically promoted GC cells proliferation, invasion, and migration. Mechanism study demonstrated that phosphorylation of STAT3 was inhibited by silencing PTPRD expression and the according changes including inhibition of cell migration and invasion were observed. CONCLUSION This study supports PTPRD as a tumor suppressor and could be served as a marker for prognostic of GC. Silencing PTPRD could be a potential therapeutic in GC.
Collapse
|
38
|
Zhang L, Qiao X, Chen M, Li P, Wen X, Sun M, Ma X, Hou Y, Yang J. Ilexgenin A prevents early colonic carcinogenesis and reprogramed lipid metabolism through HIF1α/SREBP-1. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 63:153011. [PMID: 31301538 DOI: 10.1016/j.phymed.2019.153011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 06/21/2019] [Accepted: 07/01/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND Ilexgenin A (IA), the main bioactive compound from Ilex hainanensis Merr., has significant hypolipidemic activities. However, the effects of IA on colitis-associated colorectal cancer (CRC) and its mechanisms are still unknown. PURPOSE The study was designed to evaluate the effect of IA on CRC and explore its underlying mechanisms. STUDY DESIGN The effect of IA on colitis related CRC were evaluated in azoxymethane (AOM)/dextran sulfate sodium (DSS) mice and the underlying mechanisms were revealed by metabolomics, which were further validated in vivo and in vitro. METHODS The Balb/c mice were treated with AOM/DSS to induce CRC model and fed with normal diet with or without 0.02% IA. After the experimental period, samples of plasma were collected and analyzed by ultra-high-performance liquid chromatography/quadrupole time off light mass spectrometry (UHPLC-Q-TOF). Multivariate statistical tools were used to identify the changes of serum metabolites associated with CRC and responses to IA treatment. HT 29 and HCT 116 cells were stimulated by palmitate (PA) and cultured under hypoxia. Western blot, Q-PCR, and Immunofluorescence staining were performed to confirm the molecular pathway in vivo and in vitro. RESULTS Our results showed IA significantly inhibited the inflammatory colitis symptoms such as disease activity index score, shortening of colon tissues and the increase of inflammatory cytokines. In metabolomic study, 31 potential metabolites associated with CRC were identified and 24 of them were reversed by IA treatment. Most of biomarkers were associated with arachidonic acid metabolism, glycerophospholipid catabolism, and phospholipid metabolism, suggesting lipid metabolism might be involved in the beneficial effect of IA on CRC. Furthermore, we also found IA could decrease the expressions of SREBP-1 and its target gene in the colon tissues of AOM/DSS mice. It could down-regulate the triglyceride (TG) content and the expressions of HIF1α, SREBP-1, FASN, and ACC in HT 29 and HCT 116 cells. The inhibitory effect of IA on SREBP-1 was also attenuated by desferrioxamine (DFX), suggesting HIF1α is involved in the regulation of IA on SREBP-1. CONCLUSION IA prevents early colonic carcinogenesis in AOM/DSS mice and reprogramed lipid metabolism partly through HIF1α/SREBP-1.
Collapse
Affiliation(s)
- Li Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Xin Qiao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Meihong Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Xiaodong Wen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Minhui Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Xiaonan Ma
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Yingjian Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Jie Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China.
| |
Collapse
|
39
|
NLRP3 inflammasome inhibitor glyburide expedites diabetic-induced impaired fracture healing. Immunobiology 2019; 224:786-791. [PMID: 31477246 DOI: 10.1016/j.imbio.2019.08.008] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 07/13/2019] [Accepted: 08/22/2019] [Indexed: 12/12/2022]
Abstract
Localized inflammation is accompanied by the diabetic-induced fracture. The present study aims to investigate the therapeutic effects of glyburide, an NLRP3 inflammasome inhibitor, in a diabetic-induced fracture model. An animal model of diabetic-induced fracture was established and the mice were administrated with metformin or glyburide for 3 weeks. Quantitative polymerase chain reaction (qPCR) and Western blotting were used to evaluate the relative expressions of IFN-γ, TNF-α, and IL-6. Micro-computed tomography (μCT) scanning was applied to evaluate bone callus formation. Histopathology examinations of fractured femur sections were performed using Tartrate-resistant acid phosphatase (TRAP) staining and Alcian blue and orange G staining. Bone strength was evaluated using Torsional testing. Our results showed that treatment of glyburide significantly decreased the expressions of IFN-γ, TNF-α, and IL-6 in the fracture calluses in diabetic-induced fracture model, while bone callus volume and bone volume fraction were increased. Additionally, our results also demonstrated that treatment of glyburide rescued the increase of osteoclasts in the bone-cartilage interface. Apart from decreasing a percentage of cartilage area and increasing the percentage of bone and fibrotic tissue area, treatment of glyburide increased the maximum torque and yield torque of fractures. These results implied that glyburide might be used as a potential drug candidate for diabetic-induced fracture.
Collapse
|
40
|
Zhou A, Hong Y, Lv Y. Sulforaphane Attenuates Endometriosis in Rat Models Through Inhibiting PI3K/Akt Signaling Pathway. Dose Response 2019; 17:1559325819855538. [PMID: 31217757 PMCID: PMC6560806 DOI: 10.1177/1559325819855538] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/22/2019] [Accepted: 05/07/2019] [Indexed: 01/10/2023] Open
Abstract
Sulforaphane exerts anti-inflammatory activity in inflammatory diseases. The endometriosis (EM) is accompanied by chronic inflammation. The present study aims to explore the therapeutic effects of sulforaphane on EM and its underlying mechanism. An EM rat model was established by transplantation of autologous fragments. The rats were intragastrically administered sulforaphane (5 mg/kg, 15 mg/kg, and 30 mg/kg) for 3 weeks. The volumes of endometriotic foci and adhesion score were calculated at the end of the experiment. Levels of interleukin (IL)-6, IL-10, tumor necrosis factor (TNF)-α, interferon (IFN)-γ, and vascular endothelial growth factor (VEGF) were determined by enzyme-linked immunosorbent assay (ELISA). Expressions of VEGF, B-cell lymphoma/leukemia 2 (Bcl-2), Bax, cleaved caspase-3, PI3K, and Akt in endometrial tissue were determined by Western blotting. Relative expressions of PI3K and Akt were determined by quantitative polymerase chain reaction. Posttreatment of sulforaphane dose-dependently decreased the volumes of endometriotic foci and adhesion score in EM model. Additionally, posttreatment of sulforaphane inhibited levels of IL-6, IL-10, TNF-α, IFN-γ, and VEGF in peritoneal fluid and plasma. Posttreatment of sulforaphane regulated the expressions of VEGF, bcl-2, Bax, and cleaved Caspase-3 in EM model. The underlying mechanism revealed that sulforaphane attenuated EM in the rat model by inhibition of PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Aixiu Zhou
- Department of Gynaecology and Obstetrics, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Yiting Hong
- Department of Gynaecology and Obstetrics, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| | - Yuchun Lv
- Department of Gynaecology and Obstetrics, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| |
Collapse
|
41
|
Tang H, Li T, Han X, Sun J. TLR4 antagonist ameliorates combined allergic rhinitis and asthma syndrome (CARAS) by reducing inflammatory monocytes infiltration in mice model. Int Immunopharmacol 2019; 73:254-260. [PMID: 31121415 DOI: 10.1016/j.intimp.2019.05.021] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/20/2019] [Accepted: 05/10/2019] [Indexed: 01/08/2023]
Abstract
The present study aims to investigate the effects of toll-like receptor 4 (TLR4) antagonist in an ovalbumin (OVA)-induced mouse model of combined allergic rhinitis and asthma syndrome (CARAS). An OVA-induced mouse model of CARAS was established and TLR4 antagonist, TAK-242, was administrated intranasally or intraperitoneally. The number of sneezing and nasal rubbing was counted. The frequency of different cell types in the bronchoalveolar lavage fluid (BALF) and nasal lavage fluid (NLF) was analyzed using flow cytometry. Expressions of protein in nasal mucosa and lungs were determined using western blotting. Levels of interleukin (IL)-4, IL-5, and IL-13 were determined using Enzyme-linked Immunosorbent Assay (ELISA). Histological scores were applied for the assessment of lung injury. Treatment of TAK-242 downregulated CCL2 expression and reduced monocyte infiltration in nasal mucosa and lung tissues. Additionally, treatment of TAK-242 ameliorated upper airway symptoms including the sneezing and nasal rubbing by the regulation of cytokines including IL-4, IL-5, and IL-13. Furthermore, treatment of TAK-242 ameliorated lower airway symptoms including decreasing the frequency of CD45+SiglecF+CD11b+CD11c- eosinophils in BALF and IL13+ Th2 cells in the lungs. In conclusion, treatment of TAK-242 ameliorated CARAS-related lung injury by inhibiting lymphocyte infiltration, reducing monocytes infiltration, as well as regulating the frequency of eosinophils and Th2 cells.
Collapse
Affiliation(s)
- Huaping Tang
- Department of Respiratory, Qingdao Municipal Hospital (Eastern Campus), No. 5 Donghai Zhong Road, Qingdao 266071, Shandong, China
| | - Tingtian Li
- Department of Respiratory, Qingdao Municipal Hospital (Eastern Campus), School of Medicine Qingdao University, No. 5 Donghai Zhong Road, Qingdao 266071, Shandong, China
| | - Xiaolei Han
- Department of Health Care, Qingdao Municipal Hospital (Eastern Campus), No. 5 Donghai Zhong Road, Qingdao 266071, Shandong, China
| | - Jie Sun
- International Clinic, Qingdao Municipal Hospital (Eastern Campus), No. 5 Donghai Zhong Road, Qingdao 266071, Shandong, China.
| |
Collapse
|
42
|
Parveen A, Subedi L, Kim HW, Khan Z, Zahra Z, Farooqi MQ, Kim SY. Phytochemicals Targeting VEGF and VEGF-Related Multifactors as Anticancer Therapy. J Clin Med 2019; 8:E350. [PMID: 30871059 PMCID: PMC6462934 DOI: 10.3390/jcm8030350] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/27/2019] [Accepted: 03/06/2019] [Indexed: 02/06/2023] Open
Abstract
The role of vascular endothelial growth factor (VEGF) in cancer cells is not limited to angiogenesis; there are also multiple factors, such as neuropilins (non-tyrosine kinases receptors), tyrosine kinases receptors, immunodeficiencies, and integrins, that interact with VEGF signaling and cause cancer initiation. By combating these factors, tumor progression can be inhibited or limited. Natural products are sources of several bioactive phytochemicals that can interact with VEGF-promoting factors and inhibit them through various signaling pathways, thereby inhibiting cancer growth. This review provides a deeper understanding of the relation and interaction of VEGF with cancer-promoting factors and phytochemicals in order to develop multi-targeted cancer prevention and treatment.
Collapse
Affiliation(s)
- Amna Parveen
- Department of Pharmacognosy, Faculty of Pharmaceutical Science, Government College University, Faisalabad, Faisalabad 38000, Pakistan.
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
| | - Lalita Subedi
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
| | - Heung Wan Kim
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
| | - Zahra Khan
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
| | - Zahra Zahra
- Institute of Environmental Sciences and Engineering, School of Civil and Environmental Engineering, National University of Sciences and Technology, Sector H-12, Islamabad 44000, Pakistan.
| | | | - Sun Yeou Kim
- College of Pharmacy, Gachon University, No. 191, Hambakmoero, Yeonsu-gu, Incheon 21936, Korea.
- Gachon Institute of Pharmaceutical Science, Gachon University, No. 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Korea.
| |
Collapse
|
43
|
Jiang H, Li ZP, Tian GX, Pan RY, Xu CM, Zhang B, Wu JL. Liver-targeted liposomes for codelivery of curcumin and combretastatin A4 phosphate: preparation, characterization, and antitumor effects. Int J Nanomedicine 2019; 14:1789-1804. [PMID: 30880980 PMCID: PMC6413741 DOI: 10.2147/ijn.s188971] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background Recent efforts have been focused on combining two or more therapeutic approaches with different mechanisms to enhance antitumor therapy. Moreover, nanosize drug-delivery systems for codelivering two drugs with proapoptotic and antiangiogenic activities have exhibited great potential in efficient treatment of cancers. Methods Glycyrrhetinic acid (GA)–modified liposomes (GA LPs) for liver-targeted codelivery of curcumin (Cur) and combretastatin A4 phosphate (CA4P) were prepared and characterized. In vitro cellular uptake, cytotoxicity, cell migration, in vivo biodistribution, antitumor activity, and histopathological studies were performed. Results Compared with unmodified LPs (Cur-CA4P LPs), Cur-CA4P/GA LPs were taken up effectively by human hepatocellular carcinoma cells (BEL-7402) and showed higher cytotoxicity than free drugs. In vivo real-time near-infrared fluorescence–imaging results indicated that GA-targeted LPs increased accumulation in the tumor region. Moreover, Cur-CA4P/GA LPs showed stronger inhibition of tumor proliferation than Cur, Cur + CA4P, and Cur-CA4P LPs in vivo antitumor studies, which was also verified by H&E staining. Conclusion GA-modified LPs can serve as a promising nanocarrier for liver-targeted co-delivery of antitumor drugs against hepatocellular carcinoma.
Collapse
Affiliation(s)
- Hong Jiang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, China,
| | - Zhi-Peng Li
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, China,
| | - Gui-Xiang Tian
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, China,
| | - Rui-Yan Pan
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, China,
| | - Chong-Mei Xu
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China,
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, China,
| | - Jing-Liang Wu
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, China,
| |
Collapse
|
44
|
Morse MA, Sun W, Kim R, He AR, Abada PB, Mynderse M, Finn RS. The Role of Angiogenesis in Hepatocellular Carcinoma. Clin Cancer Res 2018; 25:912-920. [PMID: 30274981 DOI: 10.1158/1078-0432.ccr-18-1254] [Citation(s) in RCA: 379] [Impact Index Per Article: 54.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/17/2018] [Accepted: 09/26/2018] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) accounts for about 90% of all primary liver cancers and is the second leading cause of cancer-related deaths worldwide. The hypervascular nature of most HCC tumors underlines the importance of angiogenesis in the pathobiology of these tumors. Several angiogenic pathways have been identified as being dysregulated in HCC, suggesting they may be involved in the development and pathogenesis of HCC. These data provide practical targets for systemic treatments such as those targeting the vascular endothelial growth factor receptor and its ligand. However, the clinical relevance of other more recently identified angiogenic pathways in HCC pathogenesis or treatment remains unclear. Research into molecular profiles and validation of prognostic or predictive biomarkers will be required to identify the patient subsets most likely to experience meaningful benefit from this important class of agents.
Collapse
Affiliation(s)
- Michael A Morse
- Department of Medicine, Division of Medical Oncology, Duke University Health System, Durham, North Carolina.
| | - Weijing Sun
- Division of Medical Oncology, University of Kansas School of Medicine, Kansas City, Kansas
| | - Richard Kim
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Aiwu Ruth He
- Department of Medicine, Georgetown University Medical Center, Washington, District of Columbia
| | | | | | - Richard S Finn
- Department of Medicine, Division of Hematology/Oncology, Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
45
|
Lu YW, Zhu YC, Zhang L, Li P, Yang J, Wen XD. Ilexgenin A enhances the effects of simvastatin on non-alcoholic fatty liver disease without changes in simvastatin pharmacokinetics. Chin J Nat Med 2018; 16:436-445. [PMID: 30047465 DOI: 10.1016/s1875-5364(18)30077-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease (CVD) is the most common cause of death in patients with non-alcoholic fatty liver disease (NAFLD). New therapeutic strategies which have the potential for slowing down the evolution of NAFLD and reducing CVD-related mortality are urgently needed. Statins are well recognized in the treatment of dyslipidemia, but their use in the treatment of NAFLD is limited due to the safety concerns. Ilexgenin A (IA) is one of the main bioactive compounds in 'Shan-lv-cha', an herbal tea commonly used in China. In the present study, we investigated the possible synergistic therapeutic effects of IA and simvastatin (SV) on NAFLD. IA or SV showed beneficial effects on the rats with NAFLD by lowering the liver weight, liver index and plasma levels of alanine aminotransferase and aspartate aminotransferase, regulating abnormal metabolism of lipids and ameliorating steatosis in liver. IA significantly enhanced the hypolipidemic and anti-inflammation effects of SV. Furthermore, a sensitive, accurate, convenient and reproducible LC-MS method was developed to investigate the effects of IA on the pharmacokinetics of SV. No significant changes were observed in pharmacokinetic parameters of SV and simvastatin hydroxy acid in the IA plus SV co-treated group in comparison with those in the group treated with SV alone. The mRNA levels and activity of CYP3A1 were not altered by IA. In conclusion, the results obtained from the present study should be helpful for further clinical application of SV and IA alone or in combination.
Collapse
Affiliation(s)
- Ya-Wen Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ying-Chao Zhu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Li Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Jie Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China.
| | - Xiao-Dong Wen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
46
|
Zhang C, Wang N, Tan HY, Guo W, Li S, Feng Y. Targeting VEGF/VEGFRs Pathway in the Antiangiogenic Treatment of Human Cancers by Traditional Chinese Medicine. Integr Cancer Ther 2018; 17:582-601. [PMID: 29807443 PMCID: PMC6142106 DOI: 10.1177/1534735418775828] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Indexed: 12/11/2022] Open
Abstract
Bearing in mind the doctrine of tumor angiogenesis hypothesized by Folkman several decades ago, the fundamental strategy for alleviating numerous cancer indications may be the strengthening application of notable antiangiogenic therapies to inhibit metastasis-related tumor growth. Under physiological conditions, vascular sprouting is a relatively infrequent event unless when specifically stimulated by pathogenic factors that contribute to the accumulation of angiogenic activators such as the vascular endothelial growth factor (VEGF) family and basic fibroblast growth factor (bFGF). Since VEGFs have been identified as the principal cytokine to initiate angiogenesis in tumor growth, synthetic VEGF-targeting medicines containing bevacizumab and sorafenib have been extensively used, but prominent side effects have concomitantly emerged. Traditional Chinese medicines (TCM)-derived agents with distinctive safety profiles have shown their multitarget curative potential by impairing angiogenic stimulatory signaling pathways directly or eliciting synergistically therapeutic effects with anti-angiogenic drugs mainly targeting VEGF-dependent pathways. This review aims to summarize ( a) the up-to-date understanding of the role of VEGF/VEGFR in correlation with proangiogenic mechanisms in various tissues and cells; ( b) the elaboration of antitumor angiogenesis mechanisms of 4 representative TCMs, including Salvia miltiorrhiza, Curcuma longa, ginsenosides, and Scutellaria baicalensis; and ( c) circumstantial clarification of TCM-driven therapeutic actions of suppressing tumor angiogenesis by targeting VEGF/VEGFRs pathway in recent years, based on network pharmacology.
Collapse
Affiliation(s)
| | - Ning Wang
- The University of Hong Kong, Hong Kong
SAR
| | | | - Wei Guo
- The University of Hong Kong, Hong Kong
SAR
| | - Sha Li
- The University of Hong Kong, Hong Kong
SAR
| | - Yibin Feng
- The University of Hong Kong, Hong Kong
SAR
| |
Collapse
|
47
|
Chen Y, Wu H, Wang X, Wang C, Gan L, Zhu J, Tong J, Li Z. Huaier Granule extract inhibit the proliferation and metastasis of lung cancer cells through down-regulation of MTDH, JAK2/STAT3 and MAPK signaling pathways. Biomed Pharmacother 2018; 101:311-321. [PMID: 29499405 DOI: 10.1016/j.biopha.2018.02.028] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/05/2018] [Accepted: 02/09/2018] [Indexed: 10/17/2022] Open
Abstract
Although the effect of Huaier has been widely studied, its role and its molecular mechanism in lung cancer are not clear. In this study, we explored the inhibitory effect of Huaier on lung cancer cells and its molecular mechanism. The cell viability, migration and invasion were analyzed by CCK-8 and BrdU cell proliferation assay kits, Transwell and colony forming assay. The cell cycle and apoptosis were analyzed by flow cytometry. The experimental results showed that the viability, migration and invasion of A549 and NCI-H1650 cells were inhibited by Huaier in a dose and time-dependent manner. Huaier induced cell apoptosis and the cells were blocked in the S phase to inhibit cell proliferation. Western blotting results showed that Huaier inhibited the expression of MTDH and increased the proportion of Bax/Bcl-2, it could also promote the expression of Cleaved Caspase-3 and increase the activity of Caspase-3, promote cell apoptosis and inhibit cell proliferation. Huaier inhibited the metastasis and invasion of lung cancer cells by inhibiting the expression levels of EMT related proteins, it also inhibited the expression of JAK2/STAT3 and MAPK signaling pathways. Therefore, our results showed that Huaier may inhibit the proliferation and metastasis of lung cancer cells through multiple targets, it had the potential for treatment of lung cancer.
Collapse
Affiliation(s)
- Yang Chen
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, People's Republic of China; Anhui University of Chinese Medicine, Hefei, People's Republic of China
| | - Huan Wu
- Scientific Research Experiment Center, Anhui University of Chinese Medicine, Hefei, People's Republic of China
| | - Xinheng Wang
- Anhui University of Chinese Medicine, Hefei, People's Republic of China
| | - Chenyang Wang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, People's Republic of China
| | - Lei Gan
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, People's Republic of China
| | - Jie Zhu
- Anhui University of Chinese Medicine, Hefei, People's Republic of China
| | - Jiabing Tong
- Anhui University of Chinese Medicine, Hefei, People's Republic of China
| | - Zegeng Li
- Anhui University of Chinese Medicine, Hefei, People's Republic of China.
| |
Collapse
|
48
|
Liu C, Shen YJ, Tu QB, Zhao YR, Guo H, Wang J, Zhang L, Shi HW, Sun Y. Pedunculoside, a novel triterpene saponin extracted from Ilex rotunda, ameliorates high-fat diet induced hyperlipidemia in rats. Biomed Pharmacother 2018. [PMID: 29518607 DOI: 10.1016/j.biopha.2018.02.131] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pedunculoside (PE) is a novel triterpene saponin extracted from the dried barks of Ilex rotunda Thunb. The present study aims to explore lipid-lowering effects of PE on hyperlipidemia rat induced by high-fat diet. The rats were fed with the high-fat diet and subjected to intragastric administration of PE at doses of 30, 15, or 5 mg/kg daily for 7 weeks. The results demonstrated that treatment with PE for 7-week dramatically decreased serum total cholesterol (TC) and low-density lipoprotein cholesterol (LDL-C) and reduced liver TC in hyperlipidemia rat induced by high-fat diet. Furthermore, the results also showed that PE modulated the expression of enzymes involved in lipid metabolism including peroxisome proliferator-activated receptor α (PPAR-α), sterol regulatory element-binding protein 1 (SREBP-1), fatty acid synthase (FAS) and stearoyl CoA desaturase-1 (SCD-1) mRNA in liver. Besides, PE-treated group decreased weights and diameters of epididymal adipose hyperlipidemia rat. Mechanism study demonstrated that PE regulated PPAR-γ, CCAAT/Enhancer-binding Protein α (C/EBPα)、and SREBP-1 expression as well as inhibited phosphorylation of AMPK in MDI (methylisobutylxanthine, dexamethasone, insulin) induced-3T3L1 cells. Molecular Docking confirmed interaction between PE with proteins involving PPAR-γ, C/EBPα and SREBP-1. In summary, these findings may support that PE is a novel lipid-lowering drug candidate.
Collapse
Affiliation(s)
- Chang Liu
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China; School of Pharmacy, University of Rhode Island, RI, 02881, United States
| | - Yan-Jun Shen
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China
| | - Qing-Bo Tu
- College of Hanlin, Nanjing University of China Medicine, Taizhou 225300, Jiangsu, PR China
| | - Yan-Ran Zhao
- College of Hanlin, Nanjing University of China Medicine, Taizhou 225300, Jiangsu, PR China
| | - Hao Guo
- School of Pharmacy, University of Rhode Island, RI, 02881, United States; Department of Dermatology, No. 1 Hospital of China Medical University, 155N. Nanjing Street, Shenyang 110001, PR China
| | - Juan Wang
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China; College of Hanlin, Nanjing University of China Medicine, Taizhou 225300, Jiangsu, PR China
| | - Li Zhang
- School of Pharmacy, University of Missouri-Kansas City, MO, 64108, United States
| | - Hua-Wei Shi
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China
| | - Yun Sun
- College of Medicine, Yangzhou University, Yangzhou 225001, Jiangsu, PR China; College of Hanlin, Nanjing University of China Medicine, Taizhou 225300, Jiangsu, PR China.
| |
Collapse
|
49
|
Liu H, Chen Z, Jin W, Barve A, Wan YJY, Cheng K. Silencing of α-complex protein-2 reverses alcohol- and cytokine-induced fibrogenesis in hepatic stellate cells. LIVER RESEARCH 2017; 1:70-79. [PMID: 28966795 PMCID: PMC5613955 DOI: 10.1016/j.livres.2017.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIM α-complex protein-2 (αCP2) encoded by the poly (rC) binding protein 2(PCBP2) gene is responsible for the accumulation of type I collagen in fibrotic livers. In this study, we silenced the PCBP2 gene using a small interfering RNA (siRNA) to reverse alcohol-and cytokine-induced profibrogenic effects on hepatic stellate cells (HSCs). METHODS Primary rat HSCs and the HSC-T6 cell line were used as fibrogenic models to mimic the initiation and perpetuation stages of fibrogenesis, respectively. We previously found that a PCBP2 siRNA, which efficiently silences expression of αCP2, reduces the stability of type I collagen mRNA. We investigated the effects of the PCBP2 siRNA on cell proliferation and migration. Expression of type I collagen in HSCs was analyzed by quantitative real-time PCR and western blotting. In addition, we evaluated the effects of the PCBP2 siRNA on apoptosis and the cell cycle. RESULTS PCBP2 siRNA reversed multiple alcohol- and cytokine-induced profibrogenic effects on primary rat HSCs and HSC-T6 cells. The PCBP2 siRNA also reversed alcohol- and cytokine-induced accumulation of type I collagen as well as cell proliferation and migration. Moreover, the combination of LY2109761, a transforming growth factor-β1 inhibitor, and the PCBP2 siRNA exerted a synergistic inhibitive effect on the accumulation of type I collagen in HSCs. CONCLUSIONS Silencing of PCBP2 using siRNA could be a potential therapeutic strategy for alcoholic liver fibrosis.
Collapse
Affiliation(s)
- Hao Liu
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Zhijin Chen
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Wei Jin
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Ashutosh Barve
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Yu-Jui Yvonne Wan
- Department of Pathology and Laboratory Medicine, UC Davis Medical Center, Sacramento, CA, USA
| | - Kun Cheng
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA,Corresponding author. Kun Cheng, Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO, USA. (K. Cheng)
| |
Collapse
|
50
|
Chen Y, Tian JL, Wu JS, Sun TM, Zhou LN, Song SJ, You S. Biotransfomation of cyperenoic acid by Cunninghamella elegans AS 3.2028 and the potent anti-angiogenic activities of its metabolites. Fitoterapia 2017; 118:32-37. [DOI: 10.1016/j.fitote.2017.02.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 02/12/2017] [Accepted: 02/14/2017] [Indexed: 10/20/2022]
|