1
|
Fiorilla S, Tasso F, Clemente N, Trisciuoglio T, Boldorini R, Carini R. Monensin Inhibits Triple-Negative Breast Cancer in Mice by a Na +-Dependent Cytotoxic Action Unrelated to Cytostatic Effects. Cells 2025; 14:185. [PMID: 39936977 PMCID: PMC11817484 DOI: 10.3390/cells14030185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/11/2025] [Accepted: 01/22/2025] [Indexed: 02/13/2025] Open
Abstract
Triple-negative breast cancer (TNBC) represents the most aggressive breast cancer subtype, defined by its limited therapeutic options and poor outcomes. This study investigated the therapeutic potential of targeting Na+ homeostasis in TNBC cells to induce TNBC inhibition. For this purpose, BALB/c mice were inoculated with 4T1-Luc2 breast cancer cells and treated with the Na+ ionophore monensin (8 mg/kg) or vehicle alone. Tumor development and cellular Na+ content were assessed using vivo live imaging techniques, while intracellular Na+ variations and cytotoxicity were evaluated through live cell analysis. Monensin treatment increased Na+ levels in cancerous tissues and reduced TNBC mass (monensin: 0.146 ± 0.06; vehicle: 0.468 ± 0.2 cm3; p < 0.001). This treatment induced extensive necrosis in TNBC tumors while preserving the structural and functional integrity of healthy organs and maintaining the proliferative activity of both tumor and normal tissues. Monensin did not alter the expression of proliferating nuclear antigen (PCNA) in 4T1-Luc2 cells but triggered cytotoxicity preceded by intracellular Na+ accumulation. Na+-free conditions prevented both Na+ accumulation and 4T1-Luc2 cell death. Thus, monensin exerts its antitumor effects in TNBC through a Na+-dependent and tumor-specific cytotoxic mechanism, without inducing cytostatic effects on normal or transformed tissues. Collectively, these findings underscore the potential of Na+ ionophores as promising therapeutic agents for TNBC.
Collapse
Affiliation(s)
| | | | | | | | | | - Rita Carini
- Department of Health Science, Università del Piemonte Orientale, Via Solaroli, 17, 28100 Novara, Italy; (S.F.); (F.T.); (N.C.); (T.T.); (R.B.)
| |
Collapse
|
2
|
Hong H, Huang Y, Yang Z, Jiang X, Liu H. Pyroptosis-related lncRNAs are potential biomarkers for predicting prognoses and immune landscapes in patients with gastric adenocarcinoma. Discov Oncol 2024; 15:684. [PMID: 39565540 PMCID: PMC11579272 DOI: 10.1007/s12672-024-01579-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 11/12/2024] [Indexed: 11/21/2024] Open
Abstract
OBJECTIVES The purpose of this study was to investigate the significance of pyroptosis-related lncRNAs (PRlncRNA) in predicting prognoses and immune landscapes of patients with gastric adenocarcinoma (STAD). METHODS Transcriptomic data and clinicopathological data were obtained from The Cancer Genome Atlas database. Based on correlation analysis and univariate Cox regression, prognostic PRlncRNA were identified. Subsequently, a PRlncRNA prognostic signature (PRLPS) was generated via least absolute shrinkage and selection operator (LASSO) regression, Kaplan-Meier method, receiver operating characteristic (ROC) curves, principal component analysis, and univariate and multivariate regression. Besides, the clinicopathological characteristics, tumor microenvironment (TME) scores, the immune landscapes in different risk subgroups were explored. Moreover, based on three PRlncRNA, we constructed a competing endogenous RNA (ceRNA) network. Additionally, Gene Set Enrichment Analysis (GSEA), Kyoto Encyclopedia of Genes and Genomes pathways (KEGG) and Gene Ontology (GO) analysis were performed for biological functional analysis based on the difference between high- and low- risk groups, which also used to screen out potential STAD drugs. RESULTS 21 PRlncRNA made up the prognostic signature, which had significant value in predicting the overall survival (OS), clinicopathological features, TME, immune checkpoint genes expression, and the response to immune checkpoint inhibitors of patients with STAD. In a addition, we constructed a ceRNA network comprising 3 PRlncRNAs and 69 mRNAs. The function of PRlncRNA was related to cancer-associated pathways. Ten small molecular drugs that might improve the prognosis of patients were screened out by connectivity maps. CONCLUSIONS Using PRlncRNA as a prognostic indicator for STAD, we identified predictive biomarkers and immunotherapy targets while refreshing our understanding.
Collapse
Affiliation(s)
- Haidu Hong
- School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Yuancheng Huang
- Department of Oncology, Dongguan People's Hospital, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, 523000, Guangdong, China
- Cancer Center, Dongguan People's Hospital, The Tenth Affiliated Hospital of Southern Medical University), Dongguan, 523000, Guangdong, China
| | - Zehong Yang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Xiaotao Jiang
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, China
| | - Hong Liu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Guangdong Pharmaceutical University, Gonghexiheng Street 1, Yuexiu District, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
3
|
Zhu X, Si Y, Gai C, Li Z. Investigating the molecular mechanisms of Fuzheng Yiliu Shenji prescription in SH-SY5Y neuroblastoma cells. Front Oncol 2024; 14:1447666. [PMID: 39319058 PMCID: PMC11420165 DOI: 10.3389/fonc.2024.1447666] [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: 06/12/2024] [Accepted: 08/15/2024] [Indexed: 09/26/2024] Open
Abstract
Background Neuroblastoma is the most common extracranial solid tumor in childhood. Fuzheng Yiliu Shenji Prescription (FYSP) has shown potential in treating malignant pediatric tumors in clinical settings. This study aims to explore the molecular mechanisms behind its effects, specifically in the context of neuroblastoma cell lines. Objective To elucidate the active compounds in FYSP and their mechanisms of action in inhibiting neuroblastoma cell viability, inducing apoptosis, and affecting the cell cycle in SH-SY5Y cells through network pharmacology and empirical validation. Materials and methods We identified the major compounds in FYSP and their predicted targets, constructing a protein-protein interaction (PPI) network and performing GO and KEGG pathway analyses. The effects of FYSP were empirically validated through assays on cell viability, cell cycle, apoptosis, and protein expression in SH-SY5Y cells. Results The study identified 172 active chemical components in FYSP, with 188 common targets related to neuroblastoma. Network analysis highlighted the PI3K-Akt pathway as a significant target. Experimental validation in SH-SY5Y cells confirmed that FYSP could inhibit cell viability, induce G2/M cell cycle arrest, and promote apoptosis through modulation of the PI3K-Akt pathway, specifically upregulating caspase-3 and downregulating Bcl-2/Bax expression. Conclusion The study elucidates the molecular basis of FYSP's effects on neuroblastoma cells in vitro, demonstrating its ability to modulate key pathways involved in cell cycle and apoptosis. While these findings suggest a potential therapeutic role for FYSP, they are limited to in vitro observations, and further research, including in vivo studies, is necessary to explore its clinical applicability.
Collapse
Affiliation(s)
- Xueying Zhu
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yinchu Si
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Cong Gai
- Department of Anatomy, School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhong Li
- Department of Oncology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
4
|
Fang T, Hu S, Song X, Wang J, Zuo R, Yun S, Jiang S, Guo D. Combination of monensin and erlotinib synergistically inhibited the growth and cancer stem cell properties of triple-negative breast cancer by simultaneously inhibiting EGFR and PI3K signaling pathways. Breast Cancer Res Treat 2024; 207:435-451. [PMID: 38958784 DOI: 10.1007/s10549-024-07374-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 05/14/2024] [Indexed: 07/04/2024]
Abstract
BACKGROUND Cancer stem cells (CSCs) in triple-negative breast cancer (TNBC) are recognized as a highly challenging subset of cells, renowned for their heightened propensity for relapse and unfavorable prognosis. Monensin, an ionophoric antibiotic, has been reported to exhibit significant therapeutic efficacy against various cancers, especially CSCs. Erlotinib is classified as one of the EGFR-TKIs and has been previously identified as a promising therapeutic target for TNBC. Our research aims to assess the effectiveness of combination of monensin and erlotinib as a potential treatment strategy for TNBC. METHODS The combination of monensin and erlotinib was assessed for its potential anticancer activity through various in vitro assays, including cytotoxicity assay, colony formation assay, wound healing assay, transwell assay, mammosphere formation assay, and proportion of CSCs assay. Additionally, an in vivo study using tumor-bearing nude mice was conducted to evaluate the inhibitory effect of the monensin and erlotinib combination on tumor growth. RESULTS The results indicated that combination of monensin with erlotinib synergistically inhibited cell proliferation, the migration rate, the invasion ability and decreased the CSCs proportion, and CSC markers SOX2 and CD133 in vivo and in vitro. Furthermore, the primary proteins involved in the signaling pathways of the EGFR/ERK and PI3K/AKT are simultaneously inhibited by the combination treatment of monensin and erlotinib in vivo and in vitro. CONCLUSIONS The simultaneous inhibition of the EGFR/ERK and PI3K/AKT/mTOR signaling pathways by the combination of monensin and erlotinib exhibited a synergistic effect on suppressing tumor proliferation and cancer cell stemness in TNBC.
Collapse
Affiliation(s)
- Tian Fang
- Engineering Center of Innovativennovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China
- Department of Comparative Medicine, Affiliated Hospital of Medicine School, Nanjing Jinling Hospital, Nanjing University, Nanjing, 210002, China
| | - Shiheng Hu
- Engineering Center of Innovativennovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China
| | - Xinhao Song
- Engineering Center of Innovativennovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China
| | - Junqi Wang
- Engineering Center of Innovativennovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China
| | - Runan Zuo
- Engineering Center of Innovativennovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China
| | - Shifeng Yun
- Department of Comparative Medicine, Affiliated Hospital of Medicine School, Nanjing Jinling Hospital, Nanjing University, Nanjing, 210002, China
| | - Shanxiang Jiang
- Engineering Center of Innovativennovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China.
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China.
| | - Dawei Guo
- Engineering Center of Innovativennovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China.
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing, 210095, China.
| |
Collapse
|
5
|
Lee ZY, Lee WH, Lim JS, Ali AAA, Loo JSE, Wibowo A, Mohammat MF, Foo JB. Golgi apparatus targeted therapy in cancer: Are we there yet? Life Sci 2024; 352:122868. [PMID: 38936604 DOI: 10.1016/j.lfs.2024.122868] [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: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024]
Abstract
Membrane trafficking within the Golgi apparatus plays a pivotal role in the intracellular transportation of lipids and proteins. Dysregulation of this process can give rise to various pathological manifestations, including cancer. Exploiting Golgi defects, cancer cells capitalise on aberrant membrane trafficking to facilitate signal transduction, proliferation, invasion, immune modulation, angiogenesis, and metastasis. Despite the identification of several molecular signalling pathways associated with Golgi abnormalities, there remains a lack of approved drugs specifically targeting cancer cells through the manipulation of the Golgi apparatus. In the initial section of this comprehensive review, the focus is directed towards delineating the abnormal Golgi genes and proteins implicated in carcinogenesis. Subsequently, a thorough examination is conducted on the impact of these variations on Golgi function, encompassing aspects such as vesicular trafficking, glycosylation, autophagy, oxidative mechanisms, and pH alterations. Lastly, the review provides a current update on promising Golgi apparatus-targeted inhibitors undergoing preclinical and/or clinical trials, offering insights into their potential as therapeutic interventions. Significantly more effort is required to advance these potential inhibitors to benefit patients in clinical settings.
Collapse
Affiliation(s)
- Zheng Yang Lee
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia
| | - Wen Hwei Lee
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia
| | - Jing Sheng Lim
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia
| | - Afiqah Ali Ajmel Ali
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia
| | - Jason Siau Ee Loo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia; Digital Health and Medical Advancements Impact Lab, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
| | - Agustono Wibowo
- Faculty of Applied Science, Universiti Teknologi MARA (UiTM) Pahang, Jengka Campus, 26400 Bandar Tun Abdul Razak Jengka, Pahang, Malaysia
| | - Mohd Fazli Mohammat
- Organic Synthesis Laboratory, Institute of Science, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor, Malaysia
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, 47500 Subang Jaya, Selangor, Malaysia; Digital Health and Medical Advancements Impact Lab, Taylor's University, Subang Jaya 47500, Selangor, Malaysia
| |
Collapse
|
6
|
Song X, Lin M, Fang T, Gong J, Wang J, Gao S, Xu X, Lv X, Gao X, Zhang J, Jiang S, Guo D. Maduramicin-guided nanotherapy: A polymeric micelles for targeted drug delivery in canine mammary tumors. Biomed Pharmacother 2024; 170:116062. [PMID: 38150878 DOI: 10.1016/j.biopha.2023.116062] [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: 10/10/2023] [Revised: 12/17/2023] [Accepted: 12/21/2023] [Indexed: 12/29/2023] Open
Abstract
Canine mammary tumors (CMT) can severely compromise the life quality of the affected dogs through local recurrence, distant metastases and ultimately succumb to death. Recently, more attention has been given to the potential antimetastatic effect of maduramicin (MAD) on breast cancer. However, its poor aqueous solubility and toxicity to normal tissues limit its clinical application. Therefore, to address the drawbacks of MAD and enhance its anticancer and antimetastatic effects, MAD-loaded TPGS polymeric micelles (MAD-TPGS) were prepared by a thin-film hydration technique. The optimized MAD-TPGS exhibited excellent size distribution, stability and improved water solubility. Cellular uptake assays showed that TPGS polymer micelles could enhance drug internalization. Moreover, TPGS synergistically improved the cytotoxicity of MAD by targeting mitochondrial organelles, improving reactive oxygen species levels and reducing the mitochondrial transmembrane potential. More importantly, MAD-TPGS significantly impeded the metastasis of tumor cells. In vivo results further confirmed that, in addition to exhibiting excellent biocompatibility, MAD-TPGS exhibited greater antitumor efficacy than free MAD. Interestingly, MAD-TPGS displayed superior suppression of CMT metastasis via tail vein injection compared to oral administration, indicating its suitability for intravenous delivery. Overall, MAD-TPGS could be applied as a potential antimetastatic cancer agent for CMT.
Collapse
Affiliation(s)
- Xinhao Song
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Mengjuan Lin
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Tian Fang
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Jiahao Gong
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Junqi Wang
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Shasha Gao
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Xiaolin Xu
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Xin Lv
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Xiuge Gao
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Junren Zhang
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Shanxiang Jiang
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Dawei Guo
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China.
| |
Collapse
|
7
|
Fu B, Fang L, Wang R, Zhang X. Inhibition of Wnt/β-catenin signaling by monensin in cervical cancer. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:21-30. [PMID: 38154961 PMCID: PMC10762490 DOI: 10.4196/kjpp.2024.28.1.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 09/19/2023] [Accepted: 10/15/2023] [Indexed: 12/30/2023]
Abstract
The challenging clinical outcomes associated with advanced cervical cancer underscore the need for a novel therapeutic approach. Monensin, a polyether antibiotic, has recently emerged as a promising candidate with anti-cancer properties. In line with these ongoing efforts, our study presents compelling evidence of monensin's potent efficacy in cervical cancer. Monensin exerts a pronounced inhibitory impact on proliferation and anchorage-independent growth. Additionally, monensin significantly inhibited cervical cancer growth in vivo without causing any discernible toxicity in mice. Mechanism studies show that monensin's anti-cervical cancer activity can be attributed to its capacity to inhibit the Wnt/β-catenin pathway, rather than inducing oxidative stress. Monensin effectively reduces both the levels and activity of β-catenin, and we identify Akt, rather than CK1, as the key player involved in monensin-mediated Wnt/β-catenin inhibition. Rescue studies using Wnt activator and β-catenin-overexpressing cells confirmed that β-catenin inhibition is the mechanism of monensin's action. As expected, cervical cancer cells exhibiting heightened Wnt/β-catenin activity display increased sensitivity to monensin treatment. In conclusion, our findings provide pre-clinical evidence that supports further exploration of monensin's potential for repurposing in cervical cancer therapy, particularly for patients exhibiting aberrant Wnt/β-catenin activation.
Collapse
Affiliation(s)
- Bingbing Fu
- Department of Obstetrics and Gynaecology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, China
| | - Lixia Fang
- Department of Obstetrics and Gynaecology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, China
| | - Ranran Wang
- Department of Obstetrics and Gynaecology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, China
| | - Xueling Zhang
- Department of Obstetrics and Gynaecology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei 441000, China
| |
Collapse
|
8
|
Gao F, Xue C, Zhang T, Zhang L, Zhu GY, Ou C, Zhang YZ, Dong X. MXene-Based Functional Platforms for Tumor Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2302559. [PMID: 37142810 DOI: 10.1002/adma.202302559] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/13/2023] [Indexed: 05/06/2023]
Abstract
Recently, 2D transition metal carbide, nitride, and carbonitrides (MXenes) materials stand out in the field of tumor therapy, particularly in the construction of functional platforms for optimal antitumor therapy due to their high specific surface area, tunable performance, strong absorption of near-infrared light as well as preferable surface plasmon resonance effect. In this review, the progress of MXene-mediated antitumor therapy is summarized after appropriate modifications or integration procedures. The enhanced antitumor treatments directly performed by MXenes, the significant improving effect of MXenes on different antitumor therapies, as well as the MXene-mediated imaging-guided antitumor strategies are discussed in detail. Moreover, the existing challenges and future development directions of MXenes in tumor therapy are presented.
Collapse
Affiliation(s)
- Fan Gao
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Chun Xue
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Tian Zhang
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, China
| | - Lu Zhang
- National and Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Guo-Yin Zhu
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Changjin Ou
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Yi-Zhou Zhang
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Xiaochen Dong
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing, 211816, China
- School of Chemistry & Materials Science, Jiangsu Normal University, Xuzhou, 221116, China
| |
Collapse
|
9
|
Serter Kocoglu S, Oy C, Secme M, Sunay FB. Investigation of the anticancer mechanism of monensin via apoptosis-related factors in SH-SY5Y neuroblastoma cells. Clin Transl Sci 2023; 16:1725-1735. [PMID: 37477356 PMCID: PMC10499413 DOI: 10.1111/cts.13593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/22/2023] Open
Abstract
Monensin is an ionophore antibiotic that inhibits the growth of cancer cells. The aim of this study was to investigate the apoptosis-mediated anticarcinogenic effects of monensin in SH-SY5Y neuroblastoma cells. The effects of monensin on cell viability, invasion, migration, and colony formation were determined by XTT, matrigel-chamber, wound healing, and colony formation tests, respectively. The effects of monensin on apoptosis were determined by real-time polymerase chain reaction, TUNEL, Western blot, and Annexin V assay. We have shown that monensin suppresses neuroblastoma cell viability, invasion, migration, and colony formation. Moreover, we reported that monensin inhibits cell viability by triggering apoptosis of neuroblastoma cells. Monensin caused apoptosis by increasing caspase-3, 7, 8, and 9 expressions and decreasing Bax and Bcl-2 expressions in neuroblastoma cells. In Annexin V results, the rates of apoptotic cells were found to be 9.66 ± 0.01% (p < 0.001), 29.28 ± 0.88% (p < 0.01), and 62.55 ± 2.36% (p < 0.01) in the 8, 16, and 32 μM monensin groups, respectively. In TUNEL results, these values were, respectively; 35 ± 2% (p < 0.001), 34 ± 0.57% (p < 0.001), and 75 ± 2.51% (p < 0.001). Our results suggest that monensin may be a safe and effective therapeutic candidate for treating pediatric neuroblastoma.
Collapse
Affiliation(s)
- Sema Serter Kocoglu
- Department of Histology and EmbryologySchool of Medicine, Balikesir UniversityBalikesirTurkey
| | - Ceren Oy
- Department of Histology and EmbryologySchool of Medicine, Bursa Uludag UniversityBursaTurkey
| | - Mücahit Secme
- Department of Medical BiologySchool of Medicine, Ordu UniversityDenizliTurkey
| | - F. Bahar Sunay
- Department of Histology and EmbryologySchool of Medicine, Balikesir UniversityBalikesirTurkey
| |
Collapse
|
10
|
Serter Kocoglu S, Sunay FB, Akkaya PN. Effects of Monensin and Rapamycin Combination Therapy on Tumor Growth and Apoptosis in a Xenograft Mouse Model of Neuroblastoma. Antibiotics (Basel) 2023; 12:995. [PMID: 37370314 DOI: 10.3390/antibiotics12060995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/22/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023] Open
Abstract
Neuroblastoma is the most common pediatric solid tumor originating from the neural crest. New treatment options are needed to improve treatment outcomes and the survival of patients with neuroblastoma. Monensin is an ionophore antibiotic with antiparasitic, antibacterial, and anticancer properties isolated from Streptomyces cinnamonensis. The aim of this study was to investigate the therapeutic effects of single and combined monensin and rapamycin treatments on mTOR (mammalian target of rapamycin) signaling pathway-mediated apoptosis and tumor growth in an SH-SY5Y neuroblastoma cell xenograft model. Control, monensin, rapamycin, and monensin + rapamycin groups were formed in the xenograft neuroblastoma model obtained from CD1 nude mice, and tumor volumes and animal weights were recorded throughout the treatment. In xenograft neuroblastoma tumor tissues, apoptosis was determined by TUNEL (terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling) and cleaved-caspase 3 immunohistochemistry, and PI3K (phosphoinositide-3-kinase)/AKT/mTOR expression was determined by the immunohistochemistry and immunofluorescence methods. The combination of monensin and rapamycin was to reduce the growth of xenograft neuroblastoma tumor tissues, trigger apoptosis, and suppress the expression of PI3K/AKT/mTOR. A significant increase in apoptotic cell rate was demonstrated in the combination group, supported by cleaved-caspase 3 immunohistochemistry results. In addition, it was reported that the combination treatment regime triggered apoptosis by reducing the expression of phosphorylated PI3K/AKT/mTOR. Our preclinical results may be a precursor to develop new therapeutic approaches to treat neuroblastoma.
Collapse
Affiliation(s)
- Sema Serter Kocoglu
- Department of Histology and Embryology, Faculty of Medicine, Balikesir University, 10145 Balikesir, Türkiye
| | - Fatma Bahar Sunay
- Department of Histology and Embryology, Faculty of Medicine, Balikesir University, 10145 Balikesir, Türkiye
| | - Pakize Nur Akkaya
- Department of Histology and Embryology, Faculty of Medicine, Balikesir University, 10145 Balikesir, Türkiye
| |
Collapse
|
11
|
Seçme M, Kocoglu SS. Investigation of the TLR4 and IRF3 signaling pathway-mediated effects of monensin in colorectal cancer cells. Med Oncol 2023; 40:187. [PMID: 37219624 DOI: 10.1007/s12032-023-02055-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 05/14/2023] [Indexed: 05/24/2023]
Abstract
Monensin is an ionophore antibiotic isolated from Streptomyces cinnamonensis with very strong antibacterial and antiparasitic effects. Although monensin is known to exhibit anticancer activity in different cancer types, there are a very limited number of studies on its anti-inflammatory effects in colorectal cancer (CRC) cells. The aim of this study was to investigate the TLR4/IRF3-mediated antiproliferative and anti-inflammatory effects of monensin in colorectal cancer cells. The dose- and time-dependent antiproliferative activity of monensin in colorectal cancer cells was determined by XTT method and its effects on mRNA expression changes of Toll-like receptors and IRF3 genes were determined by RT-PCR. TLR4 and Interferon Regulatory Factor 3 (IRF3) protein expression was evaluated by immunofluorescence method. TLR4 and type 1 interferon (IRF) levels were also evaluated by ELISA. IC50 value of monensin in HT29 cells was determined as 10.7082 µM at 48 h and 12.6288 µM at 48th for HCT116 cells. Monensin treatment decreased TLR4 and TLR7 and IRF3 mRNA expression in CRC cells. Monensin treatment decreased the expression level of IRF3 induced by LPS. Our study demonstrates for the first time the TLR4/IRF3-mediated anti-inflammatory effects of monensin in colorectal cancer cells. Further studies on the effects of monensin on TLR receptors in colorectal cancer cells are needed.
Collapse
Affiliation(s)
- Mücahit Seçme
- School of Medicine, Department of Medical Biology, Ordu University, Ordu, Turkey.
| | - Sema Serter Kocoglu
- School of Medicine, Department of Histology and Embryology, Balikesir University, Balikesir, Turkey
| |
Collapse
|
12
|
Monensin, an Antibiotic Isolated from Streptomyces Cinnamonensis, Regulates Human Neuroblastoma Cell Proliferation via the PI3K/AKT Signaling Pathway and Acts Synergistically with Rapamycin. Antibiotics (Basel) 2023; 12:antibiotics12030546. [PMID: 36978413 PMCID: PMC10044236 DOI: 10.3390/antibiotics12030546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023] Open
Abstract
Neuroblastoma is the most common extracranial childhood tumor and accounts for approximately 15% of pediatric cancer-related deaths. Further studies are needed to identify potential therapeutic targets for neuroblastoma. Monensin is an ionophore antibiotic obtained from Streptomyces cinnamonensis with known antibacterial and antiparasitic effects. No study has reported the effects of monensin on SH-SY5Y neuroblastoma cells by targeting the PI3K/AKT signaling pathway. The aim of this study was to investigate the antiproliferative effects of monensin alone and in combination with rapamycin in human SH-SY5Y neuroblastoma cells mediated by the PI3K/AKT signaling pathway. The effects of single and combination applications of monensin and rapamycin on SH-SY5Y cell proliferation were investigated by XTT, and their effects on the PI3K/AKT signaling pathway by RT-PCR, immunohistochemistry, immunofluorescence, and Western blotting. The combined effects of monensin and rapamycin on SH-SY5Y proliferation were most potent at 72 h (combination index < 1). The combination of monensin and rapamycin caused a significant decrease in the expression of P21RAS, AKT, and MAPK1 genes. Single and combined administrations of monensin and rapamycin caused a significant decrease in PI3K/AKT expression. Our results showed for the first time that monensin exerts an antiproliferative effect by targeting the PI3K/AKT signaling pathway in neuroblastoma cells. It is suggested that monensin and its combination with rapamycin may be an effective therapeutic candidate for treating neuroblastoma.
Collapse
|
13
|
Zeng C, Long M, Lu Y. Monensin synergizes with chemotherapy in uveal melanoma through suppressing RhoA. Immunopharmacol Immunotoxicol 2023; 45:35-42. [PMID: 36043455 DOI: 10.1080/08923973.2022.2112219] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Uveal melanoma (UM) is the common primary cancer of the eye and new treatments are needed. Substantial evidence has shown that an antibiotic monensin is an attractive candidate for the development of anti-cancer drug. In this study, we investigated the potential of repositioning monensin for the treatment of UM in the pre-clinical setting. MATERIALS AND METHODS Cellular activity assays were performed using multiple cell lines representing UM models with different cellular origins and genetic profiling and normal cells as control. Combination studies were performed using Chou-Talalay method. Mechanism studies were performed using immunoblotting and ELISA. RESULTS Monensin was effective against all tested UM cell lines and less effective against normal fibroblast cells. Monensin induced G0/G1 arrest and thus decreased S phase, leading to UM cell growth inhibition. It also inhibited migration and induced apoptosis in UM cells. In addition, the combination of monensin and dacarbazine was synergistic in targeting UM cells. Our mechanistic studies showed that monensin specifically decreased activity of RhoA without affecting other small GTPases, such as Ras and Rac1. Consistently, monensin decreased phosphorylation of downstream effectors of RhoA signaling, including ROCK, MYPT1 and MLC. Rescue studies using RhoA activator calpeptin showed that calpeptin significantly abolished the inhibitory effects of monensin on RhoA activity, proliferation, migration and survival, confirming that RhoA is the target of monensin in UM cells. CONCLUSIONS Our study demonstrates that monensin is a potent inhibitor of UM and synergizes with chemotherapy, via suppressing RhoA activity and RhoA-mediated signaling. Our findings suggest that monensin may be a potential lead compound for further development into a drug for UM treatment.
Collapse
Affiliation(s)
- Chaoxia Zeng
- Hainan Eye Hospital and Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Haikou, PR China
| | - Mingxia Long
- Department of Nursing, Wuhan Third Hospital-Tongren Hospital of Wuhan University, Wuhan, PR China
| | - Ying Lu
- Department of Integrated Traditional Chinese and Western Medicine, Wuhan Third Hospital -Tongren Hospital of Wuhan University, Wuhan, PR China
| |
Collapse
|
14
|
Zhao J, Luan Y, Chen Y, Cheng L, Qin Q. Toxicological and transcriptomic-based analysis of monensin and sulfamethazine co-exposure on male SD rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 245:114110. [PMID: 36155339 DOI: 10.1016/j.ecoenv.2022.114110] [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: 06/29/2022] [Revised: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 06/16/2023]
Abstract
Antibiotic residue has become an emerging environmental contaminant, while the toxicological effects and underlying mechanisms caused by the co-exposure to multiple veterinary antibiotics were rarely studied. In this study, male Sprague Dawley rats were exposed to monensin (M) (1, 2, 10 mg/(kg·body weight (BW)) combined with sulfamethazine (S) (60, 120, 600 mg/(kg·BW)) or single drugs for 28 consecutive days. The body weight, hematological and blood biochemical parameters, organ coefficients, and histopathology were analyzed to discover their combined toxicity effect. Transcriptomic analysis was used to reveal the possible mechanisms of their joint toxicity. Compared with the control group, the weight gain rate was significantly reduced in the H-M+S and H-S, and alkaline phosphatase in H-M+S was significantly increased. Furthermore, relative liver and kidneys weight was significantly increased, and the liver of H-M+S showed more severe lesions in histopathological analysis. For H-M+S, H-M and H-S, transcriptomic results showed that 344, 246, and 99 genes were differentially expressed, respectively. The Gene Ontology terms mainly differ in sterol biosynthetic process and steroid hydroxylase activity. The Kyoto Encyclopedia of Genes and Genome pathways showed abnormal retinol metabolism, metabolism of xenobiotics by cytochrome P450, and drug metabolism-cytochrome 450; the common 30 genes were screened from the network of protein-protein interaction. The results showed that mixed contamination of M and S produces hepatotoxicity by interfering with linoleic acid metabolism, retinol metabolism and CYP450 enzyme-dominated drug metabolism. Further analysis showed that Cyp1a2, Cyp2c61, Ugt1a3, and Ugt1a5 might be the key genes. These findings could provide more evidence for investigating the toxic effects and metabolism of mixed antibiotics contamination in mammals.
Collapse
Affiliation(s)
- Junjie Zhao
- College of Veterinary Medicine, China Agricultural University, Beijing 100093, China.
| | - Yehui Luan
- College of Veterinary Medicine, China Agricultural University, Beijing 100093, China.
| | - Yanan Chen
- College of Veterinary Medicine, China Agricultural University, Beijing 100093, China.
| | - Linli Cheng
- College of Veterinary Medicine, China Agricultural University, Beijing 100093, China; National Reference Laboratory on Residue of Veterinary Medicine, Beijing 100093, China; Beijing Key Laboratory of Animal Source Food Safety Testing Technology, Beijing 100093, China.
| | - Qianxi Qin
- College of Veterinary Medicine, China Agricultural University, Beijing 100093, China.
| |
Collapse
|
15
|
Song X, Zhang Y, Zuo R, Zhang J, Lin M, Wang J, Hu S, Ji H, Peng L, Lv Y, Gao X, Jiang S, Guo D. Repurposing maduramicin as a novel anticancer and anti-metastasis agent for triple-negative breast cancer as enhanced by nanoemulsion. Int J Pharm 2022; 625:122091. [PMID: 35964826 DOI: 10.1016/j.ijpharm.2022.122091] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 07/10/2022] [Accepted: 08/04/2022] [Indexed: 10/15/2022]
Abstract
Triple-negative breast cancer (TNBC) is featured by aggression and metastasis and remains an unmet medical challenge due to high death rate. We aimed to repurpose maduramicin (MAD) as an effective drug against TNBC, and develop a nanoemulsion system to enhance anticancer efficacy of MAD. MDA-MB-231 and 4 T1 cells were used as in vitro model, and cell viability was determined by performing cell counting kit-8 and a colony-formation assay. Furthermore, MAD loaded nanoemulsion (MAD-NEs) was manufactured and characterized by a series of tests. The anticancer and anti-metastasis mechanism of MAD-NEs were assessed by performing cell cycle, apoptosis, wound-healing, transwell assay and Western blotting assays. Herein, MAD was firstly demonstrated to be an effective agent to suppress growth of TNBC cells. Subsequently, the optimized MAD-NEs were shown to have stability and high encapsulation efficiency, and could arrested cells in G0/G1 phase and induced apoptosis in TNBC cells. More importantly, MAD-NEs significantly impeded the metastasis of tumor cells, which was further demonstrated by the significant altered expression of epithelial-mesenchymal transition and extracellular matrix markers in vitro and in vivo. Moreover, compared to MAD, MAD-NEs exhibited higher efficacy in shrinking breast tumor size and repressing liver and lung metastasis in vivo, and showed excellent biocompatibility in tumor-bearing mice. The successfully prepared MAD-NEs are expected to be harnessed to suppress tumor growth, invasion and metastasis in the battle against malignant TNBC.
Collapse
Affiliation(s)
- Xinhao Song
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Yan Zhang
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Runan Zuo
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Jingjing Zhang
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Mengjuan Lin
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Junqi Wang
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Shiheng Hu
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Hui Ji
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Lin Peng
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Yingjun Lv
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Xiuge Gao
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Shanxiang Jiang
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China
| | - Dawei Guo
- Engineering Center of Innovative Veterinary Drugs, Center for Veterinary Drug Research and Evaluation, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, 1 Weigang, Nanjing 210095, China.
| |
Collapse
|
16
|
Sheng H, Hao Z, Zhu L, Zeng Y, He J. Construction and validation of a two-gene signature based on SUMOylation regulatory genes in non-small cell lung cancer patients. BMC Cancer 2022; 22:572. [PMID: 35606717 PMCID: PMC9125860 DOI: 10.1186/s12885-022-09575-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 04/18/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Post-translational modification plays an important role in the occurrence and development of various tumors. However, few researches were focusing on the SUMOylation regulatory genes as tumor biomarkers to predict the survival for specific patients. Here, we constructed and validated a two-gene signature to predict the overall survival (OS) of non-small cell lung cancer (NSCLC) patients. METHODS The datasets analyzed in this study were downloaded from TCGA and GEO databases. The least absolute shrinkage and selection operator (LASSO) Cox regression was used to construct the two-gene signature. Gene set enrichment analysis (GSEA) and Gene Ontology (GO) was used to identify hub pathways associated with risk genes. The CCK-8 assay, cell cycle analysis, and transwell assay was used to validate the function of risk genes in NSCLC cell lines. RESULTS Firstly, most of the SUMOylation regulatory genes were highly expressed in various tumors through the R package 'limma' in the TCGA database. Secondly, our study found that the two gene signature constructed by LASSO regression analysis, as an independent prognostic factor, could predict the OS in both the TCGA training cohort and GEO validation cohorts (GSE68465, GSE37745, and GSE30219). Furthermore, functional enrichment analysis suggests that high-risk patients defined by the risk score system were associated with the malignant phenomenon, such as DNA replication, cell cycle regulation, p53 signaling pathway. Finally, the results of the CCK-8 assay, cell cycle analysis, and transwell assay demonstrated that the two risk genes, SAE1 and UBA2, could promote proliferation and migration in non-small cell lung cancer cells. CONCLUSIONS The two-gene signature constructed in our study could predict the OS and may provide valuable clinical guidance for the treatment of NSCLC patients.
Collapse
Affiliation(s)
- Hongxu Sheng
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China National Center for Respiratory Medicine, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Zhexue Hao
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China National Center for Respiratory Medicine, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Linhai Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang, Hangzhou, China
| | - Yuan Zeng
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China National Center for Respiratory Medicine, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China
| | - Jianxing He
- Department of Thoracic Surgery and Oncology, The First Affiliated Hospital of Guangzhou Medical University, China National Center for Respiratory Medicine, China State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou, China.
| |
Collapse
|
17
|
Li Y, Sun Q, Chen S, Yu X, Jing H. Monensin inhibits anaplastic thyroid cancer via disrupting mitochondrial respiration and AMPK/mTOR signaling. Anticancer Agents Med Chem 2022; 22:2539-2547. [PMID: 35168524 DOI: 10.2174/1871520622666220215123620] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The clinical management of anaplastic thyroid cancer (ATC) remains challenging and novel treatment methods are needed. Monensin is a carboxyl polyether ionophore that potently inhibits the growth of various cancer types. Our current work investigates whether monensin has selective anti-ATC activity and systematically explores its underlying mechanisms. METHODS Proliferation and apoptosis assays were performed using a panel of thyroid cancer cell lines. Mitochondrial biogenesis profiles, ATP levels, oxidative stress, AMPK and mTOR were examined in these cells after monensin treatment. RESULTS Monensin is effective to inhibit proliferation and induce apoptosis in a number of thyroid cancer cell lines. The results are consistent across cell lines of varying cellular origins and genetic mutations. Compared to other thyroid cancer cell types, ATC cell lines are the most sensitive to monensin. Of note, monensin used at our experimental concentration affects less of normal cells. Mechanistic studies reveal that monensin acts on ATC cells through disrupting mitochondrial function, inducing oxidative stress and damage, and AMPK activation-induced mTOR inhibition. We further show mitochondrial respiration is a critical target for monensin in ATC cells. CONCLUSIONS Our pre-clinical findings demonstrate the selective anti-ATC activities of monensin. This is supported by increasing evidence monensin can to be repurposed as a potential anti-cancer drug.
Collapse
Affiliation(s)
- Yanli Li
- Department of Endocrinology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Qianshu Sun
- Department of Endocrinology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Sisi Chen
- Department of Endocrinology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Xiongjie Yu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Hongxia Jing
- Department of Ultrasound, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| |
Collapse
|
18
|
Yao S, Wang W, Zhou B, Cui X, Yang H, Zhang S. Monensin suppresses cell proliferation and invasion in ovarian cancer by enhancing MEK1 SUMOylation. Exp Ther Med 2021; 22:1390. [PMID: 34650638 PMCID: PMC8506924 DOI: 10.3892/etm.2021.10826] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 06/30/2021] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer is the most lethal gynecologic malignancy, and is usually diagnosed at an advanced stage. Most patients relapse within 12-24 months and die from progressive chemotherapy-resistant diseases. Significant progress has been made in developing new targeted therapies for human cancer, including ovarian cancer. However, an effective alternative to drug development is to repurpose drugs. The present study investigated the possibility of reusing the antibiotic monensin as an anti-ovarian cancer drug. After applying a series of titrated monensin on a panel of ovarian cancer cell lines, the growth, migration and invasion of cells were explored. Multiple signaling molecules associated with epithelial-to-mesenchymal transition were also regulated by monensin. The mitogen-activated protein kinase (MEK)-extracellular signal-regulated kinase (ERK) pathway was further found to be the key regulator affected by monensin. Additionally, monensin enhanced the MEK1 SUMOylation in vitro and in vivo, and the SUMOylation degree depended on time and dose. Xenograft studies verified that monensin effectively inhibited xenograft tumor growth by increasing the SUMOylation of MEK1. The aforementioned results suggested that monensin is a good candidate for anti-ovarian cancer by enhancing MEK1 SUMOylation and inhibiting the MEK-ERK pathway.
Collapse
Affiliation(s)
- Shujuan Yao
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China.,Department of Obstetrics and Gynecology, Affiliated Hospital of Shandong University of TCM, Jinan, Shandong 250014, P.R. China
| | - Wen Wang
- Department of Obstetrics and Gynecology, Jining Medical University Affiliated Tengzhou Central People's Hospital, Tengzhou, Shandong 277599, P.R. China
| | - Bin Zhou
- Department of Obstetrics and Gynecology, Tai'an City Central Hospital, Tai'an, Shandong 271000, P.R. China
| | - Xiujuan Cui
- Department of Obstetrics and Gynecology, Jining Medical University Affiliated Tengzhou Central People's Hospital, Tengzhou, Shandong 277599, P.R. China
| | - Hui Yang
- Department of Obstetrics and Gynecology, Jining Medical University Affiliated Tengzhou Central People's Hospital, Tengzhou, Shandong 277599, P.R. China
| | - Shiqian Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|