1
|
Thangavelu PU, Lin CY, Forouz F, Tanaka K, Dray E, Duijf PHG. The RB protein: more than a sentry of cell cycle entry. Trends Mol Med 2025:S1471-4914(25)00088-7. [PMID: 40300971 DOI: 10.1016/j.molmed.2025.04.001] [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: 12/30/2024] [Revised: 03/28/2025] [Accepted: 04/02/2025] [Indexed: 05/01/2025]
Abstract
Genomic instability is a hallmark of cancer. It fuels cancer progression and therapy resistance. As 'the guardian of the genome', the tumor suppressor protein p53 protects against genomic damage. Canonically, the retinoblastoma protein (RB) is 'the sentry of cell cycle entry', as it dictates whether a cell enters the cell cycle to divide. However, the RB pathway also controls myriad non-canonical cellular processes, including metabolism, stemness, angiogenesis, apoptosis, and immune surveillance. We discuss how frequent RB pathway inactivation and underlying mechanisms in cancers affect these processes. We focus on RB's - rather than p53's - 'guardian of the genome' functions in DNA replication, DNA repair, centrosome duplication, chromosome segregation, and chromatin organization. Finally, we review therapeutic strategies, challenges, and opportunities for targeting RB pathway alterations in cancer.
Collapse
Affiliation(s)
- Pulari U Thangavelu
- Frazer Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Cheng-Yu Lin
- Frazer Institute, University of Queensland, Translational Research Institute, Brisbane, QLD, Australia
| | - Farzaneh Forouz
- School of Pharmacy, Faculty of Health and Behavioural Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Kozo Tanaka
- Department of Molecular Oncology, Institute of Development, Aging and Cancer (IDAC), Tohoku University, 4-1 Seiryo-machi, Aoba-ku, Sendai, Miyagi 980-8575, Japan
| | - Eloïse Dray
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Mays Cancer Center at UT Health San Antonio MD Anderson, San Antonio, TX, USA; Greehey Children's Cancer Research Institute, San Antonio, TX, USA
| | - Pascal H G Duijf
- Centre for Cancer Biology, Clinical and Health Sciences, University of South Australia & SA Pathology, Adelaide, SA, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
2
|
Wang Y, Zhang W, Peng M. E2F1-Dependent CDCA5 overexpression drives cervical cancer progression and correlates with poor prognosis. J Mol Histol 2025; 56:80. [PMID: 39907709 DOI: 10.1007/s10735-025-10356-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Cervical cancer (CC) remains a leading cause of cancer-related mortality in women worldwide, highlighting the urgent need for novel therapeutic strategies. This study investigates the molecular mechanisms and clinical significance of Cell Division Cycle Associated 5 (CDCA5) in cervical cancer progression. We performed comprehensive analyses of CDCA5 expression in cervical cancer and normal tissues, correlating expression levels with clinicopathological features and patient outcomes. Functional studies using CC cell lines (SiHa, HeLa, and CaSki) examined the effects of CDCA5 manipulation on tumor cell behavior. We identified E2F1 as a key transcriptional regulator of CDCA5 and validated our findings using in vivo xenograft models. CDCA5 was significantly upregulated in CC tissues and correlated with advanced disease stages and poor survival outcomes. Mechanistically, CDCA5 depletion in SiHa and HeLa cells suppressed proliferation, migration, and invasion, while its overexpression in CaSki cells enhanced these malignant properties. We identified E2F1 as a transcriptional activator of CDCA5. Importantly, CDCA5 knockdown significantly inhibited tumor growth in nude mouse models. Our findings establish CDCA5 as a critical E2F1-regulated oncogenic factor in cervical cancer progression. The strong correlation between CDCA5 expression and poor clinical outcomes suggests its potential as both a prognostic biomarker and therapeutic target in cervical cancer treatment.
Collapse
Affiliation(s)
- Youhui Wang
- Tumor Radiotherapy and Chemotherapy Center, Ningbo University Affiliated People's Hospital, No. 251, Baizhang East Road, Ningbo, 315040, Zhejiang, China.
| | - Wuguang Zhang
- Tumor Radiotherapy and Chemotherapy Center, Ningbo University Affiliated People's Hospital, No. 251, Baizhang East Road, Ningbo, 315040, Zhejiang, China
| | - Min Peng
- Tumor Radiotherapy and Chemotherapy Center, Ningbo University Affiliated People's Hospital, No. 251, Baizhang East Road, Ningbo, 315040, Zhejiang, China
| |
Collapse
|
3
|
Wu T, Jiang F, Wu F, Zheng G, Li Y, Wu L. E2F1 and E2F7 regulate gastric cancer cell proliferation, respectively, through transcriptional activation and transcriptional repression of MYBL2. J Biol Chem 2025; 301:108027. [PMID: 39613162 PMCID: PMC11731210 DOI: 10.1016/j.jbc.2024.108027] [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: 05/30/2024] [Revised: 10/22/2024] [Accepted: 11/12/2024] [Indexed: 12/01/2024] Open
Abstract
Gastric cancer (GC) is the most common malignant tumor of the digestive tract. However, the molecular pathogenesis is not well understood. Through bioinformatic analysis and analyzing clinical tissue samples, we found that E2F1 and E2F7 as well as their potential downstream target MYBL2 were all upregulated in GC tissues and that their expressions correlated with patient prognosis. While knockdown of E2F1 or MYBL2 inhibited cell proliferation and promoted apoptosis, knockdown of E2F7 promoted cell proliferation but had no effects on apoptosis. Chromatin immunoprecipitation and dual luciferase reporter assays demonstrated that MYBL2 was transcriptionally activated and repressed by E2F1 and E2F7, respectively. Importantly, in vitro and ex vivo experiments demonstrated that the effects of E2F1 and E2F7 on GC cell proliferation were significantly attenuated by reversely modulating MYBL2 expression, indicating that MYBL2 is a direct and functionally relevant target of E2F1 and E2F7 in GC cells. Furthermore, the effects of E2F1 and E2F7 on GC cell proliferation through transcriptional regulation of MYBL2 can be mediated by the PI3K/AKT signaling pathway. Interestingly, we found differential nucleocytoplasmic distribution of E2F7 in GC cells with functional relevance. Taken together, our data suggest that targeted therapies of GC may be achieved from three different angles, E2F1, E2F7, and MYBL2 themselves, E2F1/E2F7 expression balance, and E2F7 nuclear localization.
Collapse
Affiliation(s)
- Tianyi Wu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Fengli Jiang
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China; Department of Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fan Wu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Guoliang Zheng
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China; Department of Gastric Surgery, Cancer Hospital of China Medical University (Liaoning Cancer Hospital and Institute), Shenyang, China
| | - Yang Li
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China; Department of Gynecology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lizhao Wu
- Department of Pathophysiology, College of Basic Medical Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
4
|
Wang H, Liu C, Jin K, Li X, Zheng J, Wang D. Research advances in signaling pathways related to the malignant progression of HSIL to invasive cervical cancer: A review. Biomed Pharmacother 2024; 180:117483. [PMID: 39353319 DOI: 10.1016/j.biopha.2024.117483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
The progression of high-grade squamous intraepithelial lesion (HSIL) to invasive cervical cancer (ICC) is a complex process involving persistent human papillomavirus (HPV) infection and changes in signal transduction regulation, energy and material metabolism, cell proliferation, autoimmune, and other biological process in vaginal microenvironment and immune microenviroment. Signaling pathways are a series of interacting molecules in cells that regulate various physiological functions of cells, such as growth, differentiation, metabolism, and death. In the progression of HSIL to ICC, abnormal activation or inhibition in signaling pathways plays an essensial role. This review presented some signaling pathways related to the malignant progression of HSIL to ICC, including p53, Rb, PI3K/AKT/mTOR, Wnt/β-catenin, Notch, NF-κB, MAPK, TGF-β, JAK-STAT, Hippo, and Hedgehog. The molecular mechanisms involved in the biological process of pathway regulation were also analyzed, in order to illustrate the molecular pathway of HSIL progression to ICC and provide references for the development of more effective prevention and treatment methods.
Collapse
Affiliation(s)
- Huifang Wang
- Department of Obstetrics and Gynecology, Quanzhou Medical College, Quanzhou, Fujian 362010, China
| | - Chang Liu
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; Key Clinical Specialty of Liaoning Province, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Keer Jin
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; Key Clinical Specialty of Liaoning Province, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Xiang Li
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Jiaxin Zheng
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Danbo Wang
- Department of Gynecology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China; Key Clinical Specialty of Liaoning Province, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang 110042, China; Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang 110042, China.
| |
Collapse
|
5
|
Xu Y, Jia K, Zhao X, Li Y, Zhang Z. LncRNA LINC01703 promotes the proliferation, migration, and invasion of colorectal cancer by activating PI3K/AKT pathway. J Biochem Mol Toxicol 2024; 38:e23594. [PMID: 38050438 DOI: 10.1002/jbt.23594] [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: 05/03/2023] [Revised: 11/07/2023] [Accepted: 11/20/2023] [Indexed: 12/06/2023]
Abstract
The role of LINC01703 in cancers, especially in colorectal cancer (CRC), is still largely unclear. Bioinformatics prediction, real-time quantitative polymerase chain reaction (RT-qPCR), 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide (MTT) assay, colony formation assay, Transwell assays, in vivo animal experiments, IF, luciferase reporter assay, and Western blot were carried out for the exploration of the potential involvement and underlying molecular mechanisms of LINC01703 in CRC cells. The results showed that LINC01703 appeared upregulated in CRC and was linked to poor prognosis. LINC01703 acted as an oncogene in both in vitro and in vivo CRC cell environments. LINC01703 activated the PI3K/AKT signaling pathway by mediating the miR-205-5p/E2F1 axis in CRC. In summary, LINC01703 possesses an oncogenic function and can be a possible biomarker or target to treat CRC.
Collapse
Affiliation(s)
- Yuanhui Xu
- Department of Proctology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Keliang Jia
- Department of Proctology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Xianhai Zhao
- Department of Proctology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Yuantao Li
- Department of Proctology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Zhen Zhang
- Department of Proctology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
6
|
Yang W, An L, Li Y, Qian S. A cellular senescence-related genes model allows for prognosis and treatment stratification of cervical cancer: a bioinformatics analysis and external verification. Aging (Albany NY) 2023; 15:9408-9425. [PMID: 37768206 PMCID: PMC10564413 DOI: 10.18632/aging.204981] [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: 05/17/2023] [Accepted: 07/20/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND Cervical cancer (CC) is highly lethal and aggressive with an increasing trend of mortality for females. Molecular characterization-based methods hold great promise for improving the diagnostic accuracy and for predicting treatment response. METHODS The mRNAs expression data of CC patients and cellular senescence-related genes were obtained from the Cancer Genome Atlas (TCGA) and CellAge databases, respectively. Differentially expressed genes (DEGs) of senescence related genes between tumor and normal tissues were used for Least absolute shrinkage and selection operator (LASSO) regression to construct a prognostic model. Univariate and LASSO regression analyses were applied to establish a predictive nomogram. The performance of the nomogram were evaluated by Kaplan-Meier curve, receiver operating characteristic (ROC), Harrell's concordance index (C-index), and calibration curve. GSE44001 and GSE52903 were used for external validation. RESULTS We established a cellular senescence-related genes-based stratified model, and a multivariable-based nomogram, which could accurately predict the prognosis of CC patients in the TCGA database. The Kaplan-Meier curve indicated that patients in the low-risk group had considerably better overall survival (OS, P =2.021e-05). The area under the ROC curve (AUC) of this model was 0.743 for OS. Multivariate analysis found that the 6-gene risk signature (HR=3.166, 95%CI: 1.660-6.041, P<0.001) was an independent risk factor for CC patients. We then designed an OS-associated nomogram that included the risk signature and clinicopathological factors. The AUC reached 0.860 for predicting 5-year OS. The nomogram showed excellent consistency between the predictions and actual survival observations. Two external GEO validations were corresponding to the gene expression pattern in TCGA. CONCLUSIONS Our results suggested a six-senescence related signature and established a prognostic nomogram that reliably predicted the overall survival for CC. These findings may be beneficial to personalized treatment and medical decision-making.
Collapse
Affiliation(s)
- Weiwei Yang
- Gynecology Department 2, Cangzhou Central Hospital, Yunhe District, Cangzhou 061000, Hebei Province, China
| | - Lijuan An
- Gynecology Department 2, Cangzhou Central Hospital, Yunhe District, Cangzhou 061000, Hebei Province, China
| | - Yanfei Li
- Gynecology Department 2, Cangzhou Central Hospital, Yunhe District, Cangzhou 061000, Hebei Province, China
| | - Sumin Qian
- Gynecology Department 2, Cangzhou Central Hospital, Yunhe District, Cangzhou 061000, Hebei Province, China
| |
Collapse
|