51
|
Guo Y, Lin Z, Zhou Z, Zhang W, Mao S, Shan Z, Wu P, Yao X. Oncogenic and immunological functions of USP35 in pan-cancer and its potential as a biomarker in kidney clear cell carcinoma. BMC Cancer 2025; 25:617. [PMID: 40188027 PMCID: PMC11972461 DOI: 10.1186/s12885-025-13964-w] [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: 11/01/2024] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Ubiquitin-specific protease 35 (USP35) has gained attention as a regulator in cancer progression. However, its specific role in kidney clear cell carcinoma (KIRC) remains unclear. METHODS USP35 expression in KIRC tumor and normal tissues was evaluated using TCGA data. Correlations between USP35 expression, clinical parameters, and survival outcomes were examined. Functional enrichment analyses were performed to explore the pathways associated with USP35 expression. Immune-related analyses were conducted to assess the effect of USP35 on immune cell recruitment and neoantigen presentation. Drug sensitivity analyses were used to identify potential therapeutic agents targeting USP35. RESULTS USP35 was significantly overexpressed in KIRC tumor tissues compared to normal tissues, and its high expression correlated with advanced disease stages and poor survival outcomes. Gene set enrichment analysis revealed that high USP35 expression was associated with oncogenic pathways, including glycerophospholipid and linoleic acid metabolism, while low expression linked to nitrogen and purine metabolism. USP35 also modulated immune responses, affecting immune cell recruitment and neoantigen presentation, suggesting a role in immune evasion. Drug sensitivity analysis showed that high USP35 expression correlated with increased sensitivity to paclitaxel, bosutinib, and lapatinib. In vitro knockdown of USP35 significantly reduced KIRC cell proliferation, migration, and epithelial-mesenchymal transition (EMT), further supporting its role in tumor progression. CONCLUSION USP35 is overexpressed in KIRC and associated with poor prognosis, likely promoting tumor progression through oncogenic pathways and immune modulation. Its correlation with drug sensitivity positions USP35 as a potential therapeutic target, warranting further investigation into its mechanistic functions and therapeutic applications.
Collapse
MESH Headings
- Humans
- Carcinoma, Renal Cell/immunology
- Carcinoma, Renal Cell/genetics
- Carcinoma, Renal Cell/pathology
- Carcinoma, Renal Cell/mortality
- Carcinoma, Renal Cell/drug therapy
- Carcinoma, Renal Cell/metabolism
- Kidney Neoplasms/immunology
- Kidney Neoplasms/genetics
- Kidney Neoplasms/pathology
- Kidney Neoplasms/mortality
- Kidney Neoplasms/metabolism
- Kidney Neoplasms/drug therapy
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Ubiquitin-Specific Proteases/genetics
- Ubiquitin-Specific Proteases/metabolism
- Gene Expression Regulation, Neoplastic
- Prognosis
- Cell Line, Tumor
- Cell Proliferation
- Epithelial-Mesenchymal Transition
Collapse
Affiliation(s)
- Yadong Guo
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Ziyou Lin
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Zijing Zhou
- Laboratory of Ruijin Hospitalaffiliated to, Wuxi Branchaq, Shanghai Jiaotong University School of Medicine, Wuxi, Jiangsu, China
| | - Wentao Zhang
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Shiyu Mao
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Zezhi Shan
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
| | - Pengfei Wu
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China.
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| | - Xudong Yao
- Department of Urology, School of Medicine, Shanghai Tenth People'S Hospital, Tongji University, Shanghai, China.
- Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
52
|
Liu J, Zhang X, Lv R, Zhang X, Wang R, Zeng X. Predictive value of extracellular volume fraction determined using enhanced computed tomography for pathological grading of clear cell renal cell carcinoma: a preliminary study. Cancer Imaging 2025; 25:49. [PMID: 40186299 PMCID: PMC11969730 DOI: 10.1186/s40644-025-00866-0] [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: 01/16/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
OBJECTIVE To explore the potential of using the extracellular volume fraction (ECV), measured through enhanced computed tomography (CT), as a tool for determining the pathological grade of clear cell renal cell carcinoma (ccRCC). METHODS This retrospective study, approved by the institutional review board, included 65 patients (median age: 58.40 ± 10.84 years) who were diagnosed with ccRCC based on the nucleolar grading of the International Society of Urological Pathology (ISUP). All patients underwent preoperative abdominal enhanced CT between January 2022 and August 2024. CT features from the unenhanced, corticomedullary, nephrographic, and delayed phases were analyzed, and the extracellular volume fraction (ECV) of ccRCC was calculated by measuring CT values from regions of interest in both the unenhanced and nephrographic phases. Statistical significance was evaluated for differences in these parameters across the four ISUP grades. Additionally, diagnostic efficiency was assessed using receiver operating characteristic (ROC) curve analysis. RESULTS The ECV showed significant differences across the four ISUP grades of ccRCC, its potential as an important predictor of high-grade ccRCC (P = 0.035). The ROC curve analysis indicated that ECV exhibited the highest diagnostic efficacy for assessing the lower- and higher- pathological grade of ccRCC, with an area under the ROC curve of 0.976. The optimal diagnostic threshold for ECV was determined to be 41.64%, with a sensitivity of 91.31% and a specificity of 97.62%. CONCLUSIONS ECV derived from enhanced CT has the potential to function as an in vivo biomarker for distinguishing between lower- and higher-grade ccRCC. This quantitative measure provides diagnostic value that extends beyond traditional qualitative CT features, offering a more precise and objective assessment of tumor grade.
Collapse
Affiliation(s)
- Jian Liu
- Key Laboratory of Advanced Medical Imaging and Intelligent Computing of Guizhou Province, Engineering Research Center of Text Computing & Cognitive Intelligence, State Key Laboratory of Public Big Data, College of Computer Science and Technology, Ministry of Education, Guizhou University, No. 2708, Huaxi Avenue, Guiyang, 550025, Guizhou, China
- Department of nuclear medicine, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
| | - Xunlan Zhang
- Department of nuclear medicine, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
| | - Rui Lv
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
| | - Xiaoyong Zhang
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
| | - Rongpin Wang
- Department of Radiology, International Exemplary Cooperation Base of Precision Imaging for Diagnosis and Treatment, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China
| | - Xianchun Zeng
- Department of nuclear medicine, Guizhou Provincial People's Hospital, No. 83, Zhongshan Dong Road, Guiyang, 550002, Guizhou, China.
| |
Collapse
|
53
|
Sun Z, Wang Y, Zheng C, Xiao L, Zang Y, Fang L, Cui X, Chang M, Sun Q, Li W, Ren J. NAT10 promotes the progression of clear cell renal cell carcinoma by regulating ac4C acetylation of NFE2L3 and activating AKT/GSK3β signaling pathway. Cell Death Dis 2025; 16:235. [PMID: 40169553 PMCID: PMC11962090 DOI: 10.1038/s41419-025-07528-w] [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: 06/27/2024] [Revised: 02/13/2025] [Accepted: 03/12/2025] [Indexed: 04/03/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most common histological subtype of renal cell carcinoma, and the tumour growth and metastasis of ccRCC are related to prognosis. N4-acetylcytidine (ac4C) is one of the major modifications of RNA and is known to be mediated by N-acetyltransferase 10 (NAT10). The role of NAT10 in cancer is gradually being revealed, although the role of NAT10-mediated RNA ac4C modification in ccRCC has not been reported. In this study, NAT10 was found to be upregulated in ccRCC tissues and associated with a poor prognosis in patients. HIF-1α activated NAT10 expression at the transcriptional level. CCK8, EdU, Transwell and scratch assays after NAT10 knockdown or overexpression showed that NAT10 promoted cell proliferation and migration. The results of subcutaneous xenograft and caudal vein injection showed that NAT10 promoted tumour growth and metastasis in vivo, while Remodelin inhibited tumour growth. The acRIP-seq, RIP, RNA stability and dual luciferase reporter experiments showed that NAT10 activated ac4C acetylation of NFE2L3 mRNA and promoted NFE2L3 mRNA stability. The ChIP-seq results showed that NFE2L3 regulated the expression of LASP1 and thus activated the AKT/GSK3β signalling pathway. In summary, our results suggest that NAT10 mediates ac4C acetylation of NFE2L3 mRNA, promotes its mRNA stability, regulates the LASP1-AKT/GSK3β/β-catenin axis and promotes the progression of renal clear cell carcinoma.
Collapse
Affiliation(s)
- Zenghui Sun
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Shandong University, Jinan, PR China
| | - Yuqiong Wang
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Shandong University, Jinan, PR China
| | - Chao Zheng
- Department of Urology, Qilu Hospital, Shandong University, Jinan, PR China
| | - Lixiang Xiao
- Department of Urology, Qilu Hospital, Shandong University, Jinan, PR China
| | - Yuanwei Zang
- Department of Urology, Qilu Hospital, Shandong University, Jinan, PR China
| | - Liang Fang
- Department of Urology, Qilu Hospital, Shandong University, Jinan, PR China
| | - Xixi Cui
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Shandong University, Jinan, PR China
| | - Mingjie Chang
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Shandong University, Jinan, PR China
| | - Qiyu Sun
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Shandong University, Jinan, PR China
| | - Wenjuan Li
- Key Laboratory for Experimental Teratology of Chinese Ministry of Education, The Shandong Provincial Key Laboratory of Infection and Immunology, Department of Pathogenic biology, School of basic medical sciences, Shandong University, Jinan, PR China.
| | - Juchao Ren
- Department of Urology, Qilu Hospital, Shandong University, Jinan, PR China.
| |
Collapse
|
54
|
Assal RA, Rashwan HH, Zakaria ZI, Sweillam JH, Fouda YM, Abdelhamid AM, Youness RA. Deciphering the mysteries of MEG3 LncRNA and its implications in genitourinary cancers. Front Oncol 2025; 15:1519103. [PMID: 40242248 PMCID: PMC12000830 DOI: 10.3389/fonc.2025.1519103] [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: 10/29/2024] [Accepted: 02/28/2025] [Indexed: 04/18/2025] Open
Abstract
Maternally expressed gene 3 (MEG3), a long non-coding RNA, plays a pivotal role in various biological processes, including tumorigenesis. Aberrant expression of MEG3 has been implicated in several cancers, including genitourinary malignancies. This comprehensive review explores the multifaceted functions of MEG3 in the context of genitourinary cancers through unravelling the molecular mechanisms underlying the influence of MEG3 on cellular proliferation, apoptosis, invasion, and metastasis. Additionally, we discuss the potential clinical implications of MEG3 as a biomarker and therapeutic target in genitourinary cancers. By unraveling the intricate role of MEG3 in these biological processes, this review aims to contribute to the development of novel strategies for the diagnosis and treatment of genitourinary malignancies.
Collapse
Affiliation(s)
- Reem A. Assal
- Department of Pharmacology and Toxicology, Heliopolis University for Sustainable Development (HU), Cairo, Egypt
| | - Hannah H. Rashwan
- Bioinformatics Group, Center for Informatics Science (CIS), School of Information Technology and Computer Science (ITCS), Nile University, Giza, Egypt
| | - Zeina I. Zakaria
- Faculty of Biology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Jana H. Sweillam
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University, Cairo, Egypt
| | - Yasmine M. Fouda
- Faculty of Medicine, Al-Kasr Al Ainy, Cairo University, Cairo, Egypt
| | | | - Rana A. Youness
- Molecular Biology and Biochemistry Department, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University, Cairo, Egypt
| |
Collapse
|
55
|
Montalvan L, Bernhard JC, Bensalah K, Paparel P, Bigot P, Doumerc N, Olivier J, Bruyère F, Ingels A, Rouprêt M, Audenet F, Lebacle C, Long JA, Durand X, Waeckel T, Durand M, Lang H, Pignot G, Cussenot O, Charles T, Tambwe R, Xylinas E, Boissier R, Patard JJ, Beauval JB, Mallet R, Rouget B, Nouhaud FX. Effect of obesity on histological type of renal tumor: The Uroccr study 69. THE FRENCH JOURNAL OF UROLOGY 2025; 35:102884. [PMID: 40185313 DOI: 10.1016/j.fjurol.2025.102884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 03/13/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Renal cell carcinomas (RCC) are increasing worldwide and obesity is one of its recognized causal factor. Obesity thus could be differently associated with the different histologic types of renal tumors and also impact prognosis and patient care. PURPOSE The primary objective was to evaluate an association between obesity and histological type of RCC. The secondary objective was to assess if there was an effect of obesity on tumor characteristics. METHODS A multicenter comparative descriptive study was carried out in nearly 30 French hospitals. Histological features of operated RCC of patients included between 2007 and 2020 were compared according to their BMI. Obesity was defined as a BMI≥30kg/m2, with a P<0.05. RESULTS In total, 6749 RCC were analyzed, including 1687 (25%) from obese patients. This population was more frequently diabetic (61% versus 41%, P<0.001) and hypertensive (27% versus 11%, P<0.001). Regarding histological evaluation, the obese group presented clearer cell carcinoma. Other types presented odd ratios below 1 on multivariate analysis, P<0.01 except for type II papillary RCC. Tumor size (pT) and nuclear ISUP grade were both lower when BMI was above 30kg/m2: 72.4% of size pT 1-2 versus 68.5% (P=0.026) and 50% of nuclear ISUP grade 1-2 versus 43.3% for the non-obese group (P<0.0001). There was no difference in survival due to an insufficient number of events. CONCLUSION In this large cohort study, we report novel data on a positive association between obesity and histological type of RCC, in particular clear cell RCC. LEVEL OF EVIDENCE: 3
Collapse
|
56
|
Shiota M, Nemoto S, Ikegami R, Tanegashima T, Blas L, Miyake H, Takahashi M, Oya M, Tsuchiya N, Masumori N, Kobayashi K, Obara W, Shinohara N, Fujimoto K, Nozawa M, Ohba K, Ohyama C, Hashine K, Akamatsu S, Motoshima T, Mita K, Gotoh M, Tatarano S, Fujisawa M, Tomita Y, Mukai S, Ito K, Eto M. Predictive Model of Objective Response to Nivolumab Monotherapy for Advanced Renal Cell Carcinoma by Machine Learning Using Genetic and Clinical Data: The SNiP-RCC Study. JCO Clin Cancer Inform 2025; 9:e2400167. [PMID: 40279530 DOI: 10.1200/cci-24-00167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 12/10/2024] [Accepted: 02/24/2025] [Indexed: 04/27/2025] Open
Abstract
PURPOSE Anti-PD-1 antibodies are widely used for cancer treatment, including in advanced renal cell carcinoma (RCC). However, the therapeutic response varies among patients. This study aimed to predict tumor response to nivolumab anti-PD-1 antibody treatment for advanced RCC by integrating genetic and clinical data using machine learning (ML). METHODS Clinical and single-nucleotide polymorphism (SNP) data obtained in the SNPs in nivolumab PD-1 inhibitor for RCC study, which enrolled Japanese patients treated with nivolumab monotherapy for advanced clear cell RCC, were used. A point-wise linear (PWL) algorithm, logistic regression with elastic-net regularization, and eXtreme Gradient Boosting were used in this study. AUC values for objective response and C-indices for progression-free survival (PFS) were calculated to evaluate the utility of the models. RESULTS Among the three ML algorithms, the AUC values to predict objective response were highest for the PWL algorithm among all the data sets. Three predictive models (clinical model, small SNP model, and large SNP model) were created by the PWL algorithm using the clinical data alone and using eight and 49 SNPs in addition to the clinical data. C-indices for PFS by the clinical model, small SNP model, and large SNP model were 0.522, 0.600, and 0.635, respectively. CONCLUSION The results demonstrated that the SNP models created by ML produced excellent predictions of tumor response to nivolumab monotherapy for advanced clear cell RCC and will be helpful in treatment decisions.
Collapse
Affiliation(s)
- Masaki Shiota
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shota Nemoto
- Industrial & Digital Business Unit, Hitachi, Ltd, Tokyo, Japan
| | - Ryo Ikegami
- Industrial & Digital Business Unit, Hitachi, Ltd, Tokyo, Japan
| | - Tokiyoshi Tanegashima
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Leandro Blas
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hideaki Miyake
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masayuki Takahashi
- Department of Urology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Mototsugu Oya
- Department of Urology, Keio University School of Medicine, Tokyo, Japan
| | - Norihiko Tsuchiya
- Department of Urology, Faculty of Medicine, Yamagata University, Yamagata, Japan
| | - Naoya Masumori
- Department of Urology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Keita Kobayashi
- Department of Urology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Wataru Obara
- Department of Urology, Iwate Medical University School of Medicine, Iwate, Japan
| | - Nobuo Shinohara
- Department of Renal and Genitourinary Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | | | - Masahiro Nozawa
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kojiro Ohba
- Department of Urology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Chikara Ohyama
- Department of Urology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Katsuyoshi Hashine
- Department of Urology, National Hospital Organization Shikoku Cancer Center, Ehime, Japan
| | - Shusuke Akamatsu
- Department of Urology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takanobu Motoshima
- Department of Urology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Koji Mita
- Department of Urology, Hiroshima City Asa Citizens Hospital, Hiroshima, Japan
| | - Momokazu Gotoh
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shuichi Tatarano
- Department of Urology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Masato Fujisawa
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshihiko Tomita
- Department of Urology and Molecular Oncology, Graduate School of Medicine and Dental Sciences, Niigata University, Niigata, Japan
| | - Shoichiro Mukai
- Department of Urology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Keiichi Ito
- Department of Urology, National Defense Medical College, Saitama, Japan
| | - Masatoshi Eto
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
57
|
Lu Z, Xu J, Li J. The Transcription Factor ATF2 Accelerates Clear Cell Renal Cell Carcinoma Progression Through Activating the PLEKHO1/NUS1 Pathway. Mol Carcinog 2025; 64:617-628. [PMID: 39777695 DOI: 10.1002/mc.23868] [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: 09/12/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common malignant cancer with high mortality rate. Activating transcription factor 2 (ATF2) and pleckstrin homology domain containing O1 (PLEKHO1) were reported to participate in numerous cancers. However, their roles and the detailed mechanisms in ccRCC development remain largely unknown. RT-qPCR and western blot were used to measure the levels of PLEKHO1, ATF2, and nuclear undecaprenyl pyrophosphate synthase 1 (NUS1). Cell proliferation, apoptosis, invasion, migration and stemness were evaluated using CCK-8 assay, flow cytometry, transwell invasion assay, wound-healing assay and sphere formation assay, respectively. Dual-luciferase reporter assay was conducted to verify the relationship between ATF2 and PLEKHO1. The interaction between PLEKHO1 and NUS1 was proved by Co-IP assay. Xenograft models were utilized to evaluate the tumorigenic capability of ccRCC cells upon PLEKHO1 knockdown. PLEKHO1, ATF2 and NUS1 expression were significantly elevated in ccRCC, and PLEKHO1 might be a prognosis biomarker for ccRCC. PLEKHO1 depletion significantly inhibited cell proliferation, invasion, migration, stemness, and induced cell apoptosis in ccRCC cells. ATF2 activated PLEKHO1 expression via transcription regulation, and PLEKHO1 overexpression could reverse the suppressive effects of ATF2 knockdown on the malignant behaviors of ccRCC cells. Moreover, PLEKHO1 directly bound to NUS1, and PLEKHO1 depletion markedly restrained ccRCC progression through targeting NUS1 in vitro and in vivo. Our findings suggested that ATF2 transcriptionally activated PLEKHO1 to promote the development of ccRCC via regulating NUS1 expression.
Collapse
Affiliation(s)
- Zheng Lu
- Gravel Center, Nanyang First People's Hospital, Nanyang, China
| | - Jinge Xu
- Department of Urology, The Fourth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Junyu Li
- Department of Urology, The Fourth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
58
|
Alves Â, Ferreira M, Eiras M, Lima L, Medeiros R, Teixeira AL, Dias F. Exosome-derived hsa-miR-200c-3p, hsa-miR-25-3p and hsa-miR-301a-3p as potential biomarkers and therapeutic targets for restoration of PTEN expression in clear cell renal cell carcinoma. Int J Biol Macromol 2025; 302:140607. [PMID: 39900161 DOI: 10.1016/j.ijbiomac.2025.140607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/05/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is an aggressive kidney cancer subtype with limited biomarkers and therapeutic options. Thus, the present study aimed to evaluate the biomarker and therapeutic potential of Phosphatase and Tensin Homologue (PTEN)-regulating microRNAs (miRNAs) in 2D and 3D ccRCC models. Extracellular vesicles (EVs) from four renal cell lines were characterized, and selected miRNAs (hsa-miR-200c-3p, hsa-miR-25-3p, and hsa-miR-301a-3p) were quantified in cells and EVs. PTEN mRNA levels were measured intracellularly. 786-O cells were transfected with miRNA inhibitors in both models and effects on miRNA and PTEN expression were assessed alongside phenotypic alterations. The expression of target miRNAs increased with ccRCC cell aggressiveness, both intracellularly and in EVs, while PTEN mRNA expression decreased. Combined inhibition of these miRNAs significantly increased PTEN expression, reducing tumor cell proliferation and migration in 2D models and decreasing spheroid size and metabolic capacity in 3D models. These miRNAs show potential as biomarkers for monitoring disease aggressiveness and as therapeutic targets in ccRCC, potentially leading to more effective and personalized treatment approaches for ccRCC patients.
Collapse
Affiliation(s)
- Ângela Alves
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal
| | - Mariana Ferreira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal
| | - Mariana Eiras
- School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal; Experimental Pathology and Therapeutics Group, Research Center of IPO Porto (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal
| | - Luís Lima
- Experimental Pathology and Therapeutics Group, Research Center of IPO Porto (CI-IPOP), RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; School of Medicine and Biomedical Sciences (ICBAS), University of Porto, 4050-513 Porto, Portugal; Faculty of Medicine (FMUP), University of Porto, 4200-319 Porto, Portugal; Laboratory Medicine, Clinical Pathology Department, Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal; Biomedicine Research Center (CEBIMED), Research Innovation and Development Institute (FP-I3ID), Faculty of Health Sciences, Fernando Pessoa University (UFP), 4249-004 Porto, Portugal; Research Department, Portuguese League Against Cancer Northern Branch (LPCC-NRN), 4200-172 Porto, Portugal
| | - Ana Luísa Teixeira
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal
| | - Francisca Dias
- Molecular Oncology and Viral Pathology Group, Research Center of IPO-Porto (CI-IPOP) &RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto)/Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal.
| |
Collapse
|
59
|
Wei Z, Ye Y, Liu C, Wang Q, Zhang Y, Chen K, Cheng G, Zhang X. MIER2/PGC1A elicits sunitinib resistance via lipid metabolism in renal cell carcinoma. J Adv Res 2025; 70:287-305. [PMID: 38702028 PMCID: PMC11976417 DOI: 10.1016/j.jare.2024.04.032] [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: 02/04/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024] Open
Abstract
INTRODUCTION Renal cell carcinoma (RCC) is one of the most common malignant tumors of the urinary system and accounts for more than 90 % of all renal tumors. Resistance to targeted therapy has emerged as a pivotal factor that contributes to the progressive deterioration of patients with advanced RCC. Metabolic reprogramming is a hallmark of tumorigenesis and progression, with an increasing body of evidence indicating that abnormal lipid metabolism plays a crucial role in the advancement of renal clear cell carcinoma. OBJECTIVES Clarify the precise mechanisms underlying abnormal lipid metabolism and drug resistance. METHODS Bioinformatics screening and analyses were performed to identify hub gene. qRT-PCR, western blot, chromatin immunoprecipitation (ChIP) assays, and other biological methods were used to explore and verify related pathways. Various cell line models and animal models were used to perform biological functional experiments. RESULTS In this study, we identified Mesoderm induction early response 2 (MIER2) as a novel biomarker for RCC, demonstrating its role in promoting malignancy and sunitinib resistance by influencing lipid metabolism in RCC. Mechanistically, MIER2 facilitated P53 deacetylation by binding to HDAC1. Acetylation modification augmented the DNA-binding stability and transcriptional function of P53, while deacetylation of P53 hindered the transcriptional process of PGC1A, leading to intracellular lipid accumulation in RCC. Furthermore, Trichostatin A (TSA), an inhibitor of HDAC1, was found to impede the MIER2/HDAC1/P53/PGC1A pathway, offering potential benefits for patients with sunitinib-resistant renal cell cancer. CONCLUSION Our findings highlight MIER2 as a key player in anchoring HDAC1 and inhibiting PGC1A expression through the deacetylation of P53, thereby inducing lipid accumulation in RCC and promoting drug resistance. Lipid-rich RCC cells compensate for energy production and sustain their own growth in a glycolysis-independent manner, evading the cytotoxic effects of targeted drugs and ultimately culminating in the development of drug resistance.
Collapse
Affiliation(s)
- Zhihao Wei
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuzhong Ye
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenchen Liu
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qi Wang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunxuan Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kailei Chen
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gong Cheng
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Shenzhen Huazhong University of Science and Technology Research Institute, China.
| |
Collapse
|
60
|
Ged Y, Touma A, Meza Contreras L, Elias R, Van Galen J, Cupo O, Baraban E, Singla N, Lee CH, Pal S, Zibelman M, Kotecha RR. Multi-institutional Analysis of Immune-Oncology Combination Therapy for Metastatic MiT Family Translocation Renal Cell Carcinoma. J Immunother 2025; 48:113-117. [PMID: 39901604 PMCID: PMC11875401 DOI: 10.1097/cji.0000000000000549] [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: 08/06/2024] [Accepted: 12/03/2024] [Indexed: 02/05/2025]
Abstract
SUMMARY Metastatic translocation renal cell carcinomas (mtRCCs) are rare and aggressive tumors with limited treatment options. Recent studies demonstrated promising activity of immune-oncology (IO) combinations in mtRCC. However, the effectiveness of dual IO combinations versus IO plus VEGF-TKI combinations remains unclear. We conducted a retrospective analysis of IO combinations in mtRCC patients at 4 institutions. Eligible patients had confirmed mtRCC by genitourinary pathologist and received IO combination therapy (IO+IO or IO+VEGF-TKI). Clinical data and treatment outcomes were recorded from the start of systemic therapy. Objective response rate (ORR) was retrospectively evaluated, and time to treatment failure (TTF), and overall survival (OS) were compared for IO+IO and IO+VEGF-TKI groups. We identified 22 mtRCC patients who received IO combinations, all confirmed to have TFE3 rearrangement by FISH. Most patients were female (68%) with a median age of 41 years (16-79). Treatment breakdown included: IO+IO (n=8, 36%) and IO+VEGF-TKI (n=14, 64%). In the evaluable patients for the efficacy analysis, ORR was 14% (1/7) for the IO+IO group and 54% (6/11) for the IO+VEGF-TKI group. With a median follow-up of 32.4 months, the median TTF was 1.2 months and 6.2 months in the IO+IO and IO+VEGF-TKI groups, respectively ( P =0.12). There was no statistically significant difference in median OS between both groups, 36.7 months in the IO+IO group and 15.6 months in IO+VEGF-TKI ( P =0.9). Our findings demonstrate that IO+VEGF-TKI resulted in higher ORR and TTF point estimates without statistically detectable differences, compared with IO+IO therapy. Larger studies are needed to validate these findings and optimize treatment selection.
Collapse
Affiliation(s)
- Yasser Ged
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Amina Touma
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Luis Meza Contreras
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Roy Elias
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Joseph Van Galen
- Department of Hematology and Medical Oncology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA
| | - Olivia Cupo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ezra Baraban
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Nirmish Singla
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Chung-Han Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sumanta Pal
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Matthew Zibelman
- Department of Hematology and Medical Oncology, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA
| | - Ritesh R. Kotecha
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Medicine, Weill Cornell Medical College, New York, NY
| |
Collapse
|
61
|
Chao CL, Reddy NK, Visa M, Kundu SD, Eskandari MK. Late Survival and Long-Term Follow-Up After Radical Resection of Advanced Renal Cell Carcinoma With Associated Venous Tumor Thrombus. J Surg Oncol 2025; 131:917-924. [PMID: 39600112 PMCID: PMC12120382 DOI: 10.1002/jso.28020] [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: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND AND OBJECTIVES This study evaluates the prognostic value of venous tumor thrombus (VTT) in patients with advanced renal cell carcinoma (RCC) undergoing radical resection and inferior vena cava (IVC) thrombectomy. METHODS Retrospective review of patients with radical nephrectomy for RCC and associated VTT (2000-2024). Patients were dichotomized into Neves 0-II (infrahepatic) and Neves III-IV groups (suprahepatic) IVC involvement for univariate analysis. RESULTS A total of 64 patients (34 Neves 0-II and 30 Neves III-IV) were analyzed. No significant differences in patient or cancer characteristics. Neves III-IV was associated with greater blood loss (> 2 L) (62.1% vs. 37.9%, p = 0.02), greater intensive care unit length of stay (LOS) (4.4 vs. 1.4 days, p = 0.02), and postoperative LOS (11.0 vs. 6.5 days, p = 0.005). Overall, 30-day mortality was only 1.6% with a mean follow-up of 56.1 months. Local recurrence was 7.8% and IVC patency 96.9%. One-year survival was 82.0%, 5-year survival was 58.4%, and 15-year survival was 42.5% without significant difference between Neves levels. CONCLUSIONS Radical nephrectomy with VTT thrombectomy and primary IVC repair is safe with high early survival and low local recurrence. Extent of IVC tumor thrombus extension is not a poor prognostic factor for early or late survival.
Collapse
Affiliation(s)
- Calvin L. Chao
- Department of Surgery, Division of Vascular SurgeryNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Nidhi K. Reddy
- Department of Surgery, Division of Vascular SurgeryNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Maxime Visa
- Department of Surgery, Division of Vascular SurgeryNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Shilajit D. Kundu
- Department of Urology, Division of Urologic OncologyNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Mark K. Eskandari
- Department of Surgery, Division of Vascular SurgeryNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| |
Collapse
|
62
|
Huang Y, Eng W, Shao C, Cheng G, Qiang C, Peng W, Yang S, Liu S. Synthesis, Preclinical Characterizations and Imaging Studies of [ 18F]AlF-Labeled NY104, a CAIX-Targeting Diagnostic Agent. J Labelled Comp Radiopharm 2025; 68:e4142. [PMID: 40152075 DOI: 10.1002/jlcr.4142] [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: 09/10/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025]
Abstract
The protein carbonic anhydrase IX (CAIX) is highly expressed in clear cell renal cell carcinoma (ccRCC), and its restrictive expression in healthy tissues makes CAIX an attractive diagnostic target. The purpose of this study was to synthesize and evaluate [18F]AlF-NY104, a small-molecule CAIX-targeting PET agent in a preclinical ccRCC model. The radiolabeling parameters for [18F]AlF-NY104 have been optimized, including the solvent volume and reaction temperature. The high-purity product was synthesized by using an automated multifunctional module Mortenon M1, leading to nondecay-corrected radiochemical yields over 50% (n = 3). [18F]AlF-NY104 showed excellent tumor targeting capability and afforded high-quality microPET/CT imaging in the OS-RC-2 tumor model (15.01 ± 0.76 %ID/g [mean ± SEM]). The radiation exposure from [18F]AlF-NY104 is estimated to be 4.44 mSv for adult male and female human subjects, which is well below the FDA recommended whole-body single exposure limit of 30 mSv. Our investigation revealed that [18F]AlF-NY104 can be conveniently and efficiently radiolabeled by using an automated module. [18F]AlF-NY104 exhibited many outstanding properties, such as rapid uptake in tumor, rapid clearance through the kidney, and low uptake in most normal organs. Moreover, the high accumulation of [18F]AlF-NY104 in tumors in CAIX-positive models offers an alternative diagnostic strategy for patients with ccRCC.
Collapse
Affiliation(s)
- Yu Huang
- Hubei NO.3 People's Hospital of Jianghan University, Wuhan, China
| | - Waisi Eng
- Norroy Bioscience Co., LTD, Wuxi, China
| | | | | | | | - Wei Peng
- Norroy Bioscience Co., LTD, Wuxi, China
| | - Si Yang
- Norroy Bioscience Co., LTD, Wuxi, China
| | - Shiguo Liu
- Hubei NO.3 People's Hospital of Jianghan University, Wuhan, China
| |
Collapse
|
63
|
Xue Y, Chen T, Ma Z, Pu X, Xu J, Zhai S, Du X, Ji Y, Simon MC, Zhai W, Xue W. Osalmid sensitizes clear cell renal cell carcinoma to navitoclax through a STAT3/BCL-XL pathway. Cancer Lett 2025; 613:217514. [PMID: 39894195 DOI: 10.1016/j.canlet.2025.217514] [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: 08/22/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common and lethal urinary malignancy characterized by its resistance to apoptosis. Despite the emerging treatment options available for ccRCC, only a small proportion of patients achieve long-term survival benefits. Previous studies have demonstrated that inducing tumor cell senescence, followed by treatment using senolytics, represents a potential strategy for triggering tumor cell apoptosis. However, it remains unclear whether this strategy is suitable for the treatment of ccRCC. Using the whole-genome CRISPR screening database Dependency Map portal (DepMap), we identified ribonucleotide reductase family member 2 (RRM2), which catalyzes the conversion of ribonucleotides to deoxyribonucleotides (dNTPs), as an essential targetable gene for ccRCC. Herein, we report that the combination of the choleretic drug osalmid targeting RRM2 and the senolytic compound navitoclax targeting BCL-XL represents a novel therapeutic approach for ccRCC. Furthermore, we have validated this approach across a panel of human ccRCC cells with different genetic backgrounds and multiple preclinical models, including cell line-derived xenografts (CDX), patient-derived xenografts (PDX), and patient-derived organoids (PDO). Mechanistically, osalmid-mediated inhibition of dNTPs generation induces cellular senescence in ccRCC, concomitant with STAT3 activation and upregulation of BCL-XL, thus rendering these cells vulnerable to navitoclax, which targets the BCL-2 protein family.
Collapse
Affiliation(s)
- Yizheng Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China; Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Tianyi Chen
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Zehua Ma
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, 550001, China
| | - Xinyuan Pu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Junyao Xu
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Shuanfeng Zhai
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Xinxing Du
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China
| | - Yiyi Ji
- Department of Chemistry and Institute for Biophysical Dynamics, The University of Chicago, 929 East 57th Street, Chicago, IL, 60637, USA, Howard Hughes Medical Institute
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Wei Zhai
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China; Shanghai Immune Therapy Institute State, Key Laboratory of Oncogenes and Related Genes, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Wei Xue
- Department of Urology, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
64
|
Zhu Y, Yu S, Yang D, Yu T, Liu Y, Du W. Integrated Multi-Omics Analysis Unveils Distinct Molecular Subtypes and a Robust Immune-Metabolic Prognostic Model in Clear Cell Renal Cell Carcinoma. Int J Mol Sci 2025; 26:3125. [PMID: 40243888 PMCID: PMC11988429 DOI: 10.3390/ijms26073125] [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: 03/03/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is characterized by significant clinical and molecular heterogeneity, with immune and metabolic processes playing crucial roles in tumor progression and influencing patient outcomes. This study aims to elucidate the molecular subtypes of ccRCC by employing non-negative matrix factorization (NMF) clustering on differentially expressed genes (DEGs), thereby identifying distinct transcriptional profiles, immune cell infiltration patterns, and subsequent survival outcomes. Utilizing NMF clustering, we identified two molecular subtypes of ccRCC. We developed a prognostic model using LASSO-Cox regression, validated with multiple datasets and quantitative reverse transcription polymerase chain reaction (qRT-PCR), incorporating ten immunity- and metabolism-related genes (IMRGs) for overall survival (OS) prediction. Immune cell infiltration and tumor mutational burden (TMB) analyses were performed to explore differences between high- and low-risk groups, while Gene Set Enrichment Analysis (GSEA) provided insights into relevant biological pathways. The findings revealed that subtype C1, characterized by a "cold" tumor microenvironment, correlates with better prognostic outcomes compared to subtype C2, which exhibits an immunologically active environment and worse survival prospects. High-risk patients demonstrated poorer OS associated with alterations in immune and metabolic pathways. Immune checkpoint analysis indicated the upregulation of CTLA4, LAG3, and LGALS9 in high-risk patients, suggesting potential therapeutic targets. A nomogram integrating IMRG risk scores with clinical factors displayed high predictive accuracy for 1-, 3-, and 5-year OS. These findings provide novel insights into the molecular heterogeneity of ccRCC and emphasize the interconnected roles of immune dysregulation and metabolic alterations in tumor progression. By identifying key prognostic biomarkers and potential therapeutic targets, this study paves the way for innovative strategies aimed at harnessing immune and metabolic pathways for better clinical outcomes in ccRCC patients.
Collapse
Affiliation(s)
- Yilin Zhu
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
| | - Shihui Yu
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
| | - Dan Yang
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
| | - Tian Yu
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
| | - Yi Liu
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Wenlong Du
- Department of Bioinformatics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
- Department of Biophysics, School of Life Sciences, Xuzhou Medical University, Xuzhou 221004, China
| |
Collapse
|
65
|
Lu SH, Lui K, Qian Y, Zhou WY, Mu YY, Zhang W. Prognostic Role of SETDB2 in Clear Cell Renal Cell Carcinoma: Linking Immune Infiltration, Cuproptosis, and Tumor Suppression. Cancer Manag Res 2025; 17:675-692. [PMID: 40166493 PMCID: PMC11956738 DOI: 10.2147/cmar.s499771] [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: 11/13/2024] [Accepted: 03/20/2025] [Indexed: 04/02/2025] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is a relatively frequently diagnosed form of urological cancer that is highly malignant and associated with high rates of patient mortality. At present, there are few effective options for treating advanced cases of ccRCC, emphasizing the need to establish novel biomarkers and targets suitable for therapeutic intervention. SET domain bifurcated histone lysine methyltransferase 2 (SETDB2) belongs to the Su(var)3-9 subfamily of methyltransferases and has been linked to various forms of cancer, but the role it plays in ccRCC remains to be fully established. Methods Data on SETDB2 expression were downloaded from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Functional enrichment analyses were then used to probe the putative role that SETDB2 plays in the onset of ccRCC. The Gene Set Cancer Analysis (GSCA) platform and molecular docking analysis were utilized to investigate the relationship between gene expression and drug sensitivity. In the end, the core target and the active molecule were both given the green light for a molecular docking investigation. Functional assays and Western blotting performed with ccRCC cell lines were employed for the validation of the findings from these predictive analyses. Results SETDB2 downregulation was observed in ccRCC, and lower levels were found to linked with poor patient outcomes. Lower SETDB2 levels were associated with worse overall, progression-free, and disease-specific survival. In Functional enrichment analyses, SETDB2 was predicted to regulate key ccRCC development-associated pathways. SETDB2 levels were also significantly associated with cuproptosis induction in KIRC tissues, while in immune cell infiltration analyses, SETDB2 expression was linked with immune responses within the tumor microenvironment. Functional experiments conducted with ccRCC cell lines unveiled molecular mechanisms through which SETDB2 appears to be capable of inhibiting the development of ccRCC. Conclusion Together, these analyses highlight the utility of SETDB2 as a prognostic biomarker in ccRCC. The interactions and associated pathways detected through these analyses provide unique insight into the potential functions of SETDB2 in this cancer type, providing an evidence base for future studies.
Collapse
Affiliation(s)
- Si Hao Lu
- Department of Nephrology, Air Force Hospital of Western Theater Command, Chengdu, 610000, People’s Republic of China
| | - Kui Lui
- Department of Nephrology, Air Force Hospital of Western Theater Command, Chengdu, 610000, People’s Republic of China
| | - Yue Qian
- Department of Pathogen Biology, Guizhou Nursing Vocational College, Guiyang, Guizhou, 550000, People’s Republic of China
| | - Wei ye Zhou
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, People’s Republic of China
| | - Ying Ying Mu
- Department of Pathology, Zunyi Hospital of Traditional Chinese Medicine, Zunyi, Guizhou, 563000, People’s Republic of China
| | - Wei Zhang
- Department of Pathogen Biology, Guizhou Nursing Vocational College, Guiyang, Guizhou, 550000, People’s Republic of China
- Cell Biology Department, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, 214122, People’s Republic of China
| |
Collapse
|
66
|
Wei S, Lyu F, Qian B, Tang Y, He Q. NIPAL4 is an important marker for clear cell renal cell carcinoma prognosis and immunotherapy. Sci Rep 2025; 15:10448. [PMID: 40140659 PMCID: PMC11947290 DOI: 10.1038/s41598-025-92811-1] [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: 11/05/2024] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) poses significant clinical challenges due to its high metastatic rate at diagnosis and resistance to conventional therapies. The magnesium transporter NIPAL4, involved in lipid metabolism and magnesium ion homeostasis, has recently been implicated in cancer biology but remains understudied in ccRCC. In this study, we identifies NIPAL4 as a potential prognostic marker and therapeutic target in ccRCC. High NIPAL4 expression is associated with poor prognosis and contributes to tumor proliferation and migration. Bioinformatics analyses indicate that NIPAL4 is involved in critical pathways related to the extracellular matrix, lipid metabolism, and the TGF-β signaling pathway. Moreover, NIPAL4's association with immune cell infiltration and immune checkpoint molecules highlights its potential as an immunotherapy marker. These findings warrant further investigation into NIPAL4's role in ccRCC to develop more effective treatment strategies.
Collapse
Affiliation(s)
- Shuyan Wei
- Department of Traditional Chinese Medicine and Acupuncture, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, 442000, P.R. China
- Chengdu University of Traditional Chinese Medicine, ChengDu, China
| | - Fang Lyu
- Department of Urolgoy, Union Hospital, Tongji Medical College, Tongji Medical College, Huazhong University of Science and Technology, No.1277 jiefang Avenue, Wuhan, 430022, China
| | - Bei Qian
- Department of Thyroid and Breast Surgery, Tongji Medic al College, Union Hospital, Huazhong University of Science and Technology, 1277 jiefang Avenue, Wuhan, 430022, China
| | - Yumei Tang
- Department of Nephrology, Meishan Second People's Hospital, Renshou, Meishan, China.
| | - Qingliu He
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| |
Collapse
|
67
|
Marques-Magalhães Â, Moreira-Silva F, Graça I, Dias PC, Correia MP, Alzamora MA, Henrique R, Lopez M, Arimondo PB, Miranda-Gonçalves V, Jerónimo C. Combination of MLo-1508 with sunitinib for the experimental treatment of papillary renal cell carcinoma. Front Oncol 2025; 15:1399956. [PMID: 40196736 PMCID: PMC11973455 DOI: 10.3389/fonc.2025.1399956] [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: 03/12/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Renal cell carcinoma (RCC) is the 14th most incident cancer worldwide, and no curative therapeutic options are available for advanced and metastatic disease. Hence, new treatment alternatives are urgently needed to tackle disease management and drug resistance. Herein, we explored the use of MLo-1508 as an anti-tumoral agent in RCC and further assessed its combination with sunitinib for the treatment of papillary RCC. For that, different RCC cell lines were treated with both drugs, alone or in combination, and different phenotypic assays were performed. Moreover, global DNA methylation levels and specific DNMT3a activity were measured, and gene-specific CpG methylation and transcript levels were quantified after treatment. Finally, the combinatory potential of MLo-1508 and sunitinib were asses both in vitro and in vivo using the ACHN cell line. We found that MLo-1508 significantly decreased RCC cell viability while inducing apoptosis in a dose-dependent manner without cytotoxicity for non-malignant cells. Moreover, the treatment induced morphometric alterations and DNA damage in all RCC cell lines. MLo-1508 decreased DNMT1 and DNMT3A transcript levels in 786-O and ACHN cells, inhibited DNMT3A activity, and reduced the global DNA methylation content of ACHN cells. When combined with sunitinib, a reduction in ACHN cell viability, as well as cell cycle arrest at G2/M was observed. Importantly, MLo-1508 decreased the sunitinib effective anti-tumoral concentration against ACHN cell viability. In an in vivo ACHN CAM model, the combination induced cell necrosis. Thus, MLo-1508 might improve sensitivity to sunitinib treatment by decreasing the required concentration and delaying resistance acquisition.
Collapse
Affiliation(s)
- Ângela Marques-Magalhães
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Filipa Moreira-Silva
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Inês Graça
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Paula C. Dias
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Margareta P. Correia
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Maria Ana Alzamora
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
| | - Rui Henrique
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences of the University of Porto (ICBAS-UP), Porto, Portugal
| | - Marie Lopez
- Institut des Biomolécules Max Mousseron (IBMM), UMR 5247, CNRS-Université de Montpellier-ENSCM, Montpellier, France
| | - Paola B. Arimondo
- Epigenetic Chemical Biology, Institut Pasteur, UMR 3523CNRS, Paris, France
| | - Vera Miranda-Gonçalves
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP)/CI-IPOP@RISE (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences of the University of Porto (ICBAS-UP), Porto, Portugal
| |
Collapse
|
68
|
Yang W, Wu C, Jiang C, Jing T, Lu M, Xia D, Peng D. FDX1 overexpression inhibits the growth and metastasis of clear cell renal cell carcinoma by upregulating FMR1 expression. Cell Death Discov 2025; 11:115. [PMID: 40118855 PMCID: PMC11928736 DOI: 10.1038/s41420-025-02380-5] [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: 01/14/2025] [Revised: 02/10/2025] [Accepted: 02/27/2025] [Indexed: 03/24/2025] Open
Abstract
Kidney cancer has caused more than 150,000 deaths in 185 countries around the world and is a serious threat to human life. Clear cell renal cell carcinoma (ccRCC) is the most common type of kidney cancer. FDX1, a crucial gene for regulating copper death, plays an important role in tumors. However, its specific role in ccRCC remains unclear. In this study, by analysing data from the TCGA-KIRC and GEO databases and validation in clinical samples from our center, the expression characteristics of FDX1 and its relationship with tumor clinicopathological features and patient prognosis were clarified; the effects of FDX1 overexpression on ccRCC cell proliferation, apoptosis, migration, and invasion were determined via cell phenotype experiments and mouse orthotopic renal tumor growth models; and the downstream regulatory mechanism of FDX1 was determined via TMT proteomic sequencing, Co-IP assays, and RNA-sequencing detection. Our results confirmed that FDX1 was significantly underexpressed in ccRCC and that reduced FDX1 expression was associated with adverse clinicopathologic features and poor prognosis. FDX1 overexpression markedly inhibited the proliferation, migration, and invasion of ccRCC cells and promoted cell apoptosis in vitro. Mechanistically, FDX1 bound to the FMR1 protein and upregulated its expression, subsequently restraining Bcl-2 and N-cadherin expression and enhancing ALCAM, Cleaved Caspase-3, and E-cadherin expression. In mouse models, FDX1 overexpression significantly suppressed the growth and metastasis of renal tumors, but this inhibitory effect was markedly reversed after FMR1 expression was knocked down. Thus, our results confirmed that FDX1 expression is significantly reduced in ccRCC and serves as a prognostic marker for ccRCC patients and that its overexpression suppresses the growth and metastasis ability of ccRCC by promoting the expression of FRM1.
Collapse
Affiliation(s)
- Wuping Yang
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Cunjin Wu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Chaochao Jiang
- Department of Urology, Changxing Hospital of Traditional Chinese Medicine, Changxing, PR China
| | - Taile Jing
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Minghao Lu
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China
| | - Dan Xia
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.
| | - Ding Peng
- Department of Urology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, PR China.
| |
Collapse
|
69
|
Liu JY, Weng KQ, Gao QD, Xue XY, Xu N. Molecular subtyping of renal clear cell carcinoma based on prognostic RASSF family genes and validation of C1QL1 as a key prognostic marker. Sci Rep 2025; 15:9786. [PMID: 40118977 PMCID: PMC11928449 DOI: 10.1038/s41598-025-94978-z] [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: 11/18/2024] [Accepted: 03/18/2025] [Indexed: 03/24/2025] Open
Abstract
RASSF family proteins play crucial roles in mitosis, apoptosis, cell migration, adhesion, and immune functions, primarily acting as tumor suppressors. Their roles in renal clear cell carcinoma (ccRCC) are not fully understood. We analyzed the expression and prognostic significance of RASSF genes in 573 ccRCC samples from TCGA and GEO, identifying two molecular subtypes with distinct characteristics. We developed a RAS score to assess prognosis and molecular status and investigated the key gene C1QL1 through in vitro assays. Four RASSF genes were identified as associated with prognosis and progression in ccRCC. Based on their co-expression, we defined two patient subtypes, one with poorer prognosis. A higher RAS score correlated with advanced disease and worse outcomes but indicated a favorable response to specific inhibitors, supporting personalized treatment strategies. Additionally, VHL mutations may cause abnormal SFMBT1 expression, leading to high C1QL1 levels, which promote tumor progression via the YAP-EMT pathway. Our findings highlight the potential of RASSF-based molecular subtypes and scoring systems in personalizing ccRCC treatment. Understanding C1QL1's role may facilitate the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Jin-Yu Liu
- Department of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, The Affiliated Hospital of Putian University, Putian, 351100, China
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China
| | - Kang-Qiang Weng
- Department of Urology, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Qin-Dong Gao
- Department of Urology, Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519000, China
| | - Xue-Yi Xue
- Department of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, Binhai Campus of the First Affiliated Hospital, National Regional Medical Center, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
70
|
Zhang X, Ye J, Sun L, Xu W, He X, Bao J, Wang J. NCKAP1 Inhibits the Progression of Renal Carcinoma via Modulating Immune Responses and the PI3K/AKT/mTOR Signaling Pathway. Int J Mol Sci 2025; 26:2813. [PMID: 40141455 PMCID: PMC11942877 DOI: 10.3390/ijms26062813] [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: 02/06/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Nck-associated protein 1 (NCKAP1) is critical for cytoskeletal functions and various cellular activities, and deregulation of NCKAP1 in many cancers significantly influences the outcomes of malignant diseases. However, the functions of NCKAP1 in the progression of renal cancer are yet unknown. To investigate the specific roles of NCKAP1 in the immune regulation and tumor progression of renal cancer, the expression of NCKAP1 and genetic variations were analyzed across cancer types at different pathological stages via UALCAN and cBioPortal. Immune cell infiltration in renal cancer was also assessed by ssGSEA and single-cell gene expression data from the GEO. RNA sequencing of NCKAP1-overexpressing 769P cells further examined the impact of NCKAP1 on kidney cancer. Our pancancer analyses revealed a complex NCKAP1 expression profile across various cancer types, with reduced levels in renal cancer patients linked to patient prognosis. CIBERSORT and single-cell RNA sequencing revealed the expression patterns of NCKAP1 in different cell lineages in renal cancer and a significant correlation between NCKAP1 and immune cell infiltration in the kidney tumor microenvironment. We further verified that NCKAP1 suppressed cancer cell growth and affected tumor development in renal cancer via the PI3K/AKT/mTOR signaling pathway. Our results indicate that NCKAP1 is a potential predictive marker and treatment target for renal cancer.
Collapse
Affiliation(s)
- Xin Zhang
- Central Laboratory, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, China; (X.Z.); (L.S.); (W.X.)
| | - Jianqing Ye
- Department of Urology, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, 1665 Kongjiang Road, Shanghai 200092, China;
| | - Lixiang Sun
- Central Laboratory, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, China; (X.Z.); (L.S.); (W.X.)
| | - Wanli Xu
- Central Laboratory, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, China; (X.Z.); (L.S.); (W.X.)
| | - Xiaomeng He
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai 201508, China (J.B.)
| | - Juan Bao
- Shanghai Public Health Clinical Center, Fudan University, 2901 Caolang Road, Jinshan District, Shanghai 201508, China (J.B.)
| | - Jin Wang
- Central Laboratory, Zhongshan Hospital (Xiamen), Fudan University, Xiamen 361015, China; (X.Z.); (L.S.); (W.X.)
| |
Collapse
|
71
|
Zhang J, Liu G, Wang W. PRSS53 is a potential novel biomarker related to prognosis and immunity in clear cell renal cell carcinoma. Discov Oncol 2025; 16:362. [PMID: 40111561 PMCID: PMC11925835 DOI: 10.1007/s12672-025-02114-0] [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: 12/12/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE To analyze the expression levels, clinical significance, Immune infiltration and prognostic value of PRSS53 (Protease Serine 53) in clear cell renal cell carcinoma (ccRCC) using bioinformatics methods. METHODS Data on PRSS53 in ccRCC were extracted from databases and platforms, including The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression Project (GTEx), The Gene Expression Omnibus (GEO), Xiantao Academic Tool, Human Protein Atlas (HPA) and so on. We analyzed the relationship between PRSS53 expression and the clinical and pathological characteristics, diagnosis, immune infiltration and prognosis in ccRCC patients. Additionally, immunohistochemical analysis was performed on 9 pairs of ccRCC patient samples. RESULTS PRSS53 was significantly upregulated in ccRCC and was closely associated with the TNM stage and histological grade of ccRCC. Receiver operating characteristic (ROC) curve analysis demonstrated the excellent diagnostic performance of PRSS53 in ccRCC (AUC = 0.928). Patients with high PRSS53 expression exhibited lower overall survival (OS) and disease-specific survival (DSS). Gene set enrichment analysis (GSEA) revealed that PRSS53 is involved in cellular functions such as anchored component of membrane, basement membrane and RNA-binding involved in post-transcriptional gene silencing. Single-sample GSEA (ssGSEA) indicated a positive correlation between PRSS53 expression and T helper cells infiltration levels, and a negative correlation with T gamma delta (Tgd) cell infiltration. PRSS53 was predominantly expressed in renal proximal tubules. The immunohistochemical results and HPA database showed that PRSS53 protein expression was significantly lower in clinical ccRCC tissues compared to normal tissues. CONCLUSION PRSS53 is a new prognostic biomarker and potential therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Jiajun Zhang
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224006, China
| | - Guocheng Liu
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224006, China
| | - Wei Wang
- The Yancheng Clinical College of Xuzhou Medical University, Yancheng, 224006, China.
- Department of Urology, Yancheng No.1 People's Hospital, Yancheng, 224006, China.
| |
Collapse
|
72
|
Xia H, He T, Li X, Zhao K, Zhang Z, Zhu G, Yang H, Yan X, Wang Q, Li Z, Jiang Z, Wang K, Yin X. Study on the mechanism of BGN in progression and metastasis of ccRCC. BMC Med Genomics 2025; 18:55. [PMID: 40108593 PMCID: PMC11924620 DOI: 10.1186/s12920-025-02124-5] [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: 03/24/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
PURPOSE To investigate the role of Biglycan(BGN) in the progression and metastasis of clear cell renal cell carcinoma(ccRCC). METHODS Based on multiple public databases, we investigated the expression level of BGN in ccRCC, its clinical significance, and its association with immune cells. Real-time fluorescence quantitative polymerase chain reaction(PCR) was employed to validate BGN expression in tumor and adjacent normal tissues from ten patients. We utilized RNA sequencing results for further analysis, including differential gene analysis, GO-KEGG analysis, and GSEA analysis, to identify the signaling pathways through which BGN exerts its effects. BGN knockdown cells(786-0 and Caki-1) were generated through lentiviral transfection to examine the impact of BGN on ccRCC. Cell proliferation, migration, and invasion were assessed using CCK8, colony formation, wound healing, Transwell migration, and invasion assays, respectively. RESULTS Our findings from database analysis and PCR revealed a significant upregulation of BGN expression in kidney cancer tissues compared to normal tissues. Further analysis demonstrated a correlation between high BGN expression and ccRCC progression and immune infiltration. In vitro experiments confirmed that BGN silencing effectively inhibited cell proliferation, migration, and invasion of ccRCC. Mechanistically, these effects may be mediated through the MAPK signaling pathway. CONCLUSION BGN potentially plays a pivotal role in the progression and metastasis of ccRCC, possibly acting through the MAPK signaling pathway. Therefore, BGN holds promise as a potential therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Hanqing Xia
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Tianzhen He
- Institute of Special Environmental Medicine, Nantong University, Nantong, China
| | - Xueyu Li
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Kai Zhao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Zongliang Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Guanqun Zhu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Han Yang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Xuechuan Yan
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Qinglei Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Zhaofeng Li
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Zaiqing Jiang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Ke Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Xinbao Yin
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China.
| |
Collapse
|
73
|
Jerez-Lillo N, Tapia A, Hugo Lachos V, Ramos PL. A New Semiparametric Power-Law Regression Model With Long-Term Survival, Change-Point Detection and Regularization. Stat Med 2025; 44:e70043. [PMID: 40108812 DOI: 10.1002/sim.70043] [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: 03/13/2024] [Revised: 08/20/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025]
Abstract
Kidney cancer, a potentially life-threatening malignancy affecting the kidneys, demands early detection and proactive intervention to enhance prognosis and survival. Advancements in medical and health sciences and the emergence of novel treatments are expected to lead to a favorable response in a subset of patients. This, in turn, is anticipated to enhance overall survival and disease-free survival rates. Cure fraction models have become essential for estimating the proportion of individuals considered cured and free from adverse events. This article presents a novel piecewise power-law cure fraction model with a piecewise decreasing hazard function, deviating from the traditional piecewise constant hazard assumption. By analyzing real medical data, we evaluate various factors to explain the survival of individuals. Consistently, positive outcomes are observed, affirming the significant potential of our approach. Furthermore, we use a local influence analysis to detect potentially influential individuals and perform a postdeletion analysis to analyze their impact on our inferences.
Collapse
Affiliation(s)
- Nixon Jerez-Lillo
- Facultad de Matemáticas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandra Tapia
- Facultad de Matemáticas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Victor Hugo Lachos
- Department of Statistics, University of Connecticut, Storrs, Connecticut, USA
| | - Pedro Luiz Ramos
- Facultad de Matemáticas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
74
|
Mao W, Wu T, Barge S, Zubair M, Sanchez D, Geng J, Bhattacharya A, Chen M. Comparing oncologic outcomes of partial and radical nephrectomy for T2 renal cell carcinoma: a propensity score matching cohort study and an external multicenter validation. World J Urol 2025; 43:166. [PMID: 40072570 DOI: 10.1007/s00345-025-05561-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 03/06/2025] [Indexed: 03/14/2025] Open
Abstract
PURPOSE There is very limited evidence on the optimal surgical treatment for patients with larger T2 renal tumors. This study aims to evaluate the oncologic outcomes of partial nephrectomy (PN) and radical nephrectomy (RN) in T2 renal cell carcinoma (RCC). METHODS A retrospective data analysis was conducted on T2 RCC patients who underwent PN or RN between 2004 and 2019 using the SEER database, and validated with data from multiple centers in China from 2014 to 2019. Kaplan-Meier survival curves and multivariate Cox regression analysis were performed to assess the treatment effects of PN and RN. RESULTS The Kaplan-Meier survival curves showed that both in the SEER database and Chinese multicenter data, PN was associated with a higher overall survival (OS) compared to RN in the all patients, male and age ≤ 60 years groups after propensity score matching. The multivariate Cox regression analysis indicated that PN benefited the OS in the all patients (RN vs. PN, HR = 1.476; 95% CI, 1.113-1.957; P = 0.007), and T2a age ≤ 60 years subgroup (RN vs. PN, HR = 2.147; 95% CI, 1.228-3.754; P = 0.007). CONCLUSION PN is a viable treatment option for patients with T2 RCC, particularly for patients with T2a age ≤ 60 years, where PN is associated with a higher OS rate. However, patients with stage T2 tumors undergoing PN should be referred to high-volume centers for treatment.
Collapse
Affiliation(s)
- Weipu Mao
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Tiange Wu
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China
| | - Sagar Barge
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Muhammad Zubair
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Daniel Sanchez
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Jiang Geng
- Department of Urology, Bengbu First People's Hospital, Bengbu, China.
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China.
| | - Atrayee Bhattacharya
- Department of Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Ming Chen
- Department of Urology, Zhongda Hospital Southeast University, Nanjing, China.
| |
Collapse
|
75
|
Liu Y, Gong L, Feng J, Xiao C, Liu C, Chen B, Chen L, Jin M, Guan Y, Gao Z, Huang W. Co-delivery of axitinib and PD-L1 siRNA for the synergism of vascular normalization and immune checkpoint inhibition to boost anticancer immunity. J Nanobiotechnology 2025; 23:194. [PMID: 40059141 PMCID: PMC11892300 DOI: 10.1186/s12951-025-03170-y] [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: 11/26/2024] [Accepted: 01/27/2025] [Indexed: 05/13/2025] Open
Abstract
Immune checkpoint inhibition (ICI) has become the mainstay of immunotherapy for the treatment of renal cell carcinoma (RCC). However, only a small portion of patients exhibit a positive response to PD-1/PD-L1 blockade therapy and the key reason is that RCC belongs to a vascular-rich tumor for promoting immunosuppression. Specifically, the dysfunctional tumor vasculature hinders effector T cell infiltration and induces immunosuppressive tumor microenvironment via the release of cytokine, which attenuates the therapeutic efficacy of ICI. Therefore, regulating abnormal tumor vasculature may be a promising strategy to overcome the immunosuppressive microenvironment and enhance ICI therapy. Here, we propose an NGR peptide-modified actively targeted liposome (Axi/siRNAPD-L1@NGR-Lipo) to encapsulate the anti-angiogenic agents Axitinib and PD-L1 siRNA to promote tumor vasculature normalization and relieve immune evasion for enhanced anti-tumor immunotherapy. With NGR-mediated tumor homing and active targeting, Axi/siRNAPD-L1@NGR-Lipo could act on tumor vascular endothelial cells to inhibit neo-angiogenesis, increase pericyte coverage and vascular perfusion, and normalize the structure and function of tumor blood vessels. Meanwhile, it also enhanced immune effector T cells and NK cells infiltration and reduced the proportion of immunosuppressive T cells including MDSC cells and Tregs, thus improving the tumor immunosuppressive microenvironment. Moreover, Axi/siRNAPD-L1@NGR-Lipo reduced the expression of PD-L1 protein in tumor cells, restored the recognition and killing ability of cytotoxic T cells, and relieved immune evasion. As expected, Axi/siRNAPD-L1@NGR-Lipo displayed superior anti-tumor and anti-metastatic efficacy in mice bearing RCC. Overall, this study demonstrated the important potential of regulating abnormal tumor vasculature to reshape the immunosuppressive microenvironment and boost ICI therapy, which represents a promising avenue for the synergistic anti-tumor with cancer immunotherapy.
Collapse
Affiliation(s)
- Yanhong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Department of Pharmacy, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Gusu School of Nanjing Medical University, Suzhou, 215000, China
| | - Liming Gong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jing Feng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Congcong Xiao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Chenfei Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Bohan Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Liqing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Youyan Guan
- Department of Urology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Department of Pharmaceutics, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
76
|
Li Y, Fan C, Jiang F, Zhang J, Li Y, Jiang Y, Zhang R, Yu Z, Wang S. Identification of LIMK1 as a biomarker in clear cell renal cell carcinoma: from data mining to validation. J Cancer Res Clin Oncol 2025; 151:104. [PMID: 40056237 PMCID: PMC11890329 DOI: 10.1007/s00432-025-06146-5] [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: 10/24/2024] [Accepted: 02/20/2025] [Indexed: 03/10/2025]
Abstract
PURPOSE Clear cell renal cell carcinoma (ccRCC) is one of the most common types of renal cancer. LIM kinase 1 (LIMK1) reportedly plays an important role in tumorigenesis. However, the involvement of LIMK1 in the progression of ccRCC remains ambiguous. METHODS Based on the TCGA and CPTAC databases, the expression of LIMK1 in ccRCC was evaluated. In the TCGA-ccRCC cohort, the relationships between LIMK1 and immune cell infiltration as well as immune checkpoints were assessed. The high expression of LIMK1 in ccRCC was verified by qRT-PCR in four RCC cell lines. Immunohistochemistry was used to evaluate the expression of LIMK1 in clinical samples. The association between LIMK1 expression and survival prognosis was explored via Kaplan-Meier survival curve in the TCGA-ccRCC and local cohorts. The effects of LIMK1 knockdown on the proliferation, migration, and invasion abilities of RCC cells were evaluated via colony, CCK-8, wound healing, and Transwell assays. RESULTS Elevated expression level of LIMK1 was found in the TCGA-ccRCC cohort and was confirmed in RCC cell lines and clinical samples. Up-regulation of LIMK1 was found to be correlated with poor prognosis in TCGA-ccRCC and external cohorts. In addition, high-LIMK1 was associated with clinicopathological stage, immune cell infiltration and immune checkpoint in ccRCC. Importantly, knockdown of LIMK1 diminished the capability of proliferation, migration, and invasion in RCC cells. CONCLUSION LIMK1 may serve as a promising diagnostic and prognostic biomarker of ccRCC.
Collapse
Affiliation(s)
- Yifei Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Congcong Fan
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Feng Jiang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jingnan Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yanzhen Li
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yanjie Jiang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Rui Zhang
- Zhejiang Key Laboratory of Intelligent Cancer Biomarker Discovery and Translation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Zhixian Yu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Siqi Wang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
77
|
Li B, Pan Y, Wu J, Miao C, Wang Z. Large-scale genomic-wide CRISPR screening revealed PRC1 as a tumor essential candidate in clear cell renal cell carcinoma. Int J Med Sci 2025; 22:1658-1671. [PMID: 40093809 PMCID: PMC11905274 DOI: 10.7150/ijms.107691] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
Background: Clear cell renal cell carcinoma (ccRCC) is a prevalent and aggressive subtype of kidney cancer, often associated with metastasis and recurrence. Identifying key genes involved in ccRCC progression is critical for improving treatment strategies and patient outcomes. Methods: We performed a large-scale genome-wide CRISPR screening to identify genes crucial to ccRCC progression using the DepMap database. For discovery and validation, we integrated multi-omics data from The Cancer Genome Atlas (TCGA), GEO, and the NJMU-ccRCC clinical cohort. Bioinformatics analyses, including differential expression, pathway enrichment, and protein-protein interaction network analysis, were conducted to elucidate the biological functions. To validate our findings, we employed immunohistochemistry, qRT-PCR, and various cellular assays to investigate the role of PRC1 in ccRCC. Results: CRISPR screening identified PRC1 as a key gene significantly overexpressed in ccRCC tissues from the DepMap database. Elevated PRC1 expression was associated with poor overall survival, disease-specific survival, and progression-free interval. Silencing PRC1 in ccRCC cell lines inhibited cell proliferation, migration, and colony formation. Functional enrichment analyses revealed that PRC1 is involved in essential processes such as cell cycle regulation, mitosis, and cytokinesis. Additionally, PRC1 expression was correlated with the activation of the Wnt/β-catenin pathway, suggesting that PRC1 plays a pivotal role in tumor progression. Conclusion: PRC1 emerges as a promising biomarker and therapeutic target for ccRCC. Elevated PRC1 expression is associated with poor prognosis, and its inhibition suppresses ccRCC cell proliferation and migration. Our findings underscore the crucial role of PRC1 in ccRCC progression and highlight the need for further investigation into its molecular mechanisms and therapeutic potential.
Collapse
Affiliation(s)
| | | | - Jiajin Wu
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, Jiangsu Province 210029, China
| | - Chenkui Miao
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, Jiangsu Province 210029, China
| | - Zengjun Wang
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Street, Nanjing, Jiangsu Province 210029, China
| |
Collapse
|
78
|
Zhang M, Li Y, Quan Z, Zhou X, Meng X, Ye J, Wang Y, Wang J, Qin W, Wang J, Kang F. Value of [ 68Ga]Ga-PSMA-11 PET/CT in Reflecting the Intra- and Intertumor Heterogeneity of Neovascularization in Clear Cell Renal Cell Carcinoma. Mol Pharm 2025; 22:1529-1538. [PMID: 39912786 DOI: 10.1021/acs.molpharmaceut.4c01248] [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: 02/07/2025]
Abstract
Prostate-specific membrane antigen (PSMA) is a potential target for the diagnosis and treatment of angiogenesis in clear cell renal cell carcinoma (ccRCC). We aimed to investigate the degree of PSMA signal variability in ccRCC and assess its correlation with neovascularization in the tumor microenvironment. We included 120 patients with suspected renal tumors who underwent [68Ga]Ga-PSMA-11 positron emission tomography/computed tomography (PET/CT) scan before surgery in this retrospective study, including 98 ccRCC, 17 non-ccRCC, and 5 benign diseases. We compared the maximum standard uptake value (SUVmax) and tumor-to-liver ratio (TLR) of primary lesions in different groups and analyzed the diagnostic efficacy of PSMA imaging for ccRCC. The coefficient of variation (CV) of SUVmax, which reflects intertumor heterogeneity, and volume ratio (VR), which reflects intratumor heterogeneity, were obtained from PET imaging. We analyzed the correlation between SUVmax, PSMA immunohistiochemical (IHC) staining, microvessel density (MVD), and serum vascular endothelial growth factor (VEGF) and compared the inter- and intratumor heterogeneity of primary lesions and metastases. In our study, ccRCC showed significantly higher SUVmax and TLR compared to non-ccRCC and benign diseases (F = 14.48, p < 0.001; F = 14.49, p < 0.001). PSMA IHC staining exhibited moderate correlation with SUVmax (r = 0.421, p = 0.021) and MVD (r = 0.518, p = 0.003), but it was not correlated with serum VEGF (r = -0.003, p = 0.989). SUVmax had a moderate correlation with MVD (r = 0.448, p = 0.013) and serum VEGF (r = 0.345, p = 0.020). Serum VEGF exhibited a weak correlation with MVD (r = 0.338, p = 0.145). Based on the correlation, the SUVmax-to-angiogenesis model was validated. The mean SUVmax values of primary lesions, bone metastases, and tumor thrombi were 16.13, 18.69, and 6.02, respectively. The CV of the mean SUVmax was 58.5%, 55.9%, and 80.6%. The mean VR values of primary lesions, bone metastases, and tumor thrombi were 0.33, 0.46, and 0.75, respectively. The CV of the mean VR was 81.8%, 41.3%, and 26.7%. The SUVmax of primary lesions was significantly correlated with corresponding bone metastases and tumor thrombi (r = 0.52, p = 0.011; r = 0.87, p = 0.024). The SUVmax of primary lesion in localized ccRCC and advanced ccRCC showed no significant difference (p = 0.251), while the VR was significantly different (p = 0.049). In conclusion, [68Ga]Ga-PSMA-11 PET/CT is an effective molecular imaging tool for assessing angiogenesis and its heterogeneity and differentiating ccRCC. The SUVmax of primary lesions was significantly correlated with PSMA IHC staining, MVD, and serum VEGF. The intertumor heterogeneity of tumor thrombi was significantly higher than that of primary lesions and bone metastases. Primary lesions exhibited the highest intratumor heterogeneity, and lesions with high intratumor heterogeneity showed invasive behavior. PSMA uptake by primary lesions has a positive effect on metastasis.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032 China
| | - Yu Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032 China
- State Key Laboratory of Oral Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases Shaanxi Key Laboratory of Stomatology, Department of Operative Dentistry and Endodontics School of Stomatology, Fourth Military Medical University, Xi'an, Shaanxi 710032 China
| | - Zhiyong Quan
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032 China
| | - Xiang Zhou
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032 China
| | - Xiaoli Meng
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032 China
| | - JiaJun Ye
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032 China
| | - Yirong Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032 China
| | - Junling Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032 China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032 China
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032 China
| | - Fei Kang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032 China
| |
Collapse
|
79
|
Chang Q, Zhao S, Sun J, Guo W, Yang L, Qiu L, Zhang N, Fan Y, Liu J. Identification of a novel prognostic and therapeutic prediction model in clear cell renal carcinoma based on Renin-angiotensin system related genes. Front Endocrinol (Lausanne) 2025; 16:1521940. [PMID: 40099255 PMCID: PMC11911175 DOI: 10.3389/fendo.2025.1521940] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Background Clear cell renal cell carcinoma is the most predominant type of renal malignancies, characterized by high aggressiveness and probability of distant metastasis. Renin angiotensin system (RAS) plays a crucial role in maintaining fluid balance within the human body, and its involvement in tumorigenesis is increasingly being uncovered, while its role in ccRCC remains unclear. Methods WGCNA was used to identify RAS related genes. Machine learning was applied to screen hub genes for constructing risk model, E-MTAB-1980 dataset was used for external validation. Transwell and CCK8 assays were used to investigate the impact of SLC6A19 to ccRCC cells. Results SLC6A19, SLC16A12 and SMIM24 were eventually screened to construct risk model and the predictive efficiency for prognosis was validated by internal and external cohorts. Moreover, the differences were found in pathway enrichment, immune cell infiltration, mutational landscapes and drug prediction between high and low risk groups. Experimental results indicated that SLC6A19 could inhibit invasion and proliferation of ccRCC cells and GSEA pinpointed that SLC6A19 was intimately correlated with fatty acid metabolism and CPT1A. Conclusion The risk model based on the three RAS-related genes have a robust ability to predict the prognosis and drug sensitivity of ccRCC patients, further providing a valid instruction for clinical care.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yidong Fan
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jikai Liu
- Department of Urology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
80
|
Okubo N, Kabuto T, Kobayashi H, Kimura J, Imamura Y, Seki M, Inamura S, Taga M, Fukushima M, Terada N. Large-cell neuroendocrine carcinoma of the kidney effectively treated by nivolumab and ipilimumab. IJU Case Rep 2025; 8:89-92. [PMID: 40034907 PMCID: PMC11872206 DOI: 10.1002/iju5.12798] [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: 04/23/2024] [Accepted: 10/01/2024] [Indexed: 03/05/2025] Open
Abstract
Introduction Neuroendocrine tumors originating in the kidney are rare, and standard treatments are not established. Case presentation An 80-years-old man was referred to our hospital with renal dysfunction and a left renal mass. Based on CT and bone scintigraphy results, he was diagnosed as having a large left renal tumor with a thrombus in the inferior vena cava, harboring lymph node, liver, lung, and left iliac bone metastasis. The renal biopsy indicated a large-cell neuroendocrine carcinoma. Treatment with nivolumab + ipilimumab was introduced. The local and metastatic tumors had shrunk. Subsequently, treatment with nivolumab has remained effective for >2 years. Conclusion This case demonstrates the efficacy of treatment with the immune-checkpoint inhibitors against large-cell neuroendocrine carcinoma of the kidney.
Collapse
Affiliation(s)
- Nodoka Okubo
- Department of UrologyUniversity of Fukui Faculty of Medical SciencesYoshida‐gunFukuiJapan
| | - Takashi Kabuto
- Department of UrologyUniversity of Fukui Faculty of Medical SciencesYoshida‐gunFukuiJapan
| | - Hisato Kobayashi
- Department of UrologyUniversity of Fukui Faculty of Medical SciencesYoshida‐gunFukuiJapan
| | - Junya Kimura
- Division of Diagnostic Pathology/Surgical PathologyUniversity of FukuiYoshida‐gunFukuiJapan
| | - Yoshiaki Imamura
- Division of Diagnostic Pathology/Surgical PathologyUniversity of FukuiYoshida‐gunFukuiJapan
| | - Masaya Seki
- Department of UrologyUniversity of Fukui Faculty of Medical SciencesYoshida‐gunFukuiJapan
| | - So Inamura
- Department of UrologyUniversity of Fukui Faculty of Medical SciencesYoshida‐gunFukuiJapan
| | - Minekatsu Taga
- Department of UrologyUniversity of Fukui Faculty of Medical SciencesYoshida‐gunFukuiJapan
| | - Masato Fukushima
- Department of UrologyUniversity of Fukui Faculty of Medical SciencesYoshida‐gunFukuiJapan
| | - Naoki Terada
- Department of UrologyUniversity of Fukui Faculty of Medical SciencesYoshida‐gunFukuiJapan
| |
Collapse
|
81
|
Calhoun SR, Vass C, Myers K, Imai K, Bussberg C, Bhattacharya R, Pinto CA, Poulos C. Patient preferences for adjuvant therapy in renal cell carcinoma: a discrete-choice experiment. Future Oncol 2025; 21:843-851. [PMID: 39935407 PMCID: PMC11921160 DOI: 10.1080/14796694.2025.2463276] [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: 03/30/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
INTRODUCTION To quantify patients' preferences for adjuvant renal cell carcinoma (RCC) treatments. PATIENTS AND METHODS Preferences were elicited using a discrete-choice experiment requiring RCC patients to choose between 2 hypothetical treatments. Data were analyzed using random-parameters logit and latent-class models. RESULTS Patients (n = 250) preferred treatments that increase disease-free and overall survival (OS), are taken less frequently, require no concomitant medication, have a shorter duration, and have lower side-effect risks. The analyses also highlighted their willingness to make tradeoffs between these benefits and risks. Patients were generally tolerant of increases in the risks of treatment-related severe diarrhea, dizziness, and fatigue and were willing to accept increases in these risks in exchange for improvements in overall or disease-free survival. Latent-class analysis identified 3 classes: class 1 (37.5%) and class 2 (26.9%) preferred not to opt out of treatment and prioritized increased OS and disease-free survival, respectively; class 3 (35.5%) preferred to opt out and prioritized mode, duration, and risks. CONCLUSIONS Heterogeneity suggests patient-physician discussions are important when considering RCC treatments.
Collapse
Affiliation(s)
| | | | - Kelley Myers
- RTI Health Solutions, Research Triangle Park, NC, USA
| | | | | | | | | | | |
Collapse
|
82
|
Vass C, Pinto CA, Myers K, Imai K, Bussberg C, Bhattacharya R, Calhoun SR, Poulos C. Oncologists' and urologists' preferences for adjuvant therapy in renal cell carcinoma: a discrete-choice experiment. Future Oncol 2025; 21:833-842. [PMID: 39945436 PMCID: PMC11916416 DOI: 10.1080/14796694.2025.2464485] [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: 03/30/2024] [Accepted: 02/05/2025] [Indexed: 03/20/2025] Open
Abstract
INTRODUCTION To quantify physicians' preferences for adjuvant renal cell carcinoma (RCC) treatments. MATERIALS AND METHODS A discrete-choice experiment was administered online to board-certified/eligible physicians. Physicians chose between pairs of hypothetical adjuvant therapies for a high-risk patient who had recently undergone a radical nephrectomy. Data were analyzed using random-parameters logit and latent-class models. RESULTS Physicians (n = 250; 64% oncologists; 36% urologists) placed most importance on improvements in the chance of 5-year overall survival, followed by increased median disease-free survival and reduced risk of side effects. The analyses also highlighted their willingness to make tradeoffs between these benefits and risks. Physicians were generally tolerant of increases in the risks of treatment-related severe diarrhea, dizziness, and fatigue and were willing to accept increases in these risks in exchange for improvements in overall or disease-free survival. Subgroup analysis revealed heterogeneity between oncologists and urologists, and latent-class analysis revealed significant heterogeneity among the whole physician sample. CONCLUSIONS Most physicians in this study would recommend adjuvant therapy to a typical high-risk postnephrectomy RCC patient.
Collapse
Affiliation(s)
- Caroline Vass
- Health Preference Assessment, RTI Health Solutions, Manchester, UK
| | | | - Kelley Myers
- Health Preference Assessment, RTI Health Solutions, Research Triangle Park, NC, USA
| | | | - Cooper Bussberg
- Health Preference Assessment, RTI Health Solutions, Research Triangle Park, NC, USA
| | | | | | - Christine Poulos
- Health Preference Assessment, RTI Health Solutions, Research Triangle Park, NC, USA
| |
Collapse
|
83
|
Zhong B, Du J, Liu F, Sun S. The Role of Yes-Associated Protein in Inflammatory Diseases and Cancer. MedComm (Beijing) 2025; 6:e70128. [PMID: 40066231 PMCID: PMC11892025 DOI: 10.1002/mco2.70128] [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: 04/17/2024] [Revised: 02/02/2025] [Accepted: 02/11/2025] [Indexed: 03/17/2025] Open
Abstract
Yes-associated protein (YAP) plays a central role in the Hippo pathway, primarily governing cell proliferation, differentiation, and apoptosis. Its significance extends to tumorigenesis and inflammatory conditions, impacting disease initiation and progression. Given the increasing relevance of YAP in inflammatory disorders and cancer, this study aims to elucidate its pathological regulatory functions in these contexts. Specifically, we aim to investigate the involvement and molecular mechanisms of YAP in various inflammatory diseases and cancers. We particularly focus on how YAP activation, whether through Hippo-dependent or independent pathways, triggers the release of inflammation and inflammatory mediators in respiratory, cardiovascular, and digestive inflammatory conditions. In cancer, YAP not only promotes tumor cell proliferation and differentiation but also modulates the tumor immune microenvironment, thereby fostering tumor metastasis and progression. Additionally, we provide an overview of current YAP-targeted therapies. By emphasizing YAP's role in inflammatory diseases and cancer, this study aims to enhance our understanding of the protein's pivotal involvement in disease processes, elucidate the intricate pathological mechanisms of related diseases, and contribute to future drug development strategies targeting YAP.
Collapse
Affiliation(s)
- Bing Zhong
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Jintao Du
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Feng Liu
- Department of Otolaryngology‐Head and Neck SurgeryWest China HospitalSichuan UniversityChengduSichuanChina
| | - Silu Sun
- State Key Laboratory of Oral DiseasesNational Clinical Research Center for Oral DiseasesChinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and ManagementWest China Hospital of StomatologySichuan UniversityChengduSichuanChina
| |
Collapse
|
84
|
da Ponte TF, Fontenelle LMAR, Rodrigues CEM, Souza JC, Rodrigues EDM. Association Between Lupus Nephritis and Renal Clear-Cell Carcinoma: A Case Report and Review of the Literature. Cureus 2025; 17:e80459. [PMID: 40225477 PMCID: PMC11990667 DOI: 10.7759/cureus.80459] [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: 03/11/2025] [Indexed: 04/15/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease affecting several organs, including the kidneys, potentially leading to lupus nephritis (LN). SLE has also been associated with several neoplasias, but its relation to renal cell carcinoma (RCC) has been little explored. We report a young women diagnosed concomitantly with LN and RCC. The latter was discovered incidentally during an investigation of nephrotic syndrome and confirmed on histology and renal microscopy. The patient was submitted to partial nephrectomy and immunosuppression, with good outcome, as shown by the improvement in proteinuria and other symptoms. Our case highlights the complexity of diagnosing SLE in combination with RCC and the importance of permanent surveillance and multidisciplinary approach to optimize treatment.
Collapse
Affiliation(s)
| | | | - Carlos Ewerton Maia Rodrigues
- Medical Sciences, Medical School, University of Fortaleza, Fortaleza, BRA
- Rheumatology, Federal University of Ceará, Fortaleza, BRA
| | | | | |
Collapse
|
85
|
Li Z, Xin S, Huang L, Tian Y, Chen W, Liu X, Ye B, Bai R, Yang G, Wang W, Ye L. BEX4 inhibits the progression of clear cell renal cell carcinoma by stabilizing SH2D4A, which is achieved by blocking SIRT2 activity. Oncogene 2025; 44:665-678. [PMID: 39639172 DOI: 10.1038/s41388-024-03235-6] [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: 04/26/2024] [Revised: 11/10/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
Clear cell renal cell carcinoma (ccRCC) is one of the most common malignancies. Recently, the role of brain-expressed X-linked 4 (BEX4) in cancer progression has received increasing attention. This study aimed to investigate the function of BEX4 in ccRCC and to reveal the underlying mechanisms. We first confirmed that BEX4 was significantly downregulated in ccRCC by bioinformatics analysis and that patients with low BEX4 expression tended to have prolonged overall survival time. Subsequently, we confirmed that BEX4 inhibited ccRCC cell proliferation in vitro and tumorigenesis in vivo through a series of cell function assays and the establishment of a nude mouse xenograft model, respectively. Mechanistically, we found that BEX4 positively regulates the expression of Src homology 2 domain-containing 4A (SH2D4A), an inhibitor of the NOTCH pathway, which further promoted the tumor-suppressive effects of BEX4. In addition, our study confirmed that the promoting effect of BEX4 on SH2D4A was achieved by inhibiting the deacetylase sirtuin 2 (SIRT2) activity. On this basis, we found that there was a competition between acetylation and ubiquitination modifications at the K69 site of SH2DA4 and that BEX4-induced upregulation of acetylation at the k69 site stabilizes SH2D4A protein expression by inhibiting ubiquitination at the same site. In addition, dual-luciferase assays showed that the transcriptional activity of BEX4 was positively regulated by activation transcription factor 3 (ATF3). Our study suggests that BEX4 plays a role in inhibiting tumor progression in ccRCC and maybe a new diagnostic and therapeutic target for ccRCC patients.
Collapse
Affiliation(s)
- Ziyao Li
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
- School of Electrical Engineering of Zhengzhou University, Zhengzhou, China
- Center for Frontier Medical Engineering of Chiba University, Chiba, Japan
| | - Shiyong Xin
- Department of Urology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Liqun Huang
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ye Tian
- Department of Urology, Guizhou Provincial People's Hospital, Guiyang, China
| | - Weihua Chen
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiang Liu
- Department of Urology, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bowen Ye
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rong Bai
- Department of Pharmacy, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Guosheng Yang
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Wenwen Wang
- Department of Biotherapy, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Lin Ye
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
86
|
Huang RS, Chow R, Benour A, Chen D, Boldt G, Wallis CJD, Swaminath A, Simone CB, Lock M, Raman S. Comparative efficacy and safety of ablative therapies in the management of primary localised renal cell carcinoma: a systematic review and meta-analysis. Lancet Oncol 2025; 26:387-398. [PMID: 39922208 DOI: 10.1016/s1470-2045(24)00731-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 02/10/2025]
Abstract
BACKGROUND Non-invasive and minimally invasive ablative treatments, including stereotactic body radiotherapy (SBRT), radiofrequency ablation, microwave ablation, and cryoablation, have emerged as key treatment options for managing renal cell carcinoma, especially for patients who are unsuitable for surgery. We aimed to compare the clinical efficacy and safety of these emerging treatment methods in patients with localised renal cell carcinoma. METHODS In this systematic review and meta-analysis, we searched PubMed (MEDLINE), Embase, and the Cochrane Library for publications between Jan 1, 2000, and March 1, 2024. Eligible articles were observational studies and randomised controlled trials including at least five adult patients (age ≥18 years) with primary and localised renal cell carcinoma treated with SBRT, radiofrequency ablation, microwave ablation, or cryoablation and that reported on local control outcomes. Two reviewers independently screened titles and abstracts and then full texts of eligible studies were independently evaluated by the same reviewers, with disagreements resolved via discussion or consultation with a third reviewer. Summary estimates were extracted from published reports manually using a standardised data extraction form. The primary endpoint was local control rate at 1 year, 2 years, and 5 years after start of treatment. A meta-analysis was conducted using a DerSimonian and Laird model to summarise local control rates. Publication bias was evaluated using funnel plots and Egger's test. We also recorded the frequency and severity of adverse events after treatment on the basis of the Common Terminology Criteria for Adverse Events (version 5.0) and Clavien-Dindo complication index. The study protocol was prospectively registered with PROSPERO, CRD42024511840. FINDINGS We identified 6668 records, of which 330 were assessed via full-text review, and 133 were included in our systematic review and meta-analysis. The eligible studies included data for 8910 patients (mean age 67·9 years [SD 7·3], 2518 [31·4%] of 8018 patients with available data were female and 5500 [68·6%] were male). Local control rates for SBRT were 99% (95% CI 97-100; I2=6%) at 1 year, 97% (95-99; I2=0%) at 2 years, and 95% (89-98; I2=42%) at 5 years; for radiofrequency ablation were 96% (94-98; I2=73%) at 1 year, 95% (92-98; I2=77%) at 2 years, and 92% (88-96; I2=78%) at 5 years; for microwave ablation were 97% (95-99; I2=74%) at 1 year, 95% (92-98; I2=77%) at 2 years, and 86% (75-94; I2=66%) at 5 years; and for cryoablation were 95% (93-96; I2=61%) at 1 year, 94% (91-96; I2=69%) at 2 years, and 90% (87-93; I2=74%) at 5 years. The proportion of patients who reported grade 3-4 adverse events was 3% (121 of 3726) after cryoablation, 2% (39 of 2503) after radiofrequency ablation, 1% (22 of 2069) after microwave ablation, and 2% (11 of 612) after SBRT. Risk of bias was moderate in most studies (70 [53%] of 133) and no publication bias was observed. INTERPRETATION All investigated ablative methods continue to represent effective treatment choices in renal cell carcinoma, and these findings support multi-disciplinary discussions of these treatment methods, along with surgery and surveillance, to individualise treatment decisions in these patients. Future research should aim to conduct randomised controlled trials across larger patient populations to further elucidate the long-term oncological and survival outcomes associated with these treatments. FUNDING None.
Collapse
Affiliation(s)
- Ryan S Huang
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ronald Chow
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Ali Benour
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - David Chen
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Gabriel Boldt
- London Regional Cancer Program, London Health Sciences Centre, Schulich School of Medicine, University of Western Ontario, London, ON, Canada
| | - Christopher J D Wallis
- Division of Urology and Surgical Oncology, Department of Surgery, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Anand Swaminath
- Juravinski Cancer Centre, McMaster University, Hamilton, ON, Canada
| | - Charles B Simone
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Lock
- London Regional Cancer Program, London Health Sciences Centre, Schulich School of Medicine, University of Western Ontario, London, ON, Canada
| | - Srinivas Raman
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
87
|
Deng MH, Yang XW, Zhou YM, Xie LZ, Zou T, Ping JG. In silico research of coagulation- and fibrinolysis-related genes for predicting prognosis of clear cell renal cell carcinoma. Transl Androl Urol 2025; 14:307-324. [PMID: 40114841 PMCID: PMC11921444 DOI: 10.21037/tau-24-483] [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: 09/09/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Background Coagulation- and fibrinolysis-related genes (CFRGs) are involved in tumor progression. However, their regulatory mechanisms in clear cell renal cell carcinoma (ccRCC) remain unclear. The aim of this study was to search for genes related to coagulation and fibrinolytic systems in ccRCC and to investigate their potential role in tumor pathogenesis and progression. Methods Differentially expressed genes (DEGs) between ccRCC and control samples, as well as key module genes associated with ccRCC, were extracted from The Cancer Genome Atlas-Kidney Renal Clear Cell Carcinoma (TCGA-KIRC) dataset. Differentially expressed CFRGs (DE-CFRGs) were identified by intersecting these DEGs with CFRGs. Prognostic genes were identified through univariate Cox, least absolute shrinkage and selection operator (LASSO), and multivariate Cox analyses of DE-CFRGs. Additional independent prognostic and enrichment analyses were conducted, and potential therapeutic drugs were predicted. In addition, quantitative real-time polymerase chain reaction (RT-qPCR) was performed to validate the expression of prognostic genes. Results Sixteen DE-CFRGs were identified by intersecting 3,311 DEGs, 1,719 key module genes, and CFRGs. Four prognostic genes-TIMP1, RUNX1, BMP6, and PROS1-were found to be involved in complement and coagulation cascades and other functional pathways. The prognostic model demonstrated strong predictive power for ccRCC, with stage, risk score, and grade all correlating with prognosis. Additionally, 14 potential drugs, such as tamoxifen citrate and cytarabine, were predicted for therapeutic targeting of the identified prognostic genes. RT-qPCR confirmed that the expression levels of TIMP1, and RUNX1 were significantly upregulated in ccRCC samples, consistent with bioinformatics analysis. Conclusions A prognostic model incorporating TIMP1, RUNX1, BMP6, and PROS1 was constructed, offering new insights for prognostic evaluation and therapeutic strategies in ccRCC.
Collapse
Affiliation(s)
- Ming-Hao Deng
- Department of Urology, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Xue-Wen Yang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Nantong, China
| | - Yu-Ming Zhou
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Lv-Zhong Xie
- Department of Urology, Nantong Hospital of Traditional Chinese Medicine, Nantong, China
| | - Tao Zou
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ji-Gen Ping
- Department of Urology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
88
|
Zhang Y, Cui K, Qiang R, Wang L. FUT10 is related to the poor prognosis and immune infiltration in clear cell renal cell carcinoma. Transl Cancer Res 2025; 14:827-842. [PMID: 40104704 PMCID: PMC11912032 DOI: 10.21037/tcr-24-449] [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: 03/19/2024] [Accepted: 11/21/2024] [Indexed: 03/20/2025]
Abstract
Background Clear cell renal cell carcinoma (ccRCC), is highly metastatic with unfavorable oncologic outcomes. The metastatic dissemination and underlying mechanisms of ccRCC remain insufficiently understood. The expression of fucosyltransferases (FUTs) has been explored in multiple cancer types, which affect survival of tumor cells and oncology progress. However, the role of fucosyltransferase 10 (FUT10), a member of the FUT family, is still unclear in ccRCC. We aimed to investigate the effects of FUT10 on the prognosis and immune infiltration of ccRCC via The Cancer Genome Atlas (TCGA) database. Methods The relationship between FUT10 expression and clinical-pathologic features was evaluated by Welch's t-test, Wilcoxon signed-rank test, Dunn's test, and logistic regression based on TCGA datasets. The FUT10 expression level was converted into a categorical variable by receiver operating characteristic (ROC) and the area under the curve (AUC). The factors associated with the prognosis were determined by Kaplan-Meier method. The function of FUT10 was identified by functional enrichment analysis, gene set enrichment analysis (GSEA), gene correlation analysis, and immune infiltration analysis. At last, we verified the FUT10 messenger RNA (mRNA) expression in ccRCC and adjacent kidney tissues by quantitative real-time polymerase chain reaction (qRT-PCR). Results Downregulated FUT10 expression in ccRCC was associated with the clinical stage (P<0.001), T stage (P<0.001), M stage (P<0.001), and overall survival (OS) event (P<0.001). The ROC curve suggested that FUT10 had a certain accuracy in the diagnostic ability in ccRCC (AUC =0.787). It was shown that patient survival was prolonged in the FUT10 high-expression group. Meanwhile, multivariate analysis displayed that FUT10 was an independent risk factor for ccRCC patients (P=0.003). Moreover, we uncovered that FUT10 was involved in the phenotype of the immune response, oxidative phosphorylation (OXPHOS), arachidonic acid (AA) metabolism, and primary immunodeficiency (PID) by function enrichment analysis and GSEA. In addition, in the high FUT10 expression group, natural killer (NK) CD56bright cells exhibited lower enrichment scores, and central memory T cells exhibited higher enrichment scores. Especially, ARL8B, a key factor in NK-mediated cytotoxicity, had a certain correlation with FUT10 (r=0.590, P<0.001). Compared to the normal kidney tissues, the FUT10 mRNA expression in the ccRCC was decreased (P=0.004). Conclusions FUT10 might be a promising immune therapy target and prognostic biomarker in ccRCC.
Collapse
Affiliation(s)
- Yuqi Zhang
- Center of Medical Genetics, Northwest Women's and Children's Hospital, Xi'an, China
| | - Ke Cui
- Center of Medical Genetics, Northwest Women's and Children's Hospital, Xi'an, China
| | - Rong Qiang
- Center of Medical Genetics, Northwest Women's and Children's Hospital, Xi'an, China
| | - Lin Wang
- Center of Medical Genetics, Northwest Women's and Children's Hospital, Xi'an, China
| |
Collapse
|
89
|
Li C, Hu P, Fan C, Mi H. The prognostic and immune significance of SNHG3 in clear cell renal cell carcinoma. Transl Cancer Res 2025; 14:1008-1023. [PMID: 40104694 PMCID: PMC11912088 DOI: 10.21037/tcr-24-1509] [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: 08/25/2024] [Accepted: 12/17/2024] [Indexed: 03/20/2025]
Abstract
Background Long non-coding RNA (lncRNA) small nucleolar RNA host gene 3 (SNHG3) has been reported to be involved in the pathological process of a variety of tumors, including clear cell renal cell carcinoma (ccRCC). However, whether SNHG3 can be used as a prognostic biomarker and its correlation with immune infiltration in ccRCC remain unclear, warranting further research. This study aims to explore the relationship between SNHG3 and immune infiltration in ccRCC and confirm the potential of SNHG3 to predict survival of ccRCC patients. Methods The Cancer Genome Atlas (TCGA) database was used to assess the expression of SNHG3 in ccRCC, evaluate clinicopathological characteristics, assess prognosis, and conduct functional enrichment analysis. The ccRCC microenvironment and immune infiltration were investigated using the Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data (ESTIMATE) and Cell-type Identification By Estimating Relative Subsets Of RNA Transcripts (CIBERSORT) algorithms, respectively. We additionally investigated the relationships between SNHG3 and immunological checkpoints. Drug sensitivity of SNHG3 was investigated in R. The expression of SNHG3 was verified in the Gene Expression Omnibus (GEO) database, ccRCC cell lines, and tissues. Wound healing and Methylthiazolyldiphenyl-tetrazolium bromide (MTT) assays were used to evaluate tumor cell migration and proliferation. Fluorescence in situ hybridization (FISH) assay was conducted to localize SNHG3 in ccRCC cells. Results SNHG3 expression was significantly upregulated in ccRCC cells and tissues and associated with several clinicopathological features and poor prognosis of ccRCC patients. SNHG3 was correlated with immune cells infiltration in ccRCC and exhibited sensitivity to various targeted and chemotherapy drugs. Knockdown of SNHG3 significantly reduced the proliferation and migration of ccRCC. FISH results showed that SNHG3 was located in the cell nucleus. Conclusions Overall, this study demonstrates that SNHG3 is a prognostic biomarker correlated with immune infiltration in ccRCC.
Collapse
Affiliation(s)
- Cheng Li
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Pengnan Hu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chenglong Fan
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hua Mi
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
90
|
Tian T, Shen C, Zapała Ł, Fang X, Zheng B. Identification of a C2H2 zinc finger-related lncRNA prognostic signature and its association with the immune microenvironment in clear cell renal cell carcinoma. Transl Androl Urol 2025; 14:412-431. [PMID: 40114819 PMCID: PMC11921207 DOI: 10.21037/tau-2024-769] [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: 12/26/2024] [Accepted: 01/22/2025] [Indexed: 03/22/2025] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is the main component of renal cell carcinoma, and advanced ccRCC often predicts a poor prognosis. In recent years, research has revealed the critical role of Cys2His2 zinc finger genes (CHZFs) and long non-coding RNAs (lncRNAs) in the development of cancer. Currently, little is known about the prognostic value of the lncRNAs linked to Cys2His2 (C2H2) zinc finger proteins (ZFPs) in ccRCC. The aim of this study was to construct a prognostic model for C2H2-associated lncRNAs to assist in the selection of clinical therapy. Methods RNA-sequencing data, and related clinical and prognostic information were downloaded from The Cancer Genome Atlas (TCGA) database. Univariate and multivariate Cox regression analyses were conducted to identify Cys2His2 zinc finger-associated long non-coding RNAs (CHZFLs) and build prediction signatures. A receiver operating characteristic (ROC) curve analysis was performed to validate the risk model. The prognosis of the groups was analyzed using the Kaplan-Meier method. The independent prognostic significance of these signatures was evaluated by univariate and multivariate Cox regression analyses. The relationship between the CHZFL signature and ccRCC tumor immunity was confirmed by a differential analysis of immune function and immunological checkpoints. Results A signature composed of five lncRNAs (AL117336.2, AC026401.3, AC124854.1, DBH-AS1, and LINC02100) was constructed. The results revealed a strong correlation between the CHZFLs signature and the prognosis of ccRCC patients. Prognostic characteristics of CHZFLs are independent prognostic factors in ccRCC patients. The diagnostic efficacy of the predictive signature was higher than that of individual clinicopathologic variables, and it had a ROC area under the curve (AUC) of 0.775. The results of the clinical subgroup analysis showed that the high-risk group had shorter overall survival (OS) than the low-risk group. Common chemotherapy medications, including vinorelbine, cytarabine, epirubicin, and gemcitabine, caused increased sensitivity in the high-risk group. Additionally, the single-sample gene set enrichment analysis (ssGSEA) revealed that the immunological state of the ccRCC patients was substantially linked with the predictive parameters. Conclusions The five CHZFL signature can help predict the prognosis of ccRCC patients, and assist in selecting immunotherapy and chemotherapy regimens in clinical practice.
Collapse
Affiliation(s)
- Ting Tian
- Operating Room Nursing, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Cheng Shen
- Department of Urology, The Second Affiliated Hospital of Nantong University, Nantong, China
- Jiangsu Nantong Urological Clinical Medical Center, Nantong, China
| | - Łukasz Zapała
- Clinic of General, Oncological and Functional Urology, Medical University of Warsaw, Warsaw, Poland
| | - Xingxing Fang
- Department of Nephrology, Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Bing Zheng
- Department of Urology, The Second Affiliated Hospital of Nantong University, Nantong, China
- Jiangsu Nantong Urological Clinical Medical Center, Nantong, China
| |
Collapse
|
91
|
Page PM, Dastous SA, Richard PO, Pavic M, Nishimura T, Riazalhosseini Y, Crapoulet N, Martin M, Turcotte S. MicroRNA profiling identifies VHL/HIF-2α dependent miR-2355-5p as a key modulator of clear cell Renal cell carcinoma tumor growth. Cancer Cell Int 2025; 25:71. [PMID: 40016765 PMCID: PMC11869434 DOI: 10.1186/s12935-025-03711-3] [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: 02/13/2024] [Accepted: 02/20/2025] [Indexed: 03/01/2025] Open
Abstract
Inactivation of the von Hippel-Lindau (VHL) tumor suppressor gene is one of the first truncal events in clear cell Renal Cell Carcinoma (ccRCC) tumorigenesis. The accumulation of Hypoxia Induced Factor (HIFα) resulting from VHL loss can promote ccRCC tumorigenesis by regulating microRNA (miRNA) expression. Here, we performed miRNA profiling and high-throughput analysis to identify a panel of VHL-dependent miRNAs in ccRCC. Validation of these miRNAs revealed the overexpression of miR-2355-5p in ccRCC cell models and primary tumors. Moreover, we showed a significant increase in circulating miR-2355-5p in plasma from patients with ccRCC. Mechanistically, miR-2355-5p overexpression was confirmed to be HIF-2α dependent. Targeting miR-2355-5p with the CRISPR/Cas9 system not only negatively disrupted the ability of ccRCC cells to stimulate angiogenesis but also decreased cell proliferation and drastically reduced tumor growth in mouse xenograft models. Finally, a miR-2355-5p pulldown assay identified five tumor suppressor genes, ACO1, BTG2, CMTM4, SLIT2, and WDFY2, as potential targets. All five genes were significantly downregulated in ccRCC tumors and mouse xenograft tumors. The results from this research demonstrate the oncogenic ability of miR-2355-5p and shed light on the possible mechanism by which this miRNA controls angiogenesis and tumor growth in VHL-deficient ccRCC.
Collapse
Affiliation(s)
- Patric M Page
- Department of Chemistry and Biochemistry, Université de Moncton, 18 Antonine-Maillet, Moncton, NB, E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB, Canada
| | - Sonia A Dastous
- Department of Chemistry and Biochemistry, Université de Moncton, 18 Antonine-Maillet, Moncton, NB, E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB, Canada
| | - Patrick O Richard
- Department of Urology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Canada
- Institut de recherche sur le cancer de l'Université de Sherbrooke, Sherbrooke, Canada
| | - Michel Pavic
- Institut de recherche sur le cancer de l'Université de Sherbrooke, Sherbrooke, Canada
- Department of Hemato-Oncology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Canada
| | - Tamiko Nishimura
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- McGill Genome Centre, McGill University, Montréal, Québec, Canada
| | - Yasser Riazalhosseini
- Department of Human Genetics, McGill University, Montréal, Québec, Canada
- McGill Genome Centre, McGill University, Montréal, Québec, Canada
| | - Nicolas Crapoulet
- Laboratoire de Génétique Moléculaire, Vitalité Health Network, Moncton, Canada
| | - Mykella Martin
- Centre de formation médicale du Nouveau-Brunswick, Moncton, NB, Canada
| | - Sandra Turcotte
- Department of Chemistry and Biochemistry, Université de Moncton, 18 Antonine-Maillet, Moncton, NB, E1A 3E9, Canada.
- Atlantic Cancer Research Institute, Moncton, NB, Canada.
| |
Collapse
|
92
|
Li W, Shi J, Lv Q, Miao D, Tan D, Lu X, Xiong H, Luo Q, Xia Y, Han Y, Dong X, Huang G, Zhang X, Yang H. Identification of the LCOR-PLCL1 pathway that restrains lipid accumulation and tumor progression in clear cell renal cell carcinoma. Int J Biol Sci 2025; 21:2296-2312. [PMID: 40083699 PMCID: PMC11900815 DOI: 10.7150/ijbs.107981] [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: 12/01/2024] [Accepted: 02/11/2025] [Indexed: 03/16/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the typical pathological subtype of renal cell carcinoma (RCC), representing about 80% of RCC. Reprogramming of lipid metabolism is one of the nonnegligible pathogeneses in ccRCC. Currently, the underlying regulatory mechanisms of lipid metabolism in ccRCC remain inadequately understood. In this study, we performed bioinformatics analyses and experiments both in vivo and in vitro to explore the biological functions and specific mechanisms of the ligand dependent nuclear receptor corepressor LCOR in ccRCC. Mechanistically, RUNX1 was a transcriptional suppressor of PLCL1, LCOR could interact with RUNX1 to relieve RUNX1-mediated repression of PLCL1, leading to increased PLCL1 expression, which, in turn, inhibited the tumor progression and lipid accumulation in ccRCC. Furthermore, PLCL1 decreased lipid accumulation through UCP1-mediated lipid browning and facilitated tumor apoptosis by activating p38 phosphorylation. In conclusion, the LCOR-RUNX1-PLCL1 axis provides a novel molecular mechanism underlying the progression and lipid storage of ccRCC. LCOR modulation represents a potential therapeutic strategy for the treatment in ccRCC.
Collapse
Affiliation(s)
- Wen Li
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Shi
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingyang Lv
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Daojia Miao
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Diaoyi Tan
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojun Lu
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hairong Xiong
- College of Life Science and Technology, Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, Hubei Engineering University, Xiaogan, China
| | - Qianqian Luo
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaru Xia
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuqi Han
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuejiao Dong
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guixiao Huang
- Department of Urology, The Third Affiliated Hospital of Shenzhen University: Shenzhen Luohu Hospital Group Luohu People's Hospital, Shenzhen 518000, China
| | - Xiaoping Zhang
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Urology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
93
|
Guérin C, Vinchent A, Fernandes M, Damour I, Laratte A, Tellier R, Estevam GO, Meneboo JP, Villenet C, Descarpentries C, Fraser JS, Figeac M, Cortot AB, Rouleau E, Tulasne D. MET variants with activating N-lobe mutations identified in hereditary papillary renal cell carcinomas still require ligand stimulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.03.565283. [PMID: 37965202 PMCID: PMC10635098 DOI: 10.1101/2023.11.03.565283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
In hereditary papillary renal cell carcinoma (HPRCC), the hepatocyte growth factor receptor (MET) receptor tyrosine kinase (RTK) mutations recorded to date are located in the kinase domain and lead to constitutive MET activation. This contrasts with MET mutations identified in non-small cell lung cancer (NSCLC), which lead to exon 14 skipping and deletion of a regulatory domain: in this latter case, the mutated receptor still requires ligand stimulation. Sequencing of MET in samples from 158 HPRCC and 2808 NSCLC patients revealed ten uncharacterized mutations. Four of these, all found in HPRCC and leading to amino acid substitutions in the N-lobe of the MET kinase, proved able to induce cell transformation, which was further enhanced by hepatocyte growth factor (HGF) stimulation: His1086Leu, Ile1102Thr, Leu1130Ser and Cis1125Gly. Similar to the variant resulting in MET exon 14 skipping, the two N-lobe MET variants His1086Leu and Ile1102Thr were found to require stimulation by HGF in order to strongly activate downstream signaling pathways and epithelial cell motility. The Ile1102Thr mutation also displayed transforming potential, promoting tumor growth in a xenograft model. In addition, the N-lobe-mutated MET variants were found to trigger a common HGF-stimulation-dependent transcriptional program, consistent with an observed increase in cell motility and invasion. Altogether, this functional characterization revealed that N-lobe variants still require ligand stimulation, in contrast to other RTK variants. This suggests that HGF expression in the tumor microenvironment is important for tumor growth. The sensitivity of these variants to MET inhibitors opens the way for use of targeted therapies for patients harboring the corresponding mutations.
Collapse
|
94
|
Guo K, Ma P, Yang Q, Xu L, Zhang B, Zhang H, Zheng Z, Zhuo Z. Activation of RHO-GTPase gene pattern correlates with adverse clinical outcome and immune microenvironment in clear cell renal cell carcinoma. Clin Exp Med 2025; 25:67. [PMID: 39998699 PMCID: PMC11861022 DOI: 10.1007/s10238-025-01593-3] [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: 01/11/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC), the most prevalent renal cancer subtype, is frequently associated with poor prognosis. RHO-GTPase signaling genes have been implicated in tumor aggressiveness and unfavorable survival, but their potential in risk stratification and therapeutic guidance for ccRCC patients remains unexplored. Univariate regression identified prognostically relevant RHO-GTPase signaling genes, followed by consensus clustering for ccRCC subtype classification. LASSO regression selected key genes to construct a six-gene risk model. The model was evaluated for prognostic stratification, immune status, immunotherapy response, and chemotherapy sensitivity. Key genes were analyzed at the genomic, single-cell, and protein levels to explore underlying mechanisms. Among 62 prognostically relevant RHO-GTPase signaling genes, six (ARHGAP11B, NUF2, PLK1, CYFIP2, IQGAP2, and VAV3) were identified to form a robust prognostic signature. This model stratified patients into high- and low-risk groups, with high-risk patients demonstrating significantly worse outcomes. The model exhibited excellent predictive accuracy (AUC > 0.7 in training and validation cohorts). High-risk patients were characterized by an immunosuppressive microenvironment and reduced sensitivity to immunotherapy. Drug sensitivity analysis revealed 107 agents correlated with the risk score, underscoring therapeutic relevance. Mechanistically, the six key genes showed distinct expression patterns, cellular distribution, and positive correlation with VHL mutations, highlighting their potential as actionable drug targets. This study established a novel six-gene RHO-GTPase signaling model for predicting prognosis, immune status, and therapeutic responses in ccRCC, which offers potential for improving patient stratification and guiding personalized treatment strategies.
Collapse
Affiliation(s)
- Kehang Guo
- Department of Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Pengyue Ma
- Department of Nephrology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Yang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Lingli Xu
- Dadong Street Community Health Service Center, Guangzhou, 510080, China
| | - Biixiong Zhang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Hong Zhang
- Department of Lymphoma, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China.
| | - Zhongwen Zheng
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- Heyuan People's Hospital, Heyuan, 517001, Guangdong, China.
| | - Zewei Zhuo
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
95
|
Feng K, Li J, Li J, Li Z, Li Y. Prognostic implications of ERLncRNAs in ccRCC: a novel risk score model and its association with tumor mutation burden and immune microenvironment. Discov Oncol 2025; 16:225. [PMID: 39985635 PMCID: PMC11846825 DOI: 10.1007/s12672-025-01870-3] [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: 09/12/2024] [Accepted: 02/03/2025] [Indexed: 02/24/2025] Open
Abstract
INTRODUCTION/BACKGROUND The specific role of efferocytosis-related long noncoding RNAs (ERLncRNAs) in Clear Cell Renal Cell Carcinoma (ccRCC) has not been thoroughly examined. This study aims to identify and validate a signature of ERLncRNAs for prognostic prediction and characterization of the immune landscape in individuals with ccRCC. MATERIALS AND METHODS Analysis of ccRCC samples was conducted by utilizing clinical and RNA sequencing information obtained from The Cancer Genome Atlas (TCGA). Pearson correlation analysis was utilized to identify lncRNAs associated with efferocytosis, which was then used to create a new prognostic model through univariate Cox regression, Least Absolute Shrinkage and Selection Operator (LASSO) regression, and stepwise multivariate Cox analysis. In order to investigate the biological significance, we performed a functional enrichment analysis to assess how well the model predicts outcomes. Differences in the immune landscape were observed through a comparison of immune cell infiltration, tumor mutational burden (TMB), and tumor microenvironment (TME) characteristics. Following this, drug sensitivity analysis was conducted. RESULTS This led to the identification of a unique signature consisting of seven ERLncRNAs (LINC01615, RUNX3-AS1, FOXD2-AS1, AC002070.1, LINC02747, LINC00944, and AC092296.1). Model performance was measured by Kaplan-Meier curves and receiver operating characteristic (ROC) curves. The nomogram and C-index provided additional validation of the strong correlation between the risk signature and clinical decision-making. CONCLUSION On the whole, our innovative signature exhibits potential for prognostic prediction and assessment of immunotherapeutic response in patients with ccRCC.
Collapse
Affiliation(s)
- Kunlun Feng
- Shandong University of Traditional Chinese Medicine, Jinan, 250013, Shandong, China
| | - Jingxiang Li
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jianye Li
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhichao Li
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| | - Yahui Li
- The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| |
Collapse
|
96
|
Wu G, Chen C, Chang J, Fazlollahi F, Makary MS. Expanding the Scope of Interventional Oncology: Locoregional Therapies in Extrahepatic Malignancies. Cancers (Basel) 2025; 17:726. [PMID: 40075574 PMCID: PMC11899649 DOI: 10.3390/cancers17050726] [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: 12/26/2024] [Revised: 02/16/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Locoregional therapies (LRTs), including transarterial embolization (TAE), transarterial chemoembolization (TACE), and transarterial radioembolization (TARE), have become integral in the management of hepatocellular carcinoma (HCC) in recent decades and continue to shape evolving treatment strategies. While their role in liver tumor management is well established, their potential for treating extrahepatic malignancies is gaining increasing attention. Notably, growing research has highlighted the promising applications of TAE, TACE, and TARE in extrahepatic cancers such as glioblastoma (GBM), soft tissue sarcomas (STSs), prostate cancer (PCa), pancreatic cancer, and renal cell carcinoma (RCC). This review aims to explore these novel applications, providing a comprehensive summary of the current literature, examining clinical outcomes, and discussing future directions for integrating these techniques into broader oncologic treatment strategies. METHODS A systematic literature review was conducted focusing on LRTs such as TAE, TACE, and TARE in extrahepatic malignancies. Studies published between May 1998 and December 2024 were included, emphasizing outcomes in GBM, STS, PCa, pancreatic cancer, and RCC. Data extraction prioritized clinical outcomes, safety profiles, and procedural efficacy. RESULTS LRTs demonstrated significant potential in managing extrahepatic malignancies, with TAE, TACE, and TARE showing promising results in palliative management and tumor control. Across studies, these therapies exhibited varying degrees of success in improving progression-free survival and overall survival, with minimal systemic toxicity. CONCLUSIONS The expanding application of LRTs in extrahepatic malignancies highlights their transformative potential in interventional oncology. By offering targeted, minimally invasive treatment options, these modalities bridge critical gaps in managing tumors refractory to conventional therapies. Future research should focus on standardizing protocols, optimizing patient selection, and exploring combination therapies to maximize their clinical efficacy.
Collapse
Affiliation(s)
| | | | | | | | - Mina S. Makary
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; (G.W.); (C.C.); (J.C.)
| |
Collapse
|
97
|
Guérin C, Vinchent A, Fernandes M, Damour I, Laratte A, Tellier R, Estevam GO, Meneboo JP, Villenet C, Descarpentries C, Fraser JS, Figeac M, Cortot AB, Rouleau E, Tulasne D. MET variants with activating N-lobe mutations identified in hereditary papillary renal cell carcinomas still require ligand stimulation. Mol Oncol 2025. [PMID: 39980226 DOI: 10.1002/1878-0261.13806] [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: 03/14/2024] [Revised: 10/16/2024] [Accepted: 01/15/2025] [Indexed: 02/22/2025] Open
Abstract
In hereditary papillary renal cell carcinoma (HPRCC), the hepatocyte growth factor receptor (MET) receptor tyrosine kinase (RTK) mutations recorded to date are located in the kinase domain and lead to constitutive MET activation. This contrasts with MET mutations identified in non-small-cell lung cancer (NSCLC), which lead to exon 14 skipping and deletion of a regulatory domain: In this latter case, the mutated receptor still requires ligand stimulation. Sequencing of MET in samples from 158 HPRCC and 2808 NSCLC patients revealed 10 uncharacterized mutations. Four of these, all found in HPRCC and leading to amino acid substitutions in the N-lobe of the MET kinase, proved able to induce cell transformation, which was further enhanced by hepatocyte growth factor (HGF) stimulation: His1086Leu, Ile1102Thr, Leu1130Ser, and Cis1125Gly. Similar to the variant resulting in MET exon 14 skipping, the two N-lobe MET variants His1086Leu and Ile1102Thr were found to require stimulation by HGF in order to strongly activate downstream signaling pathways and epithelial cell motility. The Ile1102Thr mutation also displayed transforming potential, promoting tumor growth in a xenograft model. In addition, the N-lobe-mutated MET variants were found to trigger a common HGF-stimulation-dependent transcriptional program, consistent with an observed increase in cell motility and invasion. Altogether, this functional characterization revealed that N-lobe variants still require ligand stimulation, in contrast to other RTK variants. This suggests that HGF expression in the tumor microenvironment is important for tumor growth. The sensitivity of these variants to MET inhibitors opens the way for use of targeted therapies for patients harboring the corresponding mutations.
Collapse
Affiliation(s)
- Célia Guérin
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Audrey Vinchent
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Marie Fernandes
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Isabelle Damour
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Agathe Laratte
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Rémi Tellier
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Gabriella O Estevam
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Jean-Pascal Meneboo
- Univ. Lille, Plateau de génomique fonctionnelle et structurale, CHU Lille, France
| | - Céline Villenet
- Univ. Lille, Plateau de génomique fonctionnelle et structurale, CHU Lille, France
| | - Clotilde Descarpentries
- Department of Biochemistry and Molecular Biology, Hormonology Metabolism Nutrition Oncology, CHU Lille, France
| | - James S Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Martin Figeac
- Univ. Lille, Plateau de génomique fonctionnelle et structurale, CHU Lille, France
| | - Alexis B Cortot
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
- Thoracic Oncology Department, Univ. Lille, CHU Lille, France
| | - Etienne Rouleau
- Department of Medical Biology and Pathology, Cancer Genetics Laboratory, Gustave Roussy, Villejuif, France
| | - David Tulasne
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| |
Collapse
|
98
|
Liu H, Ma Z, Lih TM, Chen L, Hu Y, Wang Y, Sun Z, Huang Y, Xu Y, Zhang H. Machine Learning-Enhanced Extraction of Protein Signatures of Renal Cell Carcinoma from Proteomics Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.17.638651. [PMID: 40027663 PMCID: PMC11870591 DOI: 10.1101/2025.02.17.638651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
In this study, we generated label-free data-independent acquisition (DIA)-based liquid chromatography (LC)-mass spectrometry (MS) proteomics data from 261 renal cell carcinomas (RCC) and 195 normal adjacent tissues (NAT). The RCC tumors included 48 non-clear cell renal cell carcinomas (non-ccRCC) and 213 ccRCC. A total of 219,740 peptides and 11,943 protein groups were identified with 9,787 protein groups per sample on average. We adopted a comprehensive approach to select representative samples with different mutation sites, considering histopathological, immune, methylation, and non-negative matrix factorization (NMF)-based subtypes, along with clinical characteristics (gender, grade, and stage) to capture the complexity and diversity of ccRCC tumors. We used machine learning identified 55 protein signatures that distinguish RCC tumors from NATs. Furthermore, 39 protein signatures that differentiate different RCC tumor subtypes were also identified. Our findings offer an extensive perspective of the proteomic landscape in RCC, illuminating specific proteins that serve to distinguish RCC tumors from NATs and among various RCC tumor subtypes.
Collapse
Affiliation(s)
- Hongyi Liu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Zhuo Ma
- Krieger school of Arts and Sciences, Johns Hopkins University, MD 21218, USA
| | - T. Mamie Lih
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Lijun Chen
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yingwei Hu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yuefan Wang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Zhenyu Sun
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yuanyu Huang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Yuanwei Xu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Hui Zhang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
99
|
Zuo Y, Li T, Yang S, Chen X, Tao X, Dong D, Liu F, Zhu Y. Contribution and expression of renal drug transporters in renal cell carcinoma. Front Pharmacol 2025; 15:1466877. [PMID: 40034145 PMCID: PMC11873565 DOI: 10.3389/fphar.2024.1466877] [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: 07/18/2024] [Accepted: 12/23/2024] [Indexed: 03/05/2025] Open
Abstract
Renal cell carcinoma (RCC) is a common substantive tumor. According to incomplete statistics, RCC incidence accounts for approximately 90% of renal malignant tumors, and is the second most prevalent major malignant tumor in the genitourinary system, following bladder cancer. Only 10%-15% of chemotherapy regimens for metastatic renal cell carcinoma (mRCC) are effective, and mRCC has a high mortality. Drug transporters are proteins located on the cell membrane that are responsible for the absorption, distribution, and excretion of drugs. Lots of drug transporters are expressed in the kidneys. Changes in carrier function weaken balance, cause disease, or modify the effectiveness of drug treatment. The changes in expression of these transporters during cancer pathology results in multi-drug resistance to cancer chemotherapy. In the treatment of RCC, the study of drug transporters helps to optimize treatment regimens, improve therapeutic effects, and reduce drug side effects. In this review, we summarize advances in the role of renal drug transporters in the genesis, progression, and treatment of RCC.
Collapse
Affiliation(s)
- Yawen Zuo
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tong Li
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shilei Yang
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xuyang Chen
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Deshi Dong
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Fang Liu
- Department of Medical Oncology, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yanna Zhu
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
100
|
Wang H, Chen Y, Yang Y, Song R, Gu S, Cao X, Zhang L, Yang Y, Hou T, Qi X, Yang Y, Wang Y, Bai T, Feng D, Yang X, He J. MAGI3 enhances sensitivity to sunitinib in renal cell carcinoma by suppressing the MAS/ERK axis and serves as a prognostic marker. Cell Death Dis 2025; 16:102. [PMID: 39956807 PMCID: PMC11830799 DOI: 10.1038/s41419-025-07427-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/04/2025] [Accepted: 02/04/2025] [Indexed: 02/18/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) exhibits considerable heterogeneity, with approximately 25% of localized cases susceptible to relapse, highlighting the challenge of the absence of reliable predictive biomarkers for personalized treatment. Meanwhile, metastatic renal cell carcinoma is characterized by unfavorable survival rates, and although Sunitinib offers partial benefits, the clinical advantages are often constrained by drug resistance and adverse side effects. Here, MAGI3 was associate with ccRCC progression, as identified through comprehensive bioinformatics analysis of clinical datasets. A low level of MAGI3 emerged as a high-risk factor for ccRCC, indicating its potential as a prognostic marker. Individuals with MAGI3 expression in middle-to-low levels displayed a significantly poorer survival rate, indicating a need for additional treatment even in the early stages of ccRCC. Furthermore, patients with MAGI3 expression in middle-to-high levels exhibited increased sensitivity to Sunitinib compared to those with lower MAGI3 levels, suggesting that individuals with MAGI3 expression at middle levels may potentially benefit from Sunitinib treatment even in the early stages of ccRCC. Through its interaction with the MAS receptor, MAGI3 has been identified as a regulator of cell proliferation and a determinant of Sunitinib resistance in ccRCC, operating via the Ang-(1-7)/MAS/ERK axis. The loss of MAGI3 expression in ccRCC patients activated the ERK signaling pathway, contributing to both cancer progression and Sunitinib resistance. Therefore, our study not only highlight MAGI3's pivotal role in ccRCC progression and Sunitinib resistance, but also reinforces MAGI3's prospective value as a predictive marker.
Collapse
Affiliation(s)
- Haibo Wang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China
| | - Yibin Chen
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
| | - Ying Yang
- Core Facilities Center, Capital Medical University, Beijing, China
| | - Ran Song
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Siyu Gu
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
| | - Xuedi Cao
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
| | - Lijie Zhang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Yang Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Tianzhong Hou
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xuan Qi
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Yumeng Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Yue Wang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Tao Bai
- Department of Pathology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Duiping Feng
- Department of Interventional Radiology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaomei Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China.
| | - Junqi He
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China.
| |
Collapse
|