1
|
Yu H, Qu J, Gou H, Zhou Y. Formononetin suppresses the malignant progression of papillary thyroid carcinoma depending on downregulation of CBX4. Exp Ther Med 2024; 28:456. [PMID: 39478737 PMCID: PMC11523267 DOI: 10.3892/etm.2024.12746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 02/26/2024] [Indexed: 11/02/2024] Open
Abstract
Papillary thyroid carcinoma (PTC) is the most common malignant tumor of the endocrine system globally. Formononetin (FMNT), an isoflavonoid, exerts anti-tumorigenic effects and chromobox homolog 4 (CBX4) exerts tumor-promoting effect in specific types of tumors. Nevertheless, the predictive values and biological functions of FMNT and CBX4 in the pathological progress of PTC have not been fully understood till now. In the present study, the human PTC cell line TPC-1 was exposed to 0, 10, 30 and 100 µM FMNT for 24 h to elucidate the precise effects of FMNT on the biological behaviors of PTC cells. Moreover, FMNT-treated TPC-1 cells were transfected with oe-CBX4 to evaluate whether CBX4 was implicated in the anticarcinogenic effects of FMNT against PTC. It was demonstrated that FMNT treatment suppressed the proliferation, clone formation, migration, invasion, EMT, angiogenesis and stemness of PTC cells in a dose-dependent manner. Furthermore, it was verified that FMNT targeted CBX4 to downregulate its expression in a dose dependent manner. The suppressive effects of FMNT on the proliferation, clone formation, migration, invasion, EMT, angiogenesis and stemness of PTC cells were partially reversed by CBX4 overexpression. Upregulation of CBX4 abolished the tumor suppression effects of FMNT in the malignant progression of PTC. In conclusion, FMNT might act as a promising anti-tumorigenic agent in PTC, which depends on the downregulation of CBX4.
Collapse
Affiliation(s)
- Hongbo Yu
- Department of Traditional Chinese Medicine Traumatology and Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| | - Ji Qu
- Department of Traditional Chinese Medicine Traumatology and Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| | - Haixin Gou
- Department of Traditional Chinese Medicine Traumatology and Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, P.R. China
| | - Ying Zhou
- Department of Vascular Medicine, Shanghai Traditional Chinese Medicine-Integrated Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 200082, P.R. China
| |
Collapse
|
2
|
Aliya S, Alhammadi M, Park U, Tiwari JN, Lee JH, Han YK, Huh YS. The potential role of formononetin in cancer treatment: An updated review. Biomed Pharmacother 2023; 168:115811. [PMID: 37922652 DOI: 10.1016/j.biopha.2023.115811] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/21/2023] [Accepted: 10/26/2023] [Indexed: 11/07/2023] Open
Abstract
Currently, cancer is one of the main research topics, due to its high incidence and drug resistance to existing anti-cancer drugs. Formononetin, a natural product with phytoestrogenic properties and diverse biological functions, has attracted the attention of researchers working on anticancer drugs. Formononetin emerges as an intriguing bioactive substance compared to other isoflavones as it exhibits potent chemotherapeutic activity with less toxicity. Formononetin effectively plays a significant role in inhibiting cell proliferation, invasion, and metastatic abilities of cancer cells by targeting major signaling pathways at the junction of interconnected pathways. It also induces apoptosis and cell cycle arrest by modulating mediator proteins. It causes upregulation of key factors such as p-AKT, p38, p21, and p53 and downregulation of NF-κB. Furthermore, formononetin regulates the neoplastic microenvironment by inactivating the ERK1/2 pathway and lamin A/C signaling and has been reported to inactivate JAK/STAT, PKB or AKT, and mitogen-activated protein kinase pathways and to suppress cell migration, invasion, and angiogenesis in human cancer cells. To assist researchers in further exploring formononetin as a potential anticancer therapeutic candidate, this review focuses on both in vitro and in vivo proof of concept studies, patents, and clinical trials pertinent to formononetin's anticancer properties. Overall, this review discusses formononetin from a comprehensive perspective to highlight its potential benefits as an anticancer agent.
Collapse
Affiliation(s)
- Sheik Aliya
- Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Munirah Alhammadi
- Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Uichang Park
- Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea
| | - Jitendra N Tiwari
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 100-715, Republic of Korea
| | - Jeong-Hwan Lee
- 3D Convergence Center, Inha University, Incheon 22212, Republic of Korea; Department of Materials Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Young-Kyu Han
- Department of Energy and Materials Engineering, Dongguk University-Seoul, Seoul 100-715, Republic of Korea.
| | - Yun Suk Huh
- Department of Biological Sciences and Bioengineering, Inha University, Incheon 22212, Republic of Korea.
| |
Collapse
|
3
|
Cao X, Li Q, Li X, Liu Q, Liu K, Deng T, Weng X, Yu Q, Deng W, Yu J, Wang Q, Xiao G, Xu X. Enhancing Anticancer Efficacy of Formononetin Microspheres via Microfluidic Fabrication. AAPS PharmSciTech 2023; 24:241. [PMID: 38017231 DOI: 10.1208/s12249-023-02691-9] [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: 07/19/2023] [Accepted: 10/30/2023] [Indexed: 11/30/2023] Open
Abstract
Formononetin is a flavonoid compound with anti-tumor and anti-inflammatory properties. However, its low solubility limits its clinical use. We employed microfluidic technology to prepare formononetin-loaded PLGA-PEGDA microspheres (Degradable polymer PLGA, Crosslinking agent PEGDA), which can encapsulate and release drugs in a controlled manner. We optimized and characterized the microspheres, and evaluated their antitumor effects. The microspheres had uniform size, high drug loading efficiency, high encapsulation efficiency, and stable release for 35 days. They also inhibited the proliferation, migration, and apoptosis. The antitumor mechanism involved the induction of reactive oxygen species and modulation of Bcl-2 family proteins. These findings suggested that formononetin-loaded PLGA-PEGDA microspheres, created using microfluidic technology, could be a novel drug delivery system that can overcome the limitations of formononetin and enhance its antitumor activity.
Collapse
Affiliation(s)
- Xia Cao
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Qingwen Li
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Xiaoli Li
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Qi Liu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Kai Liu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Tianwen Deng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Xuedi Weng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Qintong Yu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Wenwen Deng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China.
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China.
| | - Gao Xiao
- College of Environment and Safety Engineering, Fuzhou University, Fuzhou, 350108, Fujian, People's Republic of China.
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China.
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China.
| |
Collapse
|
4
|
Xu X, Luo H, Chen Q, Wang Z, Chen X, Li X, Chen H, Wang M, Xu Y, Dai M, Wang J, Huang X, Wu B, Li Y. Detecting potential mechanism of vitamin D in treating rheumatoid arthritis based on network pharmacology and molecular docking. Front Pharmacol 2022; 13:1047061. [DOI: 10.3389/fphar.2022.1047061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 11/09/2022] [Indexed: 12/05/2022] Open
Abstract
Aim: Vitamin D plays a vital role in Rheumatoid arthritis (RA). However, the mechanism of vitamin D and rheumatism is still unclear. Therefore, a strategy based on network pharmacology and molecular docking was used to explore the mechanism of vitamin D and RA.Methods: The targets of RA were obtained from the GeneCards database and Therapeutic Targets Database, and the targets of vitamin D were obtained from the Drugbank database and STITCH database. Next, overlapping genes were identified by Venny, and further Gene ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and molecular docking analyses were performed.Results: A total of 1,139 targets of RA and 201 targets of vitamin D were obtained. A total of 76 overlapping genes were identified by Venny. The enrichment analysis showed that cell proliferation, immune response, and apoptotic process were the critical biological processes of vitamin D in treating RA. Antifolate resistance, osteoclast differentiation, and the nuclear factor-kappa B (NF-κB) signalling pathway are fundamental mechanisms of vitamin D in treating RA. According to further molecular docking, ALB, TNF, CASP3, and TP53 may be important punctuation points or diagnostic markers for future RA treatment.Conclusion: By analysing overlapping genes of diseases and drugs, this study confirmed that ALB, TNF, CASP3, and TP53 may be essential markers or diagnostic markers for future RA treatment.
Collapse
|
5
|
Qiu W, Sang T, Chen H, Zhou H, Wang Z, Zhou H. Wenzi Jiedu Recipe ameliorates colorectal cancer by remodeling the gut microbiota and tumor microenvironment. Front Oncol 2022; 12:915498. [PMID: 36212428 PMCID: PMC9541612 DOI: 10.3389/fonc.2022.915498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
IntroductionWenzi Jiedu Recipe (WJR), traditional Chinese medicine (TCM) formula, has been proven to be clinically useful in the treatment of colorectal cancer (CRC). However, its underlying mechanisms are still elusive, which limits its wider application. Thus, we aimed to evaluate the effect of WJR on CRC and elucidate mechanisms underlying its action.MethodsNetwork pharmacology was employed to clarify the “herb-active ingredient-target” network of WJR. The 16S rDNA sequencing method was used to analyze the changes of gut microbes mediated by WJR in tumor-bearing mice with CRC. The proportions of CD4+ T cell and CD8+ T cell were measured by flow cytometry. Levels of the cytokines interleukin (IL)-10, interferon (IFN)-γ, and tumor necrosis factor (TNF)-α were assessed by immunohistochemistry and enzyme-linked immunosorbent assay (ELISA).ResultsWJR showed significant anti-CRC effects both in vitro and in vivo. Network pharmacology revealed that WJR exerts anti-CRC therapeutic effect on multiple targets and signaling pathways. Gut microbiota analysis revealed that WJR therapy significantly enriched for Oscillibacter and Bacteroides_acidifacien. In particular, we found that WJR significantly increased the proportion of CD8+ T cells and the expression of immune-associated cytokines IL-10, IFN-γ, and TNF-α.ConclusionThe regulation of gut microbiota by WJR may be the breakthrough point to clarify its mechanism of action in the treatment of CRC, and it has a good prospect of clinical application.
Collapse
Affiliation(s)
- Wenli Qiu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Tianqing Sang
- The First Clinical Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Haibin Chen
- Science and Technology Department, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongli Zhou
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhongqiu Wang
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hongguang Zhou
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Hongguang Zhou,
| |
Collapse
|
6
|
WGCNA-Based Identification of Hub Genes and Key Pathways Involved in Nonalcoholic Fatty Liver Disease. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5633211. [PMID: 34938809 PMCID: PMC8687832 DOI: 10.1155/2021/5633211] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 11/14/2021] [Accepted: 11/23/2021] [Indexed: 12/27/2022]
Abstract
Background The morbidity of nonalcoholic fatty liver disease (NAFLD) has been rising, but the pathogenesis of NAFLD is still elusive. This study is aimed at determining NAFLD-related hub genes based on weighted gene coexpression network analysis (WGCNA). Methods GSE126848 dataset based construction of coexpression networks was performed based on WGCNA. Database for Annotation, Visualization, and Integrated Discovery (DAVID) was utilized for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Hub genes were identified and validated in independent datasets and mouse model. Results We found that the steelblue module was most significantly correlated with NAFLD. Total 15 hub genes (NDUFA9, UQCRQ, NDUFB8, COPS5, RPS17, UBL5, PSMA3, PSMA1, SF3B5, MRPL27, RPL26, PDCD5, PFDN6, SNRPD2, PSMB3) were derived from both the coexpression and PPI networks and considered “true” hub genes. Functional enrichment analysis showed that the hub genes were related to NAFLD pathway and oxidative phosphorylation. Independent dataset-based analysis and the establishment of NAFLD mouse model confirmed the involvement of two hub genes NDUFA9 and UQCRQ in the pathogenesis of NAFLD. Conclusions Oxidative phosphorylation and NAFLD pathway may be crucially involved in the pathogenesis of NAFLD, and two hub genes NDUFA9 and UQCRQ might be diagnostic biomarkers and therapeutic targets for NAFLD.
Collapse
|
7
|
Huang J, Chen X, Xie A. Formononetin ameliorates IL‑13‑induced inflammation and mucus formation in human nasal epithelial cells by activating the SIRT1/Nrf2 signaling pathway. Mol Med Rep 2021; 24:832. [PMID: 34590155 PMCID: PMC8503736 DOI: 10.3892/mmr.2021.12472] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 06/28/2021] [Indexed: 01/18/2023] Open
Abstract
Formononetin has proven to be anti‑inflammatory and able to alleviate symptoms of certain allergic diseases. The present study aimed to determine and elucidate the potential effects of formononetin in allergic rhinitis. JME/CF15 cells were pretreated with formononetin at different doses, followed by stimulation with IL‑13. Cell Counting Kit‑8 assay was performed to determine the cytotoxicity of formononetin. The expression levels of inflammation‑related proteins, histamine, IgE, TNF‑α, IL‑1β, IL‑6, granulocyte‑macrophage colony‑stimulating factor and eotaxin in IL‑13‑stimulated JME/CF15 cells were detected using ELISAs. The expression levels of phosphorylated‑NF‑κB p65, NF‑κB p65 and cyclooxygenase‑2 (Cox‑2) were analyzed using western blotting. Reverse transcription‑quantitative PCR, western blotting and immunofluorescence were performed to measure the levels of mucin 5AC oligomeric mucus/gel‑forming. Expression levels of sirtuin 1 (SIRT1) and nuclear erythroid factor 2‑related factor 2 (Nrf2) proteins were also measured using western blotting. The results of the present study revealed that formononetin exerted no cytotoxic effect on the viability of JME/CF15 cells. Following stimulation of JME/CF15 cells with IL‑13, formononetin suppressed the upregulated expression levels of proinflammatory cytokines. IL‑13‑induced formation of mucus was also attenuated by formononetin treatment. Furthermore, it was found that the SIRT1/Nrf2 signaling pathway was activated in formononetin‑treated JME/CF15 cells, whereas treatment with the SIRT1 inhibitor, EX527, reversed the effects of formononetin on IL‑13‑induced inflammation and mucus formation in JME/CF15 cells. In conclusion, the findings of the current study indicated that formononetin may activate the SIRT1/Nrf2 signaling pathway, thereby inhibiting IL‑13‑induced inflammation and mucus formation in JME/CF15 cells. These results suggested that formononetin may represent a promising agent for the treatment of allergic rhinitis.
Collapse
Affiliation(s)
- Juanjuan Huang
- Department of Traditional Chinese Medicine, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Xianfeng Chen
- Department of Traditional Chinese Medicine, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| | - Aihua Xie
- Department of Traditional Chinese Medicine, The Affiliated People's Hospital of Ningbo University, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
8
|
Network pharmacology and molecular docking analysis on molecular targets: Mechanisms of baicalin and baicalein against hyperuricemic nephropathy. Toxicol Appl Pharmacol 2021; 424:115594. [PMID: 34044073 DOI: 10.1016/j.taap.2021.115594] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 12/29/2022]
Abstract
Oxidative stress and inflammation in kidney are the main causes for hyperuricemic nephropathy (HN). Baicalin and baicalein, two flavonoids, have anti-inflammatory and anti-oxidative effects and they are interconvertible in the body. In this study, both baicalin and baicalein were administered by intragastric administration (i.g.) or intraperitoneal injection (i.p.) at the dose of 50 mg kg-1, once a day for 15 consecutive days to HN mice, a model established by i.g. of yeast extract combined with i.p. of potassium oxonate. In HN mice, baicalin and baicalein reduced serum uric acid (SUA) levels and protected kidneys by anti-inflammatory and anti-oxidative effects. Mechanistically, the effect of baicalin and baicalein on reducing SUA levels might due to their inhibitory effect on xanthine oxidase (XO) activity in vivo and in vitro. Furthermore, the mechanisms of baicalin and baicalein against HN were analyzed with network pharmacology and molecular docking technology. The network pharmacology indicated that the protective effects of baicalin and baicalein against HN were mainly related to their down-regulating effects on TLRs, NF-κB, MAPK, PI3K/AKT and NOD-like receptor signaling pathways. Molecular docking indicated high binding affinity of baicalin/baicalein to targets such as AKT1 and MAPK1. In summary, baicalin and baicalein are promising drug candidates for the treatment of HN by inhibiting XO activity, reducing inflammation and cell apoptosis through down-regulating TLRs/NLRP3/NF-κB, MAPK, PI3K/AKT/NF-κB pathways.
Collapse
|
9
|
Wang WS, Zhao CS. Formononetin exhibits anticancer activity in gastric carcinoma cell and regulating miR-542-5p. Kaohsiung J Med Sci 2021; 37:215-225. [PMID: 33231363 DOI: 10.1002/kjm2.12322] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 10/12/2020] [Accepted: 10/18/2020] [Indexed: 12/14/2022] Open
Abstract
Formononetin exhibits anti-neoplastic activities in specific types of cancers, such as colon carcinoma and breast cancer. Nevertheless, its role in suppressing gastric carcinoma (GC) growth and metastatic-associated phenotypes has not been fully understood. Here, we demonstrated that formononetin decreased the viability of GC cell line SGC-7901 and MGC-803. Furthermore, formononetin suppressed the migration and invasion abilities of GC cells. Consistent with the results in vitro, the anticancer effect of formononetin was verified using xenograft model. The expression of microRNA-542-5p (miR-542-5p), acted as an oncogene in many cancers, was identified to be upregulated in GC. Importantly, miR-542-5p might involve in formononetin exhibits anticancer activity in GC cells. Taken together, these results indicate that formononetin inhibits the growth and aggressiveness of GC cells in vitro and in vivo.
Collapse
Affiliation(s)
- Wei-Song Wang
- Department of General Surgery, Zhuji Central Hospital, Zhuji, China
| | - Can-Song Zhao
- Department of General Surgery, Zhuji People's Hospital of Zhejiang, Zhuji, China
| |
Collapse
|
10
|
Taherkhani A, Orangi A, Moradkhani S, Khamverdi Z. Molecular Docking Analysis of Flavonoid Compounds with Matrix Metalloproteinase- 8 for the Identification of Potential Effective Inhibitors. LETT DRUG DES DISCOV 2021. [DOI: 10.2174/1570180817999200831094703] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Background:
Matrix metalloproteinase-8 (MMP-8) participates in the degradation of different
types of collagens in the extracellular matrix and basement membrane. Up-regulation of the
MMP-8 has been demonstrated in many disorders including cancer development, tooth caries, periodontal/
peri-implant soft and hard tissue degeneration, and acute/chronic inflammation. Therefore,
MMP-8 has become an encouraging target for therapeutic procedures for scientists. We carried out a
molecular docking approach to study the binding affinity of 29 flavonoids, as drug candidates, with
the MMP-8. Pharmacokinetic and toxicological properties of the compounds were also studied.
Moreover, it was attempted to identify the most important amino acids participating in ligand binding
based on the degree of each of the amino acids in the ligand-amino acid interaction network for
MMP-8.
Methods:
Three-dimensional structure of the protein was gained from the RCSB database (PDB ID: 4QKZ).
AutoDock version 4.0 and Cytoscape 3.7.2 were used for molecular docking and network analysis,
respectively. Notably, the inhibitor of the protein in the crystalline structure of the 4QKZ was considered
as a control test. Pharmacokinetic and toxicological features of compounds were predicted
using bioinformatics web tools. Post-docking analyses were performed using BIOVIA Discovery
Studio Visualizer version 19.1.0.18287.
Results and Discussions:
According to results, 24 of the studied compounds were considered to be
top potential inhibitors for MMP-8 based on their salient estimated free energy of binding and inhibition
constant as compared with the control test: Apigenin-7-glucoside, nicotiflorin, luteolin,
glabridin, taxifolin, apigenin, licochalcone A, quercetin, isorhamnetin, myricetin, herbacetin,
kaemferol, epicatechin, chrysin, amentoflavone, rutin, orientin, epiafzelechin, quercetin-3-
rhamnoside, formononetin, isoliquiritigenin, vitexin, catechine, and isoquercitrin. Moreover, His-
197 was found to be the most important amino acid involved in the ligand binding for the enzyme.
Conclusion:
The results of the current study could be used in the prevention and therapeutic procedures
of a number of disorders such as cancer progression and invasion, oral diseases, and
acute/chronic inflammation. Although, in vitro and in vivo tests are inevitable in the future.
Collapse
Affiliation(s)
- Amir Taherkhani
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Athena Orangi
- Dental Research Center, Department of Restorative Dentistry, Dental School, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shirin Moradkhani
- Department of Pharmacognosy, School of Pharmacy, Medicinal Plants and Natural Product Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Zahra Khamverdi
- Dental Research Center, Department of Restorative Dentistry, Dental School, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
11
|
Li T, Zhang S, Chen F, Hu J, Yuan S, Li C, Wang X, Zhang W, Tang R. Formononetin ameliorates the drug resistance of Taxol resistant triple negative breast cancer by inhibiting autophagy. Am J Transl Res 2021; 13:497-514. [PMID: 33594306 PMCID: PMC7868832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/31/2020] [Indexed: 06/12/2023]
Abstract
Characterized by autophagy-associated protein disorders, autophagy participates in Taxol resistance in triple negative breast cancer (TNBC). As an evolutionarily conserved serine/threonine protein kinase with complex signaling pathway, mammalian target of rapamycin (mTOR) can regulate various cellular functions by phosphorylation of its downstream target proteins after activation. A large number of references have demonstrated that mTOR signaling pathway is related to autophagy and apoptosis. Formononetin (FMNT) has anticancer properties against breast, prostate and colon cancers. This study aimed to explore the regulatory effect of FMNT/miR-199a-3p/mTOR pathway on Taxol resistance and autophagy in breast cancer (BC). MiR-199a-3p, mTOR, LC3 and other autophagy related proteins were detected in Taxol sensitive and Taxol resistant TNBC cell lines, which were MDA-MB-231 and MDA-MB-231/Taxol, respectively. Cell viability and toxicity were determined by CCK-8 and MTT assay, respectively. The therapeutic effect of FMNT was evaluated in xenotransplantation model of nude mice. MiR-199a-3p was more highly expressed in MDA-MB-231/Taxol than in MDA-MB-231, while mTOR and p-mTOR decreased in MDA-MB-231/Taxol in comparison with MDA-MB-231, and autophagy activation and drug resistance were enhanced. In MDA-MB-231/Taxol cell line, the role of FMNT was verified to inhibit high miR-199a-3p expression. In addition, the combination therapy of FMNT and Taxol was found to be more effective in inhibiting autophagy and drug resistance. Moreover, mTOR was the target of miR-199a-3p, which was confirmed by dual luciferase reporter (DLR) gene assay. Oral administration of FMNT reduced tumor volume after MDA-MB-231/Taxol injection in vivo. Moreover, oral administration of FMNT and Taxol suppressed autophagy and Taxol resistance by restoring mTOR protein level to that of the parent MDA-MB-231, suggesting that miR-199a-3p can severe as a new target to overcome Taxol resistance in TNBC.
Collapse
Affiliation(s)
- Tian Li
- Breast Surgery Department, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| | - Shiyi Zhang
- The Second School of Clinical Medicine of Guangdong Medical UniversityDongguan 523000, China
| | - Fengsong Chen
- Nantong Haimen People’s HospitalNantong 226100, China
| | - Jun Hu
- Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| | - Shuai Yuan
- Breast Surgery Department, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| | - Chaoran Li
- Breast Surgery Department, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| | - Xiaoting Wang
- Breast Surgery Department, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| | - Weihong Zhang
- Breast Surgery Department, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| | - Runwei Tang
- Breast Surgery Department, Baoshan Branch, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai 201900, China
| |
Collapse
|
12
|
Feng Y, Dramani Maman ST, Zhu X, Liu X, Bongolo CC, Liang C, Tu J. Clinical value and potential mechanisms of LINC00221 in hepatocellular carcinoma based on integrated analysis. Epigenomics 2021; 13:299-317. [PMID: 33406920 DOI: 10.2217/epi-2020-0363] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Aims:This study aimed to unveil the functional roles of LINC00221 in hepatocellular carcinoma (HCC). Materials and methods:A discovery cohort and a validation cohort were respectively used to identify and verify the clinical value of LINC00221 in HCC. Bioinformatics analysis was performed to explore its potential mechanisms. Results:LINC00221 was upregulated in HCC tissues and serum samples. Survival analysis and receiver operating characteristic curve further revealed its prognostic and diagnostic roles. Exploration of the mechanism showed that LINC00221 might exert a pro-cancer role via the lncRNA-miRNA-mRNA network.Conclusions: Our study reveals that upregulated LINC00221 can serve as a potential diagnostic and prognostic biomarker and provides novel clues as to the role of LINC00221 in HCC.
Collapse
Affiliation(s)
- Yanlin Feng
- Program & Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Souraka Tapara Dramani Maman
- Program & Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xinyu Zhu
- Program & Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xuefang Liu
- Department of Clinical Laboratory, Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Christian Cedric Bongolo
- Program & Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chunzi Liang
- Program & Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jiancheng Tu
- Program & Department of Clinical Laboratory Medicine, Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| |
Collapse
|
13
|
Zhang L, Zhang J, Gong Y, Lv L. Systematic and experimental investigations of the anti-colorectal cancer mediated by genistein. Biofactors 2020; 46:974-982. [PMID: 32951326 DOI: 10.1002/biof.1677] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022]
Abstract
Recent evidences have suggested that genistein, a beneficial isoflavonoid, exerts marked anti-proliferative action on colorectal cancer (CRC) cells. However, the exact molecular mechanisms behind anti-CRC effect of genistein have not been elucidated. In current report, a systemic pharmacology analysis was used to disclose the anti-CRC mechanism of genistein prior to performing experimentative certification. As shown in network pharmacology findings, a total of 189 common targets and 9 hard-core targets of genistein-anti-CRC were collected and identified. And the detailed anti-CRC functions and pathways mediated by genistein were uncovered. In further certification, human CRC samples resulted in elevated protein and mRNA expressions of myeloid leukemia cell differentiation protein (MCL1), beta amyloid A4 protein (APP), and vascular endothelial growth factor receptor 2 (KDR). In animal experiment, genistein-treated tumor-transplanted nude mice exhibited reduced tumor growth, accompanied with dose-dependent down-regulations of MCL1, APP, and KDR proteins and mRNAs. Taken together, the integrated bioinformatic and experimental findings uncover the anti-CRC mechanisms and targets mediated by genistein. Significantly, parts of hard-core biotargets were experimentally verified before clinical application, including MCL1, APP, and KDR.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Emergency and Trauma Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Junzhi Zhang
- Department of Emergency and Trauma Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yizhen Gong
- Department of Emergency and Trauma Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Lv Lv
- Department of Emergency and Trauma Surgery, Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
14
|
Formononetin inhibits tumor growth by suppression of EGFR-Akt-Mcl-1 axis in non-small cell lung cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:62. [PMID: 32276600 PMCID: PMC7146989 DOI: 10.1186/s13046-020-01566-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Accepted: 03/26/2020] [Indexed: 12/22/2022]
Abstract
Background Epidermal growth factor receptor (EGFR) activating mutations play crucial roles in the tumorigenesis of human non-small cell lung cancer (NSCLC). The mechanism regarding how EGFR signaling regulates myeloid cell leukemia sequence 1 (Mcl-1) protein stability and ubiquitination remains undefined. Methods MTS assay was used for natural product library screening. The effect of formononetin (Formo) on NSCLC cells was determined by MTS assay and soft agar assay. Molecular modeling was performed to analyze the potential different binding modes between Formo and EGFR WT or mutants. Mcl-1 protein level and the inhibitory effect of Formo on EGFR signaling were examined by immunoblot, in vitro kinase assay, in vitro pulldown and ATP competition assays, co-immunoprecipitation assay, ubiquitination analysis, in vivo xenograft model, and immunohistochemical staining. Results Formo was identified as an EGFR inhibitor by a 98 commercially available natural product screening. Formo suppresses WT and mutant EGFR kinases activity in vitro, ex vivo, and in vivo. Molecular modeling indicates that Formo docks into the ATP-binding pocket of both WT and mutant EGFR. Formo inhibits EGFR-Akt signaling, which in turn activates GSK3β and promotes Mcl-1 phosphorylation in NSCLC cells. Treatment with Formo enhances the interaction between Mcl-1 and SCFFbw7, which eventually promotes Mcl-1 ubiquitination and degradation. Depletion of either GSK3β or SCFFbw7 compromised Formo-induced Mcl-1 downregulation. Finally, Formo inhibits the in vivo tumor growth in a xenograft mouse model. Conclusion This study highlights the importance of promoting ubiquitination-dependent Mcl-1 turnover might be an alternative strategy to enhance the anti-tumor efficacy of EGFR-TKI.
Collapse
|