1
|
Wang J, Li Z, Han X, Xie Z, Hou Y, Liu M, Cheng Y, Lu Q, Luo J, Wang H. Echinatin inhibits the growth and metastasis of human hepatocellular carcinoma cells through p38 and JNK signaling pathways. Tissue Cell 2025; 95:102907. [PMID: 40209402 DOI: 10.1016/j.tice.2025.102907] [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: 12/15/2024] [Revised: 03/05/2025] [Accepted: 03/31/2025] [Indexed: 04/12/2025]
Abstract
Hepatocellular carcinoma (HCC) ranks among the most prevalent cancers, with both a high incidence and a significant mortality rate. Clinical medications are highly toxic to patients and prone to resistance. Natural products are highly valued in the development of antitumour drugs. This study aimed to elucidate the anti-HCC ability and potential mechanism of Echinatin (Ecn), a natural existed flavonoid. Our findings revealed that Ecn suppressed the growth, migration, and invasion of HCC cells and demonstrated a superior inhibitory impact on the development of xenograft tumors. Moreover, Ecn was less toxic to mice and had a good drug safety. Mechanistically, Ecn was found to activate p38 and JNK signaling pathways. Accordingly, the suppressive effect of Ecn on HCC cells was attenuated by the introduction of p38 blocker SB203580 and JNK blocker SP600125. Collectively, our research suggests that Ecn might have anti-HCC properties through the activation of p38 and JNK signaling.
Collapse
Affiliation(s)
- Jiayu Wang
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ziyun Li
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xueqian Han
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Zhou Xie
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yajun Hou
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Miao Liu
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Yuhang Cheng
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qiuping Lu
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Jinyong Luo
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| | - Hongbo Wang
- Department of General Surgery, Chongqing Hospital of Traditional Chinese Medicine, Chongqing 400016, China.
| |
Collapse
|
2
|
Zhan P, Huang S, Chen D, Li Y, Chen D. Echinatin inhibits osteoarthritis through the NF-κB signaling pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:7363-7375. [PMID: 39747466 DOI: 10.1007/s00210-024-03756-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025]
Abstract
Osteoarthritis (OA) is currently the most common degenerative joint disease in China and even worldwide and is the leading cause of disability in the elderly population. So far, due to an insufficient understanding of the pathogenesis and etiology of the disease, there is still no effective targeted treatment for early OA. Pro-inflammatory cytokine interleukin-1 is an important inflammatory mediator secreted in early OA, and IL-1β plays a crucial role in the pathogenesis of OA, affecting chondrocytes and the extracellular matrix of CARTILAGE. Echinatin has been used for years as a health supplement, retaining its antioxidant, anti-inflammatory, and autophagy-promoting effects. However, whether echinatin has inhibitory effects on OA is still unknown. In this study, we used an in vitro OA model of chondrocytes induced by IL-1β and an in vivo OA model of rats induced by anterior cruciate ligament transection (ACLT), and through experiments such as western blotting and IHC, we demonstrated that echinatin can be used as a novel drug for treating OA. Mechanistically, we found that echinatin inhibits the activity of chondrocytes induced by IL-1β through the NF-kB signaling pathway. This study can provide more effective treatment options for OA patients and further diagnostic and therapeutic methods for clinical treatment.
Collapse
Affiliation(s)
- Peng Zhan
- Department of Bone and Joint Sports Medicine, Longyan First Affiliated Hospital of Fujian Medical University, No.105 Jiuyi North Road, Longyan, Fujian, 364000, China
| | - Shiming Huang
- Department of Bone and Joint Sports Medicine, Longyan First Affiliated Hospital of Fujian Medical University, No.105 Jiuyi North Road, Longyan, Fujian, 364000, China
| | - Daohua Chen
- Department of Bone and Joint Sports Medicine, Longyan First Affiliated Hospital of Fujian Medical University, No.105 Jiuyi North Road, Longyan, Fujian, 364000, China
| | - Ying Li
- Department of Bone and Joint Sports Medicine, Longyan First Affiliated Hospital of Fujian Medical University, No.105 Jiuyi North Road, Longyan, Fujian, 364000, China
| | - Dongfeng Chen
- Department of Bone and Joint Sports Medicine, Longyan First Affiliated Hospital of Fujian Medical University, No.105 Jiuyi North Road, Longyan, Fujian, 364000, China.
| |
Collapse
|
3
|
Lv Z, Zhao S, Wu H. LIMA1 inhibits cisplatin resistance and malignant biological behavior of bladder cancer cells by suppressing the Wnt/β-catenin pathway. BMC Med Genomics 2025; 18:78. [PMID: 40269880 PMCID: PMC12016094 DOI: 10.1186/s12920-025-02146-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] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 12/13/2024] [Indexed: 04/25/2025] Open
Abstract
OBJECTIVE This study aimed to explore the effect of LIM domain and actin binding 1 (LIMA1) on bladder cancer (BCa) cells and to investigate its underlying molecular mechanisms. METHODS The expression of LIMA1 gene in clinical BCa tissue samples and BCa cell models was detected using real-time quantitative PCR and western blot. Subsequently, LIMA1 knockdown experiments were performed exclusively in the BCa J82 cell line, while LIMA1 overexpression was conducted only in the cisplatin-resistant J82/CR cell line. The proliferation of the cells was assessed by colony formation assay. Cisplatin resistance was evaluated by MTT assay. Migration and invasion of the cells were tested by Transwell assay. Additionally, the levels of key proteins in the Wnt/β-catenin signaling pathway were examined by western blotting. RESULTS We found that LIMA1 was underexpressed in BCa tissues and cells (P < 0.01). Overexpression of LIMA1 inhibited the proliferation, migration, invasion, and epithelial-mesenchymal transition of BCa cells (P < 0.01) and improved their cisplatin resistance (P < 0.01), whereas knocking down LIMA1 produced opposite results (P < 0.01). Furthermore, overexpression of LIMA1 could suppress the Wnt/β-catenin signaling pathway in BCa cells (P < 0.01), and activation of this pathway partially reversed the anti-tumor effects produced by overexpression of LIMA1 (P < 0.01). CONCLUSION LIMA1 could inhibit the malignant biological behavior of BCa cells and weaken their cisplatin resistance by negatively regulating the Wnt/β-catenin signaling pathway. Our findings provide new insights for the clinical treatment of BCa.
Collapse
Affiliation(s)
- Zhong Lv
- Department of Urology, Changzhou Medical Center, Changzhou Wujin People's Hospital, Nanjing Medical University, Changzhou, 213003, China
- Department of Urology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213003, China
- Department of Urology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, 213003, China
| | - Suchen Zhao
- Department of Urology, Changzhou Medical Center, Changzhou Wujin People's Hospital, Nanjing Medical University, Changzhou, 213003, China
- Department of Urology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213003, China
- Department of Urology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, 213003, China
| | - Haoran Wu
- Department of Urology, Changzhou Medical Center, Changzhou Wujin People's Hospital, Nanjing Medical University, Changzhou, 213003, China.
- Department of Urology, Wujin Hospital Affiliated with Jiangsu University, Changzhou, 213003, China.
- Department of Urology, The Wujin Clinical College of Xuzhou Medical University, Changzhou, 213003, China.
| |
Collapse
|
4
|
Zhou T, Niu Y, Li Y. Advances in research on malignant tumors and targeted agents for TOP2A (Review). Mol Med Rep 2025; 31:50. [PMID: 39670307 DOI: 10.3892/mmr.2024.13415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/28/2024] [Indexed: 12/14/2024] Open
Abstract
The DNA topoisomerase isoform topoisomerase IIα (TOP2A) is essential for the condensation and segregation of cellular mitotic chromosomes and the structural maintenance. It has been demonstrated that TOP2A is highly expressed in various malignancies, including lung adenocarcinoma (LUAD), hepatocellular carcinoma (HCC) and breast cancer (BC), associating with poor prognosis and aggressive tumor behavior. Additionally, TOP2A has emerged as a promising target for cancer therapy, with widespread clinical application of associated chemotherapeutic agents. The present study explored the impact of TOP2A on malignant tumor growth and the advancements in research on its targeted drugs. The fundamental mechanisms of TOP2A have been detailed, its specific roles in tumor cells are analyzed, and its potential as a biomarker for tumor prognosis and therapeutic targeting is highlighted. Additionally, the present review compiles findings from the latest clinical trials of relevant targeted agents, information on newly developed inhibitors, and discusses future research directions and clinical application strategies in cancer therapy, aiming to propose novel ideas and methods.
Collapse
Affiliation(s)
- Tao Zhou
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Yiting Niu
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| | - Yanjun Li
- Department of Hepatobiliary Surgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, P.R. China
| |
Collapse
|
5
|
Gao K, Jiang H, Zhang C, Li L, Qi Y, Yang N, Li X, Wang Y, Wu B, Shao Q, Wu G. Suppressing the progression of bladder cancer using cyclovirobuxine D based on network pharmacology and bioinformatics approaches. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03754-9. [PMID: 39797986 DOI: 10.1007/s00210-024-03754-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025]
Abstract
Limited treatment options are available for bladder cancer (BCa) resulting in extremely high mortality rates. Cyclovirobuxine D (CVB-D), a naturally alkaloid, reportedly exhibits notable antitumor activity against diverse tumor types. However, its impact on CVB-D on BCa and its precise molecular targets remain unexplored. This study conducts CCK8 assay, colony formation assay, and flow cytometry experiments to demonstrate that CVB-D inhibits long-term proliferation and viability of BCa cell lines, thereby inducing apoptosis in vitro. It employs PPI networks and the CytoHubba algorithm to identify COL1A1, COL6A1, COL6A2, COL5A2, COL5A1, COL12A1, COL18A1, ITGA5, VCL, FLNA, and GSN as crucial therapeutic targets for CVB-D that can halt the malignant progression of BCa. GO and KEGG analyses indicate that the PI3K/AKT signaling pathway potentially may play a pivotal role in mediating the anti-BCa growth effects of CVB-D. The ROC curve and K-M survival analyses reveal the significant clinical value of all the 11 identified therapeutic targets, with GSN as the most effective target of CVB-D in combating BCa. This study also uncovers a potential interaction between GSN and CVB-D through molecular docking and molecular dynamics simulations. RT-qPCR and Western blotting experiments provide further evidence that CVB-D effectively suppresses GSN mRNA and protein expression in a concentration-dependent fashion. Our comprehensive study is the first report on the molecular mechanism of CVB-D against BCa, identifying GSN as a pivotal target in CVB-D-based anti-BCa therapy. We believe that our study results may help establish a theoretical basis for the possible utilization of CVB-D in cancer therapeutics.
Collapse
Affiliation(s)
- Ke Gao
- Department of Urology, Affiliated Xi'an Peoples Hospital (Xi'an Fourth Hospital) of Northwest University, Xi'an, 710000, China
| | - Hanbing Jiang
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi'an, 710038, China
| | - Chao Zhang
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi'an, 710038, China
| | - Lin Li
- Department of Immunology, School of Basic Medicine, Yan'an University, Yan'an, 716099, China
| | - Yuhong Qi
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi'an, 710038, China
| | - Nan Yang
- Department of Special Medical and Health Management Center, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710003, China
| | - Xiaoshun Li
- Department of Urology, Affiliated Xi'an Peoples Hospital (Xi'an Fourth Hospital) of Northwest University, Xi'an, 710000, China
| | - Yongshun Wang
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi'an, 710038, China
| | - Bin Wu
- Department of Urology, Affiliated Xi'an Peoples Hospital (Xi'an Fourth Hospital) of Northwest University, Xi'an, 710000, China
| | - Qiuju Shao
- Department of Radiotherapy, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi'an, 710038, China.
| | - Guojun Wu
- Department of Urology, Affiliated Xi'an Peoples Hospital (Xi'an Fourth Hospital) of Northwest University, Xi'an, 710000, China.
| |
Collapse
|
6
|
Zhu Z, Hu B, Zhu D, Li X, Chen D, Wu N, Rao Q, Zhang Z, Wang H, Zhu Y. Bromocriptine sensitivity in bromocriptine-induced drug-resistant prolactinomas is restored by inhibiting FGF19/FGFR4/PRL. J Endocrinol Invest 2025; 48:67-80. [PMID: 38926262 DOI: 10.1007/s40618-024-02408-0] [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: 12/15/2023] [Accepted: 06/03/2024] [Indexed: 06/28/2024]
Abstract
PURPOSE At present, various treatment strategies are available for pituitary adenomas, including medications, surgery and radiation. The guidelines indicate that pharmacological treatments, such as bromocriptine (BRC) and cabergoline (CAB), are important treatments for prolactinomas, but drug resistance is an urgent problem that needs to be addressed. Therefore, exploring the mechanism of drug resistance in prolactinomas is beneficial for clinical treatment. METHODS In our research, BRC-induced drug-resistant cells were established. Previous RNA sequencing data and an online database were used for preliminary screening of resistance-related genes. Cell survival was determined by Cell Counting Kit-8 (CCK-8) assay, colony formation assays and flow cytometry. Quantitative real-time polymerase chain reaction (qRT‒PCR), western blotting, immunohistochemistry, immunofluorescence and Co-immunoprecipitation (Co-IP) were used to assess the molecular changes and regulation. The therapeutic efficacy of BRC and FGFR4 inhibitor fisogatinib (FISO) combination was evaluated in drug-resistant cells and xenograft tumors in nude mice. RESULTS Consistent with the preliminary results of RNA sequencing and database screening, fibroblast growth factor 19 (FGF19) expression was elevated in drug-resistant cells and tumor samples. With FGF19 silencing, drug-resistant cells exhibited increased sensitivity to BRC and decreased intracellular phosphorylated fibroblast growth factor receptor 4 (FGFR4) levels. After confirming that FGF19 binds to FGFR4 in prolactinoma cells, we found that FGF19/FGFR4 regulated prolactin (PRL) synthesis through the ERK1/2 and JNK signaling pathways. Regarding the effect of targeting FGF19/FGFR4 on BRC efficacy, FISO and BRC synergistically inhibited the growth of tumor cells, promoted apoptosis and reduced PRL levels. CONCLUSION Overall, our study revealed FGF19/FGFR4 as a new mechanism involved in the drug resistance of prolactinomas, and combination therapy targeting the pathway could be helpful for the treatment of BRC-induced drug-resistant prolactinomas.
Collapse
Affiliation(s)
- Z Zhu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - B Hu
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - D Zhu
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - X Li
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - D Chen
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - N Wu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Q Rao
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - Z Zhang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China
| | - H Wang
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
| | - Y Zhu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-Sen University, No.74 Zhongshan Road 2, Guangzhou, 510080, Guangdong, China.
- Department of Histology and Embryology, School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
7
|
Huang J, Luo Y, Wang Y, Wang S, Huang R, An Y. Silencing CCT3 induces ferroptosis through the NOD1-NF-κB signaling pathway in bladder cancer. Sci Rep 2024; 14:26188. [PMID: 39478031 PMCID: PMC11525567 DOI: 10.1038/s41598-024-76724-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] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
Bladder cancer (BCa) is a lethal malignancy of the urinary system and exhibits a poor prognosis. Chaperonin-containing tailless complex polypeptide 1 subunit 3 (CCT3) acts as an oncogene in various tumors, whereas its effect on BCa remains unknown. We identified the ferroptosis-associated differentially expressed genes through bioinformatic analysis and selected CCT3 for further verification. The levels of cell viability, apoptosis, migration, invasion, and proliferation were measured to clarify the effect of silencing CCT3 on BCa cells. Then we evaluated the role of CCT3 knockdown in vivo. Ferroptosis was assessed by the expression detection of the ferroptosis-related proteins. The underlying mechanism was predicted by RNA sequencing and verified by an agonist for nucleotide-binding and oligomerization domain 1 (NOD1). Western blotting was conducted to detect the protein expression of NOD1, nuclear factor kappa B (NF-κB) inhibitor alpha (IκBα), and phospho-IκBα (p-IκBα). In vitro, down-regulation of CCT3 suppressed the cell viability, migration, invasion, and proliferation, as well as induced apoptosis of BCa cells. In vivo, silencing CCT3 elevated the body weight of mice and suppressed the BCa progression. In addition, CCT3 knockdown could induce ferroptosis in vitro and in vivo. CCT3 knockdown suppressed the expression of NOD1 and p-IκBα/IκBα and the NOD1 agonist could reverse the effect of CCT3 suppression on BCa in vitro and in vivo. In summary, our findings demonstrate that silencing CCT3 inhibits BCa via induction of ferroptosis and suppression of the NOD1-NF-κB pathway.
Collapse
Affiliation(s)
- Jianlin Huang
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Section 2, 1st Ring Road, Qingyang District, Chengdu City, 610072, Sichuan Province, China
| | - Yizhao Luo
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Section 2, 1st Ring Road, Qingyang District, Chengdu City, 610072, Sichuan Province, China
| | - Yu Wang
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Section 2, 1st Ring Road, Qingyang District, Chengdu City, 610072, Sichuan Province, China
| | - Shize Wang
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Section 2, 1st Ring Road, Qingyang District, Chengdu City, 610072, Sichuan Province, China
| | - Runhua Huang
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Section 2, 1st Ring Road, Qingyang District, Chengdu City, 610072, Sichuan Province, China.
| | - Yu An
- Department of Urology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 32, West Section 2, 1st Ring Road, Qingyang District, Chengdu City, 610072, Sichuan Province, China.
| |
Collapse
|
8
|
Duan M, Jie J, Li C, Bai X, Hua S, Tang M, Li D. Echinatin alleviates sepsis severity through modulation of the NF-κB and MEK/ERK signaling pathways. Biomed Pharmacother 2024; 179:117359. [PMID: 39236479 DOI: 10.1016/j.biopha.2024.117359] [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: 06/20/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Abstract
Sepsis, a frequently fatal condition, emerges from an exaggerated inflammatory response to infection, resulting in multi-organ dysfunction and alarmingly high mortality rates. Despite the urgent need for effective treatments, current therapeutic options remain limited to antibiotics, with no other efficacious alternatives available. Echinatin (Ecn), a potent bioactive compound extracted from the roots and rhizomes of licorice, has gained significant attention for its broad pharmacological properties, particularly its ability to combat oxidative stress. Recent research highlights the crucial role that oxidative stress plays in the onset and progression of sepsis further emphasizing the potential therapeutic value of Ecn in this context. In this study, we explored the protective effects of Ecn in a murine model of sepsis induced by cecal ligation and puncture (CLP). Ecn demonstrated a significant reduction in the levels of inflammatory cytokines and reactive oxygen species (ROS) in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Network pharmacology analysis identified 41 targets and top 15 pathways involved in the Ecn-mediated signaling network, revealing that Ecn might exert its effects through key targets including the NF-κB and MAPK signaling pathways. Molecular docking studies suggested a strong affinity between Ecn and MEK, with kinetic simulations and binding energy calculations confirming a stable interaction. Mechanistically, Ecn treatment inhibited NF-κB and the MEK/ERK signaling pathway, as evidenced by decreased phosphorylation of IκBα and nuclear p65, along with reduced phosphorylation of MEK and ERK in both LPS-stimulated RAW 264.7 macrophages and septic mice. Furthermore, the administration of MEK signaling agonists reversed the anti-inflammatory effects of Ecn, indicating the involvement of this signaling pathway in Ecn's protective mechanism. Notably, our investigation revealed that Ecn did not affect bacterial proliferation either in vivo or in vitro, underscoring its specific immunomodulatory effects rather than direct antimicrobial activity. In summation, our findings underscored the potential of Ecn as an innovative therapeutic remedy for sepsis-induced injury, particularly through the regulation of the NF-κB and MEK/ERK signaling pathway. This exploration unveiled a promising therapeutic approach for treating sepsis, supplementing existing interventions and addressing their constraints.
Collapse
Affiliation(s)
- Meina Duan
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, China
| | - Jing Jie
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, China
| | - Chunxiuli Li
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, China
| | - Xiaoxue Bai
- Department of General Practice, The First Hospital of Jilin University, Changchun, 130021, China
| | - Shucheng Hua
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| | - Mingbo Tang
- Department of Thoracic Surgery, The First Hospital of Jilin University, Changchun 130021, China.
| | - Dan Li
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
9
|
Liu J, Huang M, Yang Y, Zeng Y, Yang Y, Guo Q, Liu W, Guo L. Screening potential antileukemia agents from duckweed: Integration of chemical profiling, network pharmacology, and experimental validation. PHYTOCHEMICAL ANALYSIS : PCA 2024; 35:1633-1648. [PMID: 38924240 DOI: 10.1002/pca.3407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 06/06/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024]
Abstract
INTRODUCTION The identification of active dietary flavonoids in food is promising for novel drug discovery. The active ingredients of duckweed (a widely recognized food and herb with abundant flavonoids) that are associated with acute myeloid leukemia (AML) have yet to be identified, and their underlying mechanisms have not been elucidated. OBJECTIVES The objective of this study was to identify novel constituents exhibiting antileukemia activity in duckweed through the integration of chemical profiling, network pharmacology, and experimental validation. METHODS First, high performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) was used to characterize the primary constituents of duckweed. Subsequently, AML cell-xenograft tumor models were used to validate the anticancer effect of duckweed extract. Furthermore, network pharmacology analysis was conducted to predict the potential active compounds and drug targets against AML. Lastly, based on these findings, two monomers (apiin and luteoloside) were selected for experimental validation. RESULTS A total of 17 compounds, all of which are apigenin and luteolin derivatives, were identified in duckweed. The duckweed extract significantly inhibited AML cell growth in vivo. Furthermore, a total of 88 targets for duckweed against AML were predicted, with key targets including PTGS2, MYC, MDM2, VEGFA, CTNNB1, CASP3, EGFR, TP53, HSP90AA1, CCND1, MMP9, TNF, and MAPK1. GO and KEGG pathway enrichment analyses indicated that these targets were primarily involved in the apoptotic signaling pathway. Lastly, both apiin and luteoloside effectively induced apoptosis through CASP3 activation, and this effect could be partially reversed by a caspase inhibitor (Z-VAD). CONCLUSION Duckweed extract has an antileukemic effect, and apiin derived from duckweed shows potential as a treatment for AML.
Collapse
Affiliation(s)
- Jing Liu
- Department of Pediatrics, Children Hematological Oncology and Birth Defects Laboratory, Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, People's Republic of China
| | - Mengjun Huang
- National-Local Joint Engineering Research Center for Innovative Targeted Drugs, Chongqing University of Arts and Sciences, Chongqing, People's Republic of China
| | - Yan Yang
- Department of Pediatrics, Children Hematological Oncology and Birth Defects Laboratory, Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, People's Republic of China
| | - Yan Zeng
- Department of Pediatrics, Children Hematological Oncology and Birth Defects Laboratory, Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, People's Republic of China
| | - You Yang
- Department of Pediatrics, Children Hematological Oncology and Birth Defects Laboratory, Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, People's Republic of China
| | - Qulian Guo
- Department of Pediatrics, Children Hematological Oncology and Birth Defects Laboratory, Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, People's Republic of China
| | - Wenjun Liu
- Department of Pediatrics, Children Hematological Oncology and Birth Defects Laboratory, Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, People's Republic of China
| | - Ling Guo
- Department of Pediatrics, Children Hematological Oncology and Birth Defects Laboratory, Affiliated Hospital of Southwest Medical University, Sichuan Clinical Research Center for Birth Defects, Luzhou, Sichuan, People's Republic of China
| |
Collapse
|
10
|
Lai F, He L, Lia T, Yang Z, Huang C. Identification and validation of basement membrane-related genes predicting prognosis and immune infiltration associated with bladder cancer. Medicine (Baltimore) 2024; 103:e38858. [PMID: 39029072 PMCID: PMC11398827 DOI: 10.1097/md.0000000000038858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 06/17/2024] [Indexed: 07/21/2024] Open
Abstract
Bladder cancer (BC) is fatal during muscle invasion and treatment progress is limited. In this study, we aimed to construct and validate basement membrane (BM)-associated gene prognosis to predict BC progression and tumor immune infiltration correlation. We choreographed BM-related genes in the Cancer Genome Atlas (TCGA) database using COX regression and least absolute shrinkage and selection operator (LASSO) analysis, and the predictive value of BM-related genes was further validated by the GSE32548, GSE129845, and immunohistochemistry staining. All analyses were performed with R-version 4.2.2, and its appropriate packages. Three genes were identified to construct a gene signature to predictive of BC prognosis. We divided the TCGA database into 2 groups, and patients in the high-risk group had worse overall survival (OS) than those in the low-risk group. In GSE32548, we confirmed that patients in the high-risk group had a poorer prognosis compared to those in the low-risk group in terms of OS. Immunohistochemical staining of EPEMP1, GPC2, and ITGA3 showed significantly higher expression at the protein level in BC tissues than in normal tissues. The Spearman analysis showed risk score was positively correlated with B cell naïve, Macrophages M2, and Mast cells resting. stromal score, immune score, and ESTIMATE scores were significantly higher in the high-risk group. drugs sensitivity analysis showed IC50 of Cisplatin, Gemcitabine, and Methotrexate in the high-risk group was significantly higher than that in the low-risk group. We identified 3 prognostic genes from a novel perspective of BM genes as effective risk stratification tools for BC patients.
Collapse
Affiliation(s)
- Fie Lai
- Department of Urology Surgery, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Lin He
- Department of Pathology, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Thongher Lia
- Department of Urology Surgery, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Zhen Yang
- Department of Urology Surgery, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| | - Chaoyou Huang
- Department of Urology Surgery, Chengdu Second People’s Hospital, Chengdu, Sichuan, China
| |
Collapse
|
11
|
Saikia L, Gogoi B, Sen S, Tonk RK, Kumar D, Dutta PP. The recent update and advancements of natural products in targeting the Wnt/β-Catenin pathway for cancer prevention and therapeutics. Med Oncol 2024; 41:164. [PMID: 38816663 DOI: 10.1007/s12032-024-02387-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/19/2024] [Indexed: 06/01/2024]
Abstract
The Wnt/β-Catenin pathway (Wnt/β-CatP) is implicated in accelerating carcinogenesis and cancer progression, contributing to increased morbidity and treatment resistance. Even though it holds promise as a focus for cancer treatment, its intricate nature and diverse physiological effects pose significant challenges. Recent years have witnessed significant advancements in this domain, with numerous natural products demonstrating promising preclinical anti-tumor effects and identified as inhibitors of the Wnt/β-CatP through various upstream and downstream mechanisms. This study provides a comprehensive overview of the current landscape of Wnt/β-Cat-targeted cancer therapy, examining the impact of natural products on Wnt/β-Cat signaling in both cancer prevention and therapeutic contexts. A comprehensive search was conducted on scientific databases like SciFinder, PubMed, and Google Scholar to retrieve relevant literature on Wnt-signaling, natural products, β-Catenin (β-Cat), and cancer from 2020 to January 2024. As per the analysis of the relevant reference within the specified period, it has been noted that a total of 58 phytoconstituents, predominantly phenolics, followed by triterpenoids and several other classes, along with a limited number of plant extracts, have exhibited activity targeting the Wnt/β-CatP. Most β-Cat regulating modulators restrict cancer cell development by suppressing β-Cat expression, facilitating proteasomal degradation, and inhibiting nuclear translocation. Multiple approaches have been devised to block the activity of β-Cat in cancer therapy, a key factor in cancer progression, leading to the discovery of various Wnt/β-CatP regulators. However, their exploration remains limited, necessitating further research using clinical models for potential clinical use in cancer prevention and therapeutics.
Collapse
Affiliation(s)
- Lunasmrita Saikia
- Faculty of Pharmaceutical Science, Assam Down Town University, Guwahati, Assam, 781026, India
| | - Bhaskarjyoti Gogoi
- Department of Biotechnology, The Assam Royal Global University, Guwahati, Assam, 781035, India
| | - Saikat Sen
- Faculty of Pharmaceutical Science, Assam Down Town University, Guwahati, Assam, 781026, India
| | - Rajiv K Tonk
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Deepak Kumar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, 173229, India.
| | - Partha Pratim Dutta
- Faculty of Pharmaceutical Science, Assam Down Town University, Guwahati, Assam, 781026, India.
| |
Collapse
|
12
|
Xie L, Liang S, Jiwa H, Zhang L, Lu Q, Wang X, Luo L, Xia H, Li Z, Wang J, Luo X, Luo J. Securinine inhibits the tumor growth of human bladder cancer cells by suppressing Wnt/β-catenin signaling pathway and activating p38 and JNK signaling pathways. Biochem Pharmacol 2024; 223:116125. [PMID: 38484850 DOI: 10.1016/j.bcp.2024.116125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/28/2024] [Accepted: 03/11/2024] [Indexed: 03/18/2024]
Abstract
Bladder cancer (BC) is the most common malignant tumor in urinary system. Although chemotherapy is one of the most important adjuvant treatments for BC, drug resistance, non-specific toxicity and severe side effects are the major obstacles to BC chemotherapy. Natural products have always been a leading resource of antitumor drug discovery, with the advantages of excellent effectiveness, low toxicity, multi-targeting potency and easy availability. In this study, we evaluated the potential anti-tumor effect of securinine (SEC), a natural alkaloid from Securinega suffruticosa, on BC cells in vitro and in vivo, and delineated the underlying mechanism. We found that SEC inhibited the proliferation, migration and invasion, induced the apoptosis of BC cells in vitro, and retarded the xenograft tumor growth of BC cell in vivo. Notably, SEC had a promising safety profile because it presented no or low toxicity on normal cells and mice. Mechanistically, SEC inactivated Wnt/β-catenin signaling pathway while activated p38 and JNK signaling pathway. Moreover, β-catenin overexpression, the p38 inhibitor SB203580 and the JNK inhibitor SP600125 both mitigated the inhibitory effect of SEC on BC cells. Furthermore, we demonstrated a synergistic inhibitory effect of SEC and gemcitabine (GEM) on BC cells in vitro and in vivo. Taken together, our findings suggest that SEC may exert anti-BC cell effect at least through the activation of p38 and JNK signaling pathways, and the inhibition of Wnt/β-catenin signaling pathway. More meaningfully, the findings indicate that GEM-induced BC cell killing can be enhanced by combining with SEC.
Collapse
Affiliation(s)
- Liping Xie
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Shiqiong Liang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Habu Jiwa
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Lulu Zhang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Qiuping Lu
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiaoxuan Wang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Lijuan Luo
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Haichao Xia
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Ziyun Li
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Jiayu Wang
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China
| | - Xiaoji Luo
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Jinyong Luo
- Key Laboratory of Diagnostic Medicine Designated by the Chinese Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
13
|
You Y, Zhou X, Tang Q, Zhao T, Wang J, Huang H, Chen J, Qi Z, Li F. Echinatin mitigates sevoflurane-induced neurotoxicity through regulation of ferroptosis and iron homeostasis. Aging (Albany NY) 2024; 16:4670-4683. [PMID: 38446592 PMCID: PMC10968708 DOI: 10.18632/aging.205622] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/27/2023] [Indexed: 03/08/2024]
Abstract
Surgery and anesthesia are vital medical interventions, but concerns over their potential cognitive side effects, particularly with the use of inhalational anesthetics like sevoflurane, have surfaced. This study delves into the neuroprotective potential of Echinatin against sevoflurane-induced neurotoxicity and the underlying mechanisms. Echinatin, a natural compound, has exhibited anti-inflammatory, antioxidant, and anticancer properties. Sevoflurane, while a popular anesthetic, is associated with perioperative neurocognitive disorders (PND) and neurotoxicity. Our investigation began with cellular models, where Echinatin demonstrated a significant reduction in sevoflurane-induced apoptosis. Mechanistically, we identified ferroptosis, a novel form of programmed cell death characterized by iron accumulation and lipid peroxidation, as a key player in sevoflurane-induced neuronal injury. Echinatin notably suppressed ferroptosis in sevoflurane-exposed cells, suggesting a pivotal role in neuroprotection. Expanding our research to a murine model, we observed perturbations in iron homeostasis, inflammatory cytokines, and antioxidants due to sevoflurane exposure. Echinatin treatment effectively restored iron balance, mitigated inflammation, and preserved antioxidant levels in vivo. Behavioral assessments using the Morris water maze further confirmed Echinatin's neuroprotective potential, as it ameliorated sevoflurane-induced spatial learning and memory impairments. In conclusion, our study unveils Echinatin as a promising candidate for mitigating sevoflurane-induced neurotoxicity. Through the regulation of ferroptosis, iron homeostasis, and inflammation, Echinatin demonstrates significant neuroprotection both in vitro and in vivo. These findings illuminate the potential for Echinatin to enhance the safety of surgical procedures involving sevoflurane anesthesia, minimizing the risk of cognitive deficits and neurotoxicity.
Collapse
Affiliation(s)
- Yanqiu You
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Xudong Zhou
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Qiuqin Tang
- Ruikang Hospital Affiliated of Guangxi University of Chinese Medicine, Nanning 530011, China
| | - Tianshou Zhao
- Ruikang Hospital Affiliated of Guangxi University of Chinese Medicine, Nanning 530011, China
| | - Juan Wang
- Ruikang Hospital Affiliated of Guangxi University of Chinese Medicine, Nanning 530011, China
| | - Hanqin Huang
- Ruikang Hospital Affiliated of Guangxi University of Chinese Medicine, Nanning 530011, China
| | - Jibing Chen
- Ruikang Hospital Affiliated of Guangxi University of Chinese Medicine, Nanning 530011, China
| | - Zhongquan Qi
- Guangxi Key Laboratory of Special Biomedicine, School of Medicine, Guangxi University, Nanning 530004, China
| | - Fujun Li
- Ruikang Hospital Affiliated of Guangxi University of Chinese Medicine, Nanning 530011, China
| |
Collapse
|