1
|
Zhang S, Wang YS, Li Y, To KI, Zhang ET, Jin YH. Annexin A2 binds the 3'-UTR of H2AX mRNA and regulates histone-H2AX-derived hypoxia-inducible factor 1-alpha activation. Cell Signal 2025; 132:111781. [PMID: 40164417 DOI: 10.1016/j.cellsig.2025.111781] [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: 12/28/2024] [Revised: 03/21/2025] [Accepted: 03/28/2025] [Indexed: 04/02/2025]
Abstract
Annexin A2 (Anxa2), a multifunctional protein with RNA-binding capabilities, is frequently overexpressed in various tumors, and its expression is highly correlated with malignant progression. In this study, we demonstrate for the first time that Anxa2 was co-expressed with glycolytic genes, suggesting its potential role as a regulator of glycolysis. RNA-protein interaction assay revealed that Anxa2 interacted with 3'-UTR of H2AX mRNA and protected it from miRNA-mediated degradation. Up-regulated Histone-H2AX enhances the expression of glycolytic genes including GLUT1, HK2, PGK1, ENO1, PKM2, GAPDH and LDHA via stabilizing hypoxia-inducible factor 1-alpha (HIF1α), thereby accelerating lactic acid production and secretion. (20S) G-Rh2, a natural compound targeting Anxa2, significantly interfered the Anxa2-H2AX mRNA interaction, and inhibited subsequent glycolysis progression. We propose that Anxa2 acts as a novel regulator in glycolysis via enhancing H2AX expression, and (20S) G-Rh2 may exert its anti-cancer activity by targeting Anxa2-H2AX-HIF1α-glycolysis axis in human hepatoma HepG2 cells.
Collapse
Affiliation(s)
- Shiyin Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yu-Shi Wang
- Department of Criminal Science and Technology, Jilin Police College, Changchun 130117, China
| | - Yang Li
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Kwang-Il To
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - En-Ting Zhang
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Ying-Hua Jin
- Key Laboratory for Molecular Enzymology and Engineering of the Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China.
| |
Collapse
|
2
|
Chen X, Su W, Chen J, Ouyang P, Gong J. RNF123 inhibits cell viability, cell cycle and colony formation of breast cancer by inhibiting glycolysis via ubiquitination of PFKP. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:7219-7233. [PMID: 39725718 DOI: 10.1007/s00210-024-03723-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
E3 ubiquitin ligases have the potential to modulate key oncogenic pathways. RING finger protein 123 (RNF123), as an E3 ubiquitin ligase, has been functioned as a tumor suppressor. This study was designed to explore the role of RNF123 in breast cancer. Immunohistochemistry was applied to examine protein expression in breast cancer tissues. Western blot and Quantitative Real-time PCR were performed to gauge protein and mRNA levels. Lentivirus transduction was used to overexpress or silence genes of interest. Cell Counting Kit-8, flow cytometry, and colony formation assays were used to assess cell viability, cell cycle, and colony formation. Extracellular acidification rate, lactic acid and adenosine triphosphate were used for glycolysis assay. Co-immunoprecipitation (Co-IP) and ubiquitination analysis were used to explore the interaction between RNF123 and 6-Phosphofructo-2-kinase (PFKP). In vivo experiments were performed with xenograft tumor models. RNF123 was downregulated in tumor tissues and cells, overexpression of which significantly decreased the viability and colony-forming ability of tumor cells, suppressed the progression of the cell cycle and glycolytic activity, and suppressed tumor growth in vivo. Co-IP and ubiquitination analysis revealed that there was an interaction between RNF123 and PFKP, and RNF123 could induce ubiquitination of PFKP. PFKP could reverse the effects of RNF123 on tumor cells. RNF123 inhibited cell viability, cell cycle and colony formation of breast cancer cells by inhibiting glycolysis via ubiquitination of PFKP.
Collapse
Affiliation(s)
- Xiaoqing Chen
- Department of Breast Medicine, The affiliated Foshan Women and Children Hospital, Guangdong Medical University, Foshan, 528000, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, 613 West of Huangpu Avenue, Guangzhou, 510630, China
| | - Weijie Su
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200011, China
| | - Jiewen Chen
- Department of Breast Medicine, The affiliated Foshan Women and Children Hospital, Guangdong Medical University, Foshan, 528000, China
| | - Peng Ouyang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, 613 West of Huangpu Avenue, Guangzhou, 510630, China
| | - Jin Gong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, 613 West of Huangpu Avenue, Guangzhou, 510630, China.
| |
Collapse
|
3
|
Xu M, Pan G, Zhang Q, Huang J, Wu Y, Ashan Y. FOXM1 boosts glycolysis by upregulating SQLE to inhibit anoikis in breast cancer cells. J Cancer Res Clin Oncol 2025; 151:162. [PMID: 40360780 PMCID: PMC12075401 DOI: 10.1007/s00432-025-06174-1] [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: 12/03/2024] [Accepted: 03/15/2025] [Indexed: 05/15/2025]
Abstract
BACKGROUND Resisting anoikis is a prerequisite for cancer to spread and invade and a major cause of cancer-related deaths. Yet, the intricate mechanisms of how cancer cells evade anoikis remain largely unknown. There is a significant need to explore how these mechanisms play out in breast cancer (BC). METHODS Bioinformatics analysis revealed the expression levels of SQLE and FOXM1 in BC tissue, along with their correlation. The enrichment pathways of SQLE were also explored. qPCR detected the expression of SQLE and FOXM1 in BC cells. CCK-8 assessed cell viability, while flow cytometry measured anoikis. Western blot was employed to examine the protein expression of key genes in glycolytic metabolism and apoptosis-related proteins. Extracellular acidification rate was quantified, and corresponding kits evaluated glucose consumption, lactate production, and adenosine triphosphate levels in cells. Dual-luciferase reporter assays and chromatin immunoprecipitation tests unveiled the binding relationship between FOXM1 and SQLE. RESULTS SQLE was found to be highly expressed in BC and enriched in pathways associated with anoikis and glycolysis. SQLE curbed anoikis in BC via the aerobic glycolysis pathway. There was also a direct binding between FOXM1 and SQLE and a positive correlation between their expression. Recovery experiments substantiated that FOXM1 targeted SQLE to suppress anoikis in BC cells. CONCLUSION FOXM1 upregulates SQLE, which in turn mediates glycolysis to suppress anoikis in BC. The FOXM1/SQLE axis is a promising therapeutic target for BC treatment.
Collapse
Affiliation(s)
- Mei Xu
- Radiotherapy Department of Changji Hui Autonomous Prefecture People's Hospital, No. 303 Yan'an North Road, Changji City, Xinjiang Province, 831100, China.
| | - Guozhi Pan
- Cardiothoracic Surgery Department, Changji Hui Autonomous Prefecture People's Hospital, Changji, China
| | - Qian Zhang
- Radiotherapy Department of Changji Hui Autonomous Prefecture People's Hospital, No. 303 Yan'an North Road, Changji City, Xinjiang Province, 831100, China
| | - Jiangming Huang
- Department of Surgical Oncology, Changji People's Hospital, Changji, China
| | - Yehua Wu
- General Surgery Department of Changji Hui Autonomous Prefecture People's Hospital, Changji, China
| | - Yashengjiang Ashan
- Radiotherapy Department of Changji Hui Autonomous Prefecture People's Hospital, No. 303 Yan'an North Road, Changji City, Xinjiang Province, 831100, China
| |
Collapse
|
4
|
Zhu S, Liu Y, Chen H, Zhu X, Liu X, Xu K, Sang Y, Shang L, Chong W, Li L. Mechanism and Therapeutic Progress of One-Carbon Metabolic Key Enzyme: Serine Hydroxymethyltransferase 2 in Cancer. Clin Med Insights Oncol 2025; 19:11795549251331755. [PMID: 40337354 PMCID: PMC12056339 DOI: 10.1177/11795549251331755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 03/13/2025] [Indexed: 05/09/2025] Open
Abstract
Serine hydroxymethyltransferase 2 (SHMT2) is a crucial mitochondrial enzyme in 1-carbon (1C) metabolism. It catalyzes the conversion of serine to glycine, generating 1C units essential for purine and pyrimidine synthesis, thereby supporting DNA replication and repair. Abnormally high expression is associated with malignant progression and treatment tolerance in various cancers. This review systematically summarizes the functions of SHMT2 in different types of cancer, underscoring on its roles in metabolism, immune microenvironment, and key signaling pathways (PI3K/AKT/mTOR, JAK-STAT, etc.) and outlines its epigenetic regulation and posttranslational modification mechanisms. Compared with the existing research, we focused on the latest regulatory mechanisms of SHMT2 and its potential in cancer treatment, such as the development and application of small-molecule inhibitors (SHIN2 and AGF347).
Collapse
Affiliation(s)
- Siqiang Zhu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yuan Liu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hao Chen
- Clinical Research Center of Shandong University, Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China
| | - Xingyu Zhu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xinyu Liu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Kang Xu
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Yaodong Sang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Liang Shang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Wei Chong
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Shandong Provincial Laboratory of Translational Medicine Engineering for Digestive Tumors, Shandong Provincial Hospital, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
5
|
Li C, Chen T, Li Y, Zhou C, Du J, Li X, Tang C, Ma C, Deng N, Cui H. Impact of diabetes and metformin on cuproptosis and ferroptosis in breast cancer patients: an immunohistochemical analysis. Discov Oncol 2025; 16:634. [PMID: 40299142 PMCID: PMC12040783 DOI: 10.1007/s12672-025-02425-2] [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: 12/09/2024] [Accepted: 04/18/2025] [Indexed: 04/30/2025] Open
Abstract
OBJECTIVES Breast cancer patients with diabetes are often associated with poor prognosis. This study aims to investigate the role of metformin in ferroptosis and cuproptosis in diabetic breast cancer patients and explore its potential impact on clinical outcomes. METHODS We retrospectively analyzed tissue samples from 16 breast cancer patients, including 5 non-diabetic and 11 diabetic patients (6 treated with metformin). Immunohistochemistry (IHC) staining was performed for cuproptosis (FDX1, DLAT), ferroptosis (ACSL4, GPX4), and glycolysis markers (LDHA, PKM2). Statistical analysis used quantitative results from immunohistochemistry. RESULTS Patients treated with metformin showed significantly higher expression of FDX1 and ACSL4, along with a significant decrease in GPX4 compared to other groups. Kaplan-Meier survival analysis revealed that high FDX1 expression was associated with longer survival in breast cancer patients. Correlation analysis showed a positive association between ACSL4 and FDX1 (R = 0.51, P = 0.045), suggesting a relationship between these markers. CONCLUSIONS Metformin may simultaneously enhance both cuproptosis and ferroptosis in breast cancer. FDX1 expression could serve as a prognostic marker for survival, especially in diabetic patients, providing insights into targeting metabolic and cell death pathways in breast cancer therapy.
Collapse
Affiliation(s)
- Changwen Li
- The Affiliated Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, 221009, Jiangsu, People's Republic of China
- Department of Breast Surgery, Xuzhou Central Hospital, No 199 South Jiefang Road, Xuzhou, 221009, Jiangsu, People's Republic of China
| | - Tao Chen
- The Affiliated Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, 221009, Jiangsu, People's Republic of China
- Department of Breast Surgery, Xuzhou Central Hospital, No 199 South Jiefang Road, Xuzhou, 221009, Jiangsu, People's Republic of China
| | - Yuanyuan Li
- The Affiliated Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, 221009, Jiangsu, People's Republic of China
- Department of Breast Surgery, Xuzhou Central Hospital, No 199 South Jiefang Road, Xuzhou, 221009, Jiangsu, People's Republic of China
| | - Chunyan Zhou
- The Affiliated Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, 221009, Jiangsu, People's Republic of China
| | - Jing Du
- School of Life Sciences, Jiangsu Normal University, Xuzhou, 221116, Jiangsu, People's Republic of China
| | - Xiaoxin Li
- Department of Pathology, Xuzhou Central Hospital, The Affiliated Xuzhou Hospital of Medical College of Southeast University, Xuzhou, 221009, Jiangsu, People's Republic of China
| | - Chuangang Tang
- Department of Breast Surgery, Xuzhou Central Hospital, No 199 South Jiefang Road, Xuzhou, 221009, Jiangsu, People's Republic of China
| | - Cheng Ma
- Department of Gastrointestinal Surgery, Xuzhou Central Hospital, Xuzhou, 221009, Jiangsu, People's Republic of China
| | - Na Deng
- Department of Breast Surgery, Xuzhou Central Hospital, No 199 South Jiefang Road, Xuzhou, 221009, Jiangsu, People's Republic of China.
| | - Huaixin Cui
- The Affiliated Xuzhou Clinical College of Xuzhou Medical University, Xuzhou, 221009, Jiangsu, People's Republic of China.
- Department of Breast Surgery, Xuzhou Central Hospital, No 199 South Jiefang Road, Xuzhou, 221009, Jiangsu, People's Republic of China.
| |
Collapse
|
6
|
Sakalauskaite G, Weingartner M, Ebert S, Boot G, Bock T, Birk J, Tsachaki M, Gallon JW, Piscuoglio S, Odermatt A. A BioID-based approach uncovers the interactome of hexose-6-phosphate dehydrogenase in breast cancer cells and identifies anterior gradient protein 2 as an interacting partner. Cell Biosci 2025; 15:54. [PMID: 40281598 PMCID: PMC12032772 DOI: 10.1186/s13578-025-01388-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Hexose-6-phosphate dehydrogenase (H6PD) catalyzes the first two steps of the pentose-phosphate-pathway (PPP) within the endoplasmic reticulum, generating NADPH. H6PD modulates essential physiological processes, including energy and redox metabolism. Its sole reported interacting partner is 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1), utilizing NADPH to reactivate glucocorticoids, linking energy status with hormonal response. Previous studies showed that loss of H6PD affects breast cancer cell properties, independent of 11β-HSD1. It remains unknown whether this is due to impaired concentrations of NADPH or PPP products downstream of H6PD. To gain insight into novel roles and pathways influenced by this enzyme, we aimed to assess the H6PD interactome. RESULTS We adapted the proximity-dependent Biotin Identification (BioID) method to identify novel H6PD interacting partners. First, we validated the method and confirmed the known interaction between H6PD and 11β-HSD1. Next, we constructed a triple-negative breast cancer MDA-MB-231 cell clone stably expressing a H6PD-biotin ligase fusion protein. Enriched biotinylated proteins were analyzed by mass-spectrometry and potential candidates assessed further by co-immunoprecipitation and functional assays. The resulting interactome revealed proteins of the calreticulin/calnexin cycle, unfolded-protein response (UPR) and chaperone activation pathways. Due to its known association with breast cancer, we examined the PDI Anterior gradient protein 2 (AGR2) as H6PD interacting partner. Gene set enrichment analysis revealed multiple overlapping pathways enriched in breast cancer tissues with relatively high H6PD and AGR2 expression. These included glycolysis, fatty acid metabolism, hypoxia, angiogenesis and epithelial to mesenchymal transition. Co-immunoprecipitation (Co-IP) from MCF7 cells confirmed a physical interaction between H6PD and AGR2. ARG2 knockdown in these cells increased H6PD protein levels but decreased activity. Coexpression with AGR2 in HEK-293 cells did not affect expression but enhanced H6PD activity. CONCLUSION BioID was successfully applied in the endoplasmic reticulum to identify AGR2 as H6PD interactor. This was confirmed using Co-IP from MCF7 cells endogenously expressing both proteins. The results indicate that AGR2 controls H6PD protein expression and enhances its activity. Whether higher H6PD activity due to increased AGR2 expression promotes a more aggressive cancer cell phenotype, for example by altering energy metabolism, Ca2+-related processes or UPR and chaperone activation pathways, warrants further investigations.
Collapse
Affiliation(s)
- Gabriele Sakalauskaite
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Michael Weingartner
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Sophie Ebert
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Gina Boot
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Thomas Bock
- Proteomics Core Facility, Biozentrum, University of Basel, 4056, Basel, Switzerland
| | - Julia Birk
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - Maria Tsachaki
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland
| | - John W Gallon
- Visceral Surgery and Precision Medicine Research Laboratory, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Alex Odermatt
- Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056, Basel, Switzerland.
| |
Collapse
|
7
|
Zhang C, Chen L. 6-Methoxyflavone inhibits glycolytic energy metabolism in HeLa cells. BMC Cancer 2025; 25:719. [PMID: 40247232 PMCID: PMC12004806 DOI: 10.1186/s12885-025-14133-9] [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: 01/16/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Enhanced glycolytic levels in cancer cells are a common characteristic of many cancer types. Modulation of glycolytic metabolism is crucial for enhancing the efficacy of cancer therapy. The specific role of 6-methoxyflavone in regulating glycolytic metabolism in cancer cells remains unclear. This study aimed to elucidate the impact of 6-methoxyflavone on glycolytic metabolism in cervical cancer cells and its clinical relevance. METHODS The tandem mass tag (TMT) proteomic analysis was used to identify significantly enriched biological processes and pathways in HeLa cells after treatment with 6-methoxyflavone. Additionally, the differential expression of glycolysis-related proteins was validated using parallel reaction monitoring (PRM) proteomics. Untargeted and targeted metabolomics analyses were used to identify differentially expressed glycolysis-related metabolites. Furthermore, alternative splicing, new transcripts, and domain analyses were used to detect the effects of 6-methoxyflavone on the structures of glycolysis-related genes and proteins. Subcellular localization, molecular docking, and non-covalent interaction analyses were used to detect the subcellular localization, affinity of 6-methoxyflavone for glycolysis-related proteins, and sites of non-covalent interactions. Clinical characteristics and immunological correlation analyses were used to elucidate the relationships between glycolysis-related genes and clinicopathological characteristics, survival, prognosis, and immune-related indicators of patients with cervical cancer. Finally, glycolysis stress tests and enzyme activity assays were used to verify the effect of 6-methoxyflavone on glycolysis in HeLa cells. RESULTS TMT and PRM proteomics, as well as untargeted and targeted metabolomics results, showed that 6-methoxyflavone downregulated the expression levels of glycolysis-related proteins and metabolites in HeLa cells, and that the structures and functions of glycolysis-related genes and proteins in the cytoplasm underwent changes. 6-Methoxyflavone had a good affinity for nine glycolysis-related proteins, all of which had non-covalent interaction sites. Clinical characteristics and immune correlation analyses showed relationships between 6-methoxyflavone and five clinical characteristics, survival prognosis, and four immune-related indicators in patients with cervical cancer. After treatment with 6-methoxyflavone, the basal glycolytic level, maximum glycolytic capacity, and glycolytic reserve of HeLa cells were downregulated. Additionally, 6-methoxyflavone inhibited the activity of pyruvate kinase. CONCLUSION 6-Methoxyflavone inhibited energy metabolism in HeLa cells through the glycolysis pathway. 6-Methoxyflavone may be related to five clinical characteristics, prognosis, tumor microenvironment, immune cells, immune checkpoints, and immunotherapy efficacy in patients with cervical cancer.
Collapse
Affiliation(s)
- Chaihong Zhang
- Department of Obstetrics and Gynecology, Shaanxi Provincial People's Hospital, 256 Youyi West Road, Xi'an City, Shaanxi Province, 710000, China
| | - Lihong Chen
- Department of Obstetrics and Gynecology, Shaanxi Provincial People's Hospital, 256 Youyi West Road, Xi'an City, Shaanxi Province, 710000, China.
| |
Collapse
|
8
|
Liu L, Xing G, Guo X, Chen H, Li J, Wang J, Li Y, Liang G, Liu M. Inhibition of colorectal cancer cell growth by downregulation of M2-PK and reduction of aerobic glycolysis by clove active ingredients. Front Pharmacol 2025; 16:1552486. [PMID: 40308769 PMCID: PMC12041220 DOI: 10.3389/fphar.2025.1552486] [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: 12/28/2024] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
Exploring the anti-tumor molecular mechanisms of traditional Chinese medicines has become an important strategy to develop novel anti-tumor drugs in the clinic. Several pharmacological studies have reported the antioxidant, antibacterial, anti-inflammatory, and anti-tumor effects of clove. Previously, we have shown that the active fraction from clove (AFC) can inhibit the growth of tumor cells, particularly colon cancer cells, in vitro. However, the mechanism of action regarding the anti-colon cancer activity of AFC, especially in aerobic glycolysis, has not been adequately investigated. In this study, we found that AFC significantly inhibited the growth of five types of colon cancer cells, downregulated the mRNA and protein levels of M2-type pyruvate kinase (PKM2), and reduced aerobic glycolysis capacity. Transfection of PKM2-siRNA mimicked the inhibitory effects of AFC on aerobic glycolysis in colon cancer cells. Furthermore, the highly expressed, tumor-specific targets c-myc and cyclin D1 in cells were also found to be downregulated following the action of AFC. In the HCT116 cell xenograft nude mice models, the results after AFC administration were consistent with those of the cellular experiments, while AFC caused less liver injury and weight loss than the conventional chemotherapeutic agent 5- fluorouracil (5-FU). In conclusion, AFC inhibits colon cancer growth by downregulating PKM2 to inhibit aerobic glycolysis and reduce the tumor-specific high expression of c-myc and cyclin D1. Future work should explore how it downregulates pyruvate kinase (PK) in the first place, along with the intrinsic mechanism between the downregulation of PKM2 and the downregulation of c-myc.
Collapse
Affiliation(s)
- Lin Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
- Drug Dispending Department, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, China
| | - Gang Xing
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaoyi Guo
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jian Li
- Pharmacy Department, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Jian Wang
- Discipline Construction Office, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Yaling Li
- Pharmacy Department, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Gang Liang
- Pharmacy Department, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Minghua Liu
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
9
|
Pan Z, Liu Y, Li H, Qiu H, Zhang P, Li Z, Wang X, Tian Y, Feng Z, Zhu S, Wang X. The role and mechanism of aerobic glycolysis in nasopharyngeal carcinoma. PeerJ 2025; 13:e19213. [PMID: 40191756 PMCID: PMC11971989 DOI: 10.7717/peerj.19213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
This review delves into the pivotal role and intricate mechanisms of aerobic glycolysis in nasopharyngeal carcinoma (NPC). NPC, a malignancy originating from the nasopharyngeal epithelium, displays distinct geographical and clinical features. The article emphasizes the significance of aerobic glycolysis, a pivotal metabolic alteration in cancer cells, in NPC progression. Key enzymes such as hexokinase 2, lactate dehydrogenase A, phosphofructokinase 1, and pyruvate kinase M2 are discussed for their regulatory functions in NPC glycolysis through signaling pathways like PI3K/Akt and mTOR. Further, the article explores how oncogenic signaling pathways and transcription factors like c-Myc and HIF-1α modulate aerobic glycolysis, thereby affecting NPC's proliferation, invasion, metastasis, angiogenesis, and immune evasion. By elucidating these mechanisms, the review aims to advance research and clinical practice in NPC, informing the development of targeted therapeutic strategies that enhance treatment precision and reduce side effects. Overall, this review offers a broad understanding of the multifaceted role of aerobic glycolysis in NPC and its potential impact on therapeutic outcomes.
Collapse
Affiliation(s)
- Zhiyong Pan
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuyi Liu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Hui Li
- Department of Ophthalmology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Huisi Qiu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Pingmei Zhang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhiying Li
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xinyu Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuxiao Tian
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhengfu Feng
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Song Zhu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xin Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| |
Collapse
|
10
|
Zhang C, Liu C, Wang Z, Wang D, Chen W, Li J, Lu Q, Zhou J, Chang Y, Wang P, Gao Y, Wang J, Zhi H, Ning S. Comprehensive characterization of respiratory genes based on a computational framework in pan-cancer to develop stratified treatment strategies. PLoS Comput Biol 2025; 21:e1012963. [PMID: 40202958 PMCID: PMC11981224 DOI: 10.1371/journal.pcbi.1012963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
Abnormal cellular respiration plays a critical role in carcinogenesis. However, the molecular mechanisms underlying dysregulation of respiratory gene expression across different cancer types remain unclear. Here, we developed a computational framework that provides an analytical approach for exploring the molecular alterations and clinical relevance of respiratory genes in pan-cancer. We identified a total of 53 gene signatures in the three stages of respiration (including glycolysis, tricarboxylic acid cycle, and oxidative phosphorylation) through this framework and found that they were broadly differentially expressed and genetically altered across 33 cancer types. Pathway analysis manifested that the expression levels of almost all respiratory gene signatures were remarkably associated with the activation or inhibition of numerous oncogenic pathways, such as metabolism, angiogenesis, cell proliferation, and apoptosis. Survival analysis highlighted the oncogenic or tumor suppressor potential of the respiratory gene signatures. In particular, VCAN has shown significant oncogenic features in multiple cancer types. Finally, we identified a number of respiratory gene signatures that could be potential therapeutic targets, including VCAN. We also predicted small-molecule compounds targeting respiratory gene signatures or components of pathways regulated by them. Overall, our comprehensive analysis has greatly enhanced the understanding of molecular alterations of respiratory genes in tumorigenesis and progression, and provided insights into developing new therapeutic strategies.
Collapse
Affiliation(s)
- Caiyu Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Chenyu Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhuoru Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Di Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wenli Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jian Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Qianyi Lu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiajun Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yetong Chang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Peng Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yue Gao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Junwei Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hui Zhi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shangwei Ning
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
11
|
Lim CH, Lee JH, Lee J, Park SB. Predictive value of 18F-fluorodeoxyglucose uptake for axillary lymph node metastasis in operable breast cancer: impact of molecular subtypes. Ann Nucl Med 2025; 39:315-322. [PMID: 39623100 DOI: 10.1007/s12149-024-02002-7] [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: 08/26/2024] [Accepted: 11/10/2024] [Indexed: 03/18/2025]
Abstract
OBJECTIVES To evaluate the predictive value of standardized uptake value (SUV) in both primary tumors and axillary lymph nodes (ALNs) using FDG PET/CT for lymph node metastasis in breast cancer patients, and to assess the influence of molecular subtypes on this predictive performance. METHODS This retrospective study included 287 patients with invasive ductal carcinoma (IDC) who underwent FDG PET/CT prior to surgery between September 2016 and December 2019. The maximum standardized uptake value (SUVmax) of primary tumors (SUV-B) and ALNs (SUV-LN) were analyzed. Molecular subtypes were classified as hormone receptor-positive, HER2-positive, and triple-negative breast cancer (TNBC). Receiver operating characteristic (ROC) curve analysis was performed to assess and compare the diagnostic performance of SUV-B and SUV-LN for predicting ALN metastasis. RESULTS Among the 287 patients, 62 (21.6%) had confirmed ALN metastasis. The median SUV-LN was significantly higher in patients with metastasis compared to those without metastasis (1.5 vs. 0.9; P < 0.001). SUV-LN demonstrated good discriminative performance for ALN metastasis (AUC: 0.796), whereas SUV-B did not show significant predictive value (AUC: 0.536). The SUV_LN demonstrated significantly lower predictive performance for ALN metastasis in the hormone-positive group (AUC: 0.796) compared to the excellent discriminative performance in the HER2-positive (AUC: 0.923, P = 0.018) and TNBC (AUC: 0.940, P = 0.004) groups. Hormone receptor-positive tumors also exhibited lower FDG uptake in metastatic lymph nodes compared to HER2-positive and TNBC subtypes (P = 0.031). CONCLUSION FDG PET/CT SUV-LN effectively predicts ALN metastasis in HER2-positive and TNBC subtypes. Hormone receptor-positive breast cancers show lower FDG uptake in metastatic ALNs, reducing diagnostic accuracy. This finding may aid in selecting the most appropriate diagnostic modality based on tumor characteristics in the era of personalized medicine.
Collapse
Affiliation(s)
- Chae Hong Lim
- Department of Nuclear Medicine, Soonchunhyang University Seoul Hospital, Seoul, 04401, Republic of Korea
| | - Jun-Hee Lee
- Department of Surgery, Soonchunhyang University Seoul Hospital, Seoul, 04401, Republic of Korea
| | - Jihyoun Lee
- Department of Surgery, Soonchunhyang University Seoul Hospital, Seoul, 04401, Republic of Korea
| | - Soo Bin Park
- Department of Nuclear Medicine, Soonchunhyang University Seoul Hospital, Seoul, 04401, Republic of Korea.
| |
Collapse
|
12
|
Venkatesh D, Sarkar S, Kandasamy T, Ghosh SS. In-silico identification and validation of Silibinin as a dual inhibitor for ENO1 and GLUT4 to curtail EMT signaling and TNBC progression. Comput Biol Chem 2025; 115:108312. [PMID: 39689434 DOI: 10.1016/j.compbiolchem.2024.108312] [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: 07/20/2024] [Revised: 11/29/2024] [Accepted: 12/06/2024] [Indexed: 12/19/2024]
Abstract
The aberrant metabolic reprogramming endows TNBC cells with sufficient ATP and lactate required for survival and metastasis. Hence, the intervention of the metabolic network represents a promising avenue to alleviate the Warburg effect in TNBC cells to impair their invasive and metastatic potential. Multitudinous in-silico analysis identified Enolase1 (ENO1) and the surface transporter protein, GLUT4 to be the potential targets for the abrogation of the metabolic network. The expression profiles of ENO1 and GLUT4 genes showed anomalous expression in various cancers, including breast cancer. Subsequently, the functional and physiological interactions of the target proteins were analyzed from the protein-protein interaction network. The pathway enrichment analysis identified the prime cancer signaling pathways in which these proteins are involved. Further, docking results bestowed Silibinin as the concurrent inhibitor of ENO1 and GLUT4. Moreover, the stable interaction of Silibinin with both proteins deciphered the binding free energies values of -48.86 and -104.31 KJ/mol from MMPBSA analysis and MD simulation, respectively. Furthermore, the cell viability, ROS assay, and live-dead imaging underscored the pronounced cytotoxicity of Silibinin, illuminating its capacity to incur apoptosis within TNBC cells. Additionally, glycolysis assay and gene expression analysis demonstrated the silibinin-mediated inhibition of the glycolysis pathway. Eventually, a lipidomic reprogramming towards fatty acid metabolism was established from the elevated lipid droplet accumulation, exogenous fatty acid uptake and de-novo lipogenesis. Nevertheless, repression of EMT and Wnt pathway progression by Silibinin was perceived from the gene expression studies. Overall, the current study highlights the tweaking of intricate signaling crosstalk between glycolysis and the Wnt pathway in TNBC cells through inhibiting ENO1 and GLUT4.
Collapse
Affiliation(s)
- Dheepika Venkatesh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India
| | - Shilpi Sarkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India
| | - Thirukumaran Kandasamy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India; Centre for Nanotechnology, Indian Institute of Technology Guwahati, 39, Guwahati, Assam, India.
| |
Collapse
|
13
|
Geng C, Yan L, Li Y, Li H, Ji Y, Xiao Y, Wang Z, Chen X, Chen C, Yang Q, Tang B, Wang W. Layered Double Hydroxide Nanoparticles Loaded with Resveratrol Inhibit Glycolysis and Show Efficacy in the Treatment of Breast Cancer. Int J Nanomedicine 2025; 20:3423-3444. [PMID: 40125434 PMCID: PMC11927503 DOI: 10.2147/ijn.s492145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
Background Breast cancer is one of the most common cancers among women. Tumor cell proliferation is highly dependent on aerobic glycolysis, so regulating aerobic glycolysis in breast cancer cells is a promising therapeutic strategy. Resveratrol (Res), as a potential new anti-breast cancer drug, has been shown to regulate the glycolysis of cancer cells and inhibit the metastasis and recurrence of breast cancer. The nano drug delivery system can regulate the aerobic glycolysis metabolism by targeting the signaling factors and reaction products of the tumor aerobic glycolysis process to enhance the anti-tumor effect. Methods A new albumin-modified layered double hydroxide resveratrol dosage form (BSA@LDHs-Res) was synthesized by hydrothermal co-precipitation. Characterization was carried out to determine the successful synthesis of the nanocarrier system. The bioactivity, glycolytic activity and biocompatibility were examined by in vitro cellular assays; in vivo experiments were performed to further evaluate the anti-tumor effects of the BSA@LDHs-Res dosage form for breast cancer. Results In this study, we obtained for the first time a bovine serum albumin-modified BSA@LDHs-Res loaded dosage form, which was able to enter breast cancer cells SKBR3 and MDA-MB-231 via endocytosis and successfully escaped from lysosomal capture. BSA@LDHs-Res inhibited the proliferation, migration, and invasion of two types of breast cancer cells, induced apoptosis, and promoted the reduction of mitochondrial membrane potential and ROS. BSA@LDHs-Res inhibited the expression and viability of the key enzymes of glycolysis, hexokinase 2 (HK2), pyruvate kinase (PK), and lactate dehydrogenase, resulting in decreased glucose consumption, decreased lactate accumulation, and decreased intracellular ATP levels. BSA@LDHs-Res was examined in the mouse model with good anti-tumor effects. Conclusion BSA@LDHs-Res is an efficient nanoreagent for the treatment of breast cancer. The albumin-modified resveratrol layered double hydroxide delivery system developed in this study will provide some theoretical references for further research and clinical application of tumor aerobic glycolysis.
Collapse
Affiliation(s)
- Chenchen Geng
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- School of Life Sciences, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
| | - Liuyang Yan
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- School of Life Sciences, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
| | - Yunhao Li
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- School of Basic Courses, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
| | - Houcong Li
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- School of Basic Courses, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
| | - Yuxin Ji
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
| | - Yuhan Xiao
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
| | - Zhifa Wang
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- School of Life Sciences, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
| | - Xiaoqi Chen
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
| | - Changjie Chen
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- Department of Biochemistry and Molecular Biology, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
| | - Qingling Yang
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- Clinical Testing and Diagnose Experimental Center, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- Department of Biochemistry and Molecular Biology, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
| | - Baoding Tang
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- School of Life Sciences, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
| | - Wenrui Wang
- Anhui Provincial Key Laboratory of Tumor Evolution and Intelligent Diagnosis and Treatment, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- School of Life Sciences, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
- Anhui Engineering Research Center for Neural Regeneration Technology and Medical New Materials, Bengbu Medical University, Bengbu, Anhui, People’s Republic of China
| |
Collapse
|
14
|
Li X, Liu B, Wang S, Dong Q, Li J. EDNRB negatively regulates glycolysis to exhibit anti-tumor functions in prostate cancer by cGMP/PKG pathway. Mol Cell Endocrinol 2025; 598:112459. [PMID: 39788311 DOI: 10.1016/j.mce.2025.112459] [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: 10/22/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Prostate cancer (PCa) is the most prevalent cancer in men and the leading cause of cancer-related mortality. Recent studies have highlighted the pivotal role of glycolysis in tumor progression. This study aimed to investigate the involvement of the EDNRB gene and its ligand endothelin 3 (EDN3) in glycolysis in PCa and to elucidate its underlying molecular mechanism. Quantitative reverse transcription PCR (RT-qPCR) and methylation-specific PCR (MSP) were used to probe EDNRB expression and methylation in PCa tissues. Cell proliferation and glycolysis in PCa cells were evaluated using Cell Counting Kit-8 (CCK-8), EDU staining, Seahorse assay, and biochemical kits to analyze the effects of EDN3/EDNRB. The underlying molecular mechanism was further explored through Western blotting. The in vivo effect of EDNRB on tumor growth was examined using a xenograft tumor model. Our findings revealed that EDNRB was hypermethylated and downregulated in PCa tissues and cell lines. Overexpression of EDNRB or EDN3 led to reduced cell proliferation and downregulation of glycolytic markers. EDNRB also decreased the extracellular acidification rate (ECAR) baseline and increased the oxygen consumption rate (OCR) baseline, indicating a shift away from glycolysis. Additionally, the anticancer effects of EDNRB or EDN3 was reversed upon inhibition of the cGMP/PKG pathway. In vivo, enhanced EDNRB expression significantly suppressed tumor growth. Therefore, EDNRB or EDN3 possess anticancer potential in PCa, primarily through the regulation of glycolysis via the cGMP/PKG pathway.
Collapse
Affiliation(s)
- Xun Li
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, 830001, China
| | - Bide Liu
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, 830001, China
| | - Shuheng Wang
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, 830001, China
| | - Qiang Dong
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, 830001, China
| | - Jiuzhi Li
- Department of Urology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, 830001, China.
| |
Collapse
|
15
|
Gao H, Wang J, Liu J, Wang H, Wang T, Li S, Niu L, Wei Y. FOXD1 activates KIFC1 to modulate aerobic glycolysis and reinforce cisplatin resistance of breast cancer. Reprod Biol 2025; 25:100969. [PMID: 39541848 DOI: 10.1016/j.repbio.2024.100969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/24/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Breast cancer (BC) is the most prevalent invasive malignant tumor. Cisplatin (DDP) is a prototype of platinum-based chemotherapy drugs, its resistance severely hinders its clinical application. This project intended to figure out the exact mechanism of KIFC1 in the DDP resistance of BC. METHODS The levels of KIFC1 and FOXD1 in BC as well as their binding sites were investigated by bioinformatics analysis. The signaling pathways regulated by FOXD1 were analyzed. Chromatin immunoprecipitation (ChIP) and dual-luciferase reporter assays verified the binding relationship between the two. Through quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot (WB), we assessed the expression of FOXD1, KIFC1, and glycolysis-related genes. CCK-8 assay was applied in the determination of cell viability to assess the efficacy of DDP resistance. Extracellular acidification rate (ECAR), glucose consumption, lactate synthesis, Adenosine triphosphate (ATP) content, and oxygen consumption rate (OCR) were measured to evaluate glycolysis. RESULTS FOXD1 and KIFC1 were significantly upregulated in BC, with KIFC1 being significantly enriched in the glycolysis pathway. Overexpression of KIFC1 significantly enhanced the DDP resistance of BC cells, while promoting aerobic glycolysis. Mechanistically, FOXD1 was bound to the promoter of KIFC1 to activate its transcription. Its overexpression counteracted the inhibitory effect of KIFC1 knockdown on the DDP resistance of BC cells. CONCLUSION FOXD1 activates the glycolysis pathway by upregulating KIFC1, thereby facilitating BC cells' DDP resistance. Therefore, the FOXD1/KIFC1 axis linked the glycolysis pathway to DDP resistance and may be a promising new target for reinforcing DDP resistance in BC.
Collapse
Affiliation(s)
- Haitao Gao
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Jing Wang
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Jiacai Liu
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Huihua Wang
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Tiantian Wang
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Sha Li
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Lili Niu
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China
| | - Ya Wei
- General Surgery Department, The Affiliated Anyang Tumor Hospital of Henan University of Science and Technology, Anyang 455000, China.
| |
Collapse
|
16
|
Liu T, Jin C, Sun J, Zhu L, Wang C, Xiao F, Liu X, Lv L, Yang X, Zhou W, Tan C, Wang X, Wei W. Paroxetine alleviates dendritic cell and T lymphocyte activation via GRK2-mediated PI3K-AKT signaling in rheumatoid arthritis. Chin Med J (Engl) 2025; 138:441-451. [PMID: 38879805 PMCID: PMC11845201 DOI: 10.1097/cm9.0000000000003165] [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: 12/07/2023] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND G protein-coupled receptor kinase 2 (GRK2) could participate in the regulation of diverse cells via interacting with non-G-protein-coupled receptors. In the present work, we explored how paroxetine, a GRK2 inhibitor, modulates the differentiation and activation of immune cells in rheumatoid arthritis (RA). METHODS The blood samples of healthy individuals and RA patients were collected between July 2021 and March 2022 from the First Affiliated Hospital of Anhui Medical University. C57BL/6 mice were used to induce the collagen-induced arthritis (CIA) model. Flow cytometry analysis was used to characterize the differentiation and function of dendritic cells (DCs)/T cells. Co-immunoprecipitation was used to explore the specific molecular mechanism. RESULTS In patients with RA, high expression of GRK2 in peripheral blood lymphocytes, accompanied by the increases of phosphatidylinositol 3 kinase (PI3K), protein kinase B (AKT), and mammalian target of rapamycin (mTOR). In animal model, a decrease in regulatory T cells (T regs ), an increase in the cluster of differentiation 8 positive (CD8 + ) T cells, and maturation of DCs were observed. Paroxetine, when used in vitro and in CIA mice, restrained the maturation of DCs and the differentiation of CD8 + T cells, and induced the proportion of T regs . Paroxetine inhibited the secretion of pro-inflammatory cytokines, the expression of C-C motif chemokine receptor 7 in DCs and T cells. Simultaneously, paroxetine upregulated the expression of programmed death ligand 1, and anti-inflammatory cytokines. Additionally, paroxetine inhibited the PI3K-AKT-mTOR metabolic pathway in both DCs and T cells. This was associated with a reduction in mitochondrial membrane potential and changes in the utilization of glucose and lipids, particularly in DCs. Paroxetine reversed PI3K-AKT pathway activation induced by 740 Y-P (a PI3K agonist) through inhibiting the interaction between GRK2 and PI3K in DCs and T cells. CONCLUSION Paroxetine exerts an immunosuppressive effect by targeting GRK2, which subsequently inhibits the metabolism-related PI3K-AKT-mTOR pathway of DCs and T cells in RA.
Collapse
Affiliation(s)
- Tingting Liu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230022, China
| | - Chao Jin
- Department of Pharmacy, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Jing Sun
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230022, China
| | - Lina Zhu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Chun Wang
- Institute of Clinical Pharmacology, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, China
| | - Feng Xiao
- Institute of Clinical Pharmacology, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, China
| | - Xiaochang Liu
- Department of Gastroenterology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Liying Lv
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Xiaoke Yang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Wenjing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230022, China
| | - Chao Tan
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
- The Grade 3 Pharmaceutical Chemistry Laboratory of State Administration of Traditional Chinese Medicine, Hefei, Anhui 230022, China
| | - Xianli Wang
- Department of Pharmacy, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai 200090, China
| | - Wei Wei
- Institute of Clinical Pharmacology, Anhui Medical University, The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, Anhui 230032, China
| |
Collapse
|
17
|
Huang R, Li Y, Lin K, Zheng L, Zhu X, Huang L, Ma Y. A novel glycolysis-related gene signature for predicting prognosis and immunotherapy efficacy in breast cancer. Front Immunol 2025; 16:1512859. [PMID: 40046063 PMCID: PMC11880812 DOI: 10.3389/fimmu.2025.1512859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/29/2025] [Indexed: 05/13/2025] Open
Abstract
Background Previous studies have shown that glycolysis-related genes (GRGs) are associated with the development of breast cancer (BC), and the prognostic significance of GRGs in BC has been reported. Considering the heterogeneity of BC patients, which makes prognosis difficult to predict, and the fact that glycolysis is regulated by multiple genes, it is important to establish and evaluate new glycolysis-related prediction models in BC. Methods In total, 170 GRGs were selected from the GeneCards database. We analyzed data from the Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) database as a training set and data from the Gene Expression Omnibus (GEO) database as a validation cohort. Based on the overall survival data and the expression levels of GRGs, Cox regression analyses were applied to develop a glycolysis-related prognostic gene (GRPGs)-based prediction model. Kaplan (KM) survival and ROC analyses were performed to assess the performance of this model. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were used to identify the potential biological functions of GRPGs. cBioPortal database was used to explore the tumor mutation burden (TMB). The tumor immune dysfunction and exclusion indicator (TIDE) was used to estimate the patient response to immune checkpoint blockade (ICB). The levels of tumor-infiltrating immune cells (TICs) and stromal cells were quantitatively analyzed based on gene expression profiles. Results We constructed a prediction model of 10 GRPGs (ADPGK, HNRNPA1, PGAM1, PIM2, YWHAZ, PTK2, VDAC1, CS, PGK1, and GAPDHS) to predict the survival outcomes of patients with BC. Patients were divided into low- and high-risk groups based on the gene signature. The AUC values of the ROC curves were 0.700 (1-year OS), 0.714 (3-year OS), 0.681 (5-year OS). TMB and TIDE analyses showed that patients in the high-risk group might respond better to ICB. Additionally, by combining the GRPGs signature and clinical characteristics of patients, a novel nomogram was constructed. The AUC values for this combined prediction model were 0.827 (1-year OS), 0.792 (3-year OS), and 0.783 (5-year OS), indicating an outstanding predictive performance. Conclusion A new GRPGs based prediction model was built to predict the OS and immunotherapeutic response of patients with BC.
Collapse
Affiliation(s)
- Rui Huang
- Clinical Laboratory, Jinan Children’s Hospital, Jinan, Shandong, China
- Clinical Laboratory, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yi Li
- The First Clinical College of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Kaige Lin
- The 960 Hospital of the Chinese People's Liberation Army (PLA) Joint Logistics Support Force, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Luming Zheng
- Department of General Surgery, the 960 Hospital of the Chinese People's Liberation Army (PLA) Joint Logistics Support Force, Jinan, Shandong, China
| | - Xiaoru Zhu
- Clinical Laboratory, Jinan Children’s Hospital, Jinan, Shandong, China
- Clinical Laboratory, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Leqiu Huang
- Clinical Laboratory, Jinan Children’s Hospital, Jinan, Shandong, China
- Clinical Laboratory, Children’s Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Yunhan Ma
- Department of General Surgery, the 960 Hospital of the Chinese People's Liberation Army (PLA) Joint Logistics Support Force, Jinan, Shandong, China
| |
Collapse
|
18
|
He M, Zhang Y, Zhai Y, Li Y, Yang G, Yu S, Xiao H, Song Y. Trilobatin regulates glucose metabolism by ameliorating oxidative stress and insulin resistance in vivo and in vitro. J Pharm Pharmacol 2025; 77:236-248. [PMID: 38642915 DOI: 10.1093/jpp/rgae035] [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: 11/22/2023] [Accepted: 03/15/2024] [Indexed: 04/22/2024]
Abstract
OBJECTIVES Trilobatin, a glycosylated dihydrochalcone, has been reported to have anti-diabetic properties. However, the underlying mechanism remains unexplained. METHODS In this investigation, the regulation of trilobatin on glucose metabolism of insulin resistance (IR)-HepG2 cells and streptozocin (STZ)-induced mice and its mechanism were evaluated. KEY FINDINGS Different doses of trilobatin (5, 10 and 20 μM) increased glucose consumption, glycogen content, hexokinase (HK), and pyruvate kinase (PK) activity in IR-HepG2 cells. Among them, the HK and PK activity in IR-HepG2 cells treated with 20 μM trilobatin were 1.84 and 2.05 times than those of the IR-group. The overeating, body and tissue weight, insulin levels, liver damage, and lipid accumulation of STZ-induced mice were improved after feeding with different doses of trilobatin (10, 50, and 100 mg/kg/d) for 4 weeks. Compared with STZ-induced mice, fasting blood glucose decreased by 61.11% and fasting insulin (FINS) increased by 48.6% after feeding trilobatin (100 mg/kg/d). Meanwhile, data from quantitative real-time polymerase chain reaction (qRT-PCR) revealed trilobatin ameliorated glycogen synthesis via the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/glycogen synthase kinase-3β (GSK-3β) signaling pathway in IR-HepG2 cells and in STZ-induced mice. Furthermore, in vitro and in vivo experiments showed that trilobatin ameliorated oxidative stress by regulating the mRNA expression of nuclear erythroid-2 related factor 2 (Nrf2)/kelch-like ECH associated protein-1 (Keap-1) pathway as well as heme oxygenase-1 (HO-1) and NAD(P)H: quinone oxidoreductase-1 (NQO-1). CONCLUSIONS Our research reveals a novel pharmacological activity of trilobatin: regulating glucose metabolism through PI3K/Akt/GSK-3β and Nrf2/Keap-1 signaling pathways, improving insulin resistance and reducing oxidative stress. Trilobatin can be used as a reliable drug resource for the treatment of glucose metabolism disorders.
Collapse
Affiliation(s)
- Ming He
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Yuqing Zhang
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Yuhan Zhai
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Yaping Li
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Guorui Yang
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Shaoxuan Yu
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Haifang Xiao
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| | - Yuanda Song
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, Shandong 255049, China
| |
Collapse
|
19
|
Jin Z, Yun L, Cheng P. Tanshinone I reprograms glycolysis metabolism to regulate histone H3 lysine 18 lactylation (H3K18la) and inhibits cancer cell growth in ovarian cancer. Int J Biol Macromol 2025; 291:139072. [PMID: 39710022 DOI: 10.1016/j.ijbiomac.2024.139072] [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: 10/22/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Salvia miltiorrhiza, the anticancer properties of these components are multifaceted, encompassing the inhibition of tumor growth, prevention of the metastatic spread of cancer cells, enhancement of the sensitivity of cancer cells to chemotherapy and radiation therapy, and the suppression of angiogenesis, which is crucial for tumor growth and survival. In the context of our recent study, we have discovered that tanshinone I, one of the active components of Salvia miltiorrhiza, possesses the ability to inhibit the proliferation of ovarian cancer cells, both in laboratory settings and within living organisms. To further understand the molecular mechanisms behind this effect, we conducted a comprehensive transcriptomic analysis. Our findings indicated that tanshinone I exerts its inhibitory action by downregulating the expression of genes associated with glycolysis. Specifically, tanshinone I decreased the expression of glycolysis-related genes such as HK2 (hexokinase 2), PFK (phosphofructokinase), ENO2 (enolase 2), and LDHA (lactate dehydrogenase A). Inhibiting lactate production by tanshinone I application reduced the level of histone H3 lysine 18 lactylation (H3K18la), which reduced the expression of tumor-associated genes, such as TTK, PDGFRβ, YTHDF2 and RUBCNL. In addition, tanshinone I alleviated the immunosuppressive tumor microenvironment. In summary, tanshinone I blocks glycolysis to regulate histone H3 lysine 18 lactylation (H3K18la), which inhibits ovarian cancer cell growth, revealing the anticancer mechanism of tanshinone I.
Collapse
Affiliation(s)
- Zhou Jin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lin Yun
- Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Peng Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China; Basic Medical College, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
20
|
Adnan SF, Najim Al-Abady ZN. Therapeutic Targeting of PARP Expression and Glycolysis Rate-Limiting Enzymes in Breast Cancer Patients. Asian Pac J Cancer Prev 2025; 26:611-617. [PMID: 40022708 PMCID: PMC12118002 DOI: 10.31557/apjcp.2025.26.2.611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/22/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND Breast cancer is a heterogeneous disease characterized by diverse biochemical, histological, and clinical features. PARP1 and glycolysis rate-limiting enzymes play critical roles in cancer progression, making them promising therapeutic targets. AIM This study aimed to evaluate the expression levels of PARP1 and key glycolytic enzymes (HK, PFK, and PK) in breast cancer patients and assess their potential as therapeutic indicators. MATERIALS AND METHODS A total of 120 participants (60 breast cancer patients and 60 healthy controls) were included in the study. Blood samples were collected to measure PARP1 expression and the levels of glycolytic enzymes using ELISA. Statistical analyses were performed to compare the two groups. RESULTS PARP1 expression and glycolytic enzyme levels (HK, PFK, and PK) were significantly higher in breast cancer patients compared to healthy controls (p < 0.0001). CONCLUSION The overexpression of PARP1 and key glycolytic enzymes indicates their involvement in breast cancer progression and underscores their potential as therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Shams Firas Adnan
- Department of Chemistry, Faculty of Sciences, University of Al-Qadisiyah, Iraq.
| | | |
Collapse
|
21
|
Mao C, Liu X, Guo SW. Reduced endometrial glycolysis concomitant with increased lesional fibrosis in patients with adenomyosis who complained of heavy menstrual bleeding. Reprod Biomed Online 2025; 50:104406. [PMID: 39523182 DOI: 10.1016/j.rbmo.2024.104406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 11/16/2024]
Abstract
RESEARCH QUESTION What role, if any, does the extent of lesional fibrosis play in impaired glycolysis leading to adenomyosis-associated heavy menstrual bleeding (ADM-HMB)? DESIGN Forty-eight patients with ADM-HMB were recruited, among them 25 reported moderate to heavy bleeding (MHB), and the remaining 23, excessive bleeding (EXB). The full-thickness uterine tissue columns were processed for Masson trichrome staining and immunohistochemistry analyses. The expression levels of HIF-1α, GLUT1, HK2, PFKFB3 and PKM2 proteins that are critically involved in glycolysis in endometrial epithelial cells cultured on substrates of different stiffness, and the levels of glycolysis were quantitated. A mouse experiment with induced adenomyosis and simulated menstrual bleeding was conducted to assess the effect of adenomyosis on immunoexpression of proteins involved in glycolysis and inflammation as well as on endometrial repair and bleeding. RESULTS The endometrial staining of HIF-1α, GLUT1, HK2, PFKFB3 and PKM2 was significantly lower in the EXB group as compared with MHB patients, concomitant with higher extent of fibrosis. The expression of HIF-1α, GLUT1, HK2, PFKFB3 and PKM2 was significantly reduced when endometrial epithelial cells were cultured in stiff substrate, concomitant with reduced glycolysis. Mice with induced adenomyosis had reduced immunoexpression of Hif-1α, as well as those proteins each of which plays a vital, rate-limiting role in different steps of the glycolysis pathway, such as Glut1, Hk2, Pfkfb3 and Pkm2, and elevated fibrosis in endometrium, concomitant with disrupted endometrial repair and more bleeding. CONCLUSIONS Lesional fibrosis results in reduced endometrial glycolysis in eutopic endometrium and subsequent imbalance in pro-inflammatory and anti-inflammatory response, leading to ADM-HMB.
Collapse
Affiliation(s)
- Chenyu Mao
- Department of Gynecology, Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Xishi Liu
- Department of Gynecology, Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China.; Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| | - Sun-Wei Guo
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China.; Research Institute, Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China.
| |
Collapse
|
22
|
Zhang Y, Xu H, Han X, Yu Q, Zheng L, Xiao H. PMAIP1-mediated glucose metabolism and its impact on the tumor microenvironment in breast cancer: Integration of multi-omics analysis and experimental validation. Transl Oncol 2025; 52:102267. [PMID: 39740516 PMCID: PMC11750568 DOI: 10.1016/j.tranon.2024.102267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Glucose metabolism in breast cancer has a potential effect on tumor progression and is related to the immune microenvironment. Thus, this study aimed to develop a glucose metabolism-tumor microenvironment score to provide new perspectives on breast cancer treatment. METHOD Data were acquired from the Gene Expression Omnibus and UCSC Xena databases, and glucose-metabolism-related genes were acquired from the Gene Set Enrichment Analysis database. Genes with significant prognostic value were identified, and immune infiltration analysis was conducted, and a prognostic model was constructed based on the results of these analyses. The results were validated by in vitro experiments with MCF-7 and MCF-10A cell lines, including expression validation, functional experiments, and bulk sequencing. Single-cell analysis was also conducted to explore the role of specific cell clusters in breast cancer, and Bayes deconvolution was used to further investigate the associations between cell clusters and tumor phenotypes of breast cancer. RESULTS Four significant prognostic genes (PMAIP1, PGK1, SIRT7, and SORBS1) were identified, and, through immune infiltration analysis, a combined prognostic model based on glucose metabolism and immune infiltration was established. The model was used to classify clinical subtypes of breast cancer, and PMAIP1 was identified as a potential critical gene related to glucose metabolism in breast cancer. Single-cell analysis and Bayes deconvolution jointly confirmed the protective role of the PMAIP1+ luminal cell cluster.
Collapse
Affiliation(s)
- Yidong Zhang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China; School of Pharmacy, Queen's University of Belfast, Belfast, Northern Ireland, United Kingdom
| | - Hang Xu
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China; Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xuedan Han
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Qiyi Yu
- Mellon College of Science, Carnage Mellon University, Pittsburgh, Pennsylvania, USA
| | - Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China.
| | - Hua Xiao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
23
|
Mei B, Zeng Z, Xia Q, Liu M, Zhang Y. The role of the circ_DOCK1-miR-1297-HOXA9 regulatory network in the development of oral squamous cell carcinoma. Pathol Res Pract 2025; 266:155752. [PMID: 39721095 DOI: 10.1016/j.prp.2024.155752] [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: 06/17/2024] [Revised: 10/24/2024] [Accepted: 12/01/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE Oral squamous cell carcinoma (OSCC) is a public health concern. The current study aimed to explore the role of circRNA Dedicator of Cytokinesis 1 (circ_DOCK1) and associated action mode in OSCC. METHODS The expression of circ_DOCK1 and microRNA-1297 (miR-1297) was measured by quantitative real-time polymerase chain reaction (qRT-PCR). EdU assay, colony formation assay, transwell assay and glycolysis stress test were applied for functional analyses. The expression level of Homeobox A9 (HOXA9) was detected by western blot. The interaction between miR-1297 and circ_DOCK1 or HOXA9 was verified by dual-luciferase reporter assay. Xenograft model was established to determine the role of circ_DOCK1 in vivo. RESULTS Circ_DOCK1 was highly expressed in OSCC tumor tissues and cell lines. Circ_DOCK1 knockdown suppressed colony formation, migration, invasion and glycolysis of OSCC cells. MiR-1297 was targeted by circ_DOCK1, and its inhibition reversed the anticancer effects of circ_DOCK1 knockdown. HOXA9 was a target of miR-1297, and its overexpression recovered miR-1297 reintroduction-evoked inhibition of colony formation, migration, invasion and glycolysis in OSCC cells. Furthermore, circ_DOCK1 knockdown repressed tumor growth in vivo. CONCLUSION Circ_DOCK1 exerted its carcinogenic role in OSCC partially via the circ_DOCK1-miR-1297-HOXA9 regulatory network, which will broaden our insights to understand the pathogenesis of OSCC and provide promising biomarkers for the diagnosis and treatment of OSCC.
Collapse
Affiliation(s)
- Bingxin Mei
- Department of Stomatology, The First Affiliated Hospital Of Gannan Medical University, Ganzhou, China
| | - Zhimei Zeng
- Department of Stomatology, The First Affiliated Hospital Of Gannan Medical University, Ganzhou, China
| | - Qinmin Xia
- Department of Stomatology, The First Affiliated Hospital Of Gannan Medical University, Ganzhou, China
| | - Ming Liu
- Department of Stomatology, The First Affiliated Hospital Of Gannan Medical University, Ganzhou, China
| | - Ying Zhang
- Department of Stomatology, The First Affiliated Hospital Of Gannan Medical University, Ganzhou, China.
| |
Collapse
|
24
|
Lian W, Hong C, Chen D, Wang C. USP35 promotes breast cancer progression by regulating PFK-1 ubiquitination to mediate glycolysis. Am J Physiol Cell Physiol 2025; 328:C355-C366. [PMID: 39714773 DOI: 10.1152/ajpcell.00733.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 12/24/2024]
Abstract
Ubiquitin-specific protease 35 (USP35) was found to be involved in various tumor progression, but its role in breast cancer remains largely unknown. USP35 mRNA and protein expression in breast cancer tissues and cells were evaluated by quantitative real-time PCR and Western blot, respectively. Subsequently, flow cytometry and 5-ethynyl-2'-deoxyuridine labeling were used to evaluate breast cancer cell apoptosis and proliferation. Cellular glycolytic function was analyzed using the Seahorse assay and various kits. Furthermore, co-immunoprecipitation (Co-IP) and immunoprecipitation assays were utilized to validate the deubiquitylation mechanism of USP35. Finally, a subcutaneous human xenograft tumor model was established in nude mice to verify the effect of USP35 in vivo. By examining the clinical samples and cell lines, we found that USP35 expression was significantly upregulated in breast cancer. Further functional studies showed that knockdown USP35 expression inhibited cell proliferation and promoted apoptosis. In addition, knockdown of USP35 decreased phosphofructokinase1 (PFK-1) expression and was associated with lower extracellular acidification rate and oxygen consumption rate compared with sh-Control. Co-IP assays identified PFK-1 as a direct deubiquitiation target of USP35. Importantly, we demonstrated that PFK-1 is an essential mediator for USP35-induced cell proliferation and glycolysis in vitro and in vivo. This study identified that USP35 regulates the proliferation and glycolysis of breast cancer cells by mediating the ubiquitination level of PFK-1. The USP35/PFK-1 axis offers novel insight for the treatment of breast cancer.NEW & NOTEWORTHY This study identified that USP35 regulates the proliferation and glycolysis of breast cancer cells by mediating the ubiquitination level of PFK-1. The USP35/PFK-1 axis offers novel insight for the treatment of breast cancer.
Collapse
Affiliation(s)
- Weibin Lian
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
- Department of Breast Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| | - Chengye Hong
- Department of Breast Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| | - Debo Chen
- Department of Breast Surgery, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, People's Republic of China
| | - Chuan Wang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, People's Republic of China
| |
Collapse
|
25
|
Kul Köprülü T, Gezer B, Erkal Çam B. The impact of apoptosis-inducing MAPK and glycolytic pathways modulated by Aloe vera and royal jelly in lung and colorectal cancer. Med Oncol 2025; 42:51. [PMID: 39838121 PMCID: PMC11750912 DOI: 10.1007/s12032-025-02606-7] [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: 10/15/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025]
Abstract
Lung and colon cancer are among the most commonly diagnosed and fatal cancer types in the world. Due to their metastatic properties, they complicate the treatment process and pose a great threat to human health. These aggressive types of cancer are resistant to chemotherapy drugs. Therefore, it is extremely important to investigate the therapeutic effects of natural compounds. In our previous study, effective doses of Royal Jelly (RJ) (100 mg/mL) and Aloe vera (AVE) (20 µg/mL) were determined and tested separately and in combination on lung and colorectal cancer cells. Glycolytic capacities were determined using the Seahorse XFe24 Analyzer, total transcriptome profiles were sequenced using NovaSeq 6000, and BAX and BCL-2 gene levels were determined using RT-qPCR. It was seen that RJ and RJ + AVE affected glycolytic capacity and more genes in lung cancer cells. In HT29, AVE alone was seen to reduce glycolytic capacity and RJ + AVE combination was seen to reduce the expression level of genes related to cell proliferation and cycle. After RJ + AVE treatments, the apoptotic process which is triggered via MAPK pathway was found in lung cancer. Moreover, BAX levels increased and BCL-2 levels decreased both lung and colorectal cancer cells. It was observed that the combination of RJ and AVE affected the glycolysis process, cell cycle, proliferation and apoptosis on lung and colorectal cancer. In particular, the combination of RJ + AVE was found to be more effective on lung cancer.
Collapse
Affiliation(s)
- Tuğba Kul Köprülü
- Experimental Medicine Application and Research Center, Validebağ Research Park, University of Health Sciences, Altunizade, Kalfaçeşme Street, Üsküdar, 34662, İstanbul, Turkey.
- Department of Molecular Medicine, Hamidiye Institute of Health Sciences, University of Health Sciences, 34668, İstanbul, Turkey.
| | - Bahar Gezer
- Department of Molecular Medicine, Hamidiye Institute of Health Sciences, University of Health Sciences, 34668, İstanbul, Turkey
| | - Burçin Erkal Çam
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Yıldız Technical University, Esenler, İstanbul, Turkey
| |
Collapse
|
26
|
Huang L, Zhao W, Sun L, Niu D, Zhu X, Jin C. Research progresses and hotspots on glucose metabolic reprogramming in breast cancer: a bibliometric analysis over the past two decades. Front Oncol 2025; 14:1493996. [PMID: 39876898 PMCID: PMC11772165 DOI: 10.3389/fonc.2024.1493996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025] Open
Abstract
BACKGROUND Abnormal energy metabolism is a prominent characteristic of cancers. Increasing evidence has suggested the involvement of glucose metabolism reprogramming in the progression of breast cancer (BC). This article aims to provide a comprehensive overview of glucose metabolism reprogramming in BC through a bibliometric analysis. METHODS Relevant literatures published from 2004 to 2024 were searched in the Web of Science Core Collection database, and a bibliometric analysis was conducted using VOSviewer, CiteSpace, and Bibliometrix. RESULTS In total, 957 publications reporting glucose metabolism reprogramming in BC were included, showing an increasing trend in the annual publication outputs. China ranked first in publication outputs, and the United States of America (USA) had a dominant place in citation counts. The research achievements of Thomas Jefferson University in the USA were at the forefront and widely cited. Lisanti, Michael P., and Sotgia, Federica were the most productive authors. Keyword analysis suggested that the mechanisms of glucose metabolism reprogramming in BC and related therapeutic strategies were the research hotspots. CONCLUSION This study, for the first time, elucidated the progresses and hotspots of in the research on glucose metabolism reprogramming in BC, highlighting its potential role in treating BC. Considering that the glycolytic reprogramming of BC is a complex biological process, it is imperative for countries to enhance cooperation in the pursuit of effective antimetabolic therapies to overcome challenges in BC treatment.
Collapse
Affiliation(s)
| | | | | | | | | | - Chunhui Jin
- Department of Oncology, Wuxi Affiliated Hospital of Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
27
|
Panda VK, Mishra B, Mahapatra S, Swain B, Malhotra D, Saha S, Khanra S, Mishra P, Majhi S, Kumari K, Nath AN, Saha S, Jena S, Kundu GC. Molecular Insights on Signaling Cascades in Breast Cancer: A Comprehensive Review. Cancers (Basel) 2025; 17:234. [PMID: 39858015 PMCID: PMC11763662 DOI: 10.3390/cancers17020234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/27/2024] [Accepted: 01/01/2025] [Indexed: 01/27/2025] Open
Abstract
The complex signaling network within the breast tumor microenvironment is crucial for its growth, metastasis, angiogenesis, therapy escape, stem cell maintenance, and immunomodulation. An array of secretory factors and their receptors activate downstream signaling cascades regulating breast cancer progression and metastasis. Among various signaling pathways, the EGFR, ER, Notch, and Hedgehog signaling pathways have recently been identified as crucial in terms of breast cancer proliferation, survival, differentiation, maintenance of CSCs, and therapy failure. These receptors mediate various downstream signaling pathways such as MAPK, including MEK/ERK signaling pathways that promote common pro-oncogenic signaling, whereas dysregulation of PI3K/Akt, Wnt/β-catenin, and JAK/STAT activates key oncogenic events such as drug resistance, CSC enrichment, and metabolic reprogramming. Additionally, these cascades orchestrate an intricate interplay between stromal cells, immune cells, and tumor cells. Metabolic reprogramming and adaptations contribute to aggressive breast cancer and are unresponsive to therapy. Herein, recent insights into the novel signaling pathways operating within the breast TME that aid in their advancement are emphasized and current developments in practices targeting the breast TME to enhance treatment efficacy are reviewed.
Collapse
Affiliation(s)
- Venketesh K. Panda
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
- School of Applied Sciences, KIIT Deemed to Be University, Bhubaneswar 751024, India
| | - Barnalee Mishra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Samikshya Mahapatra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Biswajit Swain
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Diksha Malhotra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Suryendu Saha
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sinjan Khanra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Priyanka Mishra
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sambhunath Majhi
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Kavita Kumari
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Angitha N. Nath
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Swarnali Saha
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Sarmistha Jena
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
| | - Gopal C. Kundu
- School of Biotechnology, KIIT Deemed to Be University, Bhubaneswar 751024, India; (V.K.P.); (B.M.); (S.M.); (B.S.); (D.M.); (S.S.); (S.K.); (P.M.); (S.M.); (K.K.); (A.N.N.); (S.S.); (S.J.)
- School of Applied Sciences, KIIT Deemed to Be University, Bhubaneswar 751024, India
- Kalinga Institute of Medical Sciences (KIMS), KIIT Deemed to Be University, Bhubaneswar 751024, India
| |
Collapse
|
28
|
Wang Y, Xu S, Liu J, Qi P. A Novel Peroxisome-Related Gene Signature Predicts Breast Cancer Prognosis and Correlates with T Cell Suppression. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:887-911. [PMID: 39678026 PMCID: PMC11639899 DOI: 10.2147/bctt.s490154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/03/2024] [Indexed: 12/17/2024]
Abstract
Background Peroxisomes are increasingly linked to cancer development, yet the prognostic role of peroxisome-related genes (PRGs) in breast cancer remains unclear. Objective This study aimed to construct a prognostic model based on PRG expression in breast cancer to clarify their prognostic value and clinical implications. Methods Transcriptomic data from TCGA and GEO were used for training and validation cohorts. TME characteristics were analyzed with ESTIMATE, MCP-counter, and CIBERSORT algorithms. qPCR validated mRNA expression levels of risk genes, and data analysis was conducted in R. Results Univariate and multivariate Cox regression identified a 7-gene PRG risk signature (ACBD5, ACSL5, DAO, NOS2, PEX3, PEX10, and SLC27A2) predicting breast cancer prognosis in training (n=1069), internal validation (n=327), and external validation (merged from four GEO datasets, n=640) datasets. While basal and Her2 subtypes had higher risk scores than luminal subtypes, a significant prognostic impact of the PRG risk signature was seen only in luminal subtypes. The high-risk subgroup exhibited a higher frequency of focal synonymous copy number alterations (SCNAs), arm-level amplifications and deletions, and single nucleotide variations. These increased genomic aberrations were associated with greater immune suppression and reduced CD8+ T cell infiltration. Bulk RNA sequencing and single-cell analyses revealed distinct expression patterns of peroxisome-related genes (PRGs) in the breast cancer TME: PEX3 was primarily expressed in malignant and stromal cells, while ACSL5 showed high expression in T cells. Additionally, the PRG risk signature demonstrated efficacy comparable to that of well-known biomarkers for predicting immunotherapy responses. Drug sensitivity analysis revealed that the PRG high-risk subgroup was sensitive to inhibitors of BCL-2 family proteins (BCL-2, BCL-XL, and MCL1) and other kinases (PLK1, PLK1, BTK, CHDK1, and EGFR). Conclusion The PRG risk signature serves as a promising biomarker for evaluating peroxisomal activity, prognosis, and responsiveness to immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Yunxiang Wang
- Head and Neck Breast Department, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, 453000, People’s Republic of China
| | - Sheng Xu
- Head and Neck Breast Department, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, 453000, People’s Republic of China
| | - Junfeng Liu
- Head and Neck Breast Department, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, 453000, People’s Republic of China
| | - Pan Qi
- Head and Neck Breast Department, Xinxiang Central Hospital, The Fourth Clinical College of Xinxiang Medical University, Xinxiang, Henan, 453000, People’s Republic of China
| |
Collapse
|
29
|
Miao W, Wang Z, Gao J, Ohno Y. Polyphyllin II inhibits breast cancer cell proliferation via the PI3K/Akt signaling pathway. Mol Med Rep 2024; 30:224. [PMID: 39364737 PMCID: PMC11465422 DOI: 10.3892/mmr.2024.13348] [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: 07/04/2024] [Accepted: 09/05/2024] [Indexed: 10/05/2024] Open
Abstract
Paridis Rhizoma saponins (PRS) are significant components of Rhizoma Paridis and have inhibitory effects on various tumors, such as bladder, breast, liver and colon cancer. Polyphyllin II (PPII), one of the PRS, has an unclear effect on breast cancer. The present study aimed to explore the effect and mechanism of PPII in breast cancer. A network pharmacology approach was employed to predict the core components and breast cancer‑related targets of PRS. Moreover, a xenograft tumor model was established to determine the anti‑breast cancer effect of PPII in vivo. The viability of MDA‑MB‑231 cells was determined by a Cell Counting Kit‑8 assay. Apoptosis was analyzed using annexin V/PI double staining. Additionally, Transwell and scratch assays were performed to evaluate invasion and migration. The potential mechanism was predicted by Kyoto Encyclopedia of Genes and Genomes enrichment analysis and molecular docking analysis and verified by western blot analysis. The effect of PPII on aerobic glycolysis in breast cancer cells was detected by lactic acid and pyruvate kits and Western blotting of glycolytic rate‑limiting enzymes. Network pharmacology analysis revealed 26 core targets involved in breast cancer and that PPII was the core active component of PRS. The in vivo studies showed that PPII could inhibit the growth of breast cancer in mice. In vitro experiments confirmed that PPII induced cancer cell apoptosis and inhibited invasion and migration. Furthermore, PPII was capable of suppressing the expression of key proteins in the PI3K/Akt signaling pathway, reducing the generation of aerobic glycolytic products, and diminishing the protein expression levels of hexokinase 2 and pyruvate kinase M2. The results indicated that PPII inhibited aerobic glycolysis in breast cancer cells through the PI3K/Akt signaling pathway, thereby inhibiting breast cancer growth.
Collapse
Affiliation(s)
- Weiwei Miao
- Department of Health Science, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- College of Pharmacy, Shanghai University of Medicine & Health Sciences, Shanghai 201318, P.R. China
| | - Zhixiong Wang
- College of Health Management, Shanghai Jian Qiao University, Shanghai 201306, P.R. China
| | - Jianwen Gao
- College of Health Management, Shanghai Jian Qiao University, Shanghai 201306, P.R. China
| | - Yuko Ohno
- Department of Health Science, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
30
|
He L, Lin C, Zhuang L, Sun Y, Li Y, Ye Z. Targeting Hepatocellular Carcinoma: Schisandrin A Triggers Mitochondrial Disruption and Ferroptosis. Chem Biol Drug Des 2024; 104:e70010. [PMID: 39668608 PMCID: PMC11638659 DOI: 10.1111/cbdd.70010] [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: 03/05/2024] [Revised: 07/26/2024] [Accepted: 09/18/2024] [Indexed: 12/14/2024]
Abstract
The main focus of this research was to examine SchA's role in the hepatocellular carcinoma (HCC) development. LO2 and Huh7 cell viability were assessed using the MTT assay. The experiments included flow cytometry, colony formation, transwell, wound healing, and immunofluorescence assays to evaluate apoptosis levels, cells colony-forming ability, ROS levels, invasion and migration ability, and mitochondrial membrane potential. Biochemical kits was utilized for checking the ATP, mitochondrial DNA, MDA, GSH, and Fe2+ levels in the Huh7 cells, and western blot for measuring the ferroptosis and AMPK/mTOR related-protein expression levels. The MTT assay demonstrated that SchA significantly reduced the vitality of Huh7 cells ranging from 10 to 50 μM, whereas it exhibited no discernible impact on LO2 cells. Additionally, SchA significantly inhibited colony-forming ability, invasion ability, and migration ability within the concentration range of 10 to 50 μM, with a reduction of 68% in colony formation at 50 μM. SchA also induced apoptosis in a dose-dependent manner. Moreover, SchA was observed to significantly elevate ROS levels dose-dependently, down-regulate mitochondrial membrane potential (JC-1) at 20 and 50 μM, and reduce the levels of ATP and mtDNA dose-dependently. Various concentrations of SchA resulted in a notable elevation in MDA and Fe2+ levels as well as ACSL4 protein expression, accompanied by a reduction in GSH level and the protein expression of GPX4 and SLC7A11. Furthermore, SchA induced the activation of the AMPK/mTOR pathway in Huh7 cells, as evidenced by the increased phosphorylation level of AMPK and decreased phosphorylation level of mTOR. SchA might inhibit the progress of HCC through mitochondrial ferroptosis and dysfunction mediated by AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Lin‐wei He
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySouzhouJiangsuChina
| | - Chang‐jie Lin
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySouzhouJiangsuChina
| | - Lin‐jun Zhuang
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySouzhouJiangsuChina
| | - Yi‐hui Sun
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySouzhouJiangsuChina
| | - Ye‐cheng Li
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySouzhouJiangsuChina
| | - Zhen‐yu Ye
- Department of General SurgeryThe Second Affiliated Hospital of Soochow UniversitySouzhouJiangsuChina
| |
Collapse
|
31
|
Ge L, Feng T, Cao S, Yin H, Zhang Y, Pan J, Chen W, Cheng K, Wang L. Enhancing Student Comprehension of Glucose Metabolism Visualization Through Virtual Simulation Platform: An Educational Approach. Pharmacol Res Perspect 2024; 12:e70042. [PMID: 39601766 PMCID: PMC11600983 DOI: 10.1002/prp2.70042] [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: 07/09/2024] [Revised: 10/22/2024] [Accepted: 11/08/2024] [Indexed: 11/29/2024] Open
Abstract
With the rapid progression of biotechnology, the significance translational research on glycolysis in molecular pharmacology has become increasingly evident. To deepen students' understanding of glycolytic processes and facilitate their comprehension of drug action mechanisms, we have developed a visual virtual simulation platform dedicated glycolysis. The educational approach commenced with theoretical lectures on glycolysis, followed by practical laboratory sessions where students measured glycolysis-related parameters such as hexokinase, pyruvate kinase, and lactate. Students then engaged with the virtual simulation training platform to explore glycolytic stress tests and positron emission tomography/computed tomography (PET/CT) imaging, with their progress tracked through an assessment mode. The study involved 67 s-year undergraduate students majoring in biomedical sciences, all of whom had received instruction in glucose metabolism theories and completed the associated questionnaires. The results showed that the students gained a deeper understanding of glycolysis and the clinical application of PET/CT imaging in the context of glycolysis. The majority also agreed that the integration of scientific and clinical cases in teaching is beneficial and that the project sparked their interest in scientific research. These findings align with existing literature that emphasizes the importance of innovative educational tools in enhancing student engagement and understanding of the underlying theories of the curriculum. This project designed an innovative glycolytic metabolism teaching system encompassing the monitoring of traditional glycolytic indicators, glycolytic stress tests, and PET/CT imaging based on glycolysis. The visual virtual simulation platform for glycolysis can serve as an innovative educational tool in the molecular pharmacology curriculum or other courses involving glycolysis, assisting students in deeply understanding the molecular mechanisms of glycolysis and its significance in disease and drug action.
Collapse
Affiliation(s)
- Luna Ge
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Tingting Feng
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Shuo Cao
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Hang Yin
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Yuang Zhang
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Jihong Pan
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Wei Chen
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| | - Kai Cheng
- Department of PET/CT Center, Shandong Cancer Hospital and InstituteShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
| | - Lin Wang
- Biomedical Sciences College & Shandong Medicinal Biotechnology CentreShandong First Medical University, Shandong Academy of Medical SciencesJi'nanShandongChina
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences)Ji'nanShandongChina
- Key Lab for Rare & Uncommon Diseases of Shandong ProvinceJi'nanShandongChina
| |
Collapse
|
32
|
Su C, Yang J, Ding J, Ding H. Differential diagnosis of ovarian endometriosis cyst versus ovarian cystadenoma based on serum lactate dehydrogenase combined with CA-125 and CA19-9: A retrospective cohort study. Medicine (Baltimore) 2024; 103:e40776. [PMID: 39612391 PMCID: PMC11608665 DOI: 10.1097/md.0000000000040776] [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: 04/22/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024] Open
Abstract
This study aims to construct and validate a nomogram for the differential diagnosis of ovarian endometriosis cyst versus ovarian cystadenoma. We retrospectively studied the clinical characteristics of patients with ovarian endometriosis cysts and ovarian cystadenomas from January 1, 2021, to June 1, 2022. Independent risk factors for differential diagnosis were investigated using univariate and multivariate logistic regression analyses. Based on these factors, a differential diagnosis of ovarian endometriosis cyst versus ovarian cystadenoma was established. The performance of the nomogram model was assessed by internal validation using bootstrapping resampling. Decision curve analysis (DCA) was performed to evaluate the net clinical benefit of the model. Immunohistochemistry showed that lactate dehydrogenase (LDH) A was overexpressed in ectopic endometrial tissues compared to that in normal endometrial tissues. In multivariate analysis, LDH, CA-125, and CA19-9 were identified as independent risk factors for the differential diagnosis of ovarian endometriosis cyst versus ovarian cystadenoma. LDH levels >135.50 U/L combined with CA-125 levels >25.20 U/mL and CA19-9 levels >13.59 U/mL as single covariates had a high value in the differential diagnosis of ovarian endometriosis cysts versus ovarian cystadenoma. The area under the receiver operating characteristic curve (ROC) of the nomogram constructed using LDH, CA-125, and CA19-9 expression data was 0.873 (95% CI, 0.827-0.920), and the bootstrap-validated concordance index (C-index) was 0.871. Decision curve analysis confirmed that the nomogram model had excellent clinical utility. Based on serum lactate dehydrogenase combined with CA-125 and CA19-9, we constructed and validated a nomogram for the differential diagnosis of ovarian endometriosis cyst versus ovarian cystadenoma to help physicians formulate the optimal treatment strategy.
Collapse
Affiliation(s)
- Chang Su
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Anhui, Wuhu, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Anhui, Wuhu, China
| | - Jian Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Anhui, Wuhu, China
| | - Jin Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Anhui, Wuhu, China
| | - Huafeng Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wannan Medical College, Anhui, Wuhu, China
- Anhui Province Key Laboratory of Non-coding RNA Basic and Clinical Transformation, Anhui, Wuhu, China
| |
Collapse
|
33
|
Li H, Xie W, Huang X, Chen Y. FBP1 over-expression suppresses HIF-1α in papillary thyroid cancer. Sci Rep 2024; 14:29167. [PMID: 39587225 PMCID: PMC11589751 DOI: 10.1038/s41598-024-81017-6] [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: 01/25/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024] Open
Abstract
Papillary thyroid carcinoma (PTC) is generally a slow-growing disease with a favorable 10-year survival rate. However, about 10% of PTC cases show significant aggressiveness, with tendencies for local invasion or distant metastasis, the mechanisms of which remain unclear. This study aims to identify predictive indicators and explore new potential targets for clinical treatment, highlighting the need for novel biomarkers and therapeutic targets. We analyzed FBP1 expression in PTC tissues. Cell proliferation, apoptosis, and invasion were evaluated with and without FBP1 overexpression in PTC cells to assess FBP1's effects. We then investigated whether FBP1 reduces PTC cell tumorigenesis and metastasis by regulating HIF-1α expression. FBP1 expression was reduced in PTC samples and showed a negative correlation with T stage. In vitro experiments indicated that FBP1 acts as a hypoxia response inhibitor, regulating tumor cells. Additionally, FBP1 inhibited the proliferation, apoptosis, and invasion of thyroid cancer cells by modulating HIF-1α expression. Our results provide new insights into the role of FBP1 in PTC progression and indicate that targeting the FBP1-HIF-1α axis could be a promising therapeutic approach for this disease.
Collapse
Affiliation(s)
- Huashui Li
- General Surgery Department, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Wenjun Xie
- General Surgery Department, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Xiangqin Huang
- Breast Surgery Department, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China
| | - Yifan Chen
- Breast Surgery Department, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China.
| |
Collapse
|
34
|
Singh T, Sharma K, Jena L, Kaur P, Singh S, Munshi A. Mitochondrial bioenergetics of breast cancer. Mitochondrion 2024; 79:101951. [PMID: 39218051 DOI: 10.1016/j.mito.2024.101951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/05/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Breast cancer cells exhibit metabolic heterogeneity based on tumour aggressiveness. Glycolysis and mitochondrial respiration are two major metabolic pathways for ATP production. The oxygen flux, oxygen tension, proton leakage, protonmotive force, inner mitochondrial membrane potential, ECAR and electrochemical proton gradient maintain metabolic homeostasis, ATP production, ROS generation, heat dissipation, and carbon flow and are referred to as "sub-domains" of mitochondrial bioenergetics. Tumour aggressiveness is influenced by these mechanisms, especially when breast cancer cells undergo metastasis. These physiological parameters for healthy mitochondria are as crucial as energy demands for tumour growth and metastasis. The instant energy demands are already elucidated under Warburg effects, while these parameters may have dual functionality to maintain cellular bioenergetics and cellular health. The tumour cell might maintain these mitochondrial parameters for mitochondrial health or avoid apoptosis, while energy production could be a second priority. This review focuses explicitly on the crosstalk between metabolic domains and the utilisation of these parameters by breast cancer cells for their progression. Some major interventions are discussed based on mitochondrial bioenergetics that need further investigation. This review highlights the pathophysiological significance of mitochondrial bioenergetics and the regulation of its sub-domains by breast tumour cells for uncontrolled proliferation.
Collapse
Affiliation(s)
- Tashvinder Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Kangan Sharma
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Laxmipriya Jena
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Prabhsimran Kaur
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India
| | - Sandeep Singh
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India.
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda 151401, India.
| |
Collapse
|
35
|
Chen X, Su W, Chen J, Ouyang P, Gong J. ST3GAL4 promotes tumorigenesis in breast cancer by enhancing aerobic glycolysis. Hum Cell 2024; 38:1. [PMID: 39422756 DOI: 10.1007/s13577-024-01137-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: 06/07/2024] [Accepted: 09/17/2024] [Indexed: 10/19/2024]
Abstract
Sialyltransferases are enzymes that play a crucial role in regulating cancer progression by modifying glycoproteins through sialylation. In particular, the ST3 beta-galactoside alpha-2,3-sialyltransferase 4 (ST3GAL4) enzyme is known to be upregulated in breast cancer, but its specific biological functions have not been fully understood. This study aimed to investigate the impact and mechanisms of ST3GAL4 on aerobic glycolysis in breast cancer. We examined ST3GAL4 expression in tumor tissue samples and breast cancer cell lines and also manipulated ST3GAL4 expression in breast cancer cells using lentivirus transduction. The study evaluated cellular processes such as cell viability, cell cycle progression, and aerobic glycolysis by measuring parameters like extracellular acidification rate, glucose uptake, lactate production, and lactate dehydrogenase A (LDHA) expression. We found that ST3GAL4 expression was consistently increased in tumor tissues and breast cancer cell lines. High ST3GAL4 expression was associated with a poor prognosis for patients with breast cancer. Inhibiting ST3GAL4 expression decreased cell viability, disrupted cell cycle progression, and reduced aerobic glycolysis and LDHA expression. Furthermore, suppressing ST3GAL4 expression in animal models reduced tumor growth and cell proliferation. Conversely, overexpressing ST3GAL4 promoted cell viability and cell cycle progression, but these effects were reversed when an inhibitor of aerobic glycolysis was used. The study provided evidence in cells and animal models that ST3GAL4 promotes tumorigenesis in breast cancer by enhancing aerobic glycolysis. These findings suggest that targeting ST3GAL4 may be a potential strategy for the treatment of breast cancer.
Collapse
Affiliation(s)
- Xiaoqing Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, 613 West of Huangpu Avenue, Guangzhou, 510630, China
- Department of Breast Medicine, Foshan Women and Children Hospital, Foshan, 528000, China
| | - Weijie Su
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 200011, China
| | - Jiewen Chen
- Department of Breast Medicine, Foshan Women and Children Hospital, Foshan, 528000, China
| | - Peng Ouyang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, 613 West of Huangpu Avenue, Guangzhou, 510630, China
| | - Jin Gong
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Jinan University, 613 West of Huangpu Avenue, Guangzhou, 510630, China.
| |
Collapse
|
36
|
Zheng B, Chen K, Liu X, Wan Z, Wu Y, Xu L, Xiao J, Chen J. Transcription factor ETS1‑mediated ECT2 expression promotes the malignant behavior of prostate cancer cells. Oncol Lett 2024; 28:453. [PMID: 39100995 PMCID: PMC11294974 DOI: 10.3892/ol.2024.14585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/22/2024] [Indexed: 08/06/2024] Open
Abstract
Prostate cancer remains the most prevalent malignancy diagnosed in men worldwide. Epithelial cell transforming sequence 2 (ECT2) is an oncogene involved in the progression of human tumors. The present study aimed to explore the involvement of ECT2 in prostate cancer and its participation in the malignant progression of prostate cancer. ECT2 expression in prostate cancer cell lines was examined via reverse transcription-quantitative PCR and western blotting. The effects of knockdown of ECT2 expression in PC-3 cells on cellular biological behaviors, including proliferation, migration and invasion, were examined using Cell Counting Kit-8, colony formation, wound healing and Transwell assays. The glycolysis level was determined based on the lactate release, glucose uptake, oxygen consumption rate and extracellular acidification rate. The binding relationship between ECT2 and ETS1 was verified using luciferase reporter and chromatin immunoprecipitation assays. The results indicated that ECT2 was highly expressed in prostate cancer cell lines. Knockdown of ECT2 expression could inhibit cell proliferation, migration, invasion and glycolysis. In addition, the transcription factor ETS1 could directly bind to the ECT2 promoter and positively regulate ECT2 expression. These data were combined with the results of rescue experiments and demonstrated that the inhibitory effects of the knockdown of ECT2 expression on the malignant behavior and glycolysis of prostate cancer cells were partially reversed by ETS1 overexpression. In conclusion, ETS1 induced transcriptional upregulation of ECT2 and enhanced the malignant biological behaviors of prostate cancer cells, thereby promoting the progression of prostate cancer. This evidence provides a theoretical basis for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Bo Zheng
- Department of Urology, The Fifth Hospital of Xiamen City, Xiamen, Fujian 361101, P.R. China
| | - Kuifu Chen
- Department of Urology, The Fifth Hospital of Xiamen City, Xiamen, Fujian 361101, P.R. China
| | - Xin Liu
- Department of Radiology, The Second Affiliated Hospital of Xiamen Medical College, Xiamen, Fujian 361000, P.R. China
| | - Zhenghua Wan
- Department of Urology, The Fifth Hospital of Xiamen City, Xiamen, Fujian 361101, P.R. China
| | - Yulong Wu
- Department of Urology, The Fifth Hospital of Xiamen City, Xiamen, Fujian 361101, P.R. China
| | - Liming Xu
- Department of Urology, The Fifth Hospital of Xiamen City, Xiamen, Fujian 361101, P.R. China
| | - Jiguang Xiao
- Department of Urology, The Fifth Hospital of Xiamen City, Xiamen, Fujian 361101, P.R. China
| | - Jinqu Chen
- Department of Urology, The Fifth Hospital of Xiamen City, Xiamen, Fujian 361101, P.R. China
| |
Collapse
|
37
|
Zhao L, Zhang H, Ren P, Sun X. LncRNA SLC9A3-AS1 knockdown increases the sensitivity of liver cancer cell to triptolide by regulating miR-449b-5p-mediated glycolysis. Biotechnol Genet Eng Rev 2024; 40:1389-1405. [PMID: 36946780 DOI: 10.1080/02648725.2023.2193775] [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: 02/17/2023] [Accepted: 03/17/2023] [Indexed: 03/23/2023]
Abstract
Triptolide (TP) is involved in the progression of liver cancer. However, the detailed molecular network regulated through TP is still unclear. Long non-coding RNA (LncRNA) SLC9A3 exerts roles in various pathological progresses. Nevertheless, whether SLC9A3 affects the sensitivity of liver cancer cells to TP have not been uncovered. The content of SLC9A3-AS1 and miR-449b-5p was estimated by utilizing quantitative real-time polymerase-chain reaction (qRT-PCR). Cell counting kit 8 (CCK-8) assay was introduced to assess cell viability. Additionally, cell viability as well as invasion was tested via transwell assay. The direct binding between miR-449b-5p and SLC9A3-AS1 or LDHA was confirmed through luciferase reporter gene assay. Moreover, glycolysis rate was tested by calculating the uptake of glucose in addition to the production of lactate in Huh7 cells. LncRNA SLC9A3-AS1 was up-regulated in liver cancer tissue samples and cells. Knockdown of SLC9A3-AS1 notably further inhibited viability, migration as well as invasion in Huh7 cells. MiR-449b-5p was the direct downstream miRNA of SLC9A3-AS1 and was down-regulated by SLC9A3-AS1 in Huh7 cells. In addition, miR-449b-5p was reduced in liver cancer tissues and cells. Overexpressed miR-449b-5p increased the sensitivity of Huh7 cells to TP remarkably. Moreover, miR-449b-5p negatively regulated LDHA expression in Huh7 cells. This work proved that SLC9A3-AS1 increased the sensitivity of liver cancer cells to TP by regulating glycolysis rate mediated via miR-449b-5p/LDHA axis. These findings implied that TP is likely to be a potent agent for treating patients diagnosed with liver cancer.
Collapse
Affiliation(s)
- Lei Zhao
- Major of integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- Department of Thyroid Surgery, Linyi People's Hospital, Linyi, Shandong, China
| | - Houbin Zhang
- Department of Thoracic Surgery, Linyi People's Hospital, Linyi, Shandong, China
| | - Peiyou Ren
- Department of Thyroid Surgery, Linyi People's Hospital, Linyi, Shandong, China
| | - Xiangjun Sun
- Department of General Surgery, Linyi People's Hospital, Linyi, Shandong, China
| |
Collapse
|
38
|
Adem S, Rasul A, Riaz S, Sadiqa A, Ahmad M, Shahid Nazir M, Hassan M. Pyruvate kinase modulators as a therapy target: an updated patent review 2018-2023. Expert Opin Ther Pat 2024; 34:953-962. [PMID: 39279560 DOI: 10.1080/13543776.2024.2403616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 09/05/2024] [Indexed: 09/18/2024]
Abstract
INTRODUCTION Cancer cells adopt a glycolytic phenotype to fulfill their energy needs in unfavorable conditions. In metabolic rewiring, cancer cells upregulate the expression of glycolytic pathway regulators including glucose transporter 1, hexokinase 2, and PKM2 (pyruvate kinase) into its M2 splice form. Among these regulators, PKM2 plays a major role in metabolic reprogramming and is overexpressed in various diseases, including cancer. Dimerization of PKM2 causes the generation of synthetic precursors from glycolytic intermediates, which are essential for cellular growth and cancer cell proliferation. COVERED AREAS This article is focused on examining recent patents (2018-2023) on PKM2 activators, inhibitors and their biological and synthesis properties by using the advanced search service of the European Patent Office (EPO). Moreover, other databases including PubMed, Google Scholar and Elsevier were also examined for scientific data. On basis of their chemical structures, PKM2 activators and inhibitors are classified into pyrazole, pyrolidine-pyrazole, phenol, benzoxazine, isoselenazolo-pyridinium, phthalazine, and propiolylamide derivatives. EXPERT OPINION Activating PKM2 reduces proliferation and development of cells by reducing the quantity of biomolecules needed for cell formation. PKM2 activators and inhibitors are highly effective in treating many cancer pathogens. It is important to find new, more potent and selective molecules for PKM2 activation and inhibition.
Collapse
Affiliation(s)
- Sevki Adem
- Faculty of Science, Chemistry Department, Cankiri Karatekin University, Cankiri, Turkey
| | - Azhar Rasul
- Department of Zoology, Government College University, Faisalabad, Pakistan
| | - Saba Riaz
- Department of Zoology, Government College University, Faisalabad, Pakistan
| | - Ayesha Sadiqa
- Department of Zoology, Government College University, Faisalabad, Pakistan
| | - Matloob Ahmad
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| | | | - Mudassir Hassan
- Faculty of Science, Chemistry Department, Cankiri Karatekin University, Cankiri, Turkey
| |
Collapse
|
39
|
Glibetic N, Bowman S, Skaggs T, Weichhaus M. The Use of Patient-Derived Organoids in the Study of Molecular Metabolic Adaptation in Breast Cancer. Int J Mol Sci 2024; 25:10503. [PMID: 39408832 PMCID: PMC11477048 DOI: 10.3390/ijms251910503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 09/24/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Around 13% of women will likely develop breast cancer during their lifetime. Advances in cancer metabolism research have identified a range of metabolic reprogramming events, such as altered glucose and amino acid uptake, increased reliance on glycolysis, and interactions with the tumor microenvironment (TME), all of which present new opportunities for targeted therapies. However, studying these metabolic networks is challenging in traditional 2D cell cultures, which often fail to replicate the three-dimensional architecture and dynamic interactions of real tumors. To address this, organoid models have emerged as powerful tools. Tumor organoids are 3D cultures, often derived from patient tissue, that more accurately mimic the structural and functional properties of actual tumor tissues in vivo, offering a more realistic model for investigating cancer metabolism. This review explores the unique metabolic adaptations of breast cancer and discusses how organoid models can provide deeper insights into these processes. We evaluate the most advanced tools for studying cancer metabolism in three-dimensional culture models, including optical metabolic imaging (OMI), matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI), and recent advances in conventional techniques applied to 3D cultures. Finally, we explore the progress made in identifying and targeting potential therapeutic targets in breast cancer metabolism.
Collapse
Affiliation(s)
- Natalija Glibetic
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (N.G.); (S.B.); (T.S.)
- The IDeA Networks of Biomedical Research Excellence (INBRE) Program, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, HI 96816, USA
- United Nations CIFAL Honolulu Center, Chaminade University, Honolulu, HI 96816, USA
| | - Scott Bowman
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (N.G.); (S.B.); (T.S.)
- Undergraduate Program in Biochemistry, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, HI 96816, USA
| | - Tia Skaggs
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (N.G.); (S.B.); (T.S.)
- Undergraduate Program in Biology, School of Natural Sciences and Mathematics, Chaminade University, Honolulu, HI 96816, USA
| | - Michael Weichhaus
- Laboratory of Molecular Cancer Research, School of Natural Sciences and Mathematics, Chaminade University of Honolulu, Honolulu, HI 96816, USA; (N.G.); (S.B.); (T.S.)
| |
Collapse
|
40
|
Pan M, Li H, Shi X. A New Target for Hepatic Fibrosis Prevention and Treatment: The Warburg Effect. FRONT BIOSCI-LANDMRK 2024; 29:321. [PMID: 39344326 DOI: 10.31083/j.fbl2909321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 10/01/2024]
Abstract
Hepatic fibrosis is a major public health problem that endangers human wellbeing. In recent years, a number of studies have revealed the important impact of metabolic reprogramming on the occurrence and development of hepatic fibrosis. Among them, the Warburg effect, as an intracellular glucose metabolism reprogramming, can promote the occurrence and development of hepatic fibrosis by promoting the activation of hepatic stellate cells (HSCs) and inducing the polarization of liver macrophages (KC). Understanding the Warburg effect and its important role in the progression of hepatic fibrosis will assist in developing new strategies for the prevention and treatment of hepatic fibrosis. This review focuses on the Warburg effect and the specific mechanism by which it affects the progression of hepatic fibrosis by regulating HSCs activation and KC polarization. In addition, we also summarize and discuss the related experimental drugs and their mechanisms that inhibit the Warburg effect by targeting key proteins of glycolysis in order to improve hepatic fibrosis in the hope of providing more effective strategies for the clinical treatment of hepatic fibrosis.
Collapse
Affiliation(s)
- Meng Pan
- College of Basic Medical Sciences, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| | - Huanyu Li
- Second Clinical Medical College, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| | - Xiaoyan Shi
- College of Basic Medical Sciences, Shaanxi University of Chinese Medicine, 712046 Xianyang, Shaanxi, China
| |
Collapse
|
41
|
Shimi G. Dietary approaches for controlling cancer by limiting the Warburg effect: a review. Nutr Rev 2024; 82:1281-1291. [PMID: 37903372 DOI: 10.1093/nutrit/nuad130] [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] [Indexed: 11/01/2023] Open
Abstract
Cancer is a mysterious disease. Among other alterations, tumor cells, importantly, have metabolic modifications. A well-known metabolic modification commonly observed in cancer cells has been termed the Warburg effect. This phenomenon is defined as a high preference for glucose uptake, and increased lactate production from that glucose, even when oxygen is readily available. Some anti-cancer drugs target the proposed Warburg effect, and some dietary regimens can function similarly. However, the most suitable dietary strategies for treating particular cancers are not yet well understood. The aim of this review was to describe findings regarding the impact of various proposed dietary regimens targeting the Warburg effect. The evidence suggests that combining routine cancer therapies with diet-based strategies may improve the outcome in treating cancer. However, designing individualized therapies must be our ultimate goal.
Collapse
Affiliation(s)
- Ghazaleh Shimi
- Department of Cellular and Molecular Nutrition, Faculty of Nutrition Science and Food Technology, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
42
|
Xie F, Hua S, Guo Y, Wang T, Shan C, Zhang L, He T. Identification of TAT as a Biomarker Involved in Cell Cycle and DNA Repair in Breast Cancer. Biomolecules 2024; 14:1088. [PMID: 39334853 PMCID: PMC11430390 DOI: 10.3390/biom14091088] [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: 07/10/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/30/2024] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed cancer and the primary cause of cancer-related mortality in women. Treatment of triple-negative breast cancer (TNBC) remains particularly challenging due to its resistance to chemotherapy and poor prognosis. Extensive research efforts in BC screening and therapy have improved clinical outcomes for BC patients. Therefore, identifying reliable biomarkers for TNBC is of great clinical importance. Here, we found that tyrosine aminotransferase (TAT) expression was significantly reduced in BC and strongly correlated with the poor prognosis of BC patients, which distinguished BC patients from normal individuals, indicating that TAT is a valuable biomarker for early BC diagnosis. Mechanistically, we uncovered that methylation of the TAT promoter was significantly increased by DNA methyltransferase 3 (DNMT3A/3B). In addition, reduced TAT contributes to DNA replication and cell cycle activation by regulating homologous recombination repair and mismatch repair to ensure genomic stability, which may be one of the reasons for TNBC resistance to chemotherapy. Furthermore, we demonstrated that Diazinon increases TAT expression as an inhibitor of DNMT3A/3B and inhibits the growth of BC by blocking downstream pathways. Taken together, we revealed that TAT is silenced by DNMT3A/3B in BC, especially in TNBC, which promotes the proliferation of tumor cells by supporting DNA replication, activating cell cycle, and enhancing DNA damage repair. These results provide fresh insights and a theoretical foundation for the clinical diagnosis and treatment of BC.
Collapse
Affiliation(s)
- Fei Xie
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China; (F.X.); (S.H.); (Y.G.); (C.S.)
| | - Saiwei Hua
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China; (F.X.); (S.H.); (Y.G.); (C.S.)
| | - Yajuan Guo
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China; (F.X.); (S.H.); (Y.G.); (C.S.)
| | - Taoyuan Wang
- Cardiothoracic Surgery Department, Characteristic Medical Center of the Chinese People’s Armed Police Force, Tianjin 300162, China;
| | - Changliang Shan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China; (F.X.); (S.H.); (Y.G.); (C.S.)
| | - Lianwen Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, China; (F.X.); (S.H.); (Y.G.); (C.S.)
| | - Tao He
- Department of Pathology, Characteristic Medical Center of The Chinese People’s Armed Police Force, Tianjin 300162, China
| |
Collapse
|
43
|
Ni X, Lu CP, Xu GQ, Ma JJ. Transcriptional regulation and post-translational modifications in the glycolytic pathway for targeted cancer therapy. Acta Pharmacol Sin 2024; 45:1533-1555. [PMID: 38622288 PMCID: PMC11272797 DOI: 10.1038/s41401-024-01264-1] [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: 10/19/2023] [Accepted: 03/08/2024] [Indexed: 04/17/2024]
Abstract
Cancer cells largely rely on aerobic glycolysis or the Warburg effect to generate essential biomolecules and energy for their rapid growth. The key modulators in glycolysis including glucose transporters and enzymes, e.g. hexokinase 2, enolase 1, pyruvate kinase M2, lactate dehydrogenase A, play indispensable roles in glucose uptake, glucose consumption, ATP generation, lactate production, etc. Transcriptional regulation and post-translational modifications (PTMs) of these critical modulators are important for signal transduction and metabolic reprogramming in the glycolytic pathway, which can provide energy advantages to cancer cell growth. In this review we recapitulate the recent advances in research on glycolytic modulators of cancer cells and analyze the strategies targeting these vital modulators including small-molecule inhibitors and microRNAs (miRNAs) for targeted cancer therapy. We focus on the regulation of the glycolytic pathway at the transcription level (e.g., hypoxia-inducible factor 1, c-MYC, p53, sine oculis homeobox homolog 1, N6-methyladenosine modification) and PTMs (including phosphorylation, methylation, acetylation, ubiquitination, etc.) of the key regulators in these processes. This review will provide a comprehensive understanding of the regulation of the key modulators in the glycolytic pathway and might shed light on the targeted cancer therapy at different molecular levels.
Collapse
Affiliation(s)
- Xuan Ni
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215123, China
| | - Cheng-Piao Lu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Soochow University, Suzhou, 215123, China
| | - Guo-Qiang Xu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Soochow University, Suzhou, 215123, China.
- Suzhou International Joint Laboratory for Diagnosis and Treatment of Brain Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, 215123, China.
| | - Jing-Jing Ma
- Department of Pharmacy, The Fourth Affiliated Hospital of Soochow University, Suzhou Dushu Lake Hospital, Medical Center of Soochow University, Suzhou, 215123, China.
| |
Collapse
|
44
|
Yang Q, Meng D, Zhang Q, Wang J. Advances in the role of resveratrol and its mechanism of action in common gynecological tumors. Front Pharmacol 2024; 15:1417532. [PMID: 39086397 PMCID: PMC11288957 DOI: 10.3389/fphar.2024.1417532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/26/2024] [Indexed: 08/02/2024] Open
Abstract
The incidence of common gynecological malignancies remains high, with current treatments facing multiple limitations and adverse effects. Thus, continuing the search for safe and effective oncologic treatment strategies continues. Resveratrol (RES), a natural non-flavonoid polyphenolic compound, is widely found in various plants and fruits, such as grapes, Reynoutria japonica Houtt., peanuts, and berries. RES possesses diverse biological properties, including neuroprotective, antitumor, anti-inflammatory, and osteoporosis inhibition effects. Notably, RES is broadly applicable in antitumor therapy, particularly for treating gynecological tumors (cervical, endometrial, and ovarian carcinomas). RES exerts antitumor effects by promoting tumor cell apoptosis, inhibiting cell proliferation, invasion, and metastasis, regulating tumor cell autophagy, and enhancing the efficacy of antitumor drugs while minimizing their toxic side effects. However, comprehensive reviews on the role of RES in combating gynecological tumors and its mechanisms of action are lacking. This review aims to fill this gap by examining the RES antitumor mechanisms of action in gynecological tumors, providing valuable insights for clinical treatment.
Collapse
Affiliation(s)
- Qian Yang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dandan Meng
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingchen Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jin Wang
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
45
|
Hashemi M, Khosroshahi EM, Chegini MK, Asadi S, Hamyani Z, Jafari YA, Rezaei F, Eskadehi RK, Kojoori KK, Jamshidian F, Nabavi N, Alimohammadi M, Rashidi M, Mahmoodieh B, Khorrami R, Taheriazam A, Entezari M. Mechanistic insights into cisplatin response in breast tumors: Molecular determinants and drug/nanotechnology-based therapeutic opportunities. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2024; 794:108513. [PMID: 39216513 DOI: 10.1016/j.mrrev.2024.108513] [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: 05/12/2024] [Revised: 08/24/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Breast cancer continues to be a major global health challenge, driving the need for effective therapeutic strategies. Cisplatin, a powerful chemotherapeutic agent, is widely used in breast cancer treatment. However, its effectiveness is often limited by systemic toxicity and the development of drug resistance. This review examines the molecular factors that influence cisplatin response and resistance, offering crucial insights for the scientific community. It highlights the significance of understanding cisplatin resistance's genetic and epigenetic contributors, which could lead to more personalized treatment approaches. Additionally, the review explores innovative strategies to counteract cisplatin resistance, including combination therapies, nanoparticle-based drug delivery systems, and targeted therapies. These approaches are under intensive investigation and promise to enhance breast cancer treatment outcomes. This comprehensive discussion is a valuable resource to advance breast cancer therapeutics and address the challenge of cisplatin resistance.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Mohandesi Khosroshahi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrnaz Kalhor Chegini
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saba Asadi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Hamyani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Medicine, Islamic Azad University, Tehran Medical Sciences, Tehran, Iran
| | - Yasamin Alsadat Jafari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Fatemeh Rezaei
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ramtin Khodaparast Eskadehi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Kimia Kia Kojoori
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Faranak Jamshidian
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia, Canada
| | - Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Rashidi
- Department of Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Behnaz Mahmoodieh
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
46
|
Qiao Q, Hu S, Wang X. The regulatory roles and clinical significance of glycolysis in tumor. Cancer Commun (Lond) 2024; 44:761-786. [PMID: 38851859 PMCID: PMC11260772 DOI: 10.1002/cac2.12549] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/05/2024] [Accepted: 05/12/2024] [Indexed: 06/10/2024] Open
Abstract
Metabolic reprogramming has been demonstrated to have a significant impact on the biological behaviors of tumor cells, among which glycolysis is an important form. Recent research has revealed that the heightened glycolysis levels, the abnormal expression of glycolytic enzymes, and the accumulation of glycolytic products could regulate the growth, proliferation, invasion, and metastasis of tumor cells and provide a favorable microenvironment for tumor development and progression. Based on the distinctive glycolytic characteristics of tumor cells, novel imaging tests have been developed to evaluate tumor proliferation and metastasis. In addition, glycolytic enzymes have been found to serve as promising biomarkers in tumor, which could provide assistance in the early diagnosis and prognostic assessment of tumor patients. Numerous glycolytic enzymes have been identified as potential therapeutic targets for tumor treatment, and various small molecule inhibitors targeting glycolytic enzymes have been developed to inhibit tumor development and some of them are already applied in the clinic. In this review, we systematically summarized recent advances of the regulatory roles of glycolysis in tumor progression and highlighted the potential clinical significance of glycolytic enzymes and products as novel biomarkers and therapeutic targets in tumor treatment.
Collapse
Affiliation(s)
- Qiqi Qiao
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
| | - Shunfeng Hu
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
| | - Xin Wang
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
- Taishan Scholars Program of Shandong ProvinceJinanShandongP. R. China
- Branch of National Clinical Research Center for Hematologic DiseasesJinanShandongP. R. China
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuP. R. China
| |
Collapse
|
47
|
Liu Z, Li W, Wang Z, Yang Q, Chen L, Chen W, Qu X. EIF4A3-Induced CircDHTKD1 regulates glycolysis in non-small cell lung cancer via stabilizing PFKL. J Cell Mol Med 2024; 28:e18465. [PMID: 39022816 PMCID: PMC11255402 DOI: 10.1111/jcmm.18465] [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: 11/21/2023] [Revised: 05/14/2024] [Accepted: 05/17/2024] [Indexed: 07/20/2024] Open
Abstract
Lung cancer (LC) is one of the malignancies with the highest incidence and mortality in the world, approximately 85% of which is non-small cell lung cancer (NSCLC). Circular RNAs (circRNAs) exert multiple roles in NSCLC occurrence and development. The sequencing results in previous literature have illustrated that multiple circRNAs exhibit upregulation in NSCLC. We attempted to figure out which circRNA exerts an oncogenic role in NSLCL progression. RT-qPCR evaluated circDHTKD1 level in NSCLC tissue specimens and cells. Reverse transcription as well as RNase R digestion assay evaluated circDHTKD1 circular characterization in NSCLC cells. FISH determined circDHTKD1 subcellular distribution in NSCLC cells. Loss- and gain-of-function assays clarified circDHTKD1 role in NSCLC cell growth, tumour growth and glycolysis. Bioinformatics and RIP and RNA pull-down assessed association of circDHTKD1 with upstream molecule Eukaryotic initiation factor 4A-III (EIF4A3) or downstream molecule phosphofructokinase-1 liver type (PFKL) and insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) in NSCLC cells. Rescue assays assessed regulatory function of PFKL in circDHTKD1-meidated NSCLC cellular phenotypes. CircDHTKD1 exhibited upregulation and stable circular nature in NSCLC cells. EIF4A3 upregulated circDHTKD1 in NSCLC cells. CircDHTKD1 exerted a promoting influence on NSCLC cell malignant phenotypes and tumour growth. CircDHTKD1 exerted a promoting influence on NSCLC glucose metabolism. CircDHTKD1 exerts a promoting influence on NSCLC glucose metabolism through PFKL upregulation. RIP and RNA pull-down showed that circDHTKD1 could bind to IGF2BP, PFKL could bind to IGF2BP2, and circDHTKD1 promoted the binding of PFKL to IGF2BP2. In addition, RT-qPCR showed that IGF2BP2 knockdown promoted PFKL mRNA degradation, suggesting that IGF2BP2 stabilized PFKL in NSCLC cells. CircDHTKD1 exhibits upregulation in NSCLC. We innovatively validate that EIF4A3-triggered circDHTKD1 upregulation facilitates NSCLC glycolysis through recruiting m6A reader IGF2BP2 to stabilize PFKL, which may provide a new direction for seeking targeted therapy plans of NSCLC.
Collapse
Affiliation(s)
- Zhenghua Liu
- Department of Thoracic SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Wenya Li
- Department of Thoracic SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Ziyi Wang
- Department of Thoracic SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Qiwei Yang
- Department of Thoracic SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Liang Chen
- Department of Thoracic SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Weiyang Chen
- Department of Thoracic SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| | - Xiaohan Qu
- Department of Thoracic SurgeryThe First Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
48
|
Ye Y, Yang F, Gu Z, Li W, Yuan Y, Liu S, Zhou L, Han B, Zheng R, Cao Z. Fibroblast growth factor pathway promotes glycolysis by activating LDHA and suppressing LDHB in a STAT1-dependent manner in prostate cancer. J Transl Med 2024; 22:474. [PMID: 38764020 PMCID: PMC11103983 DOI: 10.1186/s12967-024-05193-9] [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: 02/29/2024] [Accepted: 04/11/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND The initiation of fibroblast growth factor 1 (FGF1) expression coincident with the decrease of FGF2 expression is a well-documented event in prostate cancer (PCa) progression. Lactate dehydrogenase A (LDHA) and LDHB are essential metabolic products that promote tumor growth. However, the relationship between FGF1/FGF2 and LDHA/B-mediated glycolysis in PCa progression is not reported. Thus, we aimed to explore whether FGF1/2 could regulate LDHA and LDHB to promote glycolysis and explored the involved signaling pathway in PCa progression. METHODS In vitro studies used RT‒qPCR, Western blot, CCK-8 assays, and flow cytometry to analyze gene and protein expression, cell viability, apoptosis, and cell cycle in PCa cell lines. Glycolysis was assessed by measuring glucose consumption, lactate production, and extracellular acidification rate (ECAR). For in vivo studies, a xenograft mouse model of PCa was established and treated with an FGF pathway inhibitor, and tumor growth was monitored. RESULTS FGF1, FGF2, and LDHA were expressed at high levels in PCa cells, while LDHB expression was low. FGF1/2 positively modulated LDHA and negatively modulated LDHB in PCa cells. The depletion of FGF1, FGF2, or LDHA reduced cell proliferation, induced cell cycle arrest, and inhibited glycolysis. LDHB overexpression showed similar inhibitory effect on PCa cells. Mechanistically, we found that FGF1/2 positively regulated STAT1 and STAT1 transcriptionally activated LDHA expression while suppressed LDHB expression. Furthermore, the treatment of an FGF pathway inhibitor suppressed PCa tumor growth in mice. CONCLUSION The FGF pathway facilitates glycolysis by activating LDHA and suppressing LDHB in a STAT1-dependent manner in PCa.
Collapse
Affiliation(s)
- Yongkang Ye
- Department of Urology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), 523059, Dongguan, China
| | - Fukan Yang
- Department of Urology, Guangdong Medical University, Graduate School, 524002, Zhanjiang, China
| | - Zhanhao Gu
- Department of Urology, Guangdong Medical University, Graduate School, 524002, Zhanjiang, China
| | - Wenxuan Li
- Department of Oncology, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), 523059, Dongguan, China
| | - Yinjiao Yuan
- Department of Oncology, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), 523059, Dongguan, China
- The First School of Clinical Medicine, Southern Medical University, 510510, Guangzhou, China
| | - Shaoqian Liu
- Department of Urology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), 523059, Dongguan, China
| | - Le Zhou
- Department of Oncology, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), 523059, Dongguan, China
- The First School of Clinical Medicine, Southern Medical University, 510510, Guangzhou, China
| | - Bo Han
- Department of Oncology, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), 523059, Dongguan, China
- The First School of Clinical Medicine, Southern Medical University, 510510, Guangzhou, China
| | - Ruinian Zheng
- Department of Oncology, Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), 523059, Dongguan, China.
- The First School of Clinical Medicine, Southern Medical University, 510510, Guangzhou, China.
| | - Zhengguo Cao
- Department of Urology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), 523059, Dongguan, China.
| |
Collapse
|
49
|
Hu Q, Wang Y, Mao W. Knockdown of Glycolysis-Related LINC01070 Inhibits the Progression of Breast Cancer. Cureus 2024; 16:e60093. [PMID: 38860098 PMCID: PMC11163994 DOI: 10.7759/cureus.60093] [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] [Accepted: 05/11/2024] [Indexed: 06/12/2024] Open
Abstract
Accumulative evidence confirms that glycolysis and long non-coding RNAs (lncRNAs) are closely associated with tumor development. The aim of this study was to construct a novel prognostic model based on glycolysis-related lncRNAs (GRLs) in breast cancer patients. By performing Pearson correlation analysis and Lasso regression analysis on differentially expressed genes and lncRNAs associated with glycolysis in the Cancer Genome Atlas (TCGA) and Gene Set Enrichment Analysis (GSEA) datasets, we identified nine GRLs and constructed associated prognostic risk signature. Kaplan-Meier survival analysis and univariate and multivariate Cox analysis showed that patients in the low-risk group had a better prognosis. The receiver operator characteristics (ROC) curves showed that the area under the curve (AUC) of the prognostic risk signature predicting patients' overall survival at 1-, 3- and 5- years was 0.78, 0.71, and 0.71, respectively. Moreover, the validation curves also showed that the signature had better diagnostic efficacy and clinical predictive power. Furthermore, clone formation assay, EdU assay, and Transwell assay showed that knockdown of LINC01070 inhibited breast cancer progression. We developed a prognostic risk-associated GRLs signature that can accurately predict the breast cancer patient's prognostic status, and LINC01070 can be used as a potential biomarker for the prognosis of breast cancer patients.
Collapse
Affiliation(s)
- Qiang Hu
- Urology, Zhongda Hospital, Southeast University, Nanjing, CHN
| | - Yiduo Wang
- Urology, Zhongda Hospital, Southeast University, Nanjing, CHN
| | - Weipu Mao
- Urology, Zhongda Hospital, Southeast University, Nanjing, CHN
| |
Collapse
|
50
|
Sathipati SY, Tsai MJ, Aimalla N, Moat L, Shukla S, Allaire P, Hebbring S, Beheshti A, Sharma R, Ho SY. An evolutionary learning-based method for identifying a circulating miRNA signature for breast cancer diagnosis prediction. NAR Genom Bioinform 2024; 6:lqae022. [PMID: 38406797 PMCID: PMC10894035 DOI: 10.1093/nargab/lqae022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/11/2024] [Accepted: 02/13/2024] [Indexed: 02/27/2024] Open
Abstract
Breast cancer (BC) is one of the most commonly diagnosed cancers worldwide. As key regulatory molecules in several biological processes, microRNAs (miRNAs) are potential biomarkers for cancer. Understanding the miRNA markers that can detect BC may improve survival rates and develop new targeted therapeutic strategies. To identify a circulating miRNA signature for diagnostic prediction in patients with BC, we developed an evolutionary learning-based method called BSig. BSig established a compact set of miRNAs as potential markers from 1280 patients with BC and 2686 healthy controls retrieved from the serum miRNA expression profiles for the diagnostic prediction. BSig demonstrated outstanding prediction performance, with an independent test accuracy and area under the receiver operating characteristic curve were 99.90% and 0.99, respectively. We identified 12 miRNAs, including hsa-miR-3185, hsa-miR-3648, hsa-miR-4530, hsa-miR-4763-5p, hsa-miR-5100, hsa-miR-5698, hsa-miR-6124, hsa-miR-6768-5p, hsa-miR-6800-5p, hsa-miR-6807-5p, hsa-miR-642a-3p, and hsa-miR-6836-3p, which significantly contributed towards diagnostic prediction in BC. Moreover, through bioinformatics analysis, this study identified 65 miRNA-target genes specific to BC cell lines. A comprehensive gene-set enrichment analysis was also performed to understand the underlying mechanisms of these target genes. BSig, a tool capable of BC detection and facilitating therapeutic selection, is publicly available at https://github.com/mingjutsai/BSig.
Collapse
Affiliation(s)
| | - Ming-Ju Tsai
- Hinda and Arthur Marcus Institute for Aging Research at Hebrew Senior Life, Boston, MA 02131, USA
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02131, USA
| | - Nikhila Aimalla
- Department of Internal Medicine-Pediatrics, Marshfield Clinic Health System, Marshfield, WI 54449, USA
| | - Luke Moat
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA
| | - Sanjay K Shukla
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA
| | - Patrick Allaire
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA
| | - Scott Hebbring
- Center for Precision Medicine Research, Marshfield Clinic Research Institute, Marshfield, WI 54449, USA
| | - Afshin Beheshti
- Blue Marble Space Institute of Science, Space Biosciences Division, NASA Ames Research Center, Moffett Field, CA94035, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Rohit Sharma
- Department of Surgical Oncology, Marshfield Clinic Health System, Marshfield, WI 54449, USA
| | - Shinn-Ying Ho
- Institute of Bioinformatics and Systems biology, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
- College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
- Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| |
Collapse
|