1
|
Song Q, He L, Feng J. SQLE promotes osteosarcoma progression via activating TGFβ/SMAD signaling pathway. Mol Cell Probes 2024; 78:101993. [PMID: 39608425 DOI: 10.1016/j.mcp.2024.101993] [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: 10/30/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND The prognosis of advanced osteosarcoma (OS) has remained stagnant in last decades, requiring the identification of novel therapeutic targets. Recently, much attention was paid to the role of squalene epoxidase (SQLE), a rate-limiting enzyme in cholesterol metabolism, in the field of oncology, while the specific role of SQLE in OS has not been sufficiently elucidated. The present study aims to investigate the role of SQLE in the progression of OS and explore the potential mechanisms. METHODS The expression levels of SQLE in OS tissues and adjacent normal tissues were compared using bioinformatic methods and experiments. Kaplan-Meier survival analysis and univariate and multivariate Cox analysis were performed to detect the association of SQLE expression and patient' prognosis. Stably cell lines with SQLE knockdown or overexpression were constructed by lentivirus infection. CCK-8, colony formation, scratch healing, and Transwell invasion assays were carried out to explore the effect of SQLE knockdown or overexpression on the proliferation, migration, and invasion of OS cells. Gene set enrichment analysis was conducted to reveal signaling pathways associated with SQLE expression. The effect of SQLE on TGFβ/SMAD signaling pathway were explored by Western blot assay. RESULTS Here, we found a notable rise of SQLE expression in OS tissues and cell lines. Survival analysis showed that individuals with high SQLE expression had a lower median overall survival time compared to those with low SQLE expression. Univariate and multivariate Cox regression analyses showed that SQLE might have the potency to serve as an independently prognostic biomarker in OS. Loss- and gain-of-function experiments indicated that silence of SQLE suppressed OS cell proliferation, migration, and invasion, while overexpression of SQLE exerted the opposite effects. Mechanistically, TGF-β signaling pathway was identified as the downstream pathway of SQLE through bioinformatic methods, and the results of Western blot assay showed that SQLE positively regulated the activity of TGFβ1/SMAD2/3 signaling in OS. Resue experiments demonstrated that SB431542, a small molecule that inhibits TGFβ/SMAD signaling, could partly reverse the promoting effects of SQLE on OS cell proliferation, migration, and invasion. CONCLUSION Our results provided preliminary evidences that SQLE was a tumor-promoting factor and prognosis predictor in OS. SQLE promoted OS cell proliferation, migration, and invasion via activating TGFβ/SMAD signaling and targeting SQLE might be a potential strategy for the treatment of OS.
Collapse
Affiliation(s)
- Qi Song
- Department of Trauma Surgery, Wuhan No. 1 Hospital, Wuhan, 430022, Hubei Province, China
| | - Lina He
- Department of Trauma Surgery, Wuhan No. 1 Hospital, Wuhan, 430022, Hubei Province, China
| | - Jing Feng
- Department of Trauma Surgery, Wuhan No. 1 Hospital, Wuhan, 430022, Hubei Province, China.
| |
Collapse
|
2
|
Yang WX, Zhang WQ, Wei MQ, Duan MH, Liu XJ, Yan C. New 3-acyl derivatives of glaucocalyxin A: designed, synthesis and in vitro antibacterial activities. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2024:1-13. [PMID: 39565838 DOI: 10.1080/10286020.2024.2429136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 11/08/2024] [Accepted: 11/09/2024] [Indexed: 11/22/2024]
Abstract
To discover novel antimicrobial drug, 22 novel acylated derivatives were synthesized by A-ring modification of glaucocalyxin A. The structures of these derivatives were confirmed by NMR and MS data. In vitro antimicrobial activity of these compounds was evaluated against E. faecium, E. faecalis, MRSA, E. coli, A. baumannii and K. pneumoniae. The results showed compound 3d against E. faecium, E. faecalis and MRSA with a minimum inhibitory concentration of 4 μg/ml. And further molecular docking revealed that compound 3d has a higher binding affinity. In conclusion, compound 3d has the potential to develop into a new drug against drug-resistant bacteria.
Collapse
Affiliation(s)
- Wei-Xian Yang
- Guizhou University of Traditional Chinese Medicine, Guiyang550000, China
- Anshun City People's Hospital, Anshun561000, China
| | | | - Mei-Qi Wei
- Guizhou University of Traditional Chinese Medicine, Guiyang550000, China
- Anshun City People's Hospital, Anshun561000, China
| | - Mei-Hui Duan
- Guizhou University of Traditional Chinese Medicine, Guiyang550000, China
- Anshun City People's Hospital, Anshun561000, China
| | - Xian-Ji Liu
- Anshun City People's Hospital, Anshun561000, China
| | - Chen Yan
- Guizhou University of Traditional Chinese Medicine, Guiyang550000, China
- Anshun City People's Hospital, Anshun561000, China
| |
Collapse
|
3
|
Yue X, Lai L, Wang R, Tan L, Wang Y, Xie Q, Li Y. DGA ameliorates severe acute pancreatitis through modulating macrophage pyroptosis. Inflamm Res 2024; 73:1803-1817. [PMID: 39231819 DOI: 10.1007/s00011-024-01931-3] [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: 06/27/2024] [Revised: 07/27/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
Severe acute pancreatitis (SAP) is an inflammatory disease with varying severity, ranging from mild local inflammation to severe systemic disease, with a high incidence rate and mortality. Current drug treatments are not ideal. Therefore, safer and more effective therapeutic drugs are urgently needed. 7α,14β-dihydroxy-ent-kaur-17-dimethylamino-3,15-dione DGA, a diterpenoid compound derivatized from glaucocalyxin A, exhibits anti-inflammatory activity. In this study, we demonstrated the therapeutic potential of DGA against SAP and elucidated the underlying mechanisms. Treatment with DGA markedly (1) inhibited death of RAW264.7 and J774a.1 cells induced by Nigericin and lipopolysaccharide, (2) alleviated edema, acinar cell vacuolation, necrosis, and inflammatory cell infiltration of pancreatic tissue in mice, and (3) inhibited the activity of serum lipase and the secretion of inflammatory factor IL-1β. DGA significantly reduced the protein expression of IL-1β and NLRP3 and inhibited the phosphorylation of NF-κB. However, DGA exhibited no inhibitory effect on the expression of caspase-1, gasdermin D (GSDMD), NF-κB, TNF-α, or apoptosis-associated speck-like protein (ASC) and on the cleavage of caspase-1 or GSDMD. Molecular docking simulation confirmed that DGA can bind to TLR4 and IL-1 receptor. In conclusion, DGA may effectively alleviate the symptoms of SAP in mice and macrophages by inhibiting the binding of TLR4 and IL-1 receptor to their ligands; therefore, DGA is a promising drug candidate for the treatment of patients with SAP.
Collapse
Affiliation(s)
- Xiyue Yue
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, 199 Ren Ai Road, Suzhou, 215123, China
| | - Lunmeng Lai
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, 199 Ren Ai Road, Suzhou, 215123, China
| | - Ruina Wang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, 199 Ren Ai Road, Suzhou, 215123, China
| | - Lulu Tan
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, 199 Ren Ai Road, Suzhou, 215123, China
| | - Yanping Wang
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, 199 Ren Ai Road, Suzhou, 215123, China
| | - Qing Xie
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, 199 Ren Ai Road, Suzhou, 215123, China.
| | - Yunsen Li
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, 199 Ren Ai Road, Suzhou, 215123, China.
| |
Collapse
|
4
|
Tang Z, Zhang D, Yao C, Jiang M, Wang C, Chen Z, Yu H, Xue C, Liu Y, Shi Y, Zhang L, Wang X, Wei Z. TIPE2 inhibits the migration and invasion of epithelial ovarian cancer cells by targeting Smad2 to reverse TGF-β1-induced EMT. FASEB J 2024; 38:e70045. [PMID: 39259551 DOI: 10.1096/fj.202401427r] [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: 06/25/2024] [Revised: 08/19/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024]
Abstract
Epithelial ovarian cancer is the deadliest gynecologic malignancy, characterized by high metastasis. Transforming growth factor-β1 (TGF-β1) drives epithelial- mesenchymal transformation (EMT), a key process in tumor metastasis. Tumor necrosis factor-α-induced protein 8 (TNFAIP8)-like 2 (TIPE2) acts as a negative regulator of innate and adaptive immunity and involves in various cancers. However, its relationship with TGF-β1 in ovarian cancer and its role in reversing TGF-β1-induced EMT remain unclear. This study examined TIPE2 mRNA and protein expression using quantitative RT-PCR (qRT-PCR), western blot and immunohistochemistry. The effects of TIPE2 overexpression and knockdown on the proliferation, migration and invasion of epithelial ovarian cancer cells were assessed through 5-ethynyl-2-deoxyuridine, colony-forming, transwell migration and invasion assays. The relationship between TIPE2 and TGF-β1 was investigated using qRT-PCR and enzyme-linked immunosorbent assay, while the interaction between TIPE2 and Smad2 was identified via co-immunoprecipitation. The results revealed that TIPE2 protein was significantly down-regulated in epithelial ovarian cancer tissues and correlated with the pathological type of tumor, patients' age, tumor differentiation degree and FIGO stage. TIPE2 and TGF-β1 appeared to play an opposite role to each other during the progression of human ovarian cancer cells. Furthermore, TIPE2 inhibited the metastasis and EMT of ovarian cancer cells by combining with Smad2 in vitro or in an intraperitoneal metastasis model. Consequently, these findings suggest that TIPE2 plays a crucial inhibitory role in ovarian cancer metastasis by modulating the TGF-β1/Smad2/EMT signaling pathway and may serve as a potential target for ovarian cancer, providing important direction for future diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Zhongyun Tang
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Derui Zhang
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Jinan Third People's Hospital, Jinan, Shandong, P.R. China
| | - Chenchen Yao
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Mengmeng Jiang
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Chongli Wang
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Zhen Chen
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Huayun Yu
- Department of Gynecology and Obstetrics, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Chenyue Xue
- Department of Gynecology and Obstetrics, Jinan Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Yuqiu Liu
- Department of Gynecology and Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| | - Yongyu Shi
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Lining Zhang
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Xiaoyan Wang
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
| | - Zengtao Wei
- Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, P.R. China
- Department of Gynecology and Obstetrics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, P.R. China
| |
Collapse
|
5
|
Li MP, Long SP, Liu WC, Long K, Gao XH. EMT-related gene classifications predict the prognosis, immune infiltration, and therapeutic response of osteosarcoma. Front Pharmacol 2024; 15:1419040. [PMID: 39170698 PMCID: PMC11335561 DOI: 10.3389/fphar.2024.1419040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024] Open
Abstract
Background Osteosarcoma (OS), a bone tumor with high ability of invasion and metastasis, has seriously affected the health of children and adolescents. Many studies have suggested a connection between OS and the epithelial-mesenchymal transition (EMT). We aimed to integrate EMT-Related genes (EMT-RGs) to predict the prognosis, immune infiltration, and therapeutic response of patients with OS. Methods We used consensus clustering to identify potential EMT-Related OS molecular subtypes. Somatic mutation, tumor immune microenvironment, and functional enrichment analyses were performed for each subtype. We next constructed an EMT-Related risk signature and evaluated it by Kaplan-Meier (K-M) analysis survival and receiver operating characteristic (ROC) curves. Moreover, we constructed a nomogram to more accurately predict OS patients' clinical outcomes. Response effects of immunotherapy in OS patients was analyzed by Tumor Immune Dysfunction and Exclusion (TIDE) analysis, while sensitivity for chemotherapeutic agents was analyzed using oncoPredict. Finally, the expression patterns of hub genes were investigated by single-cell RNA sequencing (scRNA-seq) data analysis. Results A total of 53 EMT-RDGs related to prognosis were identified, separating OS samples into two separate subgroups. The EMT-high subgroup showed favourable overall survival and more active immune response. Significant correlations were found between EMT-Related DEGs and functions as well as pathways linked to the development of OS. Additionally, a risk signature was established and OS patients were divided into two categories based on the risk scores. The signature presented a good predictive performance and could be recognized as an independent predictive factor for OS. Furthermore, patients with higher risk scores exhibited better sensitivity for five drugs, while no significant difference existed in immunotherapy response between the two risk subgroups. scRNA-seq data analysis displayed different expression patterns of the hub genes. Conclusion We developed a novel EMT-Related risk signature that can be considered as an independent predictor for OS, which may help improve clinical outcome prediction and guide personalized treatments for patients with OS.
Collapse
Affiliation(s)
- Meng-Pan Li
- Department of Orthopedics, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- The First Clinical Medical College of Nanchang University, Nanchang, China
| | - Si-Ping Long
- The Fourth Clinical Medical College of Nanchang University, Nanchang, China
| | - Wen-Cai Liu
- Department of Orthopedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun Long
- The First Clinical Medical College of Nanchang University, Nanchang, China
| | - Xing-Hua Gao
- Department of Orthopedics, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, China
| |
Collapse
|
6
|
Jihu Y, Leng R, Liu M, Ren H, Xie D, Yao C, Yan H. Angiotensin (1-7) Inhibits Transforming Growth Factor-Β1-Induced Epithelial-Mesenchymal Transition of Human Keratinocyte Hacat Cells in vitro. Clin Cosmet Investig Dermatol 2024; 17:1049-1058. [PMID: 38737946 PMCID: PMC11088851 DOI: 10.2147/ccid.s441596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 03/24/2024] [Indexed: 05/14/2024]
Abstract
Introduction Angiotensin (1-7) (Ang-(1-7)) is an emerging component of the renin-angiotensin system (RAS) with effective anti-fibrosis properties and has been shown to interfere with epithelial-mesenchymal transition (EMT) by numerous studies. In recent years, EMT has been proposed as a new therapeutic target for skin fibrotic diseases such as keloids. However, the effect of Ang-(1-7) on EMT in skin is still unclear. Hence, the purpose of this study was to explore the effect of Ang-(1-7) on Transforming growth factor-β1(TGF-β1)-induced EMT of human immortalized keratinocytes HaCaT in vitro. Methods The study involved the use of the human immortalized keratinocyte cell line (HaCaT). The cells were cultured in high-glucose DMEM medium with 10% fetal bovine serum and 1% penicillin-streptomycin. Four groups were created for experimentation: control group (Group C), TGF-β1-treated group (Group T), Ang-(1-7)-treated group (Group A), and a group treated with both TGF-β1 and Ang-(1-7) (Group A + T). Various assays were conducted, including a cell proliferation assay using CCK-8 solution, a scratch wound healing assay to evaluate cell migration, and Western blotting to detect protein expressions related to cell characteristics. Additionally, quantitative real-time polymerase chain reaction (PCR) was performed to analyze epithelial-mesenchymal transition (EMT) related gene expression levels. The study aimed to investigate the effects of TGF-β1 and Ang-(1-7) on HaCaT cells. Results We found that Ang-(1-7) not only reduced the migration of HaCaT cells induced by TGF-β1 in vitro but also reduced the expression of α-SMA and vimentin, and restored the protein expression of E-cadherin and claudin-1. Mechanistically, Ang-(1-7) inhibits the phosphorylation levels of Smad2 and Smad3 in the TGF-β1 canonical pathway, and suppresses the expression of EMT-related transcription factors (EMT-TFs) such as SNAI2, TWIST1, and ZEB1. Discussion Taken together, our findings suggest that Ang-(1-7) inhibits TGF-β1-induced EMT in HaCaT cells in vitro by disrupting the TGF-β1-Smad canonical signaling pathway. These results may be helpful in the treatment of EMT in skin fibrotic diseases such as keloids.
Collapse
Affiliation(s)
- Yueda Jihu
- Clinical College of Medicine, Southwest Medical University, Lu zhou, People’s Republic of China
- Department of Plastic and Burn Surgery, the Affiliated Hospital of Southwest Medical University, Lu zhou, People’s Republic of China
| | - Ruobing Leng
- Clinical College of Medicine, Southwest Medical University, Lu zhou, People’s Republic of China
| | - Mengchang Liu
- Clinical College of Medicine, Southwest Medical University, Lu zhou, People’s Republic of China
- Department of Plastic and Burn Surgery, the Affiliated Hospital of Southwest Medical University, Lu zhou, People’s Republic of China
| | - Hongjing Ren
- Clinical College of Medicine, Southwest Medical University, Lu zhou, People’s Republic of China
- Department of Plastic and Burn Surgery, the Affiliated Hospital of Southwest Medical University, Lu zhou, People’s Republic of China
| | - Defu Xie
- Clinical College of Medicine, Southwest Medical University, Lu zhou, People’s Republic of China
- Department of Plastic and Burn Surgery, the Affiliated Hospital of Southwest Medical University, Lu zhou, People’s Republic of China
| | - Chong Yao
- Clinical College of Medicine, Southwest Medical University, Lu zhou, People’s Republic of China
- Department of Plastic and Burn Surgery, the Affiliated Hospital of Southwest Medical University, Lu zhou, People’s Republic of China
| | - Hong Yan
- Clinical College of Medicine, Southwest Medical University, Lu zhou, People’s Republic of China
- Department of Plastic and Burn Surgery, the Affiliated Hospital of Southwest Medical University, Lu zhou, People’s Republic of China
| |
Collapse
|
7
|
Yu R, Lu G, Cheng B, Li J, Jiang Q, Lan X. Construction and validation of a novel NAD + metabolism-related risk model for prognostic prediction in osteosarcoma. J Orthop Res 2024; 42:1086-1103. [PMID: 38047487 DOI: 10.1002/jor.25757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/18/2023] [Accepted: 11/28/2023] [Indexed: 12/05/2023]
Abstract
Currently, the prognosis of osteosarcoma (OS) remains discouraging, especially in elderly/metastatic OS patients. By impairing the antitumor effect of immune cells, tumor immune microenvironment (TIME) provides an environment conducive to tumor proliferation, which highly requires accelerated nicotinamide adenine dinucleotide (NAD+) metabolism for energy. Recently, many genes involved in the sustained production of NAD+ in malignant tumors have been verified to be possible prognostic indicators and therapeutic targets. Therefore, the current study was to probe into the association of NAD+ metabolism-related genes with TIME, immunotherapeutic response, and prognosis in OS. All OS data for the study were acquired from TARGET and GEO databases. In bioinformatics analysis, we performed Cox analysis, consensus clustering, principal component analysis, t-distributed stochastic neighbor embedding, uniform manifold approximation and projection, gene set enrichment analysis, gene set variation analysis, Lasso analysis, survival and ROC curves, nomogram, immune-related analysis, drug sensitivity analysis, and single-cell RNA sequencing (scRNA-seq) analysis. Cell transfection assay, RT-qPCR, western blot analysis, as well as cell wound healing, migration, and invasion assays were performed in vitro. Bioinformatics analysis identified A&B clusters and six NAD+ metabolism-related differentially expressed genes, constructed risk model and nomogram, and performed immune-related analysis, drug susceptibility analysis, and scRNA-seq analysis to inform the clinical treatment framework. In vitro experiment revealed that CBS and INPP1 can promote migration, proliferation as well as invasion of OS cells through TGF-β1/Smad2/3 pathway. Based on bioinformatics analysis and in vitro validation, this study confirmed that NAD+ metabolism affects TIME to suggest the prognosis of OS.
Collapse
Affiliation(s)
- Ronghui Yu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Gang Lu
- Department of Orthopedics, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Banghong Cheng
- Department of Cardiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Junhong Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qiqing Jiang
- Department of Orthopedics, Jiangxi Provincial Children's Hospital, Nanchang, Jiangxi, China
| | - Xia Lan
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
8
|
Hong X, Liu X, Li B, Shi S, Xiao K, Xu T, Nie Y, Dai M, Zhu M. Glaucocalyxin A delays the progression of OA by inhibiting NF-κB and MAPK signaling pathways. J Orthop Surg Res 2024; 19:188. [PMID: 38500177 PMCID: PMC10949665 DOI: 10.1186/s13018-024-04640-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 02/21/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a common degenerative joint condition marked by inflammation and cartilage breakdown. Currently, there is a dearth of treatment medications that can clearly slow the course of OA. Glaucocalyxin A (GLA) is a diterpene chemical identified and extracted from Rabdosia japonica with antithrombotic, anticoagulant, anti-tumor, anti-inflammatory, anti-oxidant, and other pharmacological properties. Previous research has linked inflammation to abnormalities in the homeostasis of the extracellular matrix (ECM). Although GLA has been shown to have anti-inflammatory qualities, its effects on the progression of OA are unknown. As a result, the goal of this study was to see if GLA could slow the course of OA. METHODS ATDC5 cells were stimulated by IL-1β to create an inflammatory chondrocyte damage model. Quantitative polymerase chain reaction, Western Blot, high-density culture, and immunofluorescence were used to detect the expression levels of associated gene phenotypes. We also created a mouse model of OA induced by destabilization of the medial meniscus (DMM) instability, and GLA was administered intraperitoneally once every two days for eight weeks. Mice knee specimens were stained with hematoxylin-eosin, Safranin O/fast green, and immunohistochemical, and the Osteoarthritis Research Society International grade system and Mankin's score were used to assess the protective effect of GLA on cartilage. RESULTS In vitro and in vivo, we explored the effects and molecular processes of GLA as a therapy for OA. The findings demonstrated that GLA might reduce the expression of associated inflammatory mediators and protect the ECM by inhibiting the NF-κB and MAPK signaling pathways. Animal research revealed that GLA could protect against the DMM-induced OA model mice by stabilizing ECM. CONCLUSION Taken together, our findings show that GLA has a protective impact on cartilage throughout OA progression, implying that GLA could be employed as a possible therapeutic agent for OA, thus giving a new therapeutic method for the treatment of OA.
Collapse
Affiliation(s)
- Xin Hong
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Province's Artificial Joints Engineering and Technology Research Center, Nanchang, 330006, Jiangxi Province, China
| | - Xuqiang Liu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Province's Artificial Joints Engineering and Technology Research Center, Nanchang, 330006, Jiangxi Province, China
| | - Bo Li
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Province's Artificial Joints Engineering and Technology Research Center, Nanchang, 330006, Jiangxi Province, China
| | - Shoujie Shi
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Province's Artificial Joints Engineering and Technology Research Center, Nanchang, 330006, Jiangxi Province, China
| | - Kai Xiao
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Province's Artificial Joints Engineering and Technology Research Center, Nanchang, 330006, Jiangxi Province, China
| | - Tiantian Xu
- Department of Pharmacy, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Yaoyang Nie
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Province's Artificial Joints Engineering and Technology Research Center, Nanchang, 330006, Jiangxi Province, China
| | - Min Dai
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Province's Artificial Joints Engineering and Technology Research Center, Nanchang, 330006, Jiangxi Province, China.
| | - Meisong Zhu
- Orthopedic Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Province's Artificial Joints Engineering and Technology Research Center, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
9
|
Takeda T, Tsubaki M, Genno S, Tomita K, Nishida S. RANK/RANKL axis promotes migration, invasion, and metastasis of osteosarcoma via activating NF-κB pathway. Exp Cell Res 2024; 436:113978. [PMID: 38382805 DOI: 10.1016/j.yexcr.2024.113978] [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: 01/24/2024] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 02/23/2024]
Abstract
Osteosarcoma (OS) is one of the most prevalent primary bone tumors with a high degree of metastasis and poor prognosis. Epithelial-to-mesenchymal transition (EMT) is a cellular mechanism that contributes to the invasion and metastasis of cancer cells, and OS cells have been reported to exhibit EMT-like characteristics. Our previous studies have shown that the interaction between tumor necrosis factor superfamily member 11 (TNFRSF11A; also known as RANK) and its ligand TNFSF11 (also known as RANKL) promotes the EMT process in breast cancer cells. However, whether the interaction between RANK and RANKL enhances aggressive behavior by inducing EMT in OS cells has not yet been elucidated. In this study, we showed that the interaction between RANK and RANKL increased the migration, invasion, and metastasis of OS cells by promoting EMT. Importantly, we clarified that the RANK/RANKL axis induces EMT by activating the nuclear factor-kappa B (NF-κB) pathway. Furthermore, the NF-κB inhibitor dimethyl fumarate (DMF) suppressed migration, invasion, and EMT in OS cells. Our results suggest that the RANK/RANKL axis may serve as a potential tumor marker and promising therapeutic target for OS metastasis. Furthermore, DMF may have clinical applications in the treatment of lung metastasis in patients with OS.
Collapse
Affiliation(s)
- Tomoya Takeda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, Osaka, 577-8502, Japan
| | - Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, Osaka, 577-8502, Japan
| | - Shuji Genno
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, Osaka, 577-8502, Japan
| | - Kana Tomita
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, Osaka, 577-8502, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka, Osaka, 577-8502, Japan.
| |
Collapse
|
10
|
Fang L, Wang B, Yang Z, Zhao T, Hao W. PNO1 promotes the progression of osteosarcoma via TGF-β and YAP/TAZ pathway. Sci Rep 2023; 13:21827. [PMID: 38071381 PMCID: PMC10710495 DOI: 10.1038/s41598-023-49295-8] [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: 06/19/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
This study aimed to explore the potential role and mechanisms of the partner of NOB1 homolog (PNO1) in osteosarcoma. The expression of PNO1 in tumor and adjacent tissue samples was examined using western blotting. Lentiviral transfection was used to establish sh-Ctrl and sh-PNO1 osteosarcoma cell lines. MTT assay, Celigo cell cytometer count, and cell colony formation assay were used to investigate the proliferation of osteosarcoma cells in vitro, whereas xenotransplantation assay was performed for in vivo experiments. Wound-healing and Transwell assays were chosen to verify the migration and invasion of osteosarcoma cells. Flow cytometry assay and caspase-3/7 activity analysis were adopted for the analysis of cell apoptosis and cell cycle. Finally, transcriptome sequencing and bioinformatics analysis were adopted to explore the acting mechanisms. The expression of PNO1 was higher in osteosarcoma tissues than that in adjacent tissues. Down-regulation of PNO1 inhibited the proliferation, migration, and invasion, and induced cell apoptosis and cell cycle arrest of osteosarcoma cells. Furthermore, according to transcriptome sequencing and Kyoto Encyclopedia of Genes and Genomes pathway analysis, we found that PNO1 might affect the progression of osteosarcoma via TGF-β and YAP/TAZ signaling pathways. PNO1 could be a potential target for osteosarcoma treatment.
Collapse
Affiliation(s)
- Long Fang
- Department of Bone and Soft Tissue Tumors, Shandong Provincial Third Hospital, Shandong University, Jinan, 250000, China
| | - Baolong Wang
- Department of Bone and Soft Tissue Tumors, Shandong Provincial Third Hospital, Shandong University, Jinan, 250000, China
| | - Zengkun Yang
- Department of Bone and Soft Tissue Tumors, Shandong Provincial Third Hospital, Shandong University, Jinan, 250000, China
| | - Tingbao Zhao
- Department of Bone and Soft Tissue Tumors, Shandong Provincial Third Hospital, Shandong University, Jinan, 250000, China
| | - Wei Hao
- Department of Orthopedics and Traumatology, Shandong Provincial Third Hospital, Shandong University, Jinan, 250000, China.
| |
Collapse
|
11
|
Huang Z, Zhang C, Zhu K, Hu J, Xu E, Ma X, Wang Y, Zhu Y, Zhu J. (E)-SIS3 suppressed osteosarcoma progression via promoting cell apoptosis, arresting cell cycle, and regulating the tumor immune microenvironment. Drug Dev Res 2023; 84:1751-1763. [PMID: 37784254 DOI: 10.1002/ddr.22120] [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: 05/18/2023] [Revised: 09/10/2023] [Accepted: 09/22/2023] [Indexed: 10/04/2023]
Abstract
Osteosarcoma is a prevalent malignant bone tumor with a poor prognosis. Mothers against decapentaplegic homolog 3 (Smad3) present as a therapeutic target in antitumor treatment, whereas its functions in the osteosarcoma have not been well explored. In the current study, we aimed to investigate the effects of Smad3 in the progression of osteosarcoma. The tumor immune single-cell hub 2 website was used for graph-based visualization of Smad3 status in osteosarcoma single-cell database. Western Blot was applied to detect the expression of Smad3 protein in cell lines. Colony formation and cell counting kit-8 assays were used to evaluate cell proliferation. Transwell and wound healing assays were used to detect the migration and invasion abilities of cells. Cell apoptosis rates and cell cycle changes were explored by using flow cytometry analysis. The xenograft tumor growth model was applied to explore the effect in tumor growth after Smad3 blockage in vivo. Moreover, to confirm the potential mechanism of Smad3's effects on osteosarcoma, bioinformatics analysis was performed in TARGET-Osteosarcoma and GSE19276 databases. Our study found that the Smad3 protein is overexpressed in 143B and U2OS cells, suppressing the expression of Smad3 protein in osteosarcoma cells by Smad3 target inhibitor (E)-SIS3 or lentivirus can inhibit the proliferation, migration, invasion, promote cell apoptosis, arrest cell G1 cycle in osteosarcoma cells in vitro, and suppress tumor growth in vivo. Furthermore, the bioinformatics analysis demonstrated that high expression of Smad3 is closely associated with low immune status in TARGET-Osteosarcoma and GSE19276 databases. Our study suggested that Smad3 could contribute positively to osteosarcoma progression via the regulation of tumor immune microenvironment, and Smad3 may represent as an valuable potential therapeutic target in osteosarcoma therapy.
Collapse
Affiliation(s)
- Zhen Huang
- Department of Orthopaedic, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunlin Zhang
- Department of Orthopaedic, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kunpeng Zhu
- Department of Orthopaedic, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jianping Hu
- Department of Orthopaedic, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Enjie Xu
- Department of Orthopaedic, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaolong Ma
- Department of Orthopaedic, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yongjie Wang
- Department of Orthopaedic, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yurun Zhu
- Department of Orthopaedic, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiazhuang Zhu
- Department of Orthopaedic, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
12
|
Nirala BK, Yamamichi T, Petrescu DI, Shafin TN, Yustein JT. Decoding the Impact of Tumor Microenvironment in Osteosarcoma Progression and Metastasis. Cancers (Basel) 2023; 15:5108. [PMID: 37894474 PMCID: PMC10605493 DOI: 10.3390/cancers15205108] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/29/2023] Open
Abstract
Osteosarcoma (OS) is a heterogeneous, highly metastatic bone malignancy in children and adolescents. Despite advancements in multimodal treatment strategies, the prognosis for patients with metastatic or recurrent disease has not improved significantly in the last four decades. OS is a highly heterogeneous tumor; its genetic background and the mechanism of oncogenesis are not well defined. Unfortunately, no effective molecular targeted therapy is currently available for this disease. Understanding osteosarcoma's tumor microenvironment (TME) has recently gained much interest among scientists hoping to provide valuable insights into tumor heterogeneity, progression, metastasis, and the identification of novel therapeutic avenues. Here, we review the current understanding of the TME of OS, including different cellular and noncellular components, their crosstalk with OS tumor cells, and their involvement in tumor progression and metastasis. We also highlight past/current clinical trials targeting the TME of OS for effective therapies and potential future therapeutic strategies with negligible adverse effects.
Collapse
Affiliation(s)
| | | | | | | | - Jason T. Yustein
- Aflac Cancer and Blood Disorders Center, Emory University, Atlanta, GA 30322, USA; (B.K.N.); (T.Y.); (D.I.P.); (T.N.S.)
| |
Collapse
|
13
|
Qu R, Zhang W, Ma Z, Ma Q, Chen M, Lan T, Zhou L, Hu X. Glaucocalyxin A attenuates carbon tetrachloride-induced liver fibrosis and improves the associated gut microbiota imbalance. Chem Biol Drug Des 2023; 102:51-64. [PMID: 37060267 DOI: 10.1111/cbdd.14241] [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: 12/10/2022] [Revised: 02/14/2023] [Accepted: 03/31/2023] [Indexed: 04/16/2023]
Abstract
Liver fibrosis refers to the pathophysiological process of dysplasia on the connective tissue of the liver, caused by a variety of pathogenic factors. Glaucocalyxin A (GLA) has anticoagulation, antibacterial, anti-inflammation, antioxidant and antitumour properties. However, whether GLA ameliorates liver fibrosis or not is still unclear. In this study, a liver fibrosis model was established using male C57BL/6 mice. The mice were treated with 5 and 10 mg/kg GLA via intraperitoneal injection, respectively. The ones that were treated with 5 mg/kg OCA were used as the positive control group. The levels of liver function, liver fibrosis biomarkers and liver pathological changes were then evaluated. We also explored the effects of GLA on inflammatory response and liver cell apoptosis. In addition, we investigated the gut microbiota mechanisms of GLA on liver fibrosis. The results from this study that GLA could significantly decrease the level of liver function (AST, ALT, TBA) and liver fibrosis (HA, LN, PC-III, IV-C). On the other hand, a significant decrease in inflammation levels (IL-1β, TNF-α) were also noted. GLA also improves CCl4-induced pathological liver injuries and collagen deposition, in addition to decreasing apoptosis levels. In addition, an increase in the ratio of Bacteroidetes and Firmicutes in liver disease was also observed. GLA also improves the gut microbiota. In conclusion, GLA attenuates CCl4-induced liver fibrosis and improves the associated gut microbiota imbalance.
Collapse
Affiliation(s)
- Ru Qu
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wang Zhang
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zhuang Ma
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Qianwen Ma
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Mingju Chen
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Tian Lan
- Traditional Chinese Medicine Research Institute, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lin Zhou
- School of Life Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xuguang Hu
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
14
|
Regulation of the Epithelial to Mesenchymal Transition in Osteosarcoma. Biomolecules 2023; 13:biom13020398. [PMID: 36830767 PMCID: PMC9953423 DOI: 10.3390/biom13020398] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
The epithelial to mesenchymal transition (EMT) is a cellular process that has been linked to the promotion of aggressive cellular features in many cancer types. It is characterized by the loss of the epithelial cell phenotype and a shift to a more mesenchymal phenotype and is accompanied by an associated change in cell markers. EMT is highly complex and regulated via multiple signaling pathways. While the importance of EMT is classically described for carcinomas-cancers of epithelial origin-it has also been clearly demonstrated in non-epithelial cancers, including osteosarcoma (OS), a primary bone cancer predominantly affecting children and young adults. Recent studies examining EMT in OS have highlighted regulatory roles for multiple proteins, non-coding nucleic acids, and components of the tumor micro-environment. This review serves to summarize these experimental findings, identify key families of regulatory molecules, and identify potential therapeutic targets specific to the EMT process in OS.
Collapse
|
15
|
Wang J, Gong M, Xiong Z, Zhao Y, Xing D. ADAM19 and TUBB1 Correlate with Tumor Infiltrating Immune Cells and Predicts Prognosis in Osteosarcoma. Comb Chem High Throughput Screen 2023; 26:135-148. [PMID: 35388751 DOI: 10.2174/1386207325666220406112305] [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: 10/11/2021] [Revised: 01/19/2022] [Accepted: 01/29/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Osteosarcoma is the most common type of primary malignant bone tumor. INTRODUCTION This study aimed to explore potential key prognostic genes and their roles in osteosarcoma. METHODS Three microarray datasets for osteosarcoma were downloaded from the GEO database. Differentially expressed genes (DEGs) were screened by the Limma package. Functional enrichment analysis was performed based on DAVID, GeneMANIA, and Metascape databases. Prognostic value of DEGs was elevated by survival analysis. CIBERSORT was used to assess the infiltrating abundance of 22 immune cells, followed by the Pearson correlation analysis between immune cells and prognosis-related genes. Gene set enrichment analysis and drug-gene interactions prediction were performed for prognosis-related genes. RESULTS A total of 8 common up-regulated DEGs and 13 common down-regulated DEGs were screened in the GSE36001 and GSE56001 datasets. Enrichment analysis showed these DEGs were implicated in platelet activation, SMAD protein phosphorylation, lymphocyte/leukocyte/T cells activation, and cell migration. Survival analysis indicated that elevated expression of ADAM19 and TUBB1 were associated with a favorable prognosis. CIBERSORT algorithm revealed the higher infiltrating level of CD8 T cells, macrophages M0, and M2 in osteosarcoma. ADAM19 expression positively correlated with naïve B cells and negatively correlated with activated dendritic cells infiltrating abundance. TUBB1 expression positively correlated with gamma delta T cells while negatively correlated with helper follicular T cells infiltrating abundance. A total of 56 drugs were found to target TUBB1. CONCLUSION ADAM19 and TUBB1 could be prognostic biomarkers in osteosarcoma. Both their expression correlates with tumor infiltrating immune cells. TUBB1 was a multi-drug target that might be a therapeutic target in osteosarcoma.
Collapse
Affiliation(s)
- Jun Wang
- Department of Orthopedics and Trauma, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Mingzhi Gong
- Department of Orthopedics and Trauma, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Zhenggang Xiong
- Department of Orthopedics and Trauma, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Yangyang Zhao
- Department of Orthopedics and Trauma, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| | - Deguo Xing
- Department of Orthopedics and Trauma, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250033, China
| |
Collapse
|
16
|
Wang G, Zhou X, Guo Z, Huang N, Li J, Lv Y, Han L, Zheng W, Xu D, Chai D, Li H, Li L, Zheng J. The Anti-fibrosis drug Pirfenidone modifies the immunosuppressive tumor microenvironment and prevents the progression of renal cell carcinoma by inhibiting tumor autocrine TGF-β. Cancer Biol Ther 2022; 23:150-162. [PMID: 35130111 PMCID: PMC8824226 DOI: 10.1080/15384047.2022.2035629] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Transforming growth factor-β (TGF-β) plays a critical role in regulating cell growth and differentiation. Epithelial to mesenchymal transition (EMT) induced by TGF-β promotes cancer cell migration, invasion, and proliferation. Pirfenidone (5-methyl-1-phenyl-2(1 H)-pyridone, PFD), an approved drug for treating pulmonary and renal fibrosis, is a potent TGF-β inhibitor and found reduced incidence of lung cancer and alleviated renal function decline. However, whether PFD plays a role in controlling renal cancer progression is largely unknown. In the present study, we demonstrated that high TGF-β1 expression was negatively associated with ten-year overall survival of patients with renal cancer. Functionally, blockade of TGF-β signaling with PFD significantly suppressed the progression of renal cancer in a murine model. Mechanistically, we revealed that PFD significantly decreased the expression and secretion of TGF-β both in vitro and in vivo tumor mouse model, which further prevented TGF-β-induced EMT and thus cell proliferation, migration, and invasion. Importantly, the downregulation of TGF-β upon PFD treatment shaped the immunosuppressive tumor microenvironment by limiting the recruitment of tumor-infiltrating MDSCs. Therefore, our study demonstrated that PFD prevents renal cancer progression by inhibiting TGF-β production of cancer cells and downstream signaling pathway, which might be presented as a therapeutic adjuvant for renal cancer.
Collapse
Affiliation(s)
- Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaowan Zhou
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zengli Guo
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Nan Huang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Juan Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yanfang Lv
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Lulu Han
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Wei Zheng
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dandan Xu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Huizhong Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Liantao Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Junnian Zheng
- Center of Clinical Oncology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
17
|
Jin X, Yin H, Bao J, Song X, Lu F, Liang J. ML792 inhibits growth and TGF-β1-induced EMT of osteosarcoma cells via TGF-β1/Smad and PI3K/AKT pathways. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2154856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Xiangang Jin
- Taizhou Hospital of Zhejiang Procince affiliated to Wenzhou Medical University, Linhai City, People’s Republic of China
| | - Hua Yin
- Taizhou Hospital of Zhejiang Procince affiliated to Wenzhou Medical University, Linhai City, People’s Republic of China
| | - Jiaqian Bao
- Taizhou Hospital of Zhejiang Procince affiliated to Wenzhou Medical University, Linhai City, People’s Republic of China
| | - Xiaoting Song
- Taizhou Hospital of Zhejiang Procince affiliated to Wenzhou Medical University, Linhai City, People’s Republic of China
| | - Feng Lu
- Taizhou Hospital of Zhejiang Procince affiliated to Wenzhou Medical University, Linhai City, People’s Republic of China
| | - Junbo Liang
- Taizhou Hospital of Zhejiang Procince affiliated to Wenzhou Medical University, Linhai City, People’s Republic of China
| |
Collapse
|
18
|
Li H, Xue S, Zhang X, Li F, Bei S, Feng L. CircRNA PVT1 modulated cell migration and invasion through Epithelial-Mesenchymal Transition (EMT) mediation in gastric cancer through miR-423-5p/Smad3 pathway. Regen Ther 2022; 21:25-33. [PMID: 35663842 PMCID: PMC9133701 DOI: 10.1016/j.reth.2022.02.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 01/05/2022] [Accepted: 02/19/2022] [Indexed: 02/07/2023] Open
Abstract
Background Gastric cancer (GC) progression is related with gene regulations. Objectives This study explored underlying regulatory axis of circRNA PVT1 (circPVT1) in GC. Methods GC cell lines were detected for circPVT1 expression with the normal mucous epithelial cell GES-1 as control. After regulation of circPVT1, miR-423-5p and SMAD3 expression through transfection, CCK8 evaluated the cell viability, Transwell measured the migratory and invasive capability of cells. Luciferase verified the paired bindings between miR-423-5p and CircPVT1 or SMAD3. The functions of CircPVT1/miR-423-5p/SMAD3 were evaluated using RT-PCR, CCK8, Transwell assays. Western blot analyzed EMT-related proteins and phosphorylation of Smad3 in GC cells. Immunofluorescence method was used to evaluate the EMT-related proteins as well. Results CircPVT1 displayed higher expression in GC cells and knockdown led to decrease in cell growth, invasion and migration. CircPVT1 was targeted by miR-423-5p as a ceRNA of SMAD3. miR-423-5p upregulation suppressed both cicRNA PVT1 and SMAD3 in GC cells. Decrease in SMAD3 expression suppressed CircPVT1 by releasing miR-423-5p in cells, inhibiting cell growth, invasion and migration and suppressing the EMT process. Conclusion CircPVT1 modulated cell growth, invasion and migration through EMT mediation in gastric cancer through miR-423-5p/Smad3 pathway.
Collapse
|
19
|
Han X, Zhang Y, Lu F, Feng J, Zhang C, Wang G. Hypermethylated PODN represses the progression of osteosarcoma by inactivating the TGF-β/Smad2/3 pathway. Pathol Res Pract 2022; 238:154075. [PMID: 36037657 DOI: 10.1016/j.prp.2022.154075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/10/2022] [Accepted: 08/10/2022] [Indexed: 10/15/2022]
Abstract
BACKGROUND PODN is reported to be an promising biomarker for prognosis of osteosarcoma (OS), while the specific function of PODN has not been explored in OS. This study is designed to explore the function and underlying mechanism of PODN in OS. METHODS The mRNA expression of PODN was determined using qRT-PCR. Protein levels of PODN, DNMT1, DNMT3A, DNMT3B, TGF-β1, Smad2/3 and p-Smad2/3 were detected using western blot. The methylation of PODN was determined with methylation-specific PCR. Moreover, CCK-8 assay and colony formation assay were used for assessing the proliferation of OS cells. Transwell assay was used to evaluate migration and invasion abilities of OS cells. Immunohistochemical staining was performed to determine the protein expression of Ki67 and PODN in tumor tissues. For constructing a xenograft tumor model, MG-63 cells were introduced into the right side of the mouse back via subcutaneous injection. RESULTS PODN was lowly expressed and was hypermethylated in OS tissues and cells. PODN overexpression prevented OS cells from proliferating, migrating and invading, and inhibited tumorigenesis in xenograft mice. After PODN overexpression, protein levels of TGF-β1 and p-Smad2/3 were decreased in OS cells. Meantime, the suppressive effects of PODN overexpression on proliferation, migration and invasion of OS cells as well as mouse tumorigenesis were partly counteracted by TGF-β1 overexpression. CONCLUSIONS PODN overexpression inactivated the TGF-β/Smad2/3 pathway to suppress OS development in vitro and in vivo.
Collapse
Affiliation(s)
- Xiuxin Han
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yan Zhang
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Feng Lu
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jinyan Feng
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Chao Zhang
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Guowen Wang
- Department of Bone and Soft Tissue Tumors, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| |
Collapse
|
20
|
Wan R, Yang G, Liu Q, Fu X, Liu Z, Miao H, Liu H, Huang W. PKIB involved in the metastasis and survival of osteosarcoma. Front Oncol 2022; 12:965838. [PMID: 36072791 PMCID: PMC9441607 DOI: 10.3389/fonc.2022.965838] [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: 06/10/2022] [Accepted: 08/01/2022] [Indexed: 12/03/2022] Open
Abstract
Osteosarcoma is frequently metastasized at the time of diagnosis in patients. However, the underlying mechanism of osteosarcoma metastasis remains poorly understood. In this study, we evaluated DNA methylation profiles combined with gene expression profiles of 21 patients with metastatic osteosarcoma and 64 patients with non-metastatic osteosarcoma from TARGET database and identified PKIB and AIM2 as hub genes related to the metastasis of osteosarcoma. To verify the effects of PKIB on migration and invasion of osteosarcoma, we performed wound-healing assay and transwell assay. The results showed that PKIB significantly inhibited the migration and invasion of osteosarcoma cells, and the Western blot experiments showed that the protein level of E-cad was upregulated and of VIM was downregulated in 143-B cell recombinant expression PKIB. These results indicate that PKIB inhibit the metastasis of osteosarcoma. CCK-8 assay results showed that PKIB promote the proliferation of osteosarcoma. In addition, the Western blot results showed that the phosphorylation level of Akt was upregulated in 143-B cells overexpressing PKIB, indicating that PKIB promotes the proliferation of osteosarcoma probably through signaling pathway that Akt involved in. These results give us clues that PKIB was a potential target for osteosarcoma therapy. Furthermore, combined clinical profiles analysis showed that the expression of AIM2- and PKIB- related risk scores was significantly related to the overall survival of patients with osteosarcoma. Thus, we constructed a nomogram based on AIM2 and PKIB expression–related risk scores for osteosarcoma prognostic assessment to predict the 1-, 2-, 3-, and 5-year overall survival rate of patients with metastatic osteosarcoma, assisting clinicians in the diagnosis and treatment of metastatic osteosarcoma.
Collapse
Affiliation(s)
- Rongxue Wan
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Gu Yang
- Guangdong Innovation Platform for Translation of 3D Printing Application, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
| | - Qianzhen Liu
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Xiaokang Fu
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Zengping Liu
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Huilai Miao
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- The Key Laboratory of Diagnosis and Repair in Liver Injury, Guangdong Medical University, Zhanjiang, China
- *Correspondence: Huilai Miao, ; Huan Liu, ; Wenhua Huang,
| | - Huan Liu
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, China
- National Traditional Chinese Medicine Clinical Research Base, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Huilai Miao, ; Huan Liu, ; Wenhua Huang,
| | - Wenhua Huang
- Orthopaedic Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Guangdong Engineering Research Center for Translation of Medical 3D Printing Application, Guangdong Provincial Key Laboratory of Medical Biomechanics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
- Guangdong Innovation Platform for Translation of 3D Printing Application, Southern Medical University, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China
- *Correspondence: Huilai Miao, ; Huan Liu, ; Wenhua Huang,
| |
Collapse
|
21
|
Chen F, Sun F, Liu X, Shao J, Zhang B. Glaucocalyxin A Inhibits the Malignant Progression of Epithelial Ovarian Cancer by Affecting the MicroRNA-374b-5p/HMGB3/Wnt-β-Catenin Pathway Axis. Front Oncol 2022; 12:955830. [PMID: 35912216 PMCID: PMC9329791 DOI: 10.3389/fonc.2022.955830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 06/20/2022] [Indexed: 12/03/2022] Open
Abstract
Objective Glaucocalyxin A (GLA) is an ent-kaurene diterpenoid from Rabdosia japonica var possessing anti-tumor activity. This study aimed to investigate effects of GLA on epithelial ovarian cancer (EOC) and elucidate underlying mechanisms. Methods The expression of HMGB3 in EOC tissues was analyzed by GEPIA and immunohistochemistry. Cell proliferation was determined using CCK-8 and colony formation assays. Cell invasion, migration, and apoptosis were detected using Transwell, wound healing, and flow cytometry assays, respectively. Interactions between HMGB3 and miRNAs were predicted using ENCORI and validated using a dual-luciferase assay. mRNA expression levels of HMGB3 and miRNAs were measured using qPCR. Protein expression levels of HMGB3, E-cadherin, N-cadherin, Wnt3a,β-catenin, Bcl-2, and Bax were measured by western blotting. A tumor xenograft model was established to validate the efficacy and mechanism of GLA in vivo. Results HMGB3 was upregulated in EOC tissues and cells. GLA dose-dependently inhibited EOC cell proliferation and epithelial-mesenchymal transition (EMT). HMGB3 overexpression promoted proliferation, invasion, migration, and EMT, and suppressed the apoptosis of EOC cells. In addition, miR-374b-5p was targeted by HMGB3, and its overexpression hindered malignant characteristics of EOC cells. HMGB3 overexpression weakened antitumor effects of GLA and miR-374b-5p in EOC cells. Moreover, the Wnt-β-catenin pathway was inhibited by the GLA-mediated miR-374b-5p/HMGB3 axis. In vivo experiments showed that GLA inhibited EOC tumor growth, meanwhile, upregulated the miR-374b-5p level and downregulated the expression of HMGB3, Wnt3a, and β-catenin in tumor tissues. Conclusions GLA suppressed the malignant progression of EOC by regulating the miR-374b-5p/HMGB3/Wnt-β-catenin pathway axis.
Collapse
Affiliation(s)
- Feng Chen
- Department of Gynecology and Obstetrics, Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Xuzhou, China
- Department of Gynecology and Obstetrics, Xuzhou Central Hospital, Xuzhou, China
| | - Fang Sun
- Department of Gynecology and Obstetrics, Xuzhou Central Hospital, Xuzhou, China
| | - Xia Liu
- Department of Pathology, Xuzhou Central Hospital, Xuzhou, China
| | - Jing Shao
- Department of Clinical Laboratory, Xuzhou Central Hospital, Xuzhou, China
| | - Bei Zhang
- Department of Gynecology and Obstetrics, Xuzhou Central Hospital Affiliated to Nanjing University of Chinese Medicine, Xuzhou, China
- Department of Gynecology and Obstetrics, Xuzhou Central Hospital, Xuzhou, China
- *Correspondence: Bei Zhang,
| |
Collapse
|
22
|
Epithelial to Mesenchymal Transition Relevant Subtypes with Distinct Prognosis and Responses to Chemo- or Immunotherapies in Osteosarcoma. J Immunol Res 2022; 2022:1377565. [PMID: 35836470 PMCID: PMC9274235 DOI: 10.1155/2022/1377565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 11/17/2022] Open
Abstract
Objective Currently, clinical classification of osteosarcoma cannot accurately predict the survival outcomes and responses to chemo- or immunotherapies. Our goal was to classify osteosarcoma patients into clinical/biological subtypes based on EMT molecules. Methods This study retrospectively curated the RNA expression profiling of osteosarcoma patients from the TARGET and GSE21257 cohorts. Consensus clustering analyses were conducted in accordance with the expression profiling of prognostic EMT genes derived from univariate analyses. Immunological features were evaluated through immune cell infiltration, immune checkpoint expression, and activity of cancer immunity cycle. Drug sensitivity was estimated with the GDSC database. WGCNA approach was adopted to determine the EMT-derived genes. Following univariate analyses, a multivariate cox regression model was developed and externally verified. Predictive independency was evaluated with uni- and multivariate analyses. GSEA was presented to uncover relevant molecular mechanisms. Results Prognostic EMT genes across osteosarcoma patients were stratified into distinct subtypes, namely, subtypes A and B. Patients in subtype B presented desirable prognosis, high immune activation, and enhanced sensitivity to cisplatin. Meanwhile, patients in subtype A were more sensitive to gemcitabine. In total, 86 EMT-derived hub genes were determined, and an EMT score was conducted for osteosarcoma prognosis. Following external verification, this EMT score was reliably and independently predictive of patients' survival outcomes. Additionally, it was positively linked to steroid biosynthesis. Conclusion Overall, our findings proposed EMT-relevant molecular subtypes and signatures for predicting prognosis and therapeutic responses, contributing to personalized treatment and clinical implication for osteosarcoma.
Collapse
|
23
|
Huang H, Lu Q, Yuan X, Zhang P, Ye C, Wei M, Yang C, Zhang L, Huang Y, Luo X, Luo J. Andrographolide inhibits the growth of human osteosarcoma cells by suppressing Wnt/β-catenin, PI3K/AKT and NF-κB signaling pathways. Chem Biol Interact 2022; 365:110068. [PMID: 35917943 DOI: 10.1016/j.cbi.2022.110068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 07/16/2022] [Accepted: 07/19/2022] [Indexed: 11/18/2022]
Abstract
Osteosarcoma (OS) is an aggressive malignant skeletal tumor characterized by an extremely poor prognosis and a high tendency to recur. The frequently used anti-OS chemotherapy regents are often limited by drug resistance and severe adverse events. It is urgent to develop more effective, tolerable and safe drugs for the treatment of OS. Andrographolide (AG), a diterpenoid lactone isolated from Andrographis paniculata, has been proved to possess anti-tumor activity against several human cancer types. In this current study, we evaluated the inhibitory effect of AG on human OS cells and probed the possible mechanism. We found that AG inhibited the proliferation of human OS cells and blocked cell cycle at G2/M phase. Furthermore, AG impeded the migration and invasion, while promoted the apoptosis of human OS cells. Moreover, we found that AG inhibited OS growth and lung metastasis in orthotopic transplantation model. Mechanistically, we demonstrated that AG suppressed the activity of Wnt/β-catenin, PI3K/AKT and NF-κB signaling pathways. Notably, we validated that AG synergized with the inhibitors of Wnt/β-catenin, PI3K/AKT and NF-κB to suppress the proliferation, migration and invasion of human OS cells. Collectively, our study conclusively demonstrates that AG inhibits the growth of human OS cells, thus, may be a promising candidate for the treatment of OS.
Collapse
Affiliation(s)
- Huakun Huang
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, Chongqing Medical University, 400016, Chongqing, China
| | - Qiuping Lu
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, Chongqing Medical University, 400016, Chongqing, China
| | - Xiaohui Yuan
- Department of Medical Laboratory, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, 441021, Xiangyang, Hubei, China
| | - Ping Zhang
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, Chongqing Medical University, 400016, Chongqing, China
| | - Caihong Ye
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, Chongqing Medical University, 400016, Chongqing, China
| | - Mengqi Wei
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, Chongqing Medical University, 400016, Chongqing, China
| | - Chunmei Yang
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, Chongqing Medical University, 400016, Chongqing, China
| | - Lulu Zhang
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, Chongqing Medical University, 400016, Chongqing, China
| | - Yanran Huang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, 400016, Chongqing, China
| | - Jinyong Luo
- Key Laboratory of Clinical Laboratory Diagnostics, Ministry of Education, Chongqing Medical University, 400016, Chongqing, China.
| |
Collapse
|
24
|
Lin H, Peng J, Zhu T, Xiong M, Zhang R, Lei L. Exosomal miR-4800-3p Aggravates the Progression of Hepatocellular Carcinoma via Regulating the Hippo Signaling Pathway by Targeting STK25. Front Oncol 2022; 12:759864. [PMID: 35756606 PMCID: PMC9214204 DOI: 10.3389/fonc.2022.759864] [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] [Received: 08/17/2021] [Accepted: 05/17/2022] [Indexed: 12/12/2022] Open
Abstract
Background Emerging evidence has shown that exosome microRNAs (miRNAs) regulate the development of hepatocellular carcinoma (HCC). Here, the influences of miR-4800-3p on the progression of HCC were explored. Materials and Methods The expression of miR-4800-3p in the exosome derived by transforming growth factor beta 1 (TGF-β1)-treated HCC cells and the serum exosome isolated from HCC patients were identified by real-time PCR. The effects of TGF-β1 and the influences of Huh7-secreted exosomes and the effects of miR-4800-3p combined with/without STK25 on cell functions were explored using the EdU assay cloning experiments, wound healing assay, and Transwell assay. The corresponding molecular mechanisms were further detected using Western blot and real-time PCR assays. The combination of miR-4800-3p and STK25 was verified by the dual-luciferase and RNA pulldown assays. The influences of miR-4800-3p on the growth and epithelial–mesenchymal transformation (EMT) of implanted tumors were tested in vivo and further confirmed by Western blot. Results The miR-4800-3p expression was highly expressed in both exosomes derived by TGF-β1-treated HCC cells and the serum exosomes of HCC patients. In the cases of treatment with both Huh7-derived exosomes, the level of miR-4800-3p expression was highest, and the treatment of TGF-β1 could greatly promote the proliferation, stemness, migration, and invasion of HCC cells via upregulating the markers of stemness and EMT, including CD44, CD133, OCT4, N-cadherin, E-cadherin, and ZO-1. Similar results could be obtained when miR-4800-3p was overexpressed in HCC cells. Furthermore, downregulation of STK25 expression, a direct target gene of miR-4800-3p, could greatly rescue the malignant biological behaviors aggravated by overexpression of miR-4800-3p. This was achieved by suppressing the expression of CD44, CD133, OCT4, N-cadherin, and PCNA and activating the Hippo pathway while increasing E-cadherin and ZO-1. Similar results were also obtained in vivo that knockdown of miR-4800-3p expression suppressed tumor growth induced by Huh7-derived exosomes by mediating the EMT markers and the Hippo signaling pathway. Conclusion Exosomal miR-4800-3p could accelerate HCC development by regulating the Hippo signal by targeting STK25, which could be used as a new therapeutic target for HCC treatment.
Collapse
Affiliation(s)
- Haoming Lin
- Department of HBP Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jicai Peng
- Department of Emergency, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Taifeng Zhu
- Department of HBP Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Meihong Xiong
- Department of HBP Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Rui Zhang
- Department of HBP Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liming Lei
- Department of Intensive Care Unit of Cardiovascular Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Laboratory of South China Structural Heart Disease, Guangzhou, China
| |
Collapse
|
25
|
Liang H, Li L, Zhu S, Tan J, Yang B, Wang X, Wu G, Xie C, Li L, Liu Z, Li Y, Song H, Chen G, Lin L. MicroRNA-744-5p suppresses tumorigenesis and metastasis of osteosarcoma through the p38 mitogen-activated protein kinases pathway by targeting transforming growth factor-beta 1. Bioengineered 2022; 13:12309-12325. [PMID: 35593122 PMCID: PMC9276001 DOI: 10.1080/21655979.2022.2072619] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma (OS) is the most common malignant bone tumor in children and adolescents. Accumulating evidence has revealed that microRNAs (miRNAs) play a crucial role in the progression of OS. In this study, we found that miR-744-5p was the least expressed miRNA in patients with OS by analyzing GSE65071 from the GENE EXPRESSION OMNIBUS (GEO) database. Through real-time quantitative PCR (qRT-PCR), western blotting, colony formation assay, 5-Ethynyl-2-Deoxyuridine (EdU) incorporation assay, transwell migration, and invasion assays, we demonstrated its ability to inhibit the proliferation, migration, and invasion of OS cells in vitro. According to the luciferase reporter assay, transforming growth factor-β1 (TGFB1) was negatively regulated by miR-744-5p and reversed the effects of miR-744-5p on OS. Subcutaneous tumor-forming animal models and tail vein injection lung metastatic models were used in animal experiments, and it was found that miR-744-5p negatively regulated tumor growth and metastasis in vivo. Furthermore, rescue assays verified that miR-744-5p regulates TGFB1 expression in OS. Further experiments revealed that the p38 MAPK signaling pathway is involved in the miR-744-5p/TGFB1 axis. Generally, this study suggests that miR-744-5p is a negative regulator of TGFB1 and suppresses OS progression and metastasis via the p38 MAPK signaling pathway.
Collapse
Affiliation(s)
- Haofeng Liang
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China.,Department of orthopedics, Huizhou Municipal Central Hospital, Huizhou, Guangdong Province, China
| | - Lin Li
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Shuang Zhu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jianye Tan
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Bingsheng Yang
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xiaoping Wang
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Guofeng Wu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chao Xie
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lutao Li
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zhengwei Liu
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yucong Li
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Haoqiang Song
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Guoli Chen
- Department of Orthopedics, Affiliated Hospital of Putian University, Putian, Fujian Province, China
| | - Lijun Lin
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
26
|
Zhang D, Deng T, Yuan W, Chen T, Jiang S. Glaucocalyxin A induces apoptosis of NSCLC cells by inhibiting the PI3K/Akt/GSK3β pathway. Clin Exp Pharmacol Physiol 2022; 49:797-804. [PMID: 35576104 DOI: 10.1111/1440-1681.13667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 04/27/2022] [Accepted: 05/08/2022] [Indexed: 11/28/2022]
Abstract
Lung cancer is one of the fastest growing malignancies in morbidity and mortality, and current therapies are in general not sufficiently effective for this deadly disease. This study characterizes the anti-cancer effects of Glaucocalyxin A (GLA) and explores the underlying mechanisms using human non-small cell lung carcinoma (NSCLC) cells. First, our data showed that GLA suppressed the viability of cancer cells, while no effect was observed in the normal bronchial epithelial cell Bease 2B cells. Second, GLA inhibited colony formation, induced apoptosis of cancer cells. Third, GLA down-regulated the expression of B-cell lymphoma-2 (Bcl-2) protein, up-regulated the expression of Bcl2-associated X protein (Bax) , and strengthened cleavage of Caspase-3 and poly ADP-ribose polymerase (PARP). Fourth, GLA also diminished mitochondrial membrane potential and inhibited phosphatidylinositol 3-kinase (PI3K)/Akt/ glycogen synthase kinase-3β (GSK3β) pathway. In addition, injection of GLA (20 mg/kg) every two days significantly inhibited A549 xenograft tumor growth, accompanied by increased apoptosis and decreased proliferation. Together, our study provides evidence that the anticancer effect of GLA in NSCLC is mediated by inducing apoptosis through inhibiting PI3K/Akt/GSK3β pathway and suggests that GLA may be used as a promising natural medicine for NSCLC therapy. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- De Zhang
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Ting Deng
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Wa Yuan
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Tongqiang Chen
- School of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Shuping Jiang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China.,School of Basic Medicine, Gannan Medical University, Ganzhou, China.,Key Laboratory of Biomaterials and Bio-fabrication in Tissue Engineering of Jiangxi Province, Ganzhou, China
| |
Collapse
|
27
|
Glaucocalyxin A Attenuates IL-1β-Induced Inflammatory Response and Cartilage Degradation in Osteoarthritis Chondrocytes via Inhibiting the Activation of NF-κB Signaling Pathway. DISEASE MARKERS 2022; 2022:6516246. [PMID: 35256893 PMCID: PMC8898110 DOI: 10.1155/2022/6516246] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/18/2022] [Accepted: 01/26/2022] [Indexed: 11/18/2022]
Abstract
Glaucocalyxin A (GLA) is a bioactive natural compound with anti-inflammatory activity. Herein, the role of GLA in osteoarthritis (OA) was evaluated. Our results demonstrated that the IL-1β-induced inducible nitric oxide synthase (iNOS) and cyclooygenase-2 (COX-2) expression, two enzymes resulting in the release of nitric oxide (NO) and PGE2, were also prevented by GLA in chondrocytes. Moreover, GLA suppressed inflammatory cytokines production in chondrocytes. In addition, the elevated expressions of MMPs and ADAMTSs and the degradation of aggrecan and collagen II were reversed by GLA in chondrocytes. Furthermore, GLA decreased p-p65 level and suppressed the nuclear p65 accumulation in the nucleus of chondrocytes. Collectively, we concluded that GLA attenuated inflammatory response in chondrocytes via NF-κB pathway. These findings suggested that GLA might become an effective agent for OA treatment.
Collapse
|
28
|
Han YL, Luo D, Habaxi K, Tayierjiang J, Zhao W, Wang W, Aikebaier W, Wang L. COL5A2 Inhibits the TGF-β and Wnt/β-Catenin Signaling Pathways to Inhibit the Invasion and Metastasis of Osteosarcoma. Front Oncol 2022; 12:813809. [PMID: 35280775 PMCID: PMC8907856 DOI: 10.3389/fonc.2022.813809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/18/2022] [Indexed: 12/26/2022] Open
Abstract
Osteosarcoma is the most common skeletal malignancy and is the second leading cause of cancer death in adolescents. Its highly aggressive nature and high propensity to metastasize lead to an extremely poor prognosis for patients with osteosarcoma. Therefore, finding a suitable treatment has become a matter of urgency. In this study, we first divided the samples into metastatic and non-metastatic groups using the Target database and obtained 1136 differentially expressed genes (DEGs) using differential analysis. A PPI network was constructed to analyze the network of action relationships among DEGs, and the top 10 genes were derived using the MCC algorithm in Cytoscape software. A risk scoring system for 10 key genes was constructed using the LASSO-COX prognostic risk model, and genes associated with osteosarcoma prognosis were screened based on multifactorial COX. COL5A2 gene was highly expressed in metastatic osteosarcoma and led to a poor prognosis. Furthermore, qRT-PCR and immunofluorescence assays confirmed the high expression of COL5A2 in human osteosarcoma cells. CCK-8 assay and scratch WB was used to determine whether the downregulation of COL5A2 expression inhibits the TGF-β signaling and Wnt/β-Catenin signaling pathways. In this study, we screened COL5A2 for prognostic relevance to osteosarcoma through bioinformatics analysis and demonstrated that COL5A2 inhibited osteosarcoma invasion and metastasis by suppressing the TGF-β signaling and Wnt/β-Catenin signaling pathways.
Collapse
Affiliation(s)
- Yan-Long Han
- Department of Bone and Joint Surgery Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Dan Luo
- Department of Respiratory and Critical Care Medicine, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Kakeng Habaxi
- Department of Bone and Joint Surgery Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Julaiti Tayierjiang
- Department of Bone and Joint Surgery Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Wei Zhao
- Department of Bone and Joint Surgery Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Wei Wang
- Department of Bone and Joint Surgery Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Wumaierjiang Aikebaier
- Department of Bone and Joint Surgery Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Li Wang
- Department of Bone and Joint Surgery Center, People’s Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| |
Collapse
|
29
|
Tang X, Sui X, Weng L, Liu Y. SNAIL1: Linking Tumor Metastasis to Immune Evasion. Front Immunol 2021; 12:724200. [PMID: 34917071 PMCID: PMC8669501 DOI: 10.3389/fimmu.2021.724200] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
The transcription factor Snail1, a key inducer of epithelial-mesenchymal transition (EMT), plays a critical role in tumor metastasis. Its stability is strictly controlled by multiple intracellular signal transduction pathways and the ubiquitin-proteasome system (UPS). Increasing evidence indicates that methylation and acetylation of Snail1 also affects tumor metastasis. More importantly, Snail1 is involved in tumor immunosuppression by inducing chemokines and immunosuppressive cells into the tumor microenvironment (TME). In addition, some immune checkpoints potentiate Snail1 expression, such as programmed death ligand 1 (PD-L1) and T cell immunoglobulin 3 (TIM-3). This mini review highlights the pathways and molecules involved in maintenance of Snail1 level and the significance of Snail1 in tumor immune evasion. Due to the crucial role of EMT in tumor metastasis and tumor immunosuppression, comprehensive understanding of Snail1 function may contribute to the development of novel therapeutics for cancer.
Collapse
Affiliation(s)
- Xiaolong Tang
- Department of Laboratory Medicine, Binzhou Medical University, Binzhou, China
| | - Xue Sui
- Department of Laboratory Medicine, Binzhou Medical University, Binzhou, China
| | - Liang Weng
- Department of Oncology, Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology Hunan Province, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Xiangya Hospital, Central South University, Changsha, China.,Hunan Provincial Clinical Research Center for Respiratory Diseases, Xiangya Hospital, Central South University, Changsha, China.,Institute of Gerontological Cancer Research, National Clinical Research Center for Gerontology, Changsha, China.,Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha, China
| | - Yongshuo Liu
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, China.,Biomedical Pioneering Innovation Center (BIOPIC), Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| |
Collapse
|
30
|
Revealing the role of miRNA-489 as a new onco-suppressor factor in different cancers based on pre-clinical and clinical evidence. Int J Biol Macromol 2021; 191:727-737. [PMID: 34562537 DOI: 10.1016/j.ijbiomac.2021.09.089] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 01/17/2023]
Abstract
Recently, microRNAs (miRNAs) have shown to be potential therapeutic, diagnostic and prognostic targets in disease therapy. These endogenous non-coding RNAs contribute to regulation of different cellular events that are necessary for maintaining physiological condition. Dysregulation of miRNAs is correlated with development of various pathological events such as neurological disorders, cardiovascular diseases, and cancer. miRNA-489 is a new emerging miRNA and studies are extensively investigating its role in pathological conditions. Herein, potential function of miRNA-489 as tumor-suppressor in various cancers is described. miRNA-489 is able to sensitize cancer cells into chemotherapy by disrupting molecular pathways involved in cancer growth such as PI3K/Akt, and induction of apoptosis. The PROX1 and SUZ12 as oncogenic pathways, are affected by miRNA-489 in suppressing metastasis of cancer cells. Wnt/β-catenin as an oncogenic factor ensuring growth and malignancy of tumors is inhibited via miRNA-489 function. For enhancing drug sensitivity of tumors, restoring miRNA-489 expression is a promising strategy. The lncRNAs can modulate miRNA-489 expression in tumors and studies about circRNA role in miRNA-489 modulation should be performed. The expression level of miRNA-489 is a diagnostic tool for tumor detection. Besides, down-regulation of miRNA-489 in tumors provides unfavorable prognosis.
Collapse
|
31
|
Zhang S, He L, Shang J, Chen L, Xu Y, Chen X, Li X, Jiao Q, Jin S, Hu X, Liang W. Carvacrol Suppresses Human Osteosarcoma Cells via the Wnt/β-Catenin Signaling Pathway. Anticancer Agents Med Chem 2021; 22:1714-1722. [PMID: 34488595 DOI: 10.2174/1871520621666210901111932] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/16/2021] [Accepted: 07/06/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Carvacrol is a monoterpenic phenol extracted from traditional Chinese herbs, including oregano and thyme. Currently, carvacrol has been widely studied for its therapeutic role in central nervous system diseases, liver diseases and digestive system cancer. OBJECTIVE However, the role of carvacrol in osteosarcoma and its underlying molecular mechanism remain elusive. Here, we aimed to examine the anticancer effects of carvacrol on osteosarcoma. METHODS The effects of carvacrol on the osteosarcoma proliferation capacity were revealed by CCK-8 and colony formation assays. Flow cytometry and Hoechst assays were used to determine the effects of carvacrol on osteosarcoma cell apoptosis. The effect of carvacrol on migration and invasion of osteosarcoma cells was determined by wound healing and transwell tests. Protein expression was evaluated by WB assays. The suppressive effects of carvacrol on osteosarcoma in vivo were examined by a xenograft animal model, immunohistochemistry and HE staining. RESULTS We demonstrated that carvacrol treatment reduced viability and inhibited the colony formation of U2OS and 143B cells in a concentration-dependent manner. Apoptotic cell number increased after exposure to carvacrol. Meanwhile, the expression of Bax increased, and that of Bcl-2 decreased by carvacrol treatment. In addition, the MMP-9 expression and migration and invasion of 143B and U2OS cells were inhibited by carvacrol. We also found that these carvacrol-induced effects on osteosarcoma are associated with the regulation of the Wnt/β-catenin signaling pathway. CONCLUSION Our findings suggest that carvacrol suppresses proliferation, migration, invasion and promotes apoptosis in osteosarcoma cells, in part by regulating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Songou Zhang
- Shaoxing University School of Medicine, No.900 Chennan Avenue, Yuecheng District, Shaoxing 312000, Zhejiang. China
| | - Lei He
- Shaoxing People's Hospital, The First Affiliated Hospital of Shaoxing University, Zhongxing North Road, Shaoxing 312000, Zhejiang. China
| | - Jinxiang Shang
- Shaoxing People's Hospital, The First Affiliated Hospital of Shaoxing University, Zhongxing North Road, Shaoxing 312000, Zhejiang. China
| | - Long Chen
- Shaoxing People's Hospital, The First Affiliated Hospital of Shaoxing University, Zhongxing North Road, Shaoxing 312000, Zhejiang. China
| | - Yifan Xu
- Shaoxing People's Hospital, The First Affiliated Hospital of Shaoxing University, Zhongxing North Road, Shaoxing 312000, Zhejiang. China
| | - Xiaozhen Chen
- Shaoxing University School of Medicine, No.900 Chennan Avenue, Yuecheng District, Shaoxing 312000, Zhejiang. China
| | - Xinyu Li
- Shaoxing People's Hospital, The First Affiliated Hospital of Shaoxing University, Zhongxing North Road, Shaoxing 312000, Zhejiang. China
| | - Qingchuan Jiao
- Shaoxing University School of Medicine, No.900 Chennan Avenue, Yuecheng District, Shaoxing 312000, Zhejiang. China
| | - Songtao Jin
- Shaoxing People's Hospital, The First Affiliated Hospital of Shaoxing University, Zhongxing North Road, Shaoxing 312000, Zhejiang. China
| | - Xujun Hu
- Shaoxing People's Hospital, The First Affiliated Hospital of Shaoxing University, Zhongxing North Road, Shaoxing 312000, Zhejiang. China
| | - Wenqing Liang
- Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, 355 Xinqiao Road, Dinghai District, Zhoushan 316000, Zhejiang Province. China
| |
Collapse
|
32
|
Huang XL, Chen JL, Li XL, Zhao L, Cui YD, Liu JY, Morris-Natschke SL, Masuo G, Cheng YY, Lee KH, Chen DF, Zhang J. Synthesis and in vitro anticancer activities of biotinylated derivatives of glaucocalyxin A and oridonin. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2021; 23:703-711. [PMID: 32441118 DOI: 10.1080/10286020.2020.1760851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 06/11/2023]
Abstract
Fourteen glaucocalyxin A biotinylated derivatives, one glaucocalyxin C biotinylated derivative, and two oridonin biotinylated derivatives were designed and synthesized. Their structures were confirmed from 1H NMR, 13C NMR and HRMS data. The derivatives were evaluated for cytotoxic activities against lung (A549), cervical cancer cell line HeLa derivative (KB), multidrug-resistant KB subline (KB-VIN), triple-negative breast (MDA-MB-231), and estrogen receptor-positive breast (MCF-7) cancer cell lines.[Formula: see text].
Collapse
Affiliation(s)
- Xiao-Lei Huang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Jing-Lei Chen
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Xian-Lun Li
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Lei Zhao
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Ya-Dong Cui
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Jiang-Yun Liu
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| | - Susan L Morris-Natschke
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Goto Masuo
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Yung-Yi Cheng
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Kuo-Hsiung Lee
- Natural Products Research Laboratories, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Chinese Medicine Research and Development Center, China Medical University and Hospital, Taichung 20277, Taiwan
| | - Dao-Feng Chen
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jian Zhang
- College of Pharmaceutical Science, Soochow University, Suzhou 215123, China
| |
Collapse
|
33
|
Cai Y, Wang B, Li B, Huang X, Guo H, Liu Y, Chen B, Zhao S, Wu S, Li W, Wang L, Jia K, Wang H, Chen P, Jiang M, Tang X, Qi H, Dai C, Ye J, He Y. Collection on reports of molecules linked to epithelial-mesenchymal transition in the process of treating metastasizing cancer: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:946. [PMID: 34350261 PMCID: PMC8263858 DOI: 10.21037/atm-20-7002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 04/12/2021] [Indexed: 12/26/2022]
Abstract
Epithelial-mesenchymal transition (EMT) is a morphological process in which epithelial cells transform into mesenchymal cells via a specific procedure. EMT plays an important role in the cancer invasion-metastasis cascade and the current treatment of metastatic cancer, influences the migration, polarity, and adhesion of tumor cells, promotes their migration, invasiveness, anti-apoptotic ability. It contributes to the changes of the tumor microenvironment and suppresses the sensitivity of tumor cells to chemotherapy, causing cancer metastasis and worse, hindering the control and therapy of it. This paper reviews the mechanisms, detection, and treatments of cancer metastasis that have been identified and applied to date, summarizes the EMT-related biological molecules, providing a reference for EMT-targeted research and therapy. As EMT is significant in the progress of tumor metastasis, it is meaningful for the therapy and control of metastatic cancer to understand the mechanism of EMT at the molecular level. We summarized the mechanisms, detection and therapeutic implications of EMT, listed the research progress of molecules like genes, miRNAs, signaling pathways in EMT. We also discussed the prospects of EMT-targeted treatment in cancer metastasis interventions and the challenges the treatment and researches are facing. The summary is conducive to the treatment and further research of EMT and metastatic cancer.
Collapse
Affiliation(s)
- Yiyi Cai
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Boyuan Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Bingying Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Xintong Huang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Haoyue Guo
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Yu Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Bin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Sha Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Shengyu Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Wei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Lei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Keyi Jia
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Peixin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Minlin Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Medical School, Tongji University, Shanghai, China
| | - Xuzhen Tang
- Oncology and Immunology BU, Research Service Division, WuXi Apptec, Shanghai, China
| | - Hui Qi
- Oncology and Immunology BU, Research Service Division, WuXi Apptec, Shanghai, China
| | - Chunlei Dai
- Oncology and Immunology BU, Research Service Division, WuXi Apptec, Shanghai, China
| | - Junyan Ye
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
34
|
Zhang C, Ma K, Yang Y, Wang F, Li W. Glaucocalyxin A suppresses inflammatory responses and induces apoptosis in TNF-a-induced human rheumatoid arthritis via modulation of the STAT3 pathway. Chem Biol Interact 2021; 341:109451. [PMID: 33798506 DOI: 10.1016/j.cbi.2021.109451] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/14/2021] [Accepted: 03/21/2021] [Indexed: 12/12/2022]
Abstract
The pathogenesis of rheumatoid arthritis (RA) is characterized by synoviocyte hyperplasia and proinflammatory cytokine secretion, as well as the destruction of cartilage and bone. Glaucocalyxin A (GLA) is an alkaloid derived from a Chinese medicinal plant that exhibits anti-inflammatory, anti-tumor and neuroprotective properties. We investigated the effects of GLA on RA-fibroblast-like synoviocytes (FLS cells), and collagen-induced arthritis (CIA), and further explored the underlying mechanisms. GLA inhibited TNF-a-induced RA-FLS proliferation, increased apoptotic ratios and upregulated levels of caspase-3, cleaved PARP, and Bax. GLA also inhibited the expression of IL-10, IL-1β, and IL-6 in vitro. Levels of p-STAT3 were downregulated in a dose-dependent manner. Over-expression of STAT3 partly neutralized the GLA-mediated elevation of caspase-3 and cleaved PARP levels as well as the downregulation of IL-10, IL-1B and IL-6 expression levels. This suggests that GLA inactivated the STAT3 pathway. Furthermore, the production of inflammatory cytokines in RA-FLS and a CIA rat model were inhibited effectively by GLA. Taken together, our data suggest that GLA is a potential long-term therapeutic agent for patients with RA.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Apoptosis/drug effects
- Arthritis, Experimental/drug therapy
- Arthritis, Experimental/pathology
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- CD4-Positive T-Lymphocytes/drug effects
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cells, Cultured
- Cytokines/metabolism
- Diterpenes, Kaurane/pharmacology
- Humans
- Inflammation/drug therapy
- Inflammation/metabolism
- Inflammation/pathology
- Male
- Mice, Inbred DBA
- Rats, Wistar
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Synoviocytes/drug effects
- Synoviocytes/metabolism
- Synoviocytes/pathology
- Th17 Cells/drug effects
- Th17 Cells/physiology
- Tumor Necrosis Factor-alpha/pharmacology
- Mice
- Rats
Collapse
Affiliation(s)
- Chuan Zhang
- Orthopedic Hospital of Henan Province & Orthopedic Institute of Henan Province, Luoyang, Henan, 471002, PR China
| | - Kun Ma
- Orthopedic Hospital of Henan Province & Orthopedic Institute of Henan Province, Luoyang, Henan, 471002, PR China
| | - Yanmei Yang
- Orthopedic Hospital of Henan Province & Orthopedic Institute of Henan Province, Luoyang, Henan, 471002, PR China
| | - Fuqiang Wang
- Anyang Cancer Hospital, Anyang, Henan, 455000, PR China
| | - Wuyin Li
- Orthopedic Hospital of Henan Province & Orthopedic Institute of Henan Province, Luoyang, Henan, 471002, PR China.
| |
Collapse
|
35
|
Glaucocalyxin A induces apoptosis and autophagy in tongue squamous cell carcinoma cells by regulating ROS. Cancer Chemother Pharmacol 2021; 88:235-246. [PMID: 33904969 DOI: 10.1007/s00280-021-04285-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/17/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Tongue squamous cell carcinoma (TSCC) is the most common highly invasive oral cancer. Glaucocalyxin A (GLA) is a diterpenoid component isolated from Rabdosia japonica var. with anti-bacterial and anti-cancer biological properties. However, the role of GLA in human TSCC remains uncertain. The aim of this paper was to investigate the anti-cancer effect of GLA on TSCC cells as well as its underlying mechanism. METHODS Cell viability and growth were analyzed by CCK-8 assay and colony formation, respectively. DAPI staining and flow cytometry assay were used to detect the cell apoptosis. Lysotracker Red staining was used to observe the lysosomes and autolysosomes of TSCC cells. ROS fluorescent probe was used to test the intracellular ROS levels. Western blotting was used to detect the expression levels of apoptosis- and autophagy-related proteins. RESULTS GLA inhibits the cell viability and growth in TSCC cells. GLA induces TSCC cells apoptosis, autophagy and ROS production in a time- and concentration-dependent manner. In addition, GLA inhibits the viability of TSCC cells by inducing intracellular ROS production. Finally, GLA triggers ROS-dependent apoptosis and autophagy in TSCC cells. CONCLUSION Our results consistently suggested that GLA can induce apoptosis and autophagy in TSCC cells by generating ROS. GLA may serve as a promising therapeutic drug for overcoming TSCC.
Collapse
|
36
|
Chen J, Zhang W, Pan C, Fan J, Zhong X, Tang S. Glaucocalyxin A induces cell cycle arrest and apoptosis via inhibiting NF-κB/p65 signaling pathway in melanoma cells. Life Sci 2021; 271:119185. [PMID: 33577846 DOI: 10.1016/j.lfs.2021.119185] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 01/26/2021] [Accepted: 01/28/2021] [Indexed: 02/05/2023]
Abstract
AIMS Melanoma is a malignant tumor of the skin with a high metastasis rate and poor prognosis. Glaucocalyxin A (GLA), isolated from Rabdosia japonica, is a diterpenoid compound with anticancer properties. Here, we investigated the anticancer properties and explored the mechanisms underlying GLA activity in melanoma cells in vitro and in vivo. MAIN METHODS Cell Counting Kit-8 and colony formation assays were used to assess the effects of GLA on cell proliferation. Flow cytometry was used to evaluate the cell cycle, apoptosis, mitochondrial membrane potential (MMP), and reactive oxygen species (ROS), and western blot analysis and immunofluorescence staining were used to examine protein expression. Immunohistochemical analysis was performed to examine animal tissues and tumors in mice. KEY FINDINGS GLA could effectively inhibit cell proliferation and induce cell apoptosis. GLA induced an overproduction of cellular ROS, decreased MMP, and upregulated the Bax/Bcl-2 ratio, which is an indicator of apoptosis. Phosphorylation of nuclear factor κB (NF-κB)/p65 and NF-κB/p65 nuclear expression decreased after GLA treatment in vitro and in vivo, suggesting that the anticancer effects of GLA are mediated through the NF-κB/p65 pathway. Moreover, we observed that GLA was effective in inhibiting tumor growth without obvious toxicity to major organs in mice. SIGNIFICANCE This is the first study to show that GLA inhibits cell proliferation, arrests the cell cycle in the G2/M phase, and induces mitochondrial apoptosis via the NF-κB/p65 pathway in melanoma cells. Overall, our results demonstrate that GLA may be a potential anticancer agent for the treatment of melanoma.
Collapse
Affiliation(s)
- Jiasheng Chen
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, PR China
| | - Wancong Zhang
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, PR China
| | - Chen Pan
- Department of Chemistry, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Jufeng Fan
- Department of Plastic and Reconstructive Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, PR China.
| | - Xiaoping Zhong
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, PR China.
| | - Shijie Tang
- Department of Burns and Plastic Surgery, The Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, PR China.
| |
Collapse
|
37
|
Geranpayehvaghei M, Dabirmanesh B, Khaledi M, Atabakhshi-Kashi M, Gao C, Taleb M, Zhang Y, Khajeh K, Nie G. Cancer-associated-platelet-inspired nanomedicines for cancer therapy. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1702. [PMID: 33538125 DOI: 10.1002/wnan.1702] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/09/2020] [Accepted: 01/08/2021] [Indexed: 01/03/2023]
Abstract
Platelets, with hemostasis and thrombosis activities, are one of the key components in the blood circulation. As a guard, they rapidly respond to any abnormal blood vessel injury signal and release their granules' contents, which induce their adhesion and aggregation on wound site for hemostasis. Recently, increasing evidence has indicated that platelets are critically involved in the growth and metastasis of cancer cells by releasing a variety of cytokines and chemokines to stimulate cancer cell proliferation and various angiogenic regulators to accelerate tumor angiogenesis. Platelets also secrete active transforming growth factor beta (TGF-β) to promote the epithelial-mesenchymal transition of cancer cells and their extravasation from primary site, and form microthrombus on the surface of cancer cells to protect them from immune attack and high-speed shear force in the circulation. Therefore, blocking platelet-cancer cell interaction may be an attractive strategy to treat primary tumor and/or prevent cancer metastasis. However, systemic inhibition or depletion of platelets brings risk of severe bleeding complication. Cancer-associated-platelets-targeted nanomedicines and biomimetic nanomedicines coated with platelet membrane can be used for targeted anticancer drug delivery, due to their natural targeting ability to tumor cells and platelets. In the current review, we first summarized the platelet mechanisms of action in physiological condition and their multiple roles in cancer progression and conventional antiplatelet therapeutics. We then highlighted the recent progress on the design and fabrication of cancer-associated-platelet-targeted nanomedicines and platelet membrane coating nanomedicines for cancer therapy. Finally, we discussed opportunities and challenges and offered our thoughts for the future development. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Biology-Inspired Nanomaterials > Lipid-Based Structures.
Collapse
Affiliation(s)
- Marzieh Geranpayehvaghei
- Faculty of Biological Sciences, Department of Nanobiotechnology, Tarbiat Modares University, Tehran, Iran.,CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Bahareh Dabirmanesh
- Faculty of Biological Sciences, Department of Biochemistry, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Khaledi
- Faculty of Biological Sciences, Department of Biochemistry, Tarbiat Modares University, Tehran, Iran
| | - Mona Atabakhshi-Kashi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Chao Gao
- College of Pharmaceutical Science, Jilin University, Changchun, China
| | - Mohammad Taleb
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Khosro Khajeh
- Faculty of Biological Sciences, Department of Nanobiotechnology, Tarbiat Modares University, Tehran, Iran.,Faculty of Biological Sciences, Department of Biochemistry, Tarbiat Modares University, Tehran, Iran
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.,GBA Research Innovation Institute for Nanotechnology, Guangdong, China
| |
Collapse
|
38
|
Deng B, Deng J, Yi X, Zou Y, Li C. ROCK2 Promotes Osteosarcoma Growth and Glycolysis by Up-Regulating HKII via Phospho-PI3K/AKT Signalling. Cancer Manag Res 2021; 13:449-462. [PMID: 33500659 PMCID: PMC7823140 DOI: 10.2147/cmar.s279496] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 10/24/2020] [Indexed: 01/14/2023] Open
Abstract
Background Osteosarcoma (OS) is a malignant bone tumour that exhibits a high mortality. While tumours thrive in a state of malnutrition, the mechanism by which OS cells adapt to metabolic stress through metabolic reprogramming remains unclear. Methods We analysed the expression of ROCK2 in osteosarcoma tissues by RT-qPCR and Western blot. Cell proliferation were analysed using CCK8, EdU and colony formation assays. The level of cell glycolysis was detected by glucose-6 phosphate, glucose consumption, lactate production and ATP levels. Results Herein, our study showed that ROCK2 expression in OS tissues was higher than in adjacent tissues. Functional assays have demonstrated that ROCK2 contributes to the growth of OS cells by inducing aerobic glycolysis. The current study revealed that ROCK2 knockdown decreased the levels of mitochondrial hexokinase II (HKII). And also indicated that ROCK2 served as a key enzyme in glycolysis and that it served an important role in tumour growth. A significant positive correlation was identified between the mRNA and protein expressions of ROCK2 and HKII, further demonstrating that ROCK2-induced glycolysis and proliferation was dependent on HKII expression in OS cells. Mechanistically, ROCK2 promotes HKII expression by activating the phospho-PI3K/AKT signalling pathway. Conclusion Taken together, the results of the current study linked the two drivers of OS growth and aerobic glycolysis and identified a new mechanism of ROCK2 control in OS.
Collapse
Affiliation(s)
- Binbin Deng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Jianyong Deng
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Xuan Yi
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Yeqing Zou
- Jiangxi Province Key Laboratory of Molecular Medicine, Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| | - Chen Li
- Department of Orthopedics, Second Affiliated Hospital of Nanchang University, Nanchang, People's Republic of China
| |
Collapse
|
39
|
Wang L, Huang Y, Liu C, Guo M, Ma Z, He J, Wang A, Sun X, Liu Z. Deltex3 inhibits Epithelial Mesenchymal Transition in Papillary Thyroid Carcinoma via promoting ubiquitination of XRCC5 to regulate the AKT signal pathway. J Cancer 2021; 12:860-873. [PMID: 33403043 PMCID: PMC7778541 DOI: 10.7150/jca.48141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 11/02/2020] [Indexed: 01/23/2023] Open
Abstract
Background: Papillary thyroid carcinoma (PTC) is one of the most common endocrine malignant tumors. Poor prognoses such as high recurrence rate always appear in PTC patients with cervical lymph node metastasis. The process of ubiquitination plays important roles in PTC. As ubiquitin E3 ligases, Deltex (DTX) family proteins were reported to associate with multiple cancers. However, functions and mechanisms of DTX3 in PTC are currently unknown. Methods: In this study, DTX3 expressions were examined in 114 PTC and paired paracancerous normal tissues through quantitative real-time polymerase chain reaction and western blot. The clinical significances of DTX3 expressions in PTC patients were also investigated. After stable transfection with either short hairpin RNA to knock down DTX3 expression or full-length complementary DNA to upregulate DTX3 expression, changes of malignant phenotypes in two PTC cell lines K1 and TPC-1 were observed using cell viability, flow cytometry, wound healing and transwell assays. Afterwards, altered expressions of epithelial-mesenchymal transition (EMT) and AKT signal pathway related proteins were measured by western blot. Immunoprecipitation and mass spectrometry (IP-MS), immunofluorescence and Co-IP were utilized to identify the possible DTX3 interacting proteins. Results: Both mRNA and protein expressions of DTX3 were lower in PTC tissues and correlated with the presence of cervical lymph node metastasis (P<0.05). DTX3 overexpression inhibited migration and invasion of PTC cells, decreased Vimentin and phosphorylated AKT expressions, but promoted E-cadherin expression (P<0.05). Moreover, knockdown of DTX3 led to opposite changes (P<0.05). Total 46 probable DTX3 interacting proteins were identified by IP-MS. Among them, X-ray repair cross-complementing protein 5 (XRCC5) and NADH: Ubiquinone Oxidoreductase Complex Assembly Factor 5 (NDUFAF5) were verified to be associated with DTX3. Moreover, DTX3 was proved to be co-localized with XRCC5 in nucleus and promote ubiquitination of XRCC5. Conclusions: DTX3 suppresses EMT by partially facilitating ubiquitination of XRCC5 to inhibit AKT signal pathway in PTC.
Collapse
Affiliation(s)
- Lidong Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yonglian Huang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chenxi Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Mingyue Guo
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhennan Ma
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jingni He
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ailian Wang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaodan Sun
- Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Zhen Liu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
40
|
Zhou X, Ma W, Li X, Xu J. Glaucocalyxin a prevents hypoxia-induced epithelial-mesenchymal transition in human gastric cancer cells through the PI3K/Akt signaling pathway. J Recept Signal Transduct Res 2020; 42:109-116. [PMID: 33307912 DOI: 10.1080/10799893.2020.1853160] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hypoxia is a frequent occurrence in most solid tumors and associated with multiple cancer progression. Glaucocalyxin A (GLA) has been found to exhibit anti-tumor effect in several types of cancer, except gastric cancer (GC). The present study aimed to evaluate the function of GLA in GC and explore the underlying mechanism under hypoxia condition. Our results showed that GLA suppressed cell viability of MGC-803 cells in both normoxic or hypoxic conditions. MGC-803 cells were more sensitive to GLA in hypoxic condition. GLA attenuated hypoxia-induced migration and invasion of GC cells. Western blot assay proved that GLA elevated E-cadherin expression, as well reduced N-cadherin and vimentin expressions in hypoxia-induced GC cells. Moreover, we also found that GLA suppressed the expression of HIF-1α in both mRNA and protein levels. Furthermore, GLA blocked hypoxia-induced activation of PI3K/Akt pathway in GC cells. Notably, insulin like growth factor 1 (IGF-1), an activator of PI3K/Akt pathway, reversed the effects of GLA on cell migration, invasion and EMT in hypoxia-treated MGC-803 cells. In conclusion, these findings demonstrated that GLA exerted inhibitory effects on cell migration, invasion and epithelial to mesenchymal transition (EMT) via the PI3K/Akt signaling pathway in GC cells.
Collapse
Affiliation(s)
- Xihan Zhou
- Department of Gastroenterology, Wuhan Fifth Hospital, Wuhan, China
| | - Weijin Ma
- Hospital-acquired infection control department, Wuhan Fifth Hospital, Wuhan, China
| | - Xiaohui Li
- Department of Paediatrics, Wuhan Fifth Hospital, Wuhan, China
| | - Jiali Xu
- Department of supervision, Wuhan Fifth Hospital, Wuhan, China
| |
Collapse
|
41
|
Lin Z, Lu S, Xie X, Yi X, Huang H. Noncoding RNAs in drug-resistant pancreatic cancer: A review. Biomed Pharmacother 2020; 131:110768. [PMID: 33152930 DOI: 10.1016/j.biopha.2020.110768] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is the fourth-leading cause of cancer-related deaths and is expected to be the second-leading cause of cancer-related deaths in Europe and the United States by 2030. The high fatality rate of pancreatic cancer is ascribed to untimely diagnosis, early metastasis and limited responses to both chemotherapy and radiotherapy. Although gemcitabine, 5-fluorouracil and some other drugs can profoundly improve patient prognosis, most pancreatic cancer patients eventually develop drug resistance, leading to poor clinical outcomes. The underlying mechanisms of pancreatic cancer drug resistance are complicated and inconclusive. Interestingly, accumulating evidence has demonstrated that different noncoding RNAs (ncRNAs), such as microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs), play a crucial role in pancreatic cancer resistance to chemotherapy reagents. In this paper, we systematically summarize the molecular mechanism underlying the influence of ncRNAs on the generation and development of drug resistance in pancreatic cancer and discuss the potential role of ncRNAs as prognostic markers and new therapeutic targets for pancreatic cancer.
Collapse
Affiliation(s)
- Zhengjun Lin
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China.
| | - Shiyao Lu
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China.
| | - Xubin Xie
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China.
| | - Xuyang Yi
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China.
| | - He Huang
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China; State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, School of Pre-Clinical Medicine/ Second Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China.
| |
Collapse
|
42
|
Wang Z, Li L, Wang C, Piao Y, Jiang J, Li L, Yan G, Piao H. Recombinant Pyrin Domain Protein Attenuates Airway Inflammation and Alleviates Epithelial-Mesenchymal Transition by Inhibiting Crosstalk Between TGFβ1 and Notch1 Signaling in Chronic Asthmatic Mice. Front Physiol 2020; 11:559470. [PMID: 33192556 PMCID: PMC7645102 DOI: 10.3389/fphys.2020.559470] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/15/2020] [Indexed: 01/10/2023] Open
Abstract
This article aims to investigate the effects of recombinant pyrin domain (RPYD) on airway inflammation and remodeling in mice with chronic asthma. The chronic asthma BALB/c mouse model was first sensitized by ovalbumin (OVA) and then challenged by OVA nebulization. RPYD or dexamethasone was given before OVA challenge. Our results showed that RPYD significantly inhibited the increase of total cell number, eosinophils, neutrophils and lymphocytes in bronchoalveolar lavage fluid (BALF) induced by OVA, and reduced the infiltration of inflammatory cells, the proliferation of goblet cells and collagen deposition. In addition, RPYD inhibited the mRNA and protein levels of α-smooth muscle actin (α-SMA), transforming growth factor (TGF)-β1, Jagged1, Notch1, Hes1 and Smad3, as well as Smad3 phosphorylation. TGFβ1 down-regulated the level of E-cadherin and promoted the expression of α-SMA, thus inducing epithelial-mesenchymal transition (EMT) in bronchial epithelial cells. We found that RPYD reduced EMT by inhibiting TGFβ1/smad3 and Jagged1/Notch1 signaling pathways. Further overexpression of NICD showed that under the stimulation of TGFβ1, NICD enhanced the phosphorylated Smad3 and nuclear Smad3, accompanied by the increased expression of Notch1 target gene Hes1. In contrast, after treatment with smad3 siRNA, the expression of Hes1 was down regulated as the decrease of Smad3, which indicates that there is crosstalk between smad3 and NICD on Hes1 expression. In conclusion, RPYD reduces airway inflammation, improves airway remodeling and reduces EMT in chronic asthmatic mice by inhibiting the crosstalk between TGFβ1/smad3 and Jagged1/Notch1 signaling pathways.
Collapse
Affiliation(s)
- Zhiguang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji, China
| | - Liangchang Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, China
| | - Chongyang Wang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, China
| | - Yihua Piao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Intensive Care Unit, Affiliated Hospital of Yanbian University, Yanji, China
| | - Jingzhi Jiang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, China
| | - Li Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, China
| | - Guanghai Yan
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Anatomy, Histology and Embryology, Yanbian University Medical College, Yanji, China
| | - Hongmei Piao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Respiratory Medicine, Affiliated Hospital of Yanbian University, Yanji, China
| |
Collapse
|
43
|
Yang P, Liu Y, Qi YC, Lian ZH. High SENP3 Expression Promotes Cell Migration, Invasion, and Proliferation by Modulating DNA Methylation of E-Cadherin in Osteosarcoma. Technol Cancer Res Treat 2020; 19:1533033820956988. [PMID: 33030103 PMCID: PMC7549150 DOI: 10.1177/1533033820956988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
SENP3, a sentrin/SUMO2/3-specific protease, is recognized as a transcriptional factor that accumulates under cellular oxidative stress and plays a significant role in the removal of SUMO2/3 modification. In our study, we examined a TCGA dataset and found that the transcripts per million (TPM) value of SENP3 is high in sarcoma, including osteosarcoma (OS). We found that SENP3 was highly expressed in OS cancer tissues when compared with osteofibrous dysplasia tissues. The survival data of SENP3 in TCGA showed that the sarcoma patients with higher SENP3 expression levels showed poor prognosis. In vitro, SENP3 knockdown in OS cancer cells inhibited cell proliferation, migration, and invasion and induced apoptosis. In contrast, SENP3 overexpression reversed these effects. Next, we found that SENP3 inhibited the expression of E-cadherin (E-Cad) by increasing methylation of the E-Cad promoter. Finally, E-Cad expression was increased in the OS cell line MG63 following methylation, and the cell proliferation, migration, and invasion capacity were decreased. In summary, SENP3 played a significant role in OS carcinogenesis and may act as a potential biomarker in the diagnosis and treatment of OS.
Collapse
Affiliation(s)
- Pu Yang
- Postdoctoral Research Station of Clinical Medicine & Department of Plastic Surgery, The 3rd Xiangya Hospital, 504354Central South University, Changsha, Hunan, PR China
| | - Yan Liu
- Department of Plastic Surgery, The 3rd Xiangya Hospital, 504354Central South University, Changsha, PR China
| | - Yin Chao Qi
- Department of Plastic Surgery, The 3rd Xiangya Hospital, 504354Central South University, Changsha, PR China
| | - Zhang Hong Lian
- Department of Plastic Surgery, The 3rd Xiangya Hospital, 504354Central South University, Changsha, PR China
| |
Collapse
|
44
|
Xia W, Li Y, Wu Z, Wang Y, Xing N, Yang W, Wu S. Transcription factor YY1 mediates epithelial-mesenchymal transition through the TGFβ signaling pathway in bladder cancer. Med Oncol 2020; 37:93. [PMID: 32970204 DOI: 10.1007/s12032-020-01414-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/30/2020] [Indexed: 12/19/2022]
Abstract
Bladder cancer is one of the most aggressive urothelial tumors. Previous studies have suggested that epithelial-mesenchymal transition (EMT) contributes to bladder cancer progression. However, the regulatory network of EMT in bladder cancer remains elusive. In this study, we found Yin Yang 1 (YY1) is a critical regulator of EMT in bladder cancer. First, we showed that YY1 was upregulated in bladder cancer tissues than that in adjacent normal tissues. Then, we proved that YY1 promoted EMT of bladder cancer cells. Further experiments indicated that YY1 affected the EMT of bladder cancer through transforming growth factor-β (TGFβ) signaling pathway. Taken together, our study identifies YY1 as a key EMT driver in bladder cancer, suggesting it as a potential therapeutic target.
Collapse
Affiliation(s)
- Wuchao Xia
- The Affiliated Shenzhen Luohu Hospital of Anhui University of Science and Technology, Shenzhen, China
- Urology Institute of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yuqing Li
- Urology Institute of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Shenzhen, China
| | - Zhangsong Wu
- Urology Institute of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Yongqiang Wang
- Urology Institute of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Shenzhen, China
| | - Nianzeng Xing
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenzeng Yang
- Affiliated Hospital of Hebei University, Baoding, Hebei, China.
| | - Song Wu
- The Affiliated Shenzhen Luohu Hospital of Anhui University of Science and Technology, Shenzhen, China.
- Urology Institute of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China.
- Department of Urology, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Shenzhen, China.
| |
Collapse
|
45
|
Ma K, Zhang C, Li W. TGF-β is associated with poor prognosis and promotes osteosarcoma progression via PI3K/Akt pathway activation. Cell Cycle 2020; 19:2327-2339. [PMID: 32804027 PMCID: PMC7513842 DOI: 10.1080/15384101.2020.1805552] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Transforming growth factor beta (TGF-β) is a multifunctional cytokine with important functions in cell proliferation and differentiation. TGF-β is highly expressed in several types of cancers and promotes tumor invasion and metastasis. However, the role of TGF-β in osteosarcoma progression is poorly understood. In the present study, we found that TGF-β is highly expressed in osteosarcoma cells and tissues, and is associated with high Ennecking stage (P = 0.033), metastasis, and recurrence. TGF-β-knockdown osteosarcoma cell lines were established using siRNA (si-TGF-β). Cells transfected with si-TGF-β exhibited significantly reduced proliferation, migration/invasion, and colony formation abilities, and increased levels of cell apoptosis. In addition, si-TGF-β treatment reduced spheroid size, the ratio of CD133-positive cells, and expression of SOX-2, Nanog, and Oct-3/4 in osteosarcoma cells. Mechanistically, PI3K/mTOR phosphorylation is inhibited by TGF-β knockdown. Pretreatment with 25 µM LY294002, a PI3K-specific inhibitor, further enhanced the si-TGF-β-induced suppression of osteosarcoma progression. Taken together, these results reveal a novel role for TGF-β in osteosarcoma progression and modulation of stemness-related traits and indicate that TGF-β may be of value as a therapeutic target for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Kun Ma
- Luoyang Orthopaedic Hospital of Henan Province and Orthopaedic Hospital of Henan Province , Luoyang, Henan, P. R. China
| | - Chuan Zhang
- Luoyang Orthopaedic Hospital of Henan Province and Orthopaedic Hospital of Henan Province , Luoyang, Henan, P. R. China
| | - Wuyin Li
- Luoyang Orthopaedic Hospital of Henan Province and Orthopaedic Hospital of Henan Province , Luoyang, Henan, P. R. China
| |
Collapse
|
46
|
Ashrafizadeh M, Hushmandi K, Hashemi M, Akbari ME, Kubatka P, Raei M, Koklesova L, Shahinozzaman M, Mohammadinejad R, Najafi M, Sethi G, Kumar AP, Zarrabi A. Role of microRNA/Epithelial-to-Mesenchymal Transition Axis in the Metastasis of Bladder Cancer. Biomolecules 2020; 10:E1159. [PMID: 32784711 PMCID: PMC7464913 DOI: 10.3390/biom10081159] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022] Open
Abstract
Bladder cancer (BC) is the 11th most common diagnosed cancer, and a number of factors including environmental and genetic ones participate in BC development. Metastasis of BC cells into neighboring and distant tissues significantly reduces overall survival of patients with this life-threatening disorder. Recently, studies have focused on revealing molecular pathways involved in metastasis of BC cells, and in this review, we focus on microRNAs (miRNAs) and their regulatory effect on epithelial-to-mesenchymal transition (EMT) mechanisms that can regulate metastasis. EMT is a vital process for migration of BC cells, and inhibition of this mechanism restricts invasion of BC cells. MiRNAs are endogenous non-coding RNAs with 19-24 nucleotides capable of regulating different cellular events, and EMT is one of them. In BC cells, miRNAs are able to both induce and/or inhibit EMT. For regulation of EMT, miRNAs affect different molecular pathways such as transforming growth factor-beta (TGF-β), Snail, Slug, ZEB1/2, CD44, NSBP1, which are, discussed in detail this review. Besides, miRNA/EMT axis can also be regulated by upstream mediators such as lncRNAs, circRNAs and targeted by diverse anti-tumor agents. These topics are also discussed here to reveal diverse molecular pathways involved in migration of BC cells and strategies to target them to develop effective therapeutics.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran;
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1419963114, Iran;
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran;
| | - Mohammad Esmaeil Akbari
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran 1989934148, Iran;
| | - Peter Kubatka
- Department of Medical Biology and Division of Oncology—Biomedical Center Martin, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Mehdi Raei
- Health Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran 1435916471, Iran;
| | - Lenka Koklesova
- Department of Obstetrics and Gynecology, Martin University Hospital and Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Md Shahinozzaman
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA;
| | - Reza Mohammadinejad
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman 55877577, Iran;
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran;
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore;
- Cancer Science Institute of Singapore, Centre for Translational Medicine, 14 Medical Drive, #11-01M, Singapore 117599, Singapore
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, Istanbul 34956, Turkey
- Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Turkey
| |
Collapse
|
47
|
Ashrafizadeh M, Najafi M, Ang HL, Moghadam ER, Mahabady MK, Zabolian A, Jafaripour L, Bejandi AK, Hushmandi K, Saleki H, Zarrabi A, Kumar AP. PTEN, a Barrier for Proliferation and Metastasis of Gastric Cancer Cells: From Molecular Pathways to Targeting and Regulation. Biomedicines 2020; 8:E264. [PMID: 32756305 PMCID: PMC7460532 DOI: 10.3390/biomedicines8080264] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/23/2020] [Accepted: 07/23/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer is one of the life-threatening disorders that, in spite of excellent advances in medicine and technology, there is no effective cure for. Surgery, chemotherapy, and radiotherapy are extensively applied in cancer therapy, but their efficacy in eradication of cancer cells, suppressing metastasis, and improving overall survival of patients is low. This is due to uncontrolled proliferation of cancer cells and their high migratory ability. Finding molecular pathways involved in malignant behavior of cancer cells can pave the road to effective cancer therapy. In the present review, we focus on phosphatase and tensin homolog (PTEN) signaling as a tumor-suppressor molecular pathway in gastric cancer (GC). PTEN inhibits the PI3K/Akt pathway from interfering with the migration and growth of GC cells. Its activation leads to better survival of patients with GC. Different upstream mediators of PTEN in GC have been identified that can regulate PTEN in suppressing growth and invasion of GC cells, such as microRNAs, long non-coding RNAs, and circular RNAs. It seems that antitumor agents enhance the expression of PTEN in overcoming GC. This review focuses on aforementioned topics to provide a new insight into involvement of PTEN and its downstream and upstream mediators in GC. This will direct further studies for evaluation of novel signaling networks and their targeting for suppressing GC progression.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz 5166616471, Iran;
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran;
| | - Hui Li Ang
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore;
| | - Ebrahim Rahmani Moghadam
- Department of Anatomical Sciences, School of Medicine, Student Research Committee, Shiraz University of Medical Sciences, Shiraz 7134814336, Iran;
- Kazerun Health Technology Incubator, Shiraz University of Medical Sciences, Shiraz 6461665145, Iran
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan 8715988141, Iran;
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (A.K.B.); (H.S.)
| | - Leila Jafaripour
- Department of Anatomy, School of Medicine, Dezful University of Medical Sciences, Dezful 3419759811, Iran;
| | - Atefe Kazemzade Bejandi
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (A.K.B.); (H.S.)
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417414418, Iran;
| | - Hossein Saleki
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran; (A.Z.); (A.K.B.); (H.S.)
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey
- Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla 34956, Istanbul, Turkey
| | - Alan Prem Kumar
- Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore;
| |
Collapse
|
48
|
Suppression of PTTG1 inhibits cell angiogenesis, migration and invasion in glioma cells. Med Oncol 2020; 37:73. [PMID: 32725378 DOI: 10.1007/s12032-020-01398-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 07/16/2020] [Indexed: 02/07/2023]
Abstract
Pituitary tumor-transforming gene 1 (PTTG1) has been identified as an oncogene and is overexpressed in many tumor types. However, the role of PTTG1 in glioblastoma (GBM) has not been well characterized, especially in relation to angiogenesis, migration, and invasion. In the present study, our results showed that the expression of PTTG1 was significantly higher in patients with GBM. Bioinformatic analysis showed that angiogenesis and the cell migration-related process were increased in patients with high PTTG1 expression levels; meanwhile, PTTG1 was positively correlated with marker genes of angiogenesis, migration and the evasion of apoptosis. In vitro assays showed that PTTG1 knockdown dramatically suppressed angiogenesis, migration and invasion, and increased the apoptosis of GBM cells. Moreover, our results also showed that silencing PTTG1 suppressed the activity of the TGF-β/PI3K-AKT-mTOR pathway, which induced tumor deterioration in multiple organs. Overall, our findings indicate that PTTG1 is a glioma malignant factor that promotes angiogenesis, migration, invasion, and the evasion of apoptosis, and these roles may be related to the TGF-β/PI3K-AKT-mTOR pathway. Thus, the targeted inhibition of PTTG1 might be a novel therapeutic strategy and a potential diagnostic biomarker for GBM-targeted therapies.
Collapse
|
49
|
Huang Q, Fu Y, Zhang S, Zhang Y, Chen S, Zhang Z. Ethyl pyruvate inhibits glioblastoma cells migration and invasion through modulation of NF-κB and ERK-mediated EMT. PeerJ 2020; 8:e9559. [PMID: 32742812 PMCID: PMC7380274 DOI: 10.7717/peerj.9559] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/25/2020] [Indexed: 12/20/2022] Open
Abstract
Background Glioblastoma is a grade IV glioma with the highest degree of malignancy and extremely high incidence. Because of the poor therapeutic effect of surgery and radiochemotherapy, glioblastoma has a high recurrence rate and lethality, and is one of the most challenging tumors in the field of oncology. Ethyl pyruvate (EP), a stable lipophilic pyruvic acid derivative, has anti-inflammatory, antioxidant, immunomodulatory and other cellular protective effects. It has been reported that EP has potent anti-tumor effects on many types of tumors, including pancreatic cancer, prostate cancer, liver cancer, gastric cancer. However, whether EP has anti-tumor effect on glioblastoma or not is still unclear. Methods Glioblastoma U87 and U251 cells were treated with different concentrations of EP for 24 h or 48 h. CCK8 assay and Colony-Formation assay were performed to test the viability and proliferation. Wound-healing assay and Transwell assay were carried out to measure cell invasion and migration. Western blot was not only used to detect the protein expression of epithelial-mesenchymal transition (EMT)-related molecules, but also to detect the expression and activation levels of NF-κB (p65) and Extracellular Signal Regulated Kinase (ERK). Results In glioblastoma U87 and U251 cells treated with EP, the viability, proliferation, migration, invasion abilities were inhibited in a dose-dependent manner. EP inhibited EMT and the activation of NF-κB (p65) and ERK. With NF-κB (p65) and ERK activated, EMT, migration and invasion of U87 and U251 cells were promoted. However the activation of NF-κB (p65) and ERK were decreased, EMT, migration and invasion abilities were inhibited in U87 and U251 cells treated with EP. Conclusion EP inhibits glioblastoma cells migration and invasion by blocking NF-κB and ERK-mediated EMT.
Collapse
Affiliation(s)
- Qing Huang
- Department of Pathogen Biology, School of Basic Medicine, Central South University, Changsha, China
| | - Yongming Fu
- Department of Infectious Diseases and Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, China
| | - Shan Zhang
- Department of Pathogen Biology, School of Basic Medicine, Central South University, Changsha, China
| | - Youxiang Zhang
- Department of Pathogen Biology, School of Basic Medicine, Central South University, Changsha, China
| | - Simin Chen
- Department of Pathogen Biology, School of Basic Medicine, Central South University, Changsha, China
| | - Zuping Zhang
- Department of Pathogen Biology, School of Basic Medicine, Central South University, Changsha, China
| |
Collapse
|
50
|
Peng YX, Yu B, Qin H, Xue L, Liang YJ, Quan ZX. EMT-related gene expression is positively correlated with immunity and may be derived from stromal cells in osteosarcoma. PeerJ 2020; 8:e8489. [PMID: 32117617 PMCID: PMC7003688 DOI: 10.7717/peerj.8489] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/29/2019] [Indexed: 01/22/2023] Open
Abstract
Background Osteosarcoma is the most common type of bone cancer in children and young adults. Recent studies have shown a correlation between epithelial–mesenchymal transition (EMT)-related gene expression and immunity in human cancers. Here, we investigated the relationship among EMT, immune activity, stromal activity and tumor purity in osteosarcoma. Methods We defined EMT gene signatures and evaluated immune activity and stromal activity based on the gene expression and clinical data from three independent microarray datasets. These factors were evaluated by single sample Gene Set Enrichment Analyses and the ESTIMATE tool. Finally, we analyzed the key source of EMT gene expression in osteosarcoma using microarray datasets from the Gene Expression Omnibus and human samples that we collected. Results EMT-related gene expression was positively correlated with immune and stromal activity in osteosarcoma. Tumor purity was negatively correlated with EMT, immune activity and stromal cells. We further demonstrated that high EMT gene expression could significantly predict poor overall survival (OS) and recurrence-free survival (RFS) in osteosarcoma patients, while high immune activity cannot. However, combining these factors could have further prognostic value for osteosarcoma patients in terms of OS and RFS. Finally, we found that stromal cells may serve as a key source of EMT gene expression in osteosarcoma. Conclusion The results of this study reveal that the expression of EMT genes and immunity are positively correlated, but these signatures convey disparate prognostic information. Furthermore, the results indicate that EMT-related gene expression may be derived from stromal rather than epithelial cancer cells.
Collapse
Affiliation(s)
- Yin-Xiao Peng
- Department of Orthopedics, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Bin Yu
- Department of Orthopedics, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Hui Qin
- Department of Orthopedics, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Li Xue
- Department of Orthopedics, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Yi-Jian Liang
- Department of Orthopedics, The Third People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Zheng-Xue Quan
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, Chongqing, China
| |
Collapse
|