1
|
Wang Y, Lu J, Lin B, Chen J, Lin F, Zheng Q, Xue X, Wei Y, Chen S, Xu N. Integrated analysis of MIOX gene in prognosis of clear-cell renal cell carcinoma. Cell Death Dis 2025; 16:368. [PMID: 40341358 PMCID: PMC12062366 DOI: 10.1038/s41419-025-07698-7] [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: 10/26/2024] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/10/2025]
Abstract
Clear-cell renal cell carcinoma (ccRCC) is a highly aggressive malignancy that originates in the kidney. It often exhibits a limited response or can be refractory to a wide range of anti-cancer therapies, including tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors. Ferroptosis is a form of oxidative, iron-dependent cell death characterized by lipid peroxidation. Targeting ferroptosis may offer a promising alternative therapeutic strategy for cancer cells that are resistant to existing treatments. The impact of ferroptosis-related genes on the prognosis of ccRCC patients is still not fully understood. In this study, we identified 30 differentially expressed ferroptosis-related genes in ccRCC samples compared to normal tissues using data from The Cancer Genome Atlas (TCGA). Lasso regression analyses, along with Kaplan-Meier analysis, were conducted to identify genes associated with prognosis. Based on scRNA-seq and spatial transcriptome analysis, we identified specificity of MIOX in ccRCC. Furthermore, MIOX demonstrated the highest significance, highlighting its independent prognostic value as a single gene in ccRCC. Our findings suggest that MIOX could serve as potential targets for therapeutic interventions in ccRCC.
Collapse
Affiliation(s)
- Yiqiu Wang
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiayi Lu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bohan Lin
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Jiayin Chen
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Fei Lin
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qingshui Zheng
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xueyi Xue
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shaohao Chen
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
2
|
Jiang W, Liu W, Zhao J, Xu Z, Xi M, Wang X, Li B. Aberrant expression of multiple γ-glutamyltransferases is associated with tumor progression and patient outcome in prostate cancers. Front Oncol 2025; 15:1518636. [PMID: 40094020 PMCID: PMC11906340 DOI: 10.3389/fonc.2025.1518636] [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: 10/28/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction The human gamma-glutamyltransferase (GGT) is a membrane-bound extracellular glycoprotein with an enzymatic activity that cleaves gamma-glutamyl peptide bonds in glutathione and other peptides and transfers the gamma-glutamyl moiety to acceptors. It has been shown aberrant expression of GGT proteins in human cancers while their expression profiles in prostate cancers are not reported. Methods In this study, we analyzed the expression profiles of all protein-coding GGT genes using the TCGA-PRAD RNA-seq dataset derived from primary prostate cancers. GGT family gene expression profiles were also analyzed using the SU2C/PCF RNAseq dataset derived from aggressive late-stage prostate cancer patients. Androgen modulation of GGT family gene expression was analyzed using multiple NCBI/GEO datasets. Results Our results showed that prostate tissues expressed four major isoforms of GGT family genes (GGT1/5/6/7), of which GGT1 expression was upregulated but GGT6/GGT7 expression was downregulated in cancer tissues compared to benign tissues. However, GGT5 expression was increased along with tumor stage progression and associated with worse progression-free survival. GGT6 expression exhibited a superb AUC value in prostate cancer diagnosis and was associated with favorable progression-free survival. GGT1 expression was highly increased but GGT6/GGT7 expression was largely reduced in ERG-fusion-positive cases. In CRPC tumors, GGT6 expression was suppressed in patients with anti-AR therapies, which was reversed when patients were taken off the treatment. This AR-dependent modulation was confirmed in LNCaP cells and LuCaP35 xenograft models. In addition, compared to CRPC-Adeno tumors, treatment-induced NEPC tumors showed a reduced GGT1 but an elevated GGT7 level, which was in line with higher levels of GGT7 in NEPC H660 cells. Conclusion Our data suggests that GGT6 is a new AR downstream target but GGT7 is a potential NEPC biomarker.
Collapse
Affiliation(s)
- Wencong Jiang
- Department of Urology, Huadu District People's Hospital, Guangzhou, China
| | - Wang Liu
- Department of Urology, The University of Kansas Medical Center, Kansas City, MO, United States
| | - Jiang Zhao
- Department of Urology, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhijian Xu
- Department of Urology, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Ming Xi
- Department of Urology, Huadu District People's Hospital, Guangzhou, China
| | - Xiangwei Wang
- Department of Urology, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Benyi Li
- Department of Urology, The University of Kansas Medical Center, Kansas City, MO, United States
| |
Collapse
|
3
|
Huang H, Lian H, Liu W, Li B, Zhu R, Shao H. Sideroflexin family genes were dysregulated and associated with tumor progression in prostate cancers. Hum Genomics 2025; 19:10. [PMID: 39915876 PMCID: PMC11803981 DOI: 10.1186/s40246-024-00705-6] [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: 11/12/2024] [Accepted: 12/11/2024] [Indexed: 02/09/2025] Open
Abstract
Sideroflexin (SFXN) family genes encode for a group of mitochondrial proteins involved in cellular processes such as iron homeostasis, amino acid metabolism, and energy production. Recent studies showed that they were aberrantly expressed in certain human cancers. However, there is a paucity of information about their expression in prostate cancer. In this study, we took a comprehensive approach to investigate their expression profiles in benign prostate tissue, prostate-derived cell lines, and prostate cancer tissues using multiple transcriptome datasets. Our results showed that SFXN1/3/4 genes were predominantly expressed in prostate tissue and cell lines. SFXN2/4 genes were significantly upregulated while the SFXN3 expression was significantly downregulated in malignant tissues compared to benign tissues. SFXN4 expression was identified as a diagnostic biomarker and prognostic factor for unfavorite survival outcomes. In advanced prostate cancers, SFXN2/4 expressions were positively correlated with the androgen receptor signaling activity but negatively correlated with the neuroendocrinal features. Further analysis discovered that SFXN5 expression was significantly elevated in neuroendocrinal prostate cancers. In conclusion, SFXN2/4 expressions are novel biomarkers in prostate cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Hua Huang
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, The Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Huibo Lian
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, The Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China
| | - Wang Liu
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Benyi Li
- Department of Urology, The University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| | - Runzhi Zhu
- The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, Zhejiang, China
| | - Haiyan Shao
- Urology & Nephrology Center, Department of Urology, Zhejiang Provincial People's Hospital, The Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang, China.
| |
Collapse
|
4
|
Li Z, Liu J, Wang P, Zhang B, He G, Yang L. HAMP predicts a pivotal role in modulating the malignant behaviors of non-small cell lung cancer cells. Aging (Albany NY) 2024; 16:8524-8540. [PMID: 38787358 PMCID: PMC11164495 DOI: 10.18632/aging.205819] [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: 10/24/2023] [Accepted: 01/23/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Hepcidin antimicrobial peptide (HAMP) is a small peptide hormone recognized for its role in iron metabolism and cancer treatment. The purpose of this study was to examine the influence of HAMP in NSCLC. METHODS The profile of NSCLC cells and tissues was characterized via HAMP. Gain- or loss-of-function cell models of HAMP were constructed, and CCK8, colony formation, and Transwell analyses were used to confirm the influence of HAMP on NSCLC cells. Upstream and downstream HAMP mechanisms in NSCLC were also analysed. Dual-luciferase reporter and pull-down assays confirmed the associations of miR-873-5p with HAMP, miR-873-5p, and the lncRNA KCNQ1OT1/SNHG14/XIST. Moreover, a xenograft model was established in nude mice for confirming the role of HAMP in NSCLC cell growth. RESULTS In addition, HAMP expression increased in NSCLC cells and tissues. In terms of cellular functions, the HAMP-overexpressing group exhibited elevated NSCLC cell proliferation, invasion, and migration. HAMP knockdown reversed these changes. Bioinformatics analysis indicated that miR-873-5p targeted HAMP, which affected the nuclear factor kappa B (NF-κB) pathway in NSCLC. HAMP activated the NF-κB pathway, which was negatively modulated by miR-873-5p. NF-κB inhibitor JSH-23 can partly suppress the proliferation, invasion, and migration in HAMP-overexpressed cells. Moreover, miR-873-5p was the target miRNA of long noncoding RNAs (lncRNAs), which included KCNQ1OT1, SNHG14, and XIST, and these three lncRNAs promoted HAMP. CONCLUSION Noncoding RNA-mediated HAMP promotes NSCLC cell proliferation, migration, and invasion by initiating the NF-κB pathway.
Collapse
Affiliation(s)
- Zhifeng Li
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Jinglei Liu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Ping Wang
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Boyu Zhang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Guanghui He
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Liwei Yang
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050000, China
| |
Collapse
|
5
|
Han Y, Fu L, Kong Y, Jiang C, Huang L, Zhang H. STEAP3 Affects Ovarian Cancer Progression by Regulating Ferroptosis through the p53/SLC7A11 Pathway. Mediators Inflamm 2024; 2024:4048527. [PMID: 38440354 PMCID: PMC10911874 DOI: 10.1155/2024/4048527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/14/2024] [Accepted: 02/03/2024] [Indexed: 03/06/2024] Open
Abstract
Ovarian cancer (OC) is a common malignant cancer in women with a low overall survival rate, and ferroptosis may be a potential new strategy for treatment. Six-transmembrane epithelial antigen of prostate 3 (STEAP3) is a gene closely related to ferroptosis, yet the role of STEAP3 in OC has not yet been thoroughly investigated. Using biological information analysis, we first found that STEAP3 was highly expressed in OC, which was significantly associated with poor prognosis of patients and was an independent prognostic factor. Through cloning, scratch, and transwell experiments, we subsequently found that knockdown of STEAP3 significantly reduced the proliferation and migration ability of OC cells. Furthermore, we found that knockdown of STEAP3 induced ferroptosis in OC cells by detecting ferroptosis indicators. Mechanistically, we also found that knockdown of STEAP3 induced ferroptosis through the p53/SLC7A11 signaling pathway. Through tumorigenic experiments in nude mice, we finally verified that the knockdown of STEAP3 could inhibit tumor growth in vivo by promoting ferroptosis through the p53 pathway. Overall, our study identified a novel therapeutic target for ferroptosis in OC and explored its specific mechanism of action.
Collapse
Affiliation(s)
- Yi Han
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Lei Fu
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Yan Kong
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Changqing Jiang
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Liying Huang
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| | - Hualing Zhang
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao 266000, Shandong, China
| |
Collapse
|
6
|
Formica V, Riondino S, Morelli C, Guerriero S, D'Amore F, Di Grazia A, Del Vecchio Blanco G, Sica G, Arkenau HT, Monteleone G, Roselli M. HIF2α, Hepcidin and their crosstalk as tumour-promoting signalling. Br J Cancer 2023; 129:222-236. [PMID: 37081189 PMCID: PMC10338631 DOI: 10.1038/s41416-023-02266-2] [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: 10/20/2022] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/22/2023] Open
Abstract
Not all aspects of the disruption of iron homeostasis in cancer have been fully elucidated. Iron accumulation in cancer cells is frequent for many solid tumours, and this is often accompanied by the contemporary rise of two key iron regulators, HIF2α and Hepcidin. This scenario is different from what happens under physiological conditions, where Hepcidin parallels systemic iron concentrations while HIF2α levels are inversely associated to Hepcidin. The present review highlights the increasing body of evidence for the pro-tumoral effect of HIF2α and Hepcidin, discusses the possible imbalance in HIF2α, Hepcidin and iron homeostasis during cancer, and explores therapeutic options relying on these pathways as anticancer strategies.
Collapse
Affiliation(s)
- Vincenzo Formica
- Medical Oncology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy.
| | - Silvia Riondino
- Medical Oncology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Cristina Morelli
- Medical Oncology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
- PhD Program in Systems and Experimental Medicine (XXXV cycle), University of Rome Tor Vergata, Via Montpellier 1, 00133, Rome, Italy
| | - Simona Guerriero
- Medical Oncology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Federica D'Amore
- Medical Oncology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| | - Antonio Di Grazia
- Gastroenterology Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | | | - Giuseppe Sica
- Department of Surgery, University of Rome Tor Vergata, Rome, Italy
| | | | - Giovanni Monteleone
- Gastroenterology Unit, Department of Systems Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | - Mario Roselli
- Medical Oncology Unit, Department of Systems Medicine, University of Rome Tor Vergata, Viale Oxford, 81, 00133, Rome, Italy
| |
Collapse
|
7
|
Smith MM, Melrose J. Pentosan Polysulfate Affords Pleotropic Protection to Multiple Cells and Tissues. Pharmaceuticals (Basel) 2023; 16:437. [PMID: 36986536 PMCID: PMC10132487 DOI: 10.3390/ph16030437] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/18/2023] [Accepted: 03/01/2023] [Indexed: 03/16/2023] Open
Abstract
Pentosan polysulfate (PPS), a small semi-synthetic highly sulfated heparan sulfate (HS)-like molecule, shares many of the interactive properties of HS. The aim of this review was to outline the potential of PPS as an interventional therapeutic protective agent in physiological processes affecting pathological tissues. PPS is a multifunctional molecule with diverse therapeutic actions against many disease processes. PPS has been used for decades in the treatment of interstitial cystitis and painful bowel disease, it has tissue-protective properties as a protease inhibitor in cartilage, tendon and IVD, and it has been used as a cell-directive component in bioscaffolds in tissue engineering applications. PPS regulates complement activation, coagulation, fibrinolysis and thrombocytopenia, and it promotes the synthesis of hyaluronan. Nerve growth factor production in osteocytes is inhibited by PPS, reducing bone pain in osteoarthritis and rheumatoid arthritis (OA/RA). PPS also removes fatty compounds from lipid-engorged subchondral blood vessels in OA/RA cartilage, reducing joint pain. PPS regulates cytokine and inflammatory mediator production and is also an anti-tumor agent that promotes the proliferation and differentiation of mesenchymal stem cells and the development of progenitor cell lineages that have proven to be useful in strategies designed to effect repair of the degenerate intervertebral disc (IVD) and OA cartilage. PPS stimulates proteoglycan synthesis by chondrocytes in the presence or absence of interleukin (IL)-1, and stimulates hyaluronan production by synoviocytes. PPS is thus a multifunctional tissue-protective molecule of potential therapeutic application for a diverse range of disease processes.
Collapse
Affiliation(s)
- Margaret M. Smith
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Faculty of Health and Science, University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia;
| | - James Melrose
- Raymond Purves Laboratory, Institute of Bone and Joint Research, Kolling Institute of Medical Research, Faculty of Health and Science, University of Sydney at Royal North Shore Hospital, St. Leonards, NSW 2065, Australia;
- Graduate Schools of Biomedical Engineering, University of NSW, Sydney, NSW 2052, Australia
- Sydney Medical School, Northern Campus, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| |
Collapse
|