1
|
Han J, Ding R, Xiong X. Expression of NUF2 and CD52 and Their Correlation With Chemotherapy Resistance and Prognosis in Patients With Non-Small Cell Lung Cancer. J Biochem Mol Toxicol 2025; 39:e70308. [PMID: 40421783 DOI: 10.1002/jbt.70308] [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: 03/27/2025] [Revised: 03/27/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025]
Abstract
Lung cancer is one of the most common malignant tumors. This study aim to investigate the expression of cell division related gene cell division cycle related protein 2 (NUF2) and immune-related gene CD52 in patients with non-small cell lung cancer (NSCLC) and their correlation with chemotherapy resistance and clinical prognosis. The cancer tissues and adjacent tissues of 72 patients with NSCLC were collected. According to the staining results, the patients were divided into NUF2 positive and negative expression groups, and CD52 positive and negative expression groups. The relationship of NUF2 and CD52 with clinicopathological features and chemoresistance was analyzed. The patients were followed up for 5 years, and Kaplan-Meier survival curve was used to analyze the relationship. COX regression analysis was used to analyze the risk factors. The positive expression rate of NUF2 in NSCLC tissues was 66.67% (48/72), which was significantly higher than 38.89% (28/72) in adjacent tissues (p < 0.05). The positive expression rate of CD52 in NSCLC tissues was also higher than the adjacent tissues (p < 0.05). NUF2 expression was related to the degree of differentiation, TNM stage, lymph node metastasis and pleural invasion in NSCLC patients (p < 0.05). The expression of CD52 was related to TNM stage, lymph node metastasis, vascular tumor thrombus and pleural invasion (p < 0.05). The median survival time of patients with positive CD52 and NUF2 expression was lower than patients with negative expression (p < 0.05). Platinum-resistance, lymph node metastasis, pleural invasion, NUF2 and CD52 positive expression were independent risk factors for poor prognosis of NSCLC (p < 0.05). In conclusion, NUF2 and CD52 are independent risk factors for poor prognosis of patients with NSCLC. They may be used as one of the indicators to evaluate drug resistance and prognosis of patients.
Collapse
Affiliation(s)
- Jun Han
- Department of Respiratory and Critical Care Medicine, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Rui Ding
- Department of Radiotherapy, Shanxi Provincial People's Hospital, Taiyuan, China
| | - Xue Xiong
- Department of Respiratory and Critical Care Medicine, Shanxi Provincial People's Hospital, Taiyuan, China
| |
Collapse
|
2
|
Jin C, Xu J, Luo W, Guo H, Ding L, Liu Y, Liu J, Zou L, Yu Y, Hao Y, Yang B. NUF2 activated by YY1 promotes prostate cancer malignancy via p38/MAPK signaling axis and serves as a therapeutic target. Biochem Pharmacol 2025; 237:116919. [PMID: 40187571 DOI: 10.1016/j.bcp.2025.116919] [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: 12/09/2024] [Revised: 03/26/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Prostate cancer (PCa) is one of the most prevalent malignancies in the male urogenital system. Despite extensive research into its mechanisms of initiation and progression, the full scope of its pathophysiology remains insufficiently understood. This study demonstrated that NUF2 was significantly overexpressed in PCa, with its elevated levels correlating with poor patient survival outcomes. Silencing NUF2 notably impaired PCa cell proliferation and metastasis in both in vitro and in vivo models, whereas its overexpression promoted these processes. Additionally, YY1 was identified as a direct transcriptional activator of NUF2, binding to its promoter and enhancing its oncogenic effects through activation of downstream targets. Moreover, NUF2 promoted PCa progression by recruiting p38, accelerating its phosphorylation, and activating the p38/MAPK signaling pathway. Through the PubChem database, fisetin was identified as a small molecule inhibitor of NUF2. Fisetin effectively inhibited PCa cell proliferation, and NUF2 overexpression reversed this inhibitory effect. In conclusion, our results suggest that NUF2 overexpression accelerated PCa progression via the p38/MAPK pathway, regulated by YY1. The identification of fisetin as a NUF2 inhibitor offers a promising therapeutic strategy for targeting NUF2 to impede PCa growth. NUF2 may thus serve as a valuable prognostic biomarker and a potential therapeutic target for PCa.
Collapse
Affiliation(s)
- Chengqi Jin
- School of Medicine, Anhui University of Science and Technology, Huainan, China; Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Jing Xu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Wentao Luo
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Hanxu Guo
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Li Ding
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yongqiang Liu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Ji Liu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Libin Zou
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yang Yu
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yajuan Hao
- Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| | - Bin Yang
- School of Medicine, Anhui University of Science and Technology, Huainan, China; Department of Urology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China; Urologic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
3
|
Giantin M, Montanucci L, Lopparelli RM, Tolosi R, Dentini A, Grieco V, Stefanello D, Sabattini S, Marconato L, Pauletto M, Dacasto M. Expression Profile of Twelve Transcripts as a Supporting Tool for the Molecular Characterization of Canine Cutaneous Mast Cell Tumors at Diagnosis: Association with Histological Grading and Clinical Staging. Genes (Basel) 2025; 16:340. [PMID: 40149490 PMCID: PMC11942052 DOI: 10.3390/genes16030340] [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: 02/05/2025] [Revised: 02/27/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Mast cell tumors (MCTs) are the second most common malignant neoplasms in dogs. Histopathological grading and clinical staging are the main tools for estimating biological behavior and disease extent; thus, both are essential for therapeutic decision-making and prognostication. However, the biological behavior of MCTs in dogs is variable, and it sometimes deviates from expectations. In a previous study, we identified 12 transcripts whose expression profile allowed a clear distinction between Kiupel low-grade and high-grade cutaneous MCTs (cMCTs) and was associated with prognosis. Building on these findings, this study evaluated the predictive potential of these transcripts' expression profiles in classifying cMCTs into low-grade and high-grade. METHODS A logistic regression classifier based on the expression profiles of the identified transcripts and able to classify cMCTs as low- or high-grade was developed and subsequently tested on a novel dataset of 50 cMCTs whose expression profiles have been determined in this study through qPCR. RESULTS The developed logistic regression classifier reaches an accuracy of 67% and an area under the receiver operating characteristic curve (AUC) of 0.76. Interestingly, the molecular classification clearly identifies stage-IV disease (90% true positive rate). CONCLUSIONS qPCR analysis of these biomarkers combined with the machine learning-based classifier might serve as a tool to support cMCT clinical management at diagnosis.
Collapse
Affiliation(s)
- Mery Giantin
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, PD, Italy; (R.M.L.); (R.T.); (M.P.); (M.D.)
| | - Ludovica Montanucci
- Department of Neurology, Mc Govern Medical School, The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 44106, USA;
| | - Rosa Maria Lopparelli
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, PD, Italy; (R.M.L.); (R.T.); (M.P.); (M.D.)
| | - Roberta Tolosi
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, PD, Italy; (R.M.L.); (R.T.); (M.P.); (M.D.)
| | - Alfredo Dentini
- Clinica Veterinaria Tyrus, Via Aldo Bartocci 1G, I-05100 Terni, TR, Italy;
| | - Valeria Grieco
- Department of Veterinary Medicine and Animal Science, University of Milan, Via dell’Università 6, I-26900 Lodi, MI, Italy; (V.G.); (D.S.)
| | - Damiano Stefanello
- Department of Veterinary Medicine and Animal Science, University of Milan, Via dell’Università 6, I-26900 Lodi, MI, Italy; (V.G.); (D.S.)
| | - Silvia Sabattini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, Via Tolara di Sopra 50, I-40064 Ozzano dell’Emilia, BO, Italy; (S.S.); (L.M.)
| | - Laura Marconato
- Department of Veterinary Medical Sciences, Alma Mater Studiorum, University of Bologna, Via Tolara di Sopra 50, I-40064 Ozzano dell’Emilia, BO, Italy; (S.S.); (L.M.)
| | - Marianna Pauletto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, PD, Italy; (R.M.L.); (R.T.); (M.P.); (M.D.)
| | - Mauro Dacasto
- Department of Comparative Biomedicine and Food Science, University of Padua, Viale dell’Università 16, I-35020 Legnaro, PD, Italy; (R.M.L.); (R.T.); (M.P.); (M.D.)
| |
Collapse
|
4
|
Xiong Z, Huang Y, Cao S, Huang X, Zhang H. A new strategy for the treatment of advanced ovarian cancer: utilizing nanotechnology to regulate the tumor microenvironment. Front Immunol 2025; 16:1542326. [PMID: 40013141 PMCID: PMC11860879 DOI: 10.3389/fimmu.2025.1542326] [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: 12/09/2024] [Accepted: 01/30/2025] [Indexed: 02/28/2025] Open
Abstract
Advanced ovarian cancer (AOC) is prone to recurrence, which can be attributed to drug resistance. Drug resistance may be related to the tumor microenvironment (TME), including the immune and non-immune TME. In the immune TME, the immune effector cells such as dendritic cells (DCs), M1-like tumor-associated macrophages (M1-TAMs), and T cells are inhibited. In contrast, immunosuppressive cells such as M2-like tumor-associated macrophages (M2-TAMs), myeloid-derived suppressor cells (MDSCs), and regulatory T cells (Tregs) are activated. These changes make it difficult to produce immune effects and affect the efficacy of chemo-immunotherapy. In the non-immune TME, mechanisms such as apoptosis inhibition, DNA damage response (DDR), and epithelial-mesenchymal transition (EMT) can promote tumor growth, metastasis, and drug resistance. Despite the challenges posed by the TME in the treatment of AOC, the unique biological advantages of nanoparticles (NPs) make it possible to regulate the TME. NPs can stimulate the immune responses of M1-TAMs, DCs, and T cells while reducing the infiltration of immune suppressive cells such as M2-TAMs and Tregs, thereby regulating the AOC immune TME. In addition, NPs can regulate the non-immune TME by reducing apoptosis in AOC cells, inhibiting homologous recombination (HR) repair, reversing EMT, and achieving the effect of reversing drug resistance. In summary, the application of NPs provides some new venues for clinical treatment in AOC.
Collapse
Affiliation(s)
- Zixuan Xiong
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Yichun Huang
- Department of Oncology, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shulong Cao
- Department of Pathology, Songzi People’s Hospital, Jingzhou, China
| | - Xuqun Huang
- Department of Medical Oncology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| | - Haiyuan Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| |
Collapse
|
5
|
王 耀, 陈 柳, 罗 伊, 申 继, 周 素. Predictive value of NUF2 for prognosis and immunotherapy responses in pan-cancer. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2025; 45:137-149. [PMID: 39819722 PMCID: PMC11744274 DOI: 10.12122/j.issn.1673-4254.2025.01.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Indexed: 01/30/2025]
Abstract
OBJECTIVES To investigate the association of NUF2 expression with tumor prognosis and its regulatory role in tumor microenvironment. METHODS We analyzed NUF2 expression, its prognostic value, and is immune-related functions across different cancer types using datasets from the Human Protein Atlas (HPA), TCGA, GTEx, CCLE, and TIMER. RT-qPCR, Western blotting, and immunohistochemistry were used to detect NUF2 expression in liver cancer cell lines and tissue and blood samples from patients with liver cancer. GO, KEGG, and GSEA analyses were conducted to explore the molecular mechanisms of NUF2 and its related genes, and a competitive endogenous RNA (ceRNA) network for NUF2 in liver cancer was constructed. RESULTS NUF2 expression was upregulated in the tumor tissues of 27 cancers and was associated with clinical stages in several cancers. High NUF2 expressions were correlated with poor overall survival, disease-specific survival, progression-free survival, and disease-free survival of cancer patients. NUF2 expression levels were positively correlated with tumor mutational burden, microsatellite instability, infiltrating immune cells, immune cell marker genes and immune checkpoint genes in different cancers. RT-qPCR, Western blotting, and immunohistochemistry confirmed that NUF2 expression was upregulated in liver cancer cell lines and tumor tissues and blood samples of liver cancer patients, and was decreased significantly after operation. GO, KEGG and GSEA analyses indicated that NUF2 was involved in chromosome segregation and cell cycle and was associated with glycine, serine and threonine metabolism. CONCLUSIONS NUF2 expression is upregulated in 27 cancers and is associated with clinical stage and poor prognosis in some malignancies. NUF2 expression is closely correlated with immune cell infiltration in different cancers, suggesting its potential value for predicting immunotherapy response in these cancers.
Collapse
|
6
|
Lv M, Chen X, Yang Q, Huang C, Lv Y, Zhang T, Cai J. Berberine restrains non-small cell lung cancer cell growth, invasion and glycolysis via inactivating the SPC25/NUF2 pathway. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-024-03729-w. [PMID: 39755832 DOI: 10.1007/s00210-024-03729-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 12/11/2024] [Indexed: 01/06/2025]
Abstract
Berberine (BBR) has been proved to inhibit the malignant progression of non-small cell lung cancer (NSCLC), but the underlying molecular mechanism still needs to be further revealed. NSCLC cells (A549 and H1299) were treated with BBR. CCK8 assay, colony formation assay, flow cytometry, TUNEL staining and transwell assay were used to examine cell proliferation, apoptosis and invasion. The levels of spindle pole body component 25 (SPC25) and NDC80 kinetochore complex component (NUF2) were detected by qRT-PCR or western blot. The interaction between SPC25 and NUF2 was confirmed by Co-IP assay and FISH assay. Xenograft tumors were constructed to assess the anti-tumor role of BBR in vivo. BBR inhibited NSCLC cell growth, invasion and glycolysis. SPC25 was upregulated in NSCLC tissues, and BBR could reduce SPC25 expression in NSCLC cells. SPC25 knockdown repressed NSCLC cell growth, invasion and glycolysis, and its overexpression also reversed the anti-tumor effect of BBR. SPC25 could interact with NUF2, and NUF2 overexpression abolished the inhibitory effect of SPC25 knockdown or BBR on NSCLC cell behaviors. In animal experiments, BBR could suppress NSCLC tumor growth by inhibiting SPC25/NUF2 axis in vivo. BBR mainly played an anti-NSCLC role by targeting SPC25/NUF2 axis, which provided a new idea for NSCLC treatment.
Collapse
Affiliation(s)
- Meng Lv
- Department of Respiratory and Critical Care Medicine, Shenzhen Hospital of Traditional Chinese Medicine, Shenzhen, China
| | - Xiangrui Chen
- Department of Hematology and Oncology, Third People's Hospital of Zigong, Zigong, Sichuan, China
| | - Qiting Yang
- Department of Cardiopulmonary Rehabilitation and Sleep Medicine, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chushuan Huang
- Pneumology Department, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongbiao Lv
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tian Zhang
- The First Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junxiang Cai
- Department of Respiratory and Critical Care Medicine, Guangdong Provincial Hospital of Traditional Chinese Medicine, No. 111, Dade Road, Guangzhou, 510120, China.
| |
Collapse
|
7
|
Bao L, Gong Y, Che Y, Li Y, Xu T, Chen J, Wang S, Tan Z, Huang P, Pan Z, Ge M. Maintenance of magnesium homeostasis by NUF2 promotes protein synthesis and anaplastic thyroid cancer progression. Cell Death Dis 2024; 15:656. [PMID: 39242581 PMCID: PMC11379715 DOI: 10.1038/s41419-024-07041-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 08/15/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024]
Abstract
Thyroid cancer is the most frequently observed endocrine-related malignancy among which anaplastic thyroid cancer (ATC) is the most fatal subtype. The synthesis of protein is active to satisfy the rapid growth of ATC tumor, but the mechanisms regulating protein synthesis are still unknown. Our research revealed that kinetochore protein NUF2 played an essential role in protein synthesis and drove the progression of ATC. The prognosis of patients with thyroid carcinoma was positively correlated with high NUF2 expression. Depletion of NUF2 in ATC cells notably inhibited the proliferation and induced apoptosis, while overexpression of NUF2 facilitated ATC cell viability and colony formation. Deletion of NUF2 significantly suppressed the growth and metastasis of ATC in vivo. Notably, knockdown of NUF2 epigenetically inhibited the expression of magnesium transporters through reducing the abundance of H3K4me3 at promoters, thereby reduced intracellular Mg2+ concentration. Furthermore, we found the deletion of NUF2 or magnesium transporters significantly inhibited the protein synthesis mediated by the PI3K/Akt/mTOR pathway. In conclusion, NUF2 functions as an emerging regulator for protein synthesis by maintaining the homeostasis of intracellular Mg2+, which finally drives ATC progression.
Collapse
Affiliation(s)
- Lisha Bao
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yingying Gong
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yulu Che
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Ying Li
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Tong Xu
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Jinming Chen
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Shanshan Wang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Zhuo Tan
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for malignant tumor, Hangzhou, China
| | - Ping Huang
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China.
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for malignant tumor, Hangzhou, China.
| | - Zongfu Pan
- Clinical Pharmacy Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China.
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for malignant tumor, Hangzhou, China.
| | - Minghua Ge
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
- Zhejiang Key Laboratory of Precision Medicine Research on Head & Neck Cancer, Hangzhou, China.
- Zhejiang Provincial Clinical Research Center for malignant tumor, Hangzhou, China.
| |
Collapse
|
8
|
Wang Z, Ren M, Liu W, Wu J, Tang P. Role of cell division cycle-associated proteins in regulating cell cycle and promoting tumor progression. Biochim Biophys Acta Rev Cancer 2024; 1879:189147. [PMID: 38955314 DOI: 10.1016/j.bbcan.2024.189147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024]
Abstract
The cell division cycle-associated protein (CDCA) family is important in regulating cell division. High CDCA expression is significantly linked to tumor development. This review summarizes clinical and basic studies on CDCAs conducted in recent decades. Furthermore, it systematically introduces the molecular expression and function, key mechanisms, cell cycle regulation, and roles of CDCAs in tumor development, cell proliferation, drug resistance, invasion, and metastasis. Additionally, it presents the latest research on tumor diagnosis, prognosis, and treatment targeting CDCAs. These findings are pivotal for further in-depth studies on the role of CDCAs in promoting tumor development and provide theoretical support for their application as new anti-tumor targets.
Collapse
Affiliation(s)
- Zhaoyu Wang
- Department of Breast and Thyroid Surgery, Southwest Hospital, the First Affiliated Hospital of the Army Military Medical University, Chongqing 400038, China
| | - Minshijing Ren
- Department of Breast and Thyroid Surgery, Southwest Hospital, the First Affiliated Hospital of the Army Military Medical University, Chongqing 400038, China
| | - Wei Liu
- Department of Breast and Thyroid Surgery, Southwest Hospital, the First Affiliated Hospital of the Army Military Medical University, Chongqing 400038, China
| | - Jin Wu
- Department of Breast and Thyroid Surgery, Southwest Hospital, the First Affiliated Hospital of the Army Military Medical University, Chongqing 400038, China; Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China.
| | - Peng Tang
- Department of Breast and Thyroid Surgery, Southwest Hospital, the First Affiliated Hospital of the Army Military Medical University, Chongqing 400038, China.
| |
Collapse
|
9
|
Luo L, Sha T, Li H. Knockdown of NUF2-derived exosomes can inhibit the migration and autophagy of BC cells and improve resistance to doxorubicin. Tissue Cell 2024; 89:102455. [PMID: 38964084 DOI: 10.1016/j.tice.2024.102455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/06/2024] [Accepted: 06/19/2024] [Indexed: 07/06/2024]
Abstract
Breast cancer (BC) is the most common type of fatal cancer in women. New therapeutic strategies need to be explored to enhance the efficacy of doxorubicin by overcoming the resistance of BC cells. NUF2 is a component of the Ndc80 centromere complex and is a key substance in mediating mitosis and affects the progression of multiple tumors. However, the role as well as mechanisms of NUF2 resistance in BC remain unclear. This study aims to reveal the role of NUF2 in drug resistance in BC. We here revealed that NUF2 was highly expressed in human BC. NUF2 depletion-derived exosomes blocked the growth of BC cells. Further, NUF2 ablation-derived exosomes inhibited autophagy in BC cells. Also, NUF2 ablation-derived exosomes improved doxorubicin resistance in BC cells. Mechanically, NUF2 ablation-derived exosomes blocked PI3K/AKT/mTOR axis in BC cells. In summary, NUF2 ablation-derived exosomes blocked the autophagy of BC cells and improved doxorubicin resistance via mediating PI3K/AKT/mTOR axis.
Collapse
Affiliation(s)
- Lin Luo
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi City, Xinjiang Uygur Autonomous Region 830011, China
| | - Tong Sha
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi City, Xinjiang Uygur Autonomous Region 830011, China
| | - Hongtao Li
- The Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi City, Xinjiang Uygur Autonomous Region 830011, China.
| |
Collapse
|
10
|
Li F, Wu Z, Du Z, Ke Q, Fu Y, Zhan J. Comprehensive molecular analyses and experimental validation of CDCAs with potential implications in kidney renal papillary cell carcinoma prognosis. Heliyon 2024; 10:e33045. [PMID: 38988558 PMCID: PMC11234104 DOI: 10.1016/j.heliyon.2024.e33045] [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: 03/30/2024] [Revised: 05/29/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024] Open
Abstract
Previous reports have revealed that the abnormal expression of the cell division cycle-associated gene family (CDCAs) is closely associated with some human cancers. However, the precise functional roles and mechanisms of CDCAs in kidney renal papillary cell carcinoma (KIRP) remain unclear. In this study, RNA sequencing data from the Cancer Genome Atlas database and Genotype-Tissue Expression databases were utilized to perform the expression, correlation, survival, mutation, functional enrichment analysis, and immunoinfiltration analyses of CDCAs in KIRP. We found that the expression levels of CDCA genes were significantly increased in KIRP across multiple databases, as confirmed by immunohistochemistry and quantitative reverse transcription PCR (RT-qPCR). Moreover, increased expression of CDCA genes is significantly associated with poor prognosis. Univariate and multivariate Cox regression analyses demonstrated that pathologic T and N staging, NUF2, CDCA2, CDCA3, CDCA5, CBX2, CDCA7, and CDCA8 were independent prognostic factors for patients with KIRP. Utilizing these nine variables, we developed a nomogram prognostic model. Furthermore, the results of GO and KEGG functional enrichment analyses suggested that CDCA genes were associated with nuclear division, mitotic nuclear division, and chromosome segregation and were involved in the cell cycle, p53 signaling pathway, and cellular senescence. We found that the expression of NUF2, CDCA2, CDCA5, and CBX2 was closely associated with the expression of lymphocytes, immunostimulatory molecules, immunoinhibitory molecules, and chemokines. In summary, NUF2, CDCA2, CDCA3, CDCA5, CBX2, CDCA7, and CDCA8 are potential biomarkers for KIRP diagnosis and prognosis.
Collapse
Affiliation(s)
- Fuping Li
- Department of General Surgery, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- Department of the Second Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Zhenheng Wu
- Department of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhiyong Du
- Department of General Surgery, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Qiming Ke
- Department of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Yuxiang Fu
- Department of General Surgery, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Jiali Zhan
- Department of General Practice, Xiamen Fifth Hospital, Xiamen, China
| |
Collapse
|
11
|
Liu Y, Wang Y, Wang J, Jiang W, Chen Y, Shan J, Li X, Wu X. NUF2 regulated the progression of hepatocellular carcinoma through modulating the PI3K/AKT pathway via stabilizing ERBB3. Transl Oncol 2024; 44:101933. [PMID: 38507923 PMCID: PMC10966282 DOI: 10.1016/j.tranon.2024.101933] [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: 10/16/2023] [Revised: 02/08/2024] [Accepted: 03/07/2024] [Indexed: 03/22/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is among the most prevalent and lethal cancers worldwide. The NDC80 kinetochore complex component NUF2 has been previously identified as up-regulating in HCC and associated with patient prognosis. However, the pathophysiological effects and molecular mechanisms of NUF2 in tumorigenesis remain unclear. In this study, we confirmed a significant increase in NUF2 expression in HCC tissues and established a correlation between high NUF2 expression and adverse outcomes in HCC patients. Through in vitro and in vivo experiments, we demonstrated that genetic inhibition of NUF2 suppressed the proliferation of HCC cells and disrupted the cell cycle. Further investigation into the molecular mechanisms revealed that NUF2 interacted with ERBB3, inhibiting its ubiquitination degradation, thus activating the PI3K/AKT signaling pathway and influencing cell cycle regulation. Overall, this study revealed the crucial role of NUF2 in promoting the malignant progression of HCC, suggesting its potential as both a prognostic biomarker and a therapeutic target for HCC.
Collapse
Affiliation(s)
- Yiwei Liu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China; Jiangsu Provincial Medical Innovation Center; Jiangsu Provincial Medical Key Laboratory, Nanjing, China.
| | - Yuming Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China; Jiangsu Provincial Medical Innovation Center; Jiangsu Provincial Medical Key Laboratory, Nanjing, China
| | - Jifei Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China; Jiangsu Provincial Medical Innovation Center; Jiangsu Provincial Medical Key Laboratory, Nanjing, China
| | - Wangjie Jiang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China; Jiangsu Provincial Medical Innovation Center; Jiangsu Provincial Medical Key Laboratory, Nanjing, China
| | - Yananlan Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China; Jiangsu Provincial Medical Innovation Center; Jiangsu Provincial Medical Key Laboratory, Nanjing, China
| | - Jijun Shan
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China; Jiangsu Provincial Medical Innovation Center; Jiangsu Provincial Medical Key Laboratory, Nanjing, China
| | - Xiao Li
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Xiaofeng Wu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University; Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China; Jiangsu Provincial Medical Innovation Center; Jiangsu Provincial Medical Key Laboratory, Nanjing, China.
| |
Collapse
|
12
|
Aimaier R, Chung MH, Gu Y, Yu Q, Wei C, Li H, Guo Z, Long M, Li Y, Wang W, Li Q, Wang Z. FOXM1 promotes neurofibromatosis type 1-associated malignant peripheral nerve sheath tumor progression in a NUF2-dependent manner. Cancer Gene Ther 2023; 30:1390-1402. [PMID: 37488294 DOI: 10.1038/s41417-023-00645-8] [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: 09/22/2022] [Revised: 06/23/2023] [Accepted: 07/04/2023] [Indexed: 07/26/2023]
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft-tissue sarcomas characterized by poor prognosis and low drug response rates. Traditional chemo/radiotherapies show only mild benefits for patients with MPNSTs, and no targeted therapy is available in the clinic. A better understanding of the molecular background of MPNSTs is critical for the development of effective targeted therapies. Forkhead box M1 (FOXM1) has been implicated in the progression of many human malignancies, though its role in MPNSTs is unclear. In this study, using four Gene Expression Omnibus (GEO) datasets and a tissue microarray, we demonstrated that FOXM1 upregulation was associated with poor prognosis in patients with MPNSTs. FOXM1 overexpression and knockdown regulated the proliferation and colony formation of MPNST cells. Using bioinformatics analysis and luciferase reporter assays, we identified NUF2 as a direct downstream target of FOXM1. Both in vitro and in vivo experiments demonstrated that the induction of MPNST cell proliferation by FOXM1 was dependent on elevated NUF2 expression, as NUF2 knockdown abolished the FOXM1-induced proliferation of MPNST cells. Our study showed that the FOXM1-NUF2 axis mediates human MPNST progression and could be a potential therapeutic target.
Collapse
Affiliation(s)
- Rehanguli Aimaier
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Man-Hon Chung
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yihui Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingxiong Yu
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai, China
| | - Chengjiang Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haibo Li
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zizhen Guo
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Manmei Long
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuehua Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhichao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|