1
|
Saadh MJ, Allela OQB, Kareem RA, Baldaniya L, Ballal S, Vashishth R, Parmar M, Sameer HN, Hamad AK, Athab ZH, Adil M. Prognostic gene expression profile of colorectal cancer. Gene 2025; 955:149433. [PMID: 40122415 DOI: 10.1016/j.gene.2025.149433] [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: 12/20/2024] [Revised: 02/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Colorectal cancer is a major global health burden, with significant heterogeneity in clinical outcomes among patients. Identifying robust prognostic gene expression signatures can help stratify patients, guide treatment decisions, and improve clinical management. This review provides an overview of current prognostic gene expression profiles in colorectal cancer research. We have synthesized evidence from numerous published studies investigating the association between tumor gene expression patterns and patient survival outcomes. The reviewed literature reveals several promising gene signatures that have demonstrated the ability to predict disease-free survival and overall survival in CRC patients, independent of standard clinicopathological risk factors. These genes are crucial in fundamental biological processes, including cell cycle control, epithelial-mesenchymal transition, and immune regulation. The implementation of prognostic gene expression tests in clinical practice holds great potential for enabling more personalized management strategies for colorectal cancer.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Lalji Baldaniya
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences, Marwadi University, Rajkot 360003 Gujarat, India.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India.
| | - Raghav Vashishth
- Department of Surgery, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India.
| | - Manisha Parmar
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India.
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar 64001, Iraq.
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq.
| | | |
Collapse
|
2
|
Tsukamoto S. Natural products that target p53 for cancer therapy. J Nat Med 2025:10.1007/s11418-025-01906-6. [PMID: 40295432 DOI: 10.1007/s11418-025-01906-6] [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: 03/17/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025]
Abstract
Wild-type p53 acts as a tumor suppressor, but p53 is frequently mutated and inactivated in tumor cells, promoting cancer progression, invasion, and metastasis. Thus, compounds that reactivate p53 may be leveraged for cancer treatment, and the development of drugs targeting p53 reactivation is actively progressing. Notably, natural products exhibit diverse structures and biological activities and are used as therapeutic agents for various diseases worldwide. This review discusses the natural products that inhibit p53 degradation through p53-Mdm2 interaction, promote p53 reactivation by inducing conformational changes, and exhibit p53-dependent growth inhibition.
Collapse
Affiliation(s)
- Sachiko Tsukamoto
- Department of Natural Medicines, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-Honmachi, Kumamoto, 862-0973, Japan.
| |
Collapse
|
3
|
Gu W. A bibliometric analysis of programmed cell death in oral cancer literature: research patterns and emerging trends (2000-2024). Discov Oncol 2025; 16:585. [PMID: 40261469 PMCID: PMC12014878 DOI: 10.1007/s12672-025-02410-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Programmed cell death (PCD) plays a crucial role in oral cancer pathogenesis and treatment. However, a comprehensive bibliometric analysis of the global research landscape in this field has not been conducted. This study aims to analyze the evolution and current trends of PCD research in oral cancer from 2000 to 2024. METHODS Publications were retrieved from the Web of Science Core Collection database using relevant keywords related to oral cancer and PCD. VOSviewer 1.6.20 and CiteSpace 6.1R6 software were employed to conduct bibliometric analysis, including publication trends, citation analysis, co-authorship networks, keyword co-occurrence, and research hotspots. The time span was set from January 2000 to December 2024. RESULTS A total of 963 publications were identified and analyzed. The annual publication output showed a steady increase, with a significant growth rate after 2010, dividing the study period into three distinct phases. The most productive countries were China (58.42%), South Korea (12.27%), and Japan (10.04%), with China Medical University and Kaohsiung Medical University being the leading institutions. Research hotspots evolved from traditional apoptosis studies to emerging forms of PCD such as autophagy, ferroptosis, and pyroptosis. Keyword analysis revealed three major research clusters: basic molecular mechanisms (centered around ROS and oxidative stress), clinical aspects (including prognosis and cell proliferation), and cell death pathways. Citation burst analysis identified emerging trends in targeting multiple PCD pathways simultaneously for oral cancer therapy, with special focus on treatment resistance and survival. CONCLUSION This bibliometric analysis provides a comprehensive overview of global research trends in PCD and oral cancer over the past two decades. The findings highlight the shift from basic mechanistic studies focusing on apoptosis to more diverse PCD pathways and translational research. Emerging research directions include the exploration of synergistic mechanisms among multiple PCD pathways, development of AI-based personalized treatment plans, investigation of microenvironment regulation of PCD, and application of novel drug delivery systems. These trends demonstrate the field's evolution toward more integrated, personalized approaches in oral cancer treatment. This study offers valuable insights for researchers and funding agencies to identify research gaps and potential collaboration opportunities in this rapidly developing field.
Collapse
Affiliation(s)
- Wenli Gu
- Stomatological Hospital, School of Stomatology, Southern Medical University, S366 Jiangnan Boulevard, Haizhu District, Guangzhou, Guangdong, China.
| |
Collapse
|
4
|
Zhou E, Yang JI, Habowski AN, Deschênes A, Belleau P, Ha T, Tzanavaris CJ, Boyd J, Hollweg CA, Zhu X, Tuveson DA, King DA. GATA6 Amplification Is Associated With Improved Survival in TP53-Mutated Unresectable Pancreatic Cancer. Pancreas 2025; 54:e303-e309. [PMID: 40262102 DOI: 10.1097/mpa.0000000000002431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/01/2024] [Indexed: 04/24/2025]
Abstract
OBJECTIVES GATA6 expression is recognized as a favorable prognostic marker of pancreatic cancer, whereas TP53 is a poor prognostic marker. We evaluated treatment outcomes by genetic alterations in TP53 and GATA6 to determine the prognostic and predictive impact of co-alterations. MATERIALS AND METHODS A single institution retrospective analysis was performed on patients diagnosed with pancreatic ductal adenocarcinoma between 2014 and 2023. TP53 genotype and GATA6 amplification status were included in an analysis of overall survival (OS) and progression-free survival (PFS). Previously published patient-derived organoids were used to investigate correlation between genetic status and drug sensitivity. RESULTS Patients with TP53 mutations had worse OS compared with the wild-type TP53 population. Patients with GATA6 amplification had better OS and a trend toward better PFS than the nonamplified population. Among patients with a TP53 mutation, patients with GATA6 co-alteration had longer OS compared with those who were not GATA6 amplified. In contrast, among patients who were TP53 wild-type, the presence or absence of a GATA6 amplification did not impact OS or PFS. GATA6 genotype was not associated chemotherapy drug response in an organoid pharmacotyping model. CONCLUSIONS We found that GATA6 amplification appeared to attenuate poor prognosis observed in TP53-mutant patients regardless of the type of standard chemotherapy received, suggesting the GATA6 amplification is a prognostic biomarker but not a predictive biomarker of standard-of-care chemotherapy.
Collapse
Affiliation(s)
- Edward Zhou
- Northwell Health Cancer Institute, New York, NY
| | | | | | | | - Pascal Belleau
- Cancer Center, Cold Spring Harbor Laboratory, New York, NY
| | - Taehoon Ha
- Cancer Center, Cold Spring Harbor Laboratory, New York, NY
| | - Chris J Tzanavaris
- Division of Pulmonary, Critical Care, and Sleep Medicine, Northwell Health, New York, NY
| | - Jeff Boyd
- Northwell Health Cancer Institute, New York, NY
| | - Christopher A Hollweg
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, NY
| | - Xinhua Zhu
- Northwell Health Cancer Institute, New York, NY
| | | | | |
Collapse
|
5
|
Agrawal R, Sengupta S. p53 regulates DREAM complex-mediated repression in a p21-independent manner. EMBO J 2025; 44:2279-2297. [PMID: 40038454 PMCID: PMC12000331 DOI: 10.1038/s44318-025-00402-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 01/18/2025] [Accepted: 02/10/2025] [Indexed: 03/06/2025] Open
Abstract
The DREAM repressor complex regulates genes involved in the cell cycle and DNA repair, vital for maintaining genome stability. Although it mediates p53-driven repression through the canonical p53-p21-Rb axis, the potential for p53 to directly regulate DREAM targets independently of its transcriptional activity has not been explored. Here, we demonstrate that in asynchronously growing cells, p53 loss leads to greater de-repression of DREAM targets compared to p21 loss alone. Both wild-type and transactivation-deficient p53 mutants are capable of repressing DREAM targets, suggesting a transactivation-independent "non-canonical" repression mechanism. These p53 variants bind p130/p107, irrespective of their phosphorylation status, while cancer-associated p53 mutants disrupt DREAM complex function by sequestering E2F4. Re-ChIP analysis shows co-recruitment of p53 and E2F4 to known and newly identified DREAM target promoters, indicating direct repression of these targets by p53. These findings reveal a novel, transactivation-independent mechanism of p53-mediated repression, expanding our understanding of p53's tumor-suppressive functions and suggesting DREAM complex targeting as potential future avenues in cancer therapy.
Collapse
Affiliation(s)
- Ritu Agrawal
- Biotechnology Research and Innovation Council-National Institute of Immunology (BRIC-NII), Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Sagar Sengupta
- Biotechnology Research and Innovation Council-National Institute of Immunology (BRIC-NII), Aruna Asaf Ali Marg, New Delhi, 110067, India.
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics (BRIC-NIBMG), PO: NSS, Kalyani, 741251, India.
| |
Collapse
|
6
|
Kim M, Lee M, Lee A, Choi BO, Park WC, Kim SH, Lee J, Kang J. Correlating p53 immunostaining patterns with somatic TP53 mutation and functional properties of mutant p53 in triple-negative breast cancer. Histopathology 2025. [PMID: 40162573 DOI: 10.1111/his.15453] [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: 08/12/2024] [Revised: 01/21/2025] [Accepted: 03/15/2025] [Indexed: 04/02/2025]
Abstract
AIMS Immunohistochemical (IHC) staining of p53 is a potential marker for TP53 mutations in various cancers. However, criteria for predicting TP53 mutations in triple-negative breast cancer (TNBC) using p53 IHC staining are not yet established. We aim to correlate p53 IHC expression patterns with TP53 mutation status in TNBC. METHODS AND RESULTS A total of 113 TNBC cases were analysed for p53 IHC staining pattern and somatic TP53 mutation using whole-exome sequencing. Functional properties of TP53 mutations were determined using the National Cancer Institute (NCI) TP53 database. P53 IHC patterns were categorized as nuclear overexpression (n = 58), null pattern (n = 40), wildtype (n = 15), cytoplasmic (n = 5), and subclonal (n = 5). The cutoff for predictive p53 nuclear overexpression was determined to be 80%, which strongly correlated with TP53 mutations. Notably, p53 overexpression had a positive predictive value (PPV) of 83% for missense or in-frame mutations, while the null pattern showed a PPV of 85% for detecting nonsense, frameshift, or splicing mutations. P53 overexpression was significantly linked to missense mutations within the DNA-binding domain (DBD) exhibiting gain-of-function (GOF) or dominant-negative effect (DNE). Cases exhibiting cytoplasmic expression correlated with nonsense or frameshift mutations in the DBD, nuclear localization signal (NLS), or splice sites. Cases with subclonal p53 staining patterns were associated with TP53 mutations. CONCLUSION Our study proposes newly defined criteria for interpreting p53 immunostaining patterns in TNBC, potentially allowing for the prediction of TP53 mutation types and their functional implications.
Collapse
Affiliation(s)
- Meejeong Kim
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Miseon Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ahwon Lee
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Cancer Research Institute, The Catholic University of Korea, Seoul, Korea
| | - Byung-Ock Choi
- Department of Radiation Oncology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Woo-Chan Park
- Division of Breast Surgery, Department of Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung Hun Kim
- Department of Radiology, Seoul Saint Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jieun Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jun Kang
- Department of Hospital Pathology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
7
|
Wu H, Shang J, Bao Y, Liu H, Zhang H, Xiao Y, Li Y, Huang Z, Cheng X, Ma Z, Zhang W, Mo P, Wang D, Zhang M, Zhan Y. Identification of a novel prognostic marker ADGRG6 in pancreatic adenocarcinoma: multi-omics analysis and experimental validation. Front Immunol 2025; 16:1530789. [PMID: 40226617 PMCID: PMC11986822 DOI: 10.3389/fimmu.2025.1530789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/10/2025] [Indexed: 04/15/2025] Open
Abstract
Background Pancreatic adenocarcinoma (PAAD) ranks among the most lethal malignancies worldwide. Current treatment options have limited efficacy, underscoring the need for new therapeutic targets. Methods This study employed a multi-omics analytical framework to delve into the expression profiles and prognostic implications of ADGRG6 within the pan-cancer dataset of The Cancer Genome Atlas (TCGA) database, highlighting the prognostic value and potential carcinogenic role of ADGRG6 in PAAD, which was further validated using data from multiple PAAD cohorts in the Gene Expression Omnibus (GEO) database. To assess the role of ADGRG6 in the tumor microenvironment of PAAD, we evaluated immune infiltration using CIBERSORT, ssGSEA, xCell and Tracking Tumor Immunophenotype (TIP), and utilized single-cell sequencing data to explore cell-cell interactions. Further cellular and animal experiments, such as colony formation assay, transwell assay, western blot, real-time PCR, and tumor xenograft experiments, were used to investigate the effect of ADGRG6 on the proliferation, metastatic potential and immune marker expression of PAAD and the underlying mechanisms. Results ADGRG6 emerged as a potential prognostic biomarker and a therapeutic target for PAAD, which was further corroborated by data extracted from multiple PAAD cohorts archived in the GEO database. Single-cell sequencing and immune infiltration analyses predicted the positive correlation of ADGRG6 with the infiltration of immune cells and with the interaction between malignant cells and fibroblasts/macrophages within the PAAD microenvironment. In vitro cell assays demonstrated that ADGRG6 promoted the proliferation, metastatic potential and immune marker expression of PAAD cells by increasing protein level of mutated p53 (mutp53), which activated a spectrum of gain-of-functions to promote cancer progression via the EGFR, AMPK and NF-κB signaling cascades. Furthermore, subcutaneous xenograft experiments in mice demonstrated that ADGRG6 knockdown substantially suppressed the growth of engrafted PAAD tumors. Conclusions ADGRG6 may serve as a novel prognostic marker and a therapeutic targets for PAAD, playing a crucial role in the proliferation, metastasis, and immune marker regulation of PAAD through elevating protein level of mutated p53.
Collapse
Affiliation(s)
- Han Wu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Jin Shang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yuanyan Bao
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Huajie Liu
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Haoran Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Yaosheng Xiao
- Department of Infectious Disease, Xiang’an Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| | - Yangtaobo Li
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Zhaozhang Huang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xiaolei Cheng
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Zixuan Ma
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wenqing Zhang
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Pingli Mo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Science, Xiamen University, Xiamen, Fujian, China
| | - Daxuan Wang
- Provincial College of Clinical Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, China
| | - Mingqing Zhang
- Department of Gastroenterology, The 909th Hospital, School of Medicine, Xiamen University, Zhangzhou, Fujian, China
| | - Yanyan Zhan
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
8
|
Ye Z, Yuan J, Hong D, Xu P, Liu W. Multimodal diagnostic models and subtype analysis for neoadjuvant therapy in breast cancer. Front Immunol 2025; 16:1559200. [PMID: 40170854 PMCID: PMC11958217 DOI: 10.3389/fimmu.2025.1559200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 02/26/2025] [Indexed: 04/03/2025] Open
Abstract
Background Breast cancer, a heterogeneous malignancy, comprises multiple subtypes and poses a substantial threat to women's health globally. Neoadjuvant therapy (NAT), administered prior to surgery, is integral to breast cancer treatment strategies. It aims to downsize tumors, optimize surgical outcomes, and evaluate tumor responsiveness to treatment. However, accurately predicting NAT efficacy remains challenging due to the disease's complexity and the diverse responses across different molecular subtypes. Methods In this study, we harnessed multimodal data, including proteomic, genomic, MRI imaging, and clinical information, sourced from multiple cohorts such as I-SPY2, TCGA-BRCA, GSE161529, and METABRIC. Post data preprocessing, Lasso regression was utilized for feature extraction and selection. Five machine learning algorithms were employed to construct diagnostic models, with pathological complete response (pCR) as the predictive endpoint. Results Our results revealed that the multi-omics Ridge regression model achieved the optimal performance in predicting pCR, with an AUC of 0.917. Through unsupervised clustering using the R package MOVICS and nine clustering algorithms, we identified four distinct multimodal breast cancer subtypes associated with NAT. These subtypes exhibited significant differences in proteomic profiles, hallmark cancer gene sets, pathway activities, tumor immune microenvironments, transcription factor activities, and clinical characteristics. For instance, CS1 subtype, predominantly ER-positive, had a low pCR rate and poor response to chemotherapy drugs, while CS4 subtype, characterized by high immune infiltration, showed a better response to immunotherapy. At the single-cell level, we detected significant heterogeneity in the tumor microenvironment among the four subtypes. Malignant cells in different subtypes displayed distinct copy number variations, differentiation levels, and evolutionary trajectories. Cell-cell communication analysis further highlighted differential interaction patterns among the subtypes, with implications for tumor progression and treatment response. Conclusion Our multimodal diagnostic model and subtype analysis provide novel insights into predicting NAT efficacy in breast cancer. These findings hold promise for guiding personalized treatment strategies. Future research should focus on experimental validation, in-depth exploration of the underlying mechanisms, and extension of these methods to other cancers and treatment modalities.
Collapse
Affiliation(s)
- Zheng Ye
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, China
| | - Jiaqi Yuan
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Deqing Hong
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| | - Peng Xu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
- School of Computer Science of Information Technology, Qiannan Normal University for Nationalities, Duyun, Guizhou, China
| | - Wenbin Liu
- Institute of Computational Science and Technology, Guangzhou University, Guangzhou, China
| |
Collapse
|
9
|
Thoenen E, Ranjan A, Parrales A, Nishikawa S, Dixon DA, Oka S, Iwakuma T. Suppression of stress granule formation is a vulnerability imposed by mutant p53. Nat Commun 2025; 16:2365. [PMID: 40064891 PMCID: PMC11894096 DOI: 10.1038/s41467-025-57539-6] [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: 02/02/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Missense mutations in the TP53 (p53) gene have been linked to malignant progression. However, our in-silico analyses reveal that hepatocellular carcinoma (HCC) patients with mutant p53 (mutp53) have better overall survival compared to those with p53-null (p53null) HCC, unlike other cancer types. Given the historical use of sorafenib (SOR) monotherapy for advanced HCC, we hypothesize that mutp53 increases sensitivity to SOR, a multikinase inhibitor that induces endoplasmic reticulum (ER) stress. Here we show that mutp53 inhibits stress granule (SG) formation by binding to an ER stress sensor, PKR-like ER kinase (PERK), and a key SG component, GAP SH3 domain-binding protein 1 (G3BP1), contributing to increased sensitivity of SG-competent cells and xenografts to ER stress inducers including SOR. Our study identifies a unique vulnerability imposed by mutp53, suggesting mutp53 as a biomarker for ER stress-inducing agents and highlighting the importance of SG inhibition for cancer treatment.
Collapse
Affiliation(s)
- Elizabeth Thoenen
- Department of Pediatrics, Division of Hematology & Oncology, Children's Mercy Research Institute, Kansas City, MO, USA
| | - Atul Ranjan
- Department of Pediatrics, Division of Hematology & Oncology, Children's Mercy Research Institute, Kansas City, MO, USA
| | - Alejandro Parrales
- Department of Pediatrics, Division of Hematology & Oncology, Children's Mercy Research Institute, Kansas City, MO, USA
| | - Shigeto Nishikawa
- Department of Pediatrics, Division of Hematology & Oncology, Children's Mercy Research Institute, Kansas City, MO, USA
| | - Dan A Dixon
- Department of Biochemistry and Molecular Biology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sugako Oka
- Faculty of Medical Science, Kyushu University, Fukuoka, Japan
| | - Tomoo Iwakuma
- Department of Pediatrics, Division of Hematology & Oncology, Children's Mercy Research Institute, Kansas City, MO, USA.
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
10
|
Hwang J, Likasitwatanakul P, Deshmukh SK, Wu S, Kwon JJ, Toye E, Moline D, Evans MG, Elliott A, Passow R, Luo C, John E, Gandhi N, McKay RR, Heath EI, Nabhan C, Reizine N, Orme JJ, Domingo Domenech JM, Sartor O, Baca SC, Dehm SM, Antonarakis ES. Structurally Oriented Classification of FOXA1 Alterations Identifies Prostate Cancers with Opposing Clinical Outcomes and Distinct Molecular and Immunologic Subtypes. Clin Cancer Res 2025; 31:936-948. [PMID: 39745364 PMCID: PMC11873805 DOI: 10.1158/1078-0432.ccr-24-3471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/06/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025]
Abstract
PURPOSE Around 10% to 15% of prostate cancers harbor recurrent aberrations in the Forkhead Box A1 gene, FOXA1, whereby the alteration type and the effect on the forkhead (FKH) domain affect protein function. We developed a FOXA1 classification system to inform clinical management. EXPERIMENTAL DESIGN A total of 5,014 prostate cancer samples were examined using whole-exome and -transcriptome sequencing from the Caris Life Sciences database. We denoted class 1 FOXA1 alterations as missense and in-frame insertions/deletions with subclasses oriented with respect to the FKH domain. These were in the first part of the FKH domain [class 1A: amino acids (AA) 168-246], within the Wing2 region of FKH (class 1B: AA 247-269), or outside FKH (class 1C: AA 1-167, 270+). Two hotspot missense mutations at R219 were denoted class 2. Class 3 included predicted truncating mutations with subclasses partitioned based on the FKH domain (class 3A: AA 1-269 and class 3B: AA 270+). Class 4 represented FOXA1 amplifications. Real-world overall survival and therapy outcomes were determined from insurance claims. RESULTS FOXA1 alterations did not influence survival when considered in aggregate but had distinct prognostic effects when stratified by class. In primary prostate samples, class 1A alterations were associated with overall improved survival (HR, 0.57; P = 0.03); a similar trend was seen in metastatic biopsies with class 1B (HR, 0.84; P = 0.09). Conversely, in primary specimens, class 1C exhibited worse survival upon second-generation androgen receptor signaling inhibitor treatment (HR, 1.93; P < 0.001). Class 2 mutations (R219C/S) were enriched in neuroendocrine prostate cancers and were associated with overall poor survival (HR, 2.05; P < 0.001) and worse outcomes to first-line androgen-deprivation therapies (HR, 2.5; P < 0.001). Class 3A alterations indicated improved survival (HR, 0.70; P = 0.01), whereas class 3B alterations portended poor outcomes (HR, 1.50; P < 0.001). Amplifications (class 4) indicated poor outcomes in metastatic samples (HR, 1.48; P = 0.02). Molecularly, different FOXA1 alteration classes harbored distinct mutational and immunologic features as well as unique transcriptional programs. Finally, relative to European Americans, African Americans had increased class 1C alterations, whereas Asian/Pacific Islander patients had increased class 1B alterations. CONCLUSIONS FOXA1 alterations should not be interpreted in aggregate, as different classes are associated with divergent molecular features and clinical outcomes. Our revised classification schema facilitates clinical decision-making for patients with prostate cancer and uncovers important racial differences.
Collapse
Affiliation(s)
- Justin Hwang
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Pornlada Likasitwatanakul
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
- Dana Farber Cancer Institute, Boston, Massachusetts
- Department of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - Sharon Wu
- Department of Medical Affairs, CarisLifeSciences, Irving, Texas
| | - Jason J. Kwon
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | - Eamon Toye
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - David Moline
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Mark G. Evans
- Department of Medical Affairs, CarisLifeSciences, Irving, Texas
| | - Andrew Elliott
- Department of Medical Affairs, CarisLifeSciences, Irving, Texas
| | - Rachel Passow
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Christine Luo
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Emily John
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Nishant Gandhi
- Department of Medical Affairs, CarisLifeSciences, Irving, Texas
| | - Rana R. McKay
- University of California San Diego, San Diego, California
| | | | - Chadi Nabhan
- Department of Medical Affairs, CarisLifeSciences, Irving, Texas
| | | | | | | | | | | | - Scott M. Dehm
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Emmanuel S. Antonarakis
- Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota
- Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| |
Collapse
|
11
|
Alshammari QA, Alshammari SO, Alshammari A, Alfarhan M, Baali FH. Unraveling the mechanisms of glioblastoma's resistance: investigating the influence of tumor suppressor p53 and non-coding RNAs. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2569-2585. [PMID: 39476245 DOI: 10.1007/s00210-024-03564-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/22/2024] [Indexed: 01/04/2025]
Abstract
Glioblastoma (GB) is one of the most fatal CNS malignancies, and its high resistance to therapy and poor outcomes have made it one of the primary challenges in oncology. Resistance to standard therapy, i.e., radio-chemotherapy with temozolomide, is one of the principal causes of the poor prognostic outcomes of GB. Finding the molecular basis of GB resistance to therapy is key to creating effective solution approaches. The general problem of GB resistance is supervised by cancer suppressive protein, p53, and has become a very special interest in molecular research in recent decades. The principal aim of this manuscript is to perform a comprehensive survey on the complex network of interactions developed by p53 with non-coding RNAs (ncRNA) in the context of GB resistance. The present article details the functional aspects of p53 as a cellular stress response protein, including its roles in apoptosis, cell cycle regulation, and DNA repair in glioblastoma (GB), along with the disruption of p53 and its involvement in chemoresistance (CR). It also highlights several classes of ncRNAs, namely microRNAs, long ncRNAs, and circular RNAs, that manipulate p53 signaling in GB-CR. The article likewise explains how disruption in the expression of these ncRNAs can promote GB-CR and how it interacts with essential cellular functions, such as proliferation, apoptosis, and DNA repair. The manuscript also describes the potential of targeting p53 and ncRNAs with their diagnostic and prognostic potential as novel promising therapeutics for GB. Nevertheless, ncRNA-based biomarkers still present challenges for their suitability in GB resistance. However, modern research continues to discover novel prediction targets, potentially enhancing patient outcomes and therapeutic options. Therefore, the neutralization of this intricate regulatory network of GB resistance might have a primary clinical effect in fighting GB resistance therapy and thus might lead to a substantial increase in patient survival and quality of life.
Collapse
Affiliation(s)
- Qamar A Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| | - Saud O Alshammari
- Department of Pharmacognosy and Alternative Medicine, College of Pharmacy, Northern Border University, 76321, Rafha, Saudi Arabia
| | - Abdulkarim Alshammari
- Department of Pharmacy Practice, College of Pharmacy, Northern Border University, Rafha, Saudi Arabia
| | - Moaddey Alfarhan
- Department of Clinical Practice, College of Pharmacy, Jazan University, 45142, Jazan, Jizan, Saudi Arabia
| | - Fahad Hassan Baali
- Department of Clinical Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| |
Collapse
|
12
|
Sadahiro Y, Okubo M, Hitora Y, Hitora-Imamura N, Kotani S, Tsukamoto S. Pestones A and B from a Fungus Pestalotiopsis sp. Bound to Mutant p53 and Changed Its Conformation. JOURNAL OF NATURAL PRODUCTS 2025; 88:546-553. [PMID: 39952908 DOI: 10.1021/acs.jnatprod.4c01440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2025]
Abstract
Oncogenic mutant p53 is one of the targets for cancer therapy, and the development of anticancer drugs that reactivate mutant p53 is a promising strategy. The extract of fungus Pestalotiopsis sp. changed mutant p53 to wild-type-like p53 in Saos-2 (p53R175H) cells, as shown by fluorescent immunostaining, and bioassay-guided purification of the extract afforded new dimeric epoxyquinoids, pestones A and B (1 and 2), and a known compound, rosnecatrone (3). The relative and absolute configurations of 1 and 2 were determined based on the spectroscopic data and semisynthesis from 3. Compounds 1 and 2 altered the conformation of mutant p53 in Saos-2 (p53R175H) cells, as shown by immunofluorescence staining. The cellular thermal shift assay analysis showed that 1 increased the thermostability of mutant p53 in Saos-2 (p53R175H) cells, suggesting the direct binding of 1 to mutant p53. Compounds 1 and 2 exhibited cytotoxic activities against Saos-2 (p53R175H) cells with IC50 values of 1.0 and 1.1 μM, respectively. Compound 1 was found to induce apoptosis in Saos-2 (p53R175H) cells by flow cytometry analysis and decreased tumor growth in vivo using a mouse model with HuCCT1 (p53R175H) cells.
Collapse
Affiliation(s)
- Yusaku Sadahiro
- Department of Natural Medicines, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Misaki Okubo
- Department of Natural Medicines, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Yuki Hitora
- Department of Natural Medicines, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Natsuko Hitora-Imamura
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Shunsuke Kotani
- Department of Organic Chemistry, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| | - Sachiko Tsukamoto
- Department of Natural Medicines, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan
| |
Collapse
|
13
|
Masuda Y, Sadato D, Toya T, Hosoda Y, Hirama C, Shimizu H, Najima Y, Harada H, Harada Y, Doki N. Transplantation outcomes of TP53-mutant AML and MDS: a single transplantation center experience of 63 patients. Int J Hematol 2025:10.1007/s12185-025-03951-z. [PMID: 40011351 DOI: 10.1007/s12185-025-03951-z] [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: 12/12/2024] [Revised: 02/13/2025] [Accepted: 02/13/2025] [Indexed: 02/28/2025]
Abstract
Allogeneic hematopoietic stem cell transplantation is recommended for TP53-mutant acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) despite a high relapse rate and poor survival. To understand TP53 alterations on a molecular level and define stratified prognostic outcomes following transplantation, we performed targeted next-generation sequencing on 63 patients who underwent transplantation for TP53-mutant AML/MDS and profiled their molecular spectrum. Sixty-eight TP53 mutations were detected, with a median variant allele frequency of 46.8%. Copy number alterations at the TP53 locus were present in 19 patients (30%). Complex karyotype was detected in 48 patients (76%) and was significantly associated with larger TP53 clone size, bi-allelic status, and the absence of concurrent mutations, reflecting the high TP53 mutational burden. Specifically, 51 patients (81%) with the dominant TP53 clone greatly overlapped with those with the complex karyotype. Multivariable overall survival (OS) analysis identified AML (hazard ratio [HR], 2.51; P = 0.03) and TP53 clonal dominance (HR, 5.30; P = 0.002) as prognostic factors. One-year OS was worse in AML with the dominant TP53 clone than in others (13% vs 61%; P < 0.001). Our results underscore the utility of mutational profile-guided risk stratification in patients with TP53-mutant AML/MDS, and could aid in transplantation-related decision-making.
Collapse
Affiliation(s)
- Yasutaka Masuda
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| | - Daichi Sadato
- Clinical Research and Trials Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Takashi Toya
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan.
| | - Yuzuru Hosoda
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| | - Chizuko Hirama
- Clinical Research and Trials Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Hiroaki Shimizu
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| | - Yuho Najima
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| | - Hironori Harada
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yuka Harada
- Clinical Research and Trials Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| |
Collapse
|
14
|
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
|
15
|
Meng F, Qi T, Liu X, Wang Y, Yu J, Lu Z, Cai X, Li A, Jung D, Duan J. Enhanced pharmacological activities of AKR1C3-activated prodrug AST-3424 in cancer cells with defective DNA repair. Int J Cancer 2025; 156:417-430. [PMID: 39243400 DOI: 10.1002/ijc.35170] [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: 03/01/2024] [Revised: 07/28/2024] [Accepted: 08/07/2024] [Indexed: 09/09/2024]
Abstract
AST-3424 is a novel and highly tumor-selective prodrug. AST-3424 is activated by AKR1C3 to release a toxic bis-alkylating moiety, AST 2660. In this study, we have investigated the essential role of DNA repair in AST-3424 mediated pharmacological activities in vitro and in vivo. We show here that AST-3424 is effective as a single therapeutic agent against cancer cells to induce cytotoxicity, DNA damage, apoptosis and cell cycle arrest at G2 phase in a dose- and AKR1C3-dependent manner in both p53-proficient H460 (RRID:CVCL_0459) and p53-deficient HT-29 cells (RRID:CVCL_0320). The combination of abrogators of G2 checkpoint with AST-3424 was only synergistic in HT-29 but not in H460 cells. The enhanced activity of AST-3424 in HT-29 cells was due to impaired DNA repair ability via the attenuation of cell cycle G2 arrest and reduced RAD51 expression. Furthermore, we utilized a BRCA2 deficient cell line and two PDX models with BRCA deleterious mutations to study the increased activity of AST-3424. The results showed that AST-3424 exhibited enhanced in vitro cytotoxicity and superior and durable in vivo anti-tumor effects in cells deficient of DNA repair protein BRCA2. In summary, we report here that when DNA repair capacity is reduced, the in vitro and in vivo activity of AST-3424 can be further enhanced, thus providing supporting evidence for the further evaluation of AST-3424 in the clinic.
Collapse
Affiliation(s)
- Fanying Meng
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Tianyang Qi
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Xing Liu
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Yizhi Wang
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Jibing Yu
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Zhaoqiang Lu
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Xiaohong Cai
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Anrong Li
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Don Jung
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| | - Jianxin Duan
- Ascentawits Pharmaceuticals, LTD, Shenzhen, China
| |
Collapse
|
16
|
Tornesello ML. TP53 mutations in cancer: Molecular features and therapeutic opportunities (Review). Int J Mol Med 2025; 55:7. [PMID: 39450536 PMCID: PMC11554381 DOI: 10.3892/ijmm.2024.5448] [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: 07/10/2024] [Accepted: 09/03/2024] [Indexed: 10/26/2024] Open
Abstract
The tumour suppressor factor p53 plays an essential role in regulating numerous cellular processes, including the cell cycle, DNA repair, apoptosis, autophagy, cell metabolism and immune response. TP53 is the most commonly mutated gene in human cancers. These mutations are primarily non‑synonymous changes that produce mutant p53 proteins characterized by loss of function, a dominant negative effect on p53 tetramerisation and gain of function (GOF). GOF mutations not only disrupt the tumour‑suppressive activities of p53 but also endow the mutant proteins with new oncogenic properties. Recent studies analysing different pathogenic features of mutant p53 in cancer‑derived cell lines have demonstrated that restoring wild‑type p53, rather than removing GOF mutations, reduces cancer cell growth. These findings suggest that therapeutic strategies for reactivating wild‑type p53 function in cancer cells may bring a greater benefit than approaches halting mutant p53. This approach could involve the use of small molecules, gene therapy and other methods to re‑establish wild‑type p53 activity. This review describes the complexity of the biological activities of different p53 mutants and summarizes the current therapeutic approaches to restore p53 function.
Collapse
Affiliation(s)
- Maria Lina Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, I-80131 Napoli, Italy
| |
Collapse
|
17
|
Jamialahmadi K, Noruzi S. Matrix metalloproteinases, chemoresistance and cancer. PATHOPHYSIOLOGICAL ASPECTS OF PROTEASES IN CANCER 2025:385-409. [DOI: 10.1016/b978-0-443-30098-1.00023-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
18
|
Malhotra L, Singh A, Kaur P, Ethayathulla AS. Phenotypical mapping of TP53 unique missense mutations spectrum in human cancers. J Biomol Struct Dyn 2024:1-14. [PMID: 39639563 DOI: 10.1080/07391102.2024.2435060] [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: 01/11/2024] [Accepted: 04/04/2024] [Indexed: 12/07/2024]
Abstract
The p53 tumor suppressor is one of the most mutated genes responsible for tumorigenesis in most human cancers. Out of 29,891 genomic mutations reported in the TP53 Database (https://tp53.isb-cgc.org/), 1,297 are identified as unique missense somatic mutations excluding frameshift, intronic, deletion, nonsense, silent, splice, and other unknown mutations. We have comprehensively analyzed all these 1,297 unique missense mutations and created a phenotypical map based on the distribution of mutations in each domain, the functional state of the protein, and their occurrence in different types of tissues and organs. Our mutation map shows that almost 118 unique missense mutations are reported in the transactivation and proline-rich domains, 1,065 in the central DNA-binding domains, and 113 in the oligomerization and regulatory domains. Based on the phenotype, these mutations are subdivided into 46 super trans, 491 functional, 315 partially functional, and 415 non-functional mutations. The prevalence of these mutations was checked in 71 different types of tissues and found that the mutant R248Q is reported in 51 types of tissues followed by R175H and R273H in 46 types. We correlated the potential impact of mutation in target gene transcription and regulation with nucleosomal DNA and RNA-Pol II complexes. We have discussed the impact of mutation at post-translational modification sites in the structure and function of p53 highlighting the potential therapeutic drug targets with tremendous clinical applications.
Collapse
Affiliation(s)
- Lakshay Malhotra
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
- Department of Biochemistry, Sri Venkateswara College, University of Delhi, New Delhi, India
| | - Alankrita Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| | - Abdul S Ethayathulla
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
19
|
Pervushin NV, Nilov DK, Pushkarev SV, Shipunova VO, Badlaeva AS, Yapryntseva MA, Kopytova DV, Zhivotovsky B, Kopeina GS. BH3-mimetics or DNA-damaging agents in combination with RG7388 overcome p53 mutation-induced resistance to MDM2 inhibition. Apoptosis 2024; 29:2197-2213. [PMID: 39222276 PMCID: PMC11550243 DOI: 10.1007/s10495-024-02014-8] [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] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
The development of drug resistance reduces the efficacy of cancer therapy. Tumor cells can acquire resistance to MDM2 inhibitors, which are currently under clinical evaluation. We generated RG7388-resistant neuroblastoma cells, which became more proliferative and metabolically active and were less sensitive to DNA-damaging agents in vitro and in vivo, compared with wild-type cells. The resistance was associated with a mutation of the p53 protein (His193Arg). This mutation abated its transcriptional activity via destabilization of the tetrameric p53-DNA complex and was observed in many cancer types. Finally, we found that Cisplatin and various BH3-mimetics could enhance RG7388-mediated apoptosis in RG7388-resistant neuroblastoma cells, thereby partially overcoming resistance to MDM2 inhibition.
Collapse
Affiliation(s)
- N V Pervushin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, 119991, Russia
| | - D K Nilov
- Belozersky Institute of Physicochemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - S V Pushkarev
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119234, Russia
| | - V O Shipunova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- Moscow Center for Advanced Studies, Moscow, 123592, Russia
| | - A S Badlaeva
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Russian Ministry of Health, Moscow, 117513, Russia
| | - M A Yapryntseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, 119991, Russia
| | - D V Kopytova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - B Zhivotovsky
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, 119991, Russia.
- Division of Toxicology, Institute of Environmental Medicine, Karolinska Institutet, Box 210, 17177, Stockholm, Sweden.
| | - G S Kopeina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia.
- Faculty of Medicine, MV Lomonosov Moscow State University, Moscow, 119991, Russia.
| |
Collapse
|
20
|
Zhang H, Xu J, Long Y, Maimaitijiang A, Su Z, Li W, Li J. Unraveling the Guardian: p53's Multifaceted Role in the DNA Damage Response and Tumor Treatment Strategies. Int J Mol Sci 2024; 25:12928. [PMID: 39684639 DOI: 10.3390/ijms252312928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/21/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
DNA damage can lead to mutations that can alter the function of oncogenes or tumor suppressor genes, thus promoting the development of cancer. p53 plays a multifaceted and complex role in the DNA damage response and cancer progression and is known as the 'guardian of the gene'. When DNA damage occurs, p53 is activated through a series of post-translational modifications, which stabilize the protein and enhance its function as a transcription factor. It regulates processes including cell cycle checkpoints, DNA repair and apoptosis, thereby preventing the spread of damaged DNA and maintaining genome integrity. On the one hand, p53 can initiate cell cycle arrest and induce cells to enter the G1/S and G2/M checkpoints, preventing cells with damaged DNA from continuing to proliferate and gaining time for DNA repair. At the same time, p53 can promote the activation of DNA repair pathways, including base excision repair, nucleotide excision repair and other repair pathways, to ensure the integrity of genetic material. If the damage is too severe to repair, p53 will trigger the apoptosis process to eliminate potential cancer risks in time. p53 also plays a pivotal role in cancer progression. Mutations in the p53 gene are frequently found in many cancers, and the mutated p53 not only loses its normal tumor suppressor function but may even acquire pro-cancer activity. Therefore, we also discuss therapeutic strategies targeting the p53 pathway, such as the use of small-molecule drugs to restore the function of wild-type p53, the inhibition of negative regulatory factors and synthetic lethality approaches for p53-deficient tumors. This review therefore highlights the important role of p53 in maintaining genomic stability and its potential in therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Han Zhang
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Jianxiong Xu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Yuxuan Long
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Ayitila Maimaitijiang
- School of Pharmaceutical Science, Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| | - Zhengding Su
- School of Pharmaceutical Science, Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| | - Wenfang Li
- School of Pharmaceutical Science, Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| |
Collapse
|
21
|
Luo S, Xu J, Mo C, Gong W, Li C, Hou X, Ou M. High-throughput sequencing reveals twelve cell death pattern prognostic target genes as potential drug-response-associated genes in the treatment of colorectal cancer cells with palmatine hydrochloride. ONCOLOGIE 2024. [DOI: 10.1515/oncologie-2024-0378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Abstract
Objectives
Palmatine Hydrochloride (PaH), an isoquinoline alkaloid from Phellodendron amurense and Coptis chinensis, has analgesic, anti-inflammatory, and anticancer properties. This study aimed to assess PaH’s effectiveness against SW480 colorectal cancer (CRC) cells and explore its molecular mechanisms.
Methods
PaH’s effects on SW480 CRC cells were evaluated using MTT assays for proliferation, scratch assays for migration, and flow cytometry for apoptosis. Differentially expressed genes (DEGs) were identified through high-throughput sequencing. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses assessed DEG roles. Prognostic significance related to programmed cell death (PCD) was analyzed using R-Package with TCGA data. RT-qPCR validated key genes identified.
Results
PaH significantly inhibited SW480 cell growth, invasion, and apoptosis. The MTT assay showed inhibition rates increased from 5.49 % at 25 μg/mL to 52.48 % at 400 μg/mL. Scratch assays indicated reduced cell invasion over 24, 48, and 72 h. Apoptosis rose from 12.36 % in controls to 45.54 % at 400 μg/mL. Sequencing identified 3,385 significant DEGs, primarily in cancer pathways (p=0.004). Among 35 PCD-related DEGs, Lasso Cox regression highlighted 12 key genes, including TERT, TGFBR1, WNT4, and TP53. RT-qPCR confirmed TERT and TGFBR1 downregulation (0.614-fold, p=0.008; 0.41-fold, p<0.001) and TP53 and WNT4 upregulation (5.634-fold, p<0.001; 5.124-fold, p=0.002).
Conclusions
PaH inhibits CRC cell proliferation, migration, and invasion by modulating key PCD genes, suggesting its potential as a CRC therapeutic agent.
Collapse
Affiliation(s)
- Sha Luo
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Jiajun Xu
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Chune Mo
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Weiwei Gong
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Chunhong Li
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Xianliang Hou
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| | - Minglin Ou
- Laboratory Center, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
- Laboratory Center, Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases , The Second Affiliated Hospital of Guilin Medical University , Guilin, 541199 , China
| |
Collapse
|
22
|
Advani D, Kumar P. Uncovering Cell Cycle Dysregulations and Associated Mechanisms in Cancer and Neurodegenerative Disorders: A Glimpse of Hope for Repurposed Drugs. Mol Neurobiol 2024; 61:8600-8630. [PMID: 38532240 DOI: 10.1007/s12035-024-04130-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
The cell cycle is the sequence of events orchestrated by a complex network of cell cycle proteins. Unlike normal cells, mature neurons subsist in a quiescent state of the cell cycle, and aberrant cell cycle activation triggers neuronal death accompanied by neurodegeneration. The periodicity of cell cycle events is choreographed by various mechanisms, including DNA damage repair, oxidative stress, neurotrophin activity, and ubiquitin-mediated degradation. Given the relevance of cell cycle processes in cancer and neurodegeneration, this review delineates the overlapping cell cycle events, signaling pathways, and mechanisms associated with cell cycle aberrations in cancer and the major neurodegenerative disorders. We suggest that dysregulation of some common fundamental signaling processes triggers anomalous cell cycle activation in cancer cells and neurons. We discussed the possible use of cell cycle inhibitors for neurodegenerative disorders and described the associated challenges. We propose that a greater understanding of the common mechanisms driving cell cycle aberrations in cancer and neurodegenerative disorders will open a new avenue for the development of repurposed drugs.
Collapse
Affiliation(s)
- Dia Advani
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), Shahbad Daulatpur, Bawana Road, New Delhi, Delhi, 110042, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly Delhi College of Engineering), Shahbad Daulatpur, Bawana Road, New Delhi, Delhi, 110042, India.
| |
Collapse
|
23
|
Perez LM, Venugopal SV, Martin AS, Freedland SJ, Di Vizio D, Freeman MR. Mechanisms governing lineage plasticity and metabolic reprogramming in cancer. Trends Cancer 2024; 10:1009-1022. [PMID: 39218770 DOI: 10.1016/j.trecan.2024.08.001] [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: 04/19/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024]
Abstract
Dynamic alterations in cellular phenotypes during cancer progression are attributed to a phenomenon known as 'lineage plasticity'. This process is associated with therapeutic resistance and involves concurrent shifts in metabolic states that facilitate adaptation to various stressors inherent in malignant growth. Certain metabolites also serve as synthetic reservoirs for chromatin modification, thus linking metabolic states with epigenetic regulation. There remains a critical need to understand the mechanisms that converge on lineage plasticity and metabolic reprogramming to prevent the emergence of lethal disease. This review attempts to offer an overview of our current understanding of the interplay between metabolic reprogramming and lineage plasticity in the context of cancer, highlighting the intersecting drivers of cancer hallmarks, with an emphasis on solid tumors.
Collapse
Affiliation(s)
- Lillian M Perez
- Departments of Urology and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Smrruthi V Venugopal
- Departments of Urology and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Anna St Martin
- Departments of Urology and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Stephen J Freedland
- Departments of Urology and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dolores Di Vizio
- Department of Pathology and Laboratory Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Michael R Freeman
- Departments of Urology and Biomedical Sciences, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
24
|
Wang X, Yang J, Yang W, Sheng H, Jia B, Cheng P, Xu S, Hong X, Jiang C, Yang Y, Wu Z, Wang J. Multiple roles of p53 in cancer development: Regulation of tumor microenvironment, m 6A modification and diverse cell death mechanisms. J Adv Res 2024:S2090-1232(24)00481-8. [PMID: 39490612 DOI: 10.1016/j.jare.2024.10.026] [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: 07/16/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND The protein p53, encoded by the most frequently mutated gene TP53 in human cancers, has diverse functions in tumor suppression. As a best known transcription factor, p53 can regulate various fundamental cellular responses, ranging from the cell-cycle arrest, DNA repair, senescence to the programmed cell death (PCD), which includes autophagy, apoptosis, ferroptosis, cuproptosis, pyroptosis and disulfidoptosis. Accumulating evidence has indicated that the tumor microenvironment (TME), N6-methyladenosine (m6A) modification and diverse PCD are important for the progression, proliferation and metastases of cancers. AIM OF REVIEW This paper aims to systematically and comprehensively summarize the multiple roles of p53 in the development of cancers from the regulation of TME, m6A Modification and diverse PCD. KEY SCIENTIFIC CONCEPTS OF REVIEW TME, a crucial local homeostasis environment, influences every step of tumorigenesis and metastasis. m6A, the most prevalent and abundant endogenous modification in eukaryotic RNAs, plays an essential role in various biological processes, containing the progression of cancers. Additionally, PCD is an evolutionarily conserved mechanism of cell suicide and a common process in living organisms. Some forms of PCD contribute to the occurrence and development of cancer. However, the complex roles of p53 within the TME, m6A modification and diverse PCD mechanisms are still not completely understood. Presently, the function roles of p53 including the wild-type and mutant p53 in different context are summarized. Additionally, the interaction between the cancer immunity, cancer cell death and RNA m6A methylation and the p53 regulation during the development and progress of cancers were discussed. Moreover, the key molecular mechanisms by which p53 participates in the regulation of TME, m6A and diverse PCD are also explored. All the findings will facilitate the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Xiangyu Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Jianhua Yang
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Wanting Yang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Haiyang Sheng
- Global Biometrics and Data Sciences, Bristol Myers Squibb, New York City, USA
| | - Buyun Jia
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Peng Cheng
- The First Affiliated Hospital, Anhui University of Traditional Chinese Medicine, Hefei, Anhui, China
| | - Shanshan Xu
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Xinhui Hong
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Chuanwei Jiang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yinfeng Yang
- School of Medical Informatics Engineering, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| | - Ziyin Wu
- State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Jiangsu Kanion Pharmaceutical Co. Ltd, Lianyungang, Jiangsu, China.
| | - Jinghui Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| |
Collapse
|
25
|
Song J, Cui Q, Gao J. Roles of lncRNAs related to the p53 network in breast cancer progression. Front Oncol 2024; 14:1453807. [PMID: 39479021 PMCID: PMC11521785 DOI: 10.3389/fonc.2024.1453807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/30/2024] [Indexed: 11/02/2024] Open
Abstract
The p53 is a crucial tumor suppressor and transcription factor that participates in apoptosis and senescence. It can be activated upon DNA damage to regulate the expression of a series of genes. Previous studies have demonstrated that some specific lncRNAs are part of the TP53 regulatory network. To enhance our understanding of the relationship between lncRNAs and P53 in cancers, we review the localization, structure, and function of some lncRNAs that are related to the mechanisms of the p53 pathway or serve as p53 transcriptional targets.
Collapse
Affiliation(s)
| | - Qiuxia Cui
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Jidong Gao
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| |
Collapse
|
26
|
Guzman A, Kawase T, Devanny AJ, Efe G, Navaridas R, Yu K, Regunath K, Mercer IG, Avard RC, Muniz de Queiroz R, Rustgi AK, Kaufman LJ, Prives C. Mutant p53 regulates cancer cell invasion in complex three-dimensional environments through mevalonate pathway-dependent Rho/ROCK signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.618100. [PMID: 39464132 PMCID: PMC11507699 DOI: 10.1101/2024.10.13.618100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Certain mutations can confer neomorphic gain of function (GOF) activities to the p53 protein that affect cancer progression. Yet the concept of mutant p53 GOF has been challenged. Here, using various strategies to alter the status of mutant versions of p53 in different cell lines, we demonstrate that mutant p53 stimulates cancer cell invasion in three-dimensional environments. Mechanistically, mutant p53 enhances RhoA/ROCK-dependent cell contractility and cell-mediated extracellular matrix (ECM) re-organization via increasing mevalonate pathway-dependent RhoA localization to the membrane. In line with this, RhoA-dependent pro-invasive activity is also mediated by IDI-1, a mevalonate pathway product. Further, the invasion-enhancing effect of mutant p53 is dictated by the biomechanical properties of the surrounding ECM, thereby adding a cell-independent layer of regulation to mutant p53 GOF activity that is mediated by dynamic reciprocal cell-ECM interactions. Together our findings link mutant p53 metabolic GOF activity with an invasive cellular phenotype in physiologically relevant and context-dependent settings. Significance This study addresses the contribution of mutant p53 to the process of cancer cell dissemination in physiologically relevant three-dimensional environments - a key characteristic of metastatic disease. Several mutant p53 proteins display pro-oncogenic activity with respect to cancer cell invasion in 3D environments via mevalonate pathway-dependent Rho/ROCK signaling axis.
Collapse
|
27
|
Song G, Liu J, Tang X, Zhong J, Zeng Y, Zhang X, Zhou J, Zhou J, Cao L, Zhang Q, Li Y. Cell cycle checkpoint revolution: targeted therapies in the fight against malignant tumors. Front Pharmacol 2024; 15:1459057. [PMID: 39464635 PMCID: PMC11505109 DOI: 10.3389/fphar.2024.1459057] [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: 07/03/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
Malignant tumors are among the most important causes of death worldwide. The pathogenesis of a malignant tumor is complex and has not been fully elucidated. Studies have shown that such pathogenesis is related to abnormal cell cycle progression. The expression levels of cyclins, cyclin-dependent kinases (CDKs), and CDK inhibitors as well as functions of the cell cycle checkpoints determine whether the cell cycle progression is smooth. Cell-cycle-targeting drugs have the advantages of high specificity, low toxicity, low side effects, and low drug resistance. Identifying drugs that target the cell cycle and applying them in clinical treatments are expected to promote chemotherapeutic developments against malignant tumors. This article aims to review drugs targeted against the cell cycle and their action mechanisms.
Collapse
Affiliation(s)
- Guangming Song
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jue Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xing Tang
- Department of Assisted Reproductive Centre, The affiliated Zhuzhou hospital Xiangya medical college, Central South University, Zhuzhou, Hunan, China
| | - Jie Zhong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuhuan Zeng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaodi Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jianbin Zhou
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jie Zhou
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Lu Cao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Qunfeng Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yukun Li
- Department of Assisted Reproductive Centre, The affiliated Zhuzhou hospital Xiangya medical college, Central South University, Zhuzhou, Hunan, China
| |
Collapse
|
28
|
Lo CS, Alavi P, Bassey-Archibong B, Jahroudi N, Pasdar M. Differential effect of plakoglobin in restoring the tumor suppressor activities of p53-R273H vs. p53-R175H mutants. PLoS One 2024; 19:e0306705. [PMID: 39361615 PMCID: PMC11449273 DOI: 10.1371/journal.pone.0306705] [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: 02/19/2024] [Accepted: 06/21/2024] [Indexed: 10/05/2024] Open
Abstract
The six most common missense mutations in the DNA binding domain of p53 are known as "hot spots" and include two of the most frequently occurring p53 mutations (p53-R175H and p53-R273H). p53 stability and function are regulated by various post-translational modifications such as phosphorylation, acetylation, sumoylation, methylation, and interactions with other proteins including plakoglobin. Previously, using various carcinoma cell lines we showed that plakoglobin interacted with wild-type and several endogenous p53 mutants (e.g., R280K, R273H, S241F, S215R, R175H) and restored their tumor suppressor activities in vitro. Since mutant p53 function is both mutant-specific and cell context-dependent, we sought herein, to determine if plakoglobin tumor suppressive effects on exogenously expressed p53-R273H and p53-R175H mutants are similarly maintained under the same genetic background using the p53-null and plakoglobin-deficient H1299 cell line. Functional assays were performed to assess colony formation, migration, and invasion while immunoblotting and qPCR were used to examine the subcellular distribution and expression of specific proteins and genes that are typically regulated by or regulate p53 function and are altered in mutant p53-expressing cell lines and tumors. We show that though, plakoglobin interacted with both p53-R273H and p53-R175H mutants, it had a differential effect on the transcription and subcellular distribution of their gene targets and their overall oncogenic properties in vitro. Notably, we found that plakoglobin's tumor suppressive effects were significantly stronger in p53-R175H expressing cells compared to p53-R273H cells. Together, our results indicate that exploring plakoglobin interactions with p53-R175H may be useful for the development of cancer therapeutics focused on the restoration of p53 function.
Collapse
Affiliation(s)
- Chu Shiun Lo
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Parnian Alavi
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Blessing Bassey-Archibong
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Biology and Environmental Sciences Concordia University of Edmonton, Edmonton, Alberta, Canada
| | - Nadia Jahroudi
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Manijeh Pasdar
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
29
|
Chougoni KK, Neely V, Ding B, Oduah E, Lam VT, Hu B, Koblinski JE, Windle BE, Palit Deb S, Deb S, Nieva JJ, Radhakrishnan SK, Harada H, Grossman SR. Oncogenic Mutant p53 Sensitizes Non-Small Cell Lung Cancer Cells to Proteasome Inhibition via Oxidative Stress-Dependent Induction of Mitochondrial Apoptosis. CANCER RESEARCH COMMUNICATIONS 2024; 4:2685-2698. [PMID: 39302104 PMCID: PMC11474859 DOI: 10.1158/2767-9764.crc-23-0637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/17/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
SIGNIFICANCE NSCLC is the leading cause of cancer death due, in part, to a lack of active therapies in advanced disease. We demonstrate that combination therapy with a proteasome inhibitor, BH3-mimetic, and chemotherapy is an active precision therapy in NSCLC cells and tumors expressing Onc-p53 alleles.
Collapse
Affiliation(s)
- Kranthi Kumar Chougoni
- Department of Medicine, Keck School of Medicine of USC, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California.
| | - Victoria Neely
- Philips Institute for Oral Health Research, VCU School of Dentistry, Virginia Commonwealth University, Richmond, Virginia.
| | - Boxiao Ding
- Department of Medicine, Keck School of Medicine of USC, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California.
| | - Eziafa Oduah
- Division of Medical Oncology, Department of Medicine, Duke University School of Medicine, Duke Cancer Institute, Duke University, Durham, North Carolina.
| | - Vianna T. Lam
- Philips Institute for Oral Health Research, VCU School of Dentistry, Virginia Commonwealth University, Richmond, Virginia.
| | - Bin Hu
- VCU Cancer Mouse Models Core, Virginia Commonwealth University, Richmond, Virginia.
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia.
| | - Jennifer E. Koblinski
- VCU Cancer Mouse Models Core, Virginia Commonwealth University, Richmond, Virginia.
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia.
- Department of Pathology, VCU School of Medicine, Virginia Commonwealth University, Richmond, Virginia.
| | - Bradford E. Windle
- Philips Institute for Oral Health Research, VCU School of Dentistry, Virginia Commonwealth University, Richmond, Virginia.
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia.
| | - Swati Palit Deb
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia.
- Department of Biochemistry and Molecular Biology, VCU School of Medicine, VCU School of Medicine, Virginia Commonwealth University, Richmond, Virginia.
| | - Sumitra Deb
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia.
- Department of Biochemistry and Molecular Biology, VCU School of Medicine, VCU School of Medicine, Virginia Commonwealth University, Richmond, Virginia.
| | - Jorge J. Nieva
- Department of Medicine, Keck School of Medicine of USC, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California.
| | - Senthil K. Radhakrishnan
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia.
- Department of Pathology, VCU School of Medicine, Virginia Commonwealth University, Richmond, Virginia.
| | - Hisashi Harada
- Philips Institute for Oral Health Research, VCU School of Dentistry, Virginia Commonwealth University, Richmond, Virginia.
- VCU Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia.
| | - Steven R. Grossman
- Department of Medicine, Keck School of Medicine of USC, USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California.
| |
Collapse
|
30
|
Hwang E, Kruhlak M, Wong N, Chari R, Kimura T, Cheng SY. Attenuation of aggressive tumor progression of anaplastic thyroid cancer by p53. Am J Cancer Res 2024; 14:4429-4444. [PMID: 39417187 PMCID: PMC11477845 DOI: 10.62347/kxjj8824] [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: 05/30/2024] [Accepted: 07/12/2024] [Indexed: 10/19/2024] Open
Abstract
Anaplastic thyroid cancer (ATC) is the most aggressive thyroid cancer, with very limited treatment options. Mutations of p53 are associated with lethal outcomes of ATC. In this study, we tested the hypothesis that wild type p53 (WTp53) mitigates its aggressive progression. We used human 8505C cells (from human ATC tumors) as a model, harboring a BRAFV600E mutation and single of mutated p53C742G allele. We exogenously expressed WTp53 or mutant p53C742G into 8505C cells (8505C-WTp53 or 8505C-MTp53, respectively). The expressed WTp53 inhibited cell proliferation, decreased cell migration, and induced apoptosis via induction of proapoptotic WTp53 target BAX and PUMA genes in vitro. Mouse xenograft studies showed suppression of tumors induced by 8505C-WTp53 but not by 8505C-MTp53 cells. Consistent with in vitro findings, WTp53 inhibited proliferation of tumor cells, evidenced by decreased proliferation marker Ki-67 in tumors. WTp53 also induced apoptosis in xenograft tumors as shown by increased cleaved caspase-3 proteins and pro-apoptotic regulators, BAX and PUMA. Single cells RNA-sequencing (scRNA-seq) of tumors induced by 8505C, 8505C-WTp53, and 8505C-MTp53 cells demonstrated differential expression gene (DEG) patterns between 8505C-WTp53 and 8505C tumors. DEGs analysis identified alteration of multiple pathways, leading to attenuating the oncogenic actions of mutant p53. The discovery of the suppression of TNFα via NFκB pathway topped the pathways list, resulting in subduing the deleterious inflammatory responses caused by mutant p53. Our findings that exogenously expressed WTp53 could counter act the oncogenic actions of p53 has heightened the feasibility of using CRISPR/Cas9 genome editing to modify the p53 alleles for potential treatment of ATC.
Collapse
Affiliation(s)
- Eunmi Hwang
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Michael Kruhlak
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Nathan Wong
- CCR Collaborative Bioinformatics Resource, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer ResearchFrederick, MD 21702, USA
| | - Raj Chari
- Genome Modification Core, Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer ResearchFrederick, MD 21702, USA
| | - Takahito Kimura
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| | - Sheue-Yann Cheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesda, MD 20892, USA
| |
Collapse
|
31
|
Li X, Pan J, Zheng P. USP7 regulates growth and maintains the stemness of p53-mutant colorectal cancer cells via stabilizing of mutant p53. Front Oncol 2024; 14:1427663. [PMID: 39346740 PMCID: PMC11427698 DOI: 10.3389/fonc.2024.1427663] [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: 05/31/2024] [Accepted: 08/16/2024] [Indexed: 10/01/2024] Open
Abstract
Introduction TP53 is one of the most frequently mutated genes among all cancers, and TP53 mutants occur more than 40% in colorectal cancers (CRCs). Accumulation of mutant p53 may augment colorectal cancer stem cells (CCSCs) phenotype and enhance colorectal tumorigenesis. Thus, reducing the level of mutant p53 protein is an attractive anticancer strategy. Methods CSC-enriched cancer cells were obtained by tumor sphere formation assay. The effects of USP7 on the proliferation of cancer cells were determined by MTS and colony formation assays. Wound healing assay was used to test cell migratory abilities. qPCR and western blotting assays were performed to verify the mRNA and protein levels of CSC markers, USP7 and p53. Co-immunoprecipitation assay was used to test the interaction effects between USP7 and p53. Results In this study, we found that USP7 and mutant p53 were dramatically elevated in CSC-enriched colorectal cancer cells and USP7 expression was positively associated with self-renewal and maintenance of CCSCs. USP7 regulated cell growth, stemness and migration of colorectal cancer cells. USP7 depletion significantly reduced proliferation of cancer cells and suppressed the self-renewal of CSC-enriched colorectal cancer cells. Further studies indicated that USP7 knockdown could significantly decrease mutant p53 protein levels both in CRCs and CSC-enriched colorectal cancer cells. Moreover, mutant p53 was stabilized by USP7 and they interacted with each other. Furthermore, USP7 inhibitor P5091 also diminished CCSCs self-renewal and reduced mutant p53 levels. Conclusion Taken together, our findings demonstrated that USP7 involved in the modulation of CCSCs stemness, as well as a critical target for clinical treatment of cancers with different p53 mutations.
Collapse
Affiliation(s)
- Xue Li
- Department of Pharmacy, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jie Pan
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Department of Stomatology, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Pengcheng Zheng
- Department of Pharmacy, The First People’s Hospital of Yunnan Province, Kunming, Yunnan, China
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
32
|
Szlanka T, Lukacsovich T, Bálint É, Virágh E, Szabó K, Hajdu I, Molnár E, Lin YH, Zvara Á, Kelemen-Valkony I, Méhi O, Török I, Hegedűs Z, Kiss B, Ramasz B, Magdalena LM, Puskás L, Mechler BM, Fónagy A, Asztalos Z, Steinbach G, Žurovec M, Boros I, Kiss I. Dominant suppressor genes of p53-induced apoptosis in Drosophila melanogaster. G3 (BETHESDA, MD.) 2024; 14:jkae149. [PMID: 38985658 PMCID: PMC11373661 DOI: 10.1093/g3journal/jkae149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/06/2024] [Accepted: 06/15/2024] [Indexed: 07/12/2024]
Abstract
One of the major functions of programmed cell death (apoptosis) is the removal of cells that suffered oncogenic mutations, thereby preventing cancerous transformation. By making use of a Double-Headed-EP (DEP) transposon, a P element derivative made in our laboratory, we made an insertional mutagenesis screen in Drosophila melanogaster to identify genes that, when overexpressed, suppress the p53-activated apoptosis. The DEP element has Gal4-activatable, outward-directed UAS promoters at both ends, which can be deleted separately in vivo. In the DEP insertion mutants, we used the GMR-Gal4 driver to induce transcription from both UAS promoters and tested the suppression effect on the apoptotic rough eye phenotype generated by an activated UAS-p53 transgene. By DEP insertions, 7 genes were identified, which suppressed the p53-induced apoptosis. In 4 mutants, the suppression effect resulted from single genes activated by 1 UAS promoter (Pka-R2, Rga, crol, and Spt5). In the other 3 (Orct2, Polr2M, and stg), deleting either UAS promoter eliminated the suppression effect. In qPCR experiments, we found that the genes in the vicinity of the DEP insertion also showed an elevated expression level. This suggested an additive effect of the nearby genes on suppressing apoptosis. In the eukaryotic genomes, there are coexpressed gene clusters. Three of the DEP insertion mutants are included, and 2 are in close vicinity of separate coexpressed gene clusters. This raises the possibility that the activity of some of the genes in these clusters may help the suppression of the apoptotic cell death.
Collapse
Affiliation(s)
- Tamás Szlanka
- Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
- Biology Centre, Czech Academy of Sciences, 37005 České Budějovice, Czech Republic
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Tamás Lukacsovich
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Éva Bálint
- Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Erika Virágh
- Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
- Biology Centre, Czech Academy of Sciences, 37005 České Budějovice, Czech Republic
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Kornélia Szabó
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
- Department of Developmental Genetics, German Cancer Research Centre, 69120 Heidelberg, Germany
| | - Ildikó Hajdu
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Enikő Molnár
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Yu-Hsien Lin
- Biology Centre, Czech Academy of Sciences, 37005 České Budějovice, Czech Republic
| | - Ágnes Zvara
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Ildikó Kelemen-Valkony
- Cellular Imaging Laboratory, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Orsolya Méhi
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - István Török
- Department of Developmental Genetics, German Cancer Research Centre, 69120 Heidelberg, Germany
| | - Zoltán Hegedűs
- Bioinformatics Laboratory, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Brigitta Kiss
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Beáta Ramasz
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Laura M Magdalena
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - László Puskás
- Laboratory of Functional Genomics, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Bernard M Mechler
- Department of Developmental Genetics, German Cancer Research Centre, 69120 Heidelberg, Germany
| | - Adrien Fónagy
- Centre for Agricultural Sciences, Plant Protection Institute, 1022 Budapest, Hungary
| | - Zoltán Asztalos
- Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
- Aktogen Hungary Ltd., 6726 Szeged, Hungary
| | - Gábor Steinbach
- Cellular Imaging Laboratory, Core Facility, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - Michal Žurovec
- Biology Centre, Czech Academy of Sciences, 37005 České Budějovice, Czech Republic
| | - Imre Boros
- Institute of Biochemistry, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| | - István Kiss
- Institute of Genetics, HUN-REN Biological Research Centre, 6726 Szeged, Hungary
| |
Collapse
|
33
|
Wang C, Tan JYM, Chitkara N, Bhatt S. TP53 Mutation-Mediated Immune Evasion in Cancer: Mechanisms and Therapeutic Implications. Cancers (Basel) 2024; 16:3069. [PMID: 39272927 PMCID: PMC11393945 DOI: 10.3390/cancers16173069] [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: 08/01/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Mutation in p53 is the most frequent event in cancer development and a leading cause of cancer therapy resistance due to evasion of the apoptosis cascade. Beyond chemotherapies and radiation therapies, growing evidence indicates that p53-mutant tumors are resistant to a broad range of immune-based therapies, such as immune checkpoint inhibitors, chimeric antigen receptor (CAR) T, and hematopoietic stem cell transplantation (HSCT). This highlights the role of p53 mutations in driving immune evasion of tumor cells. In this review, we first summarize recent studies revealing mechanisms by which p53-mutant tumors evade immune surveillance from T cells, natural killer (NK) cells, and macrophages. We then review how these mutant tumor cells reshape the tumor microenvironment (TME), modulating bystander cells such as macrophages, neutrophils, and regulatory T (Treg) cells to foster immunosuppression. Additionally, we review clinical observations indicative of immune evasion associated with p53 loss or mutations. Finally, we discuss therapeutic strategies to enhance immune response in p53 wild-type (WT) or mutant tumors.
Collapse
Affiliation(s)
- Chuqi Wang
- Department of Pharmacy & Pharmaceutical Sciences, National University of Singapore, Singapore 117559, Singapore
| | - Jordan Yong Ming Tan
- Department of Pharmacy & Pharmaceutical Sciences, National University of Singapore, Singapore 117559, Singapore
| | | | - Shruti Bhatt
- Department of Pharmacy & Pharmaceutical Sciences, National University of Singapore, Singapore 117559, Singapore
| |
Collapse
|
34
|
Shoemaker R, Huang MF, Wu YS, Huang CS, Lee DF. Decoding the molecular symphony: interactions between the m 6A and p53 signaling pathways in cancer. NAR Cancer 2024; 6:zcae037. [PMID: 39329012 PMCID: PMC11426327 DOI: 10.1093/narcan/zcae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 08/05/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
The p53 tumor suppressor gene governs a multitude of complex cellular processes that are essential for anti-cancer function and whose dysregulation leads to aberrant gene transcription, activation of oncogenic signaling and cancer development. Although mutations can occur at any point in the genetic sequence, missense mutations comprise the majority of observed p53 mutations in cancers regardless of whether the mutation is germline or somatic. One biological process involved in both mutant and wild-type p53 signaling is the N 6-methyladenosine (m6A) epitranscriptomic network, a type of post-transcriptional modification involved in over half of all eukaryotic mRNAs. Recently, a significant number of findings have demonstrated unique interactions between p53 and the m6A epitranscriptomic network in a variety of cancer types, shedding light on a previously uncharacterized connection that causes significant dysregulation. Cross-talk between wild-type or mutant p53 and the m6A readers, writers and erasers has been shown to impact cellular function and induce cancer formation by influencing various cancer hallmarks. Here, this review aims to summarize the complex interplay between the m6A epitranscriptome and p53 signaling pathway, highlighting its effects on tumorigenesis and other hallmarks of cancer, as well as identifying its therapeutic implications for the future.
Collapse
Affiliation(s)
- Rachel Shoemaker
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Mo-Fan Huang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Ying-Si Wu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Cheng-Shuo Huang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei 114, Taiwan
| | - Dung-Fang Lee
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center, UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- Center for Stem Cell and Regenerative Medicine, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
35
|
Hu X, Kabir M, Lin Y, Xiong Y, Parsons RE, Gu W, Jin J. Design, Synthesis, and Evaluation of p53Y220C Acetylation Targeting Chimeras (AceTACs). J Med Chem 2024; 67:14633-14648. [PMID: 39169826 PMCID: PMC11378941 DOI: 10.1021/acs.jmedchem.4c01497] [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] [Indexed: 08/23/2024]
Abstract
The well-known tumor suppressor p53 is mutated in approximately half of all cancers. The Y220C mutation is one of the major p53 hotspot mutations. Several small-molecule stabilizers of p53Y220C have been developed. We recently developed a new technology for inducing targeted protein acetylation, termed acetylation targeting chimera (AceTAC), and the first p53Y220C AceTAC that effectively acetylated p53Y220C at lysine 382. Here, we report structure-activity relationship (SAR) studies of p53Y220C AceTACs, which led to the discovery of a novel p53Y220C AceTAC, compound 11 (MS182). 11 effectively acetylated p53Y220C at lysine 382 in a time- and concentration-dependent manner via inducing the ternary complex formation between p300/CBP acetyltransferase and p53Y220C. 11 was more effective than the parent p53Y220C stabilizer in suppressing the proliferation and clonogenicity in cancer cells harboring the p53Y200C mutation and was bioavailable in mice. Overall, 11 is a potentially valuable chemical tool to investigate the role of p53Y220C acetylation in cancer.
Collapse
Affiliation(s)
- Xiaoping Hu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Md Kabir
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Yindan Lin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Yan Xiong
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Ramon E Parsons
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Wei Gu
- Institute for Cancer Genetics, and Department of Pathology and Cell Biology, and Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, New York 10032, United States
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
36
|
Shah NN, Dave BP, Shah KC, Shah DD, Maheshwari KG, Chorawala MR, Parekh PS, Jani M. Disabled-2, a versatile tissue matrix multifunctional scaffold protein with multifaceted signaling: Unveiling its potential in the cancer battle. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:5533-5557. [PMID: 38502243 DOI: 10.1007/s00210-024-03037-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 03/01/2024] [Indexed: 03/21/2024]
Abstract
A multifunctional scaffold protein termed Disabled-2 (Dab2) has recently gained attention in the scientific community and has emerged as a promising candidate in the realm of cancer research. Dab2 protein is involved in a variety of signaling pathways, due to which its significance in the pathogenesis of several carcinomas has drawn considerable attention. Dab2 is essential for controlling the advancement of cancer because it engages in essential signaling pathways such as the Wnt/β-catenin, epidermal growth factor receptor (EGFR), and transforming growth factor-beta (TGF-β) pathways. Dab2 can also repress epithelial-mesenchymal transition (EMT) which is involved in tumor progression with metastatic expansion and adds another layer of significance to its possible impact on cancer spread. Furthermore, the role of Dab2 in processes such as cell growth, differentiation, apoptosis, invasion, and metastasis has been explored in certain investigative studies suggesting its significance. The present review examines the role of Dab2 in the pathogenesis of various cancer subtypes including breast cancer, ovarian cancer, gastric cancer, prostate cancer, and bladder urothelial carcinoma and also sheds some light on its potential to act as a therapeutic target and a prognostic marker in the treatment of various carcinomas. By deciphering this protein's diverse signaling, we hope to provide useful insights that may pave the way for novel therapeutic techniques and tailored treatment approaches in cancer management. Preclinical and clinical trial data on the impact of Dab2 regulation in cancer have also been included, allowing us to delineate role of Dab2 in tumor suppressor function, as well as its correlation with disease stage classification and potential therapy options. However, we observed that there is very scarce data in the form of studies on the evaluation of Dab2 role and treatment function in carcinomas, and further research into this matter could prove beneficial in the generation of novel therapeutic agents for patient-centric and tailored therapy, as well as early prognosis of carcinomas.
Collapse
Affiliation(s)
- Nidhi N Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Bhavarth P Dave
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Kashvi C Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Kunal G Maheshwari
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, Gujarat, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Navrangpura, Ahmedabad, 380009, Gujarat, India.
| | - Priyajeet S Parekh
- AV Pharma LLC, 1545 University Blvd N Ste A, Jacksonville, FL, 32211, USA
| | - Maharsh Jani
- Anand Niketan Shilaj, Ahmedabad, 380059, Gujarat, India
| |
Collapse
|
37
|
Tian X, Srinivasan PR, Tajiknia V, Sanchez Sevilla Uruchurtu AF, Seyhan AA, Carneiro BA, De La Cruz A, Pinho-Schwermann M, George A, Zhao S, Strandberg J, Di Cristofano F, Zhang S, Zhou L, Raufi AG, Navaraj A, Zhang Y, Verovkina N, Ghandali M, Ryspayeva D, El-Deiry WS. Targeting apoptotic pathways for cancer therapy. J Clin Invest 2024; 134:e179570. [PMID: 39007268 PMCID: PMC11245162 DOI: 10.1172/jci179570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Abstract
Apoptosis is a form of programmed cell death that is mediated by intrinsic and extrinsic pathways. Dysregulation of and resistance to cell death are hallmarks of cancer. For over three decades, the development of therapies to promote treatment of cancer by inducing various cell death modalities, including apoptosis, has been a main goal of clinical oncology. Apoptosis pathways also interact with other signaling mechanisms, such as the p53 signaling pathway and the integrated stress response (ISR) pathway. In addition to agents directly targeting the intrinsic and extrinsic pathway components, anticancer drugs that target the p53 and ISR signaling pathways are actively being developed. In this Review, we discuss selected and promising anticancer therapies in various stages of development, including drug targets, mechanisms, and resistance to related treatments, focusing especially on B cell lymphoma 2 (BCL-2) inhibitors, TRAIL analogues, DR5 antibodies, and strategies that target p53, mutant p53, and the ISR.
Collapse
Affiliation(s)
- Xiaobing Tian
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Praveen R. Srinivasan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Vida Tajiknia
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Ashley F. Sanchez Sevilla Uruchurtu
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
- Pathobiology Graduate Program, Brown University, Providence, Rhode Island, USA
| | - Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Benedito A. Carneiro
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
- Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, Rhode Island, USA
| | - Arielle De La Cruz
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Maximilian Pinho-Schwermann
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
- Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, Rhode Island, USA
| | - Andrew George
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Shuai Zhao
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Jillian Strandberg
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Francesca Di Cristofano
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Shengliang Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Lanlan Zhou
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Alexander G. Raufi
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
- Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, Rhode Island, USA
| | - Arunasalam Navaraj
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Yiqun Zhang
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Nataliia Verovkina
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Maryam Ghandali
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Dinara Ryspayeva
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics and
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, Providence, Rhode Island, USA
- Legorreta Cancer Center at Brown University, Providence, Rhode Island, USA
- Pathobiology Graduate Program, Brown University, Providence, Rhode Island, USA
- Hematology/Oncology Division, Department of Medicine, Lifespan Health System and Brown University, Providence, Rhode Island, USA
| |
Collapse
|
38
|
Cesca BA, Caverzan MD, Lamberti MJ, Ibarra LE. Enhancing Therapeutic Approaches in Glioblastoma with Pro-Oxidant Treatments and Synergistic Combinations: In Vitro Experience of Doxorubicin and Photodynamic Therapy. Int J Mol Sci 2024; 25:7525. [PMID: 39062770 PMCID: PMC11277534 DOI: 10.3390/ijms25147525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/27/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive brain cancer characterized by significant molecular and cellular heterogeneity, which complicates treatment efforts. Current standard therapies, including surgical resection, radiation, and temozolomide (TMZ) chemotherapy, often fail to achieve long-term remission due to tumor recurrence and resistance. A pro-oxidant environment is involved in glioma progression, with oxidative stress contributing to the genetic instability that leads to gliomagenesis. Evaluating pro-oxidant therapies in brain tumors is crucial due to their potential to selectively target and eradicate cancer cells by exploiting the elevated oxidative stress levels inherent in these malignant cells, thereby offering a novel and effective strategy for overcoming resistance to conventional therapies. This study investigates the therapeutic potential of doxorubicin (DOX) and photodynamic therapy (PDT) with Me-ALA, focusing on their effects on redox homeostasis. Basal ROS levels and antioxidant gene expression (NFE2L2, CAT, GSR) were quantitatively assessed across GBM cell lines, revealing significant variability probably linked to genetic differences. DOX and PDT treatments, both individually and in combination, were analyzed for their efficacy in inducing oxidative stress and cytotoxicity. An in silico analysis further explored the relationship between gene mutations and oxidative stress in GBM patients, providing insights into the molecular mechanisms underlying treatment responses. Our findings suggest that pro-oxidant therapies, such as DOX and PDT in combination, could selectively target GBM cells, highlighting a promising avenue for improving therapeutic outcomes in GBM.
Collapse
Affiliation(s)
- Bruno Agustín Cesca
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Rio Cuarto, Rio Cuarto X5800BIA, Argentina; (B.A.C.); (M.J.L.)
| | - Matías Daniel Caverzan
- Departamento de Patología Animal, Facultad de Agronomía y Veterinaria, Universidad Nacional de Rio Cuarto, Rio Cuarto X5800BIA, Argentina;
- Instituto de Investigaciones en Tecnologías Energéticas y Materiales Avanzados (IITEMA), Universidad Nacional de Rio Cuarto (UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rio Cuarto X5800BIA, Argentina
| | - María Julia Lamberti
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Rio Cuarto, Rio Cuarto X5800BIA, Argentina; (B.A.C.); (M.J.L.)
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Rio Cuarto (UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rio Cuarto X5800BIA, Argentina
| | - Luis Exequiel Ibarra
- Departamento de Biología Molecular, Facultad de Ciencias Exactas, Fisicoquímicas y Naturales, Universidad Nacional de Rio Cuarto, Rio Cuarto X5800BIA, Argentina; (B.A.C.); (M.J.L.)
- Instituto de Biotecnología Ambiental y Salud (INBIAS), Universidad Nacional de Rio Cuarto (UNRC), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Rio Cuarto X5800BIA, Argentina
| |
Collapse
|
39
|
Benitez DA, Cumplido-Laso G, Olivera-Gómez M, Del Valle-Del Pino N, Díaz-Pizarro A, Mulero-Navarro S, Román-García A, Carvajal-Gonzalez JM. p53 Genetics and Biology in Lung Carcinomas: Insights, Implications and Clinical Applications. Biomedicines 2024; 12:1453. [PMID: 39062026 PMCID: PMC11274425 DOI: 10.3390/biomedicines12071453] [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: 05/16/2024] [Revised: 06/20/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The TP53 gene is renowned as a tumor suppressor, playing a pivotal role in overseeing the cell cycle, apoptosis, and maintaining genomic stability. Dysregulation of p53 often contributes to the initiation and progression of various cancers, including lung cancer (LC) subtypes. The review explores the intricate relationship between p53 and its role in the development and progression of LC. p53, a crucial tumor suppressor protein, exists in various isoforms, and understanding their distinct functions in LC is essential for advancing our knowledge of this deadly disease. This review aims to provide a comprehensive literature overview of p53, its relevance to LC, and potential clinical applications.
Collapse
Affiliation(s)
- Dixan A. Benitez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain; (G.C.-L.); (M.O.-G.); (N.D.V.-D.P.); (A.D.-P.); (S.M.-N.); (A.R.-G.)
| | | | | | | | | | | | | | - Jose Maria Carvajal-Gonzalez
- Departamento de Bioquímica, Biología Molecular y Genética, Facultad de Ciencias, Universidad de Extremadura, 06006 Badajoz, Spain; (G.C.-L.); (M.O.-G.); (N.D.V.-D.P.); (A.D.-P.); (S.M.-N.); (A.R.-G.)
| |
Collapse
|
40
|
Hwang SP, Denicourt C. The impact of ribosome biogenesis in cancer: from proliferation to metastasis. NAR Cancer 2024; 6:zcae017. [PMID: 38633862 PMCID: PMC11023387 DOI: 10.1093/narcan/zcae017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/23/2024] [Accepted: 03/26/2024] [Indexed: 04/19/2024] Open
Abstract
The dysregulation of ribosome biogenesis is a hallmark of cancer, facilitating the adaptation to altered translational demands essential for various aspects of tumor progression. This review explores the intricate interplay between ribosome biogenesis and cancer development, highlighting dynamic regulation orchestrated by key oncogenic signaling pathways. Recent studies reveal the multifaceted roles of ribosomes, extending beyond protein factories to include regulatory functions in mRNA translation. Dysregulated ribosome biogenesis not only hampers precise control of global protein production and proliferation but also influences processes such as the maintenance of stem cell-like properties and epithelial-mesenchymal transition, contributing to cancer progression. Interference with ribosome biogenesis, notably through RNA Pol I inhibition, elicits a stress response marked by nucleolar integrity loss, and subsequent G1-cell cycle arrest or cell death. These findings suggest that cancer cells may rely on heightened RNA Pol I transcription, rendering ribosomal RNA synthesis a potential therapeutic vulnerability. The review further explores targeting ribosome biogenesis vulnerabilities as a promising strategy to disrupt global ribosome production, presenting therapeutic opportunities for cancer treatment.
Collapse
Affiliation(s)
- Sseu-Pei Hwang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| | - Catherine Denicourt
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center, Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
41
|
Li T, Liu J, Zhou Y, Huang S, Wang D, Chen J, Fu Y, He P. Clinical relevance of somatic mutations in Chinese lung adenocarcinoma and their prognostic implications for survival. Cancer Med 2024; 13:e7227. [PMID: 38770632 PMCID: PMC11106684 DOI: 10.1002/cam4.7227] [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: 09/11/2023] [Revised: 04/07/2024] [Accepted: 04/18/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND To comprehensively elucidate the genomic and mutational features of lung cancer cases, and lung adenocarcinoma (LUAD), it is imperative to conduct ongoing investigations into the genomic landscape. In this study, we aim to analyze the somatic mutation profile and assessed the significance of these informative genes utilizing a retrospective LUAD cohort. METHODS A total of 247 Chinese samples were analyzed to exhibit the tumor somatic genomic alterations in patients with LUAD. The Cox regression analysis was employed to identify prognosis-related genes and establish a predictive model for stratifying patients with LUAD. RESULTS In the Dianjiang People's Hospital (DPH) cohort, the top five frequent mutated genes were (Epidermal growth factor receptor) EGFR (68%), TP53 (30%), RBM10 (13%), LRP1B (9%), and KRAS (9%). Of which, EGFR is a mostly altered driver gene, and most mutation sites are located in tyrosine kinase regions. Oncogene pathway alteration and mutation signature analysis demonstrated the RTK-RAS pathway alteration, and smoking was the main carcinogenic factor of the DPH cohort. Furthermore, we identified 34 driver genes in the DPH cohort, including EGFR (68%), TP53 (30.4%), RBM10 (12.6%), KRAS (8.5%), LRP1B (8.5%), and so on, and 45 Clinical Characteristic-Related Genes (CCRGs) were found to closely related to the clinical high-risk factors. We developed a Multiple Parameter Gene Mutation (MPGM) risk model by integrating critical genes and oncogenic pathway alterations in LUAD patients from the DPH cohort. Based on publicly available LUAD datasets, we identified five genes, including BRCA2, Anaplastic lymphoma kinase (ALK), BRAF, EGFR, and Platelet-Derived Growth Factor Receptor Alpha (PDGFRA), according to the multivariable Cox regression analysis. The MPGM-low group showed significantly better overall survival (OS) compared to the MPGM-high group (p < 0.0001, area under the curve (AUC) = 0.754). The robust performance was validated in 55 LUAD patients from the DPH cohort and another LUAD dataset. Immune characteristics analysis revealed a higher proportion of primarily DCs and mononuclear cells in the MPGM-low risk group, while the MPGM-high risk group showed lower immune cells and higher tumor cell infiltration. CONCLUSION This study provides a comprehensive genomic landscape of Chinese LUAD patients and develops an MPGM risk model for LUAD prognosis stratification. Further follow-up will be performed for the patients in the DPH cohort consistently to explore the resistance and prognosis genetic features.
Collapse
Affiliation(s)
- Tongxin Li
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of ChongqingChongqingChina
| | - Jie Liu
- Department of Thoracic Surgery, Southwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| | - Yu Zhou
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of ChongqingChongqingChina
| | - Shengyuan Huang
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of ChongqingChongqingChina
| | - Dong Wang
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of ChongqingChongqingChina
| | - Jianrong Chen
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of ChongqingChongqingChina
| | - Yong Fu
- Department of Cardiothoracic SurgeryDianjiang People's Hospital of ChongqingChongqingChina
| | - Ping He
- Department of Cardiac Surgery, Southwest HospitalArmy Medical University (Third Military Medical University)ChongqingChina
| |
Collapse
|
42
|
Zhou B, Wang L, Ren Z, Liang Y, Yang S, Zhang Y, Che S, Fang W. Pyrogallol promotes growth arrest by activating the p53-mediated up-regulation of p21 and p62/SQSTM1-dependent degradation of β-catenin in nonsmall cell lung cancer cells. ENVIRONMENTAL TOXICOLOGY 2024; 39:2150-2165. [PMID: 38108618 DOI: 10.1002/tox.24099] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 11/13/2023] [Accepted: 12/01/2023] [Indexed: 12/19/2023]
Abstract
Pyrogallol (1,2,3-trihydroxybenzene), a polyphenolic natural compound, has attracted considerable attention with regard to its potential anticancer activity. However, further study is needed to elucidate the underlying mechanism related to the antiNSCLC activity of pyrogallol and provide a comprehensive theoretical basis for better clinical utilization of pyrogallol. Our current study aims to investigate the effects and potential underlying mechanisms of pyrogallol on the inhibition of NSCLC growth. Our results showed that pyrogallol treatment induced cell cycle arrest at the G2/M phase and apoptosis in two different NSCLC cell lines. Mechanistically, we found that the induction of cell cycle arrest in NSCLC cells at the G2/M phase by pyrogallol was due to the upregulation of p21 in a p53-dependent manner. And blockade of p53 and p21 effectively abolished the cell cycle arrest at the G2/M phase. Meanwhile, p53 inhibition has been found to abrogate the pyrogallol-induced apoptosis of the two NSCLC cells. Moreover, we revealed that the inhibitory effects of pyrogallol on β-catenin signaling resulted from autophagy initiation depending on p53 activation, accompanied by an increase in p62/SQSTM1 expression, thus p62 subsequently interacting with ubiquitinated β-catenin and facilitating autophagic destruction of β-catenin. Furthermore, in vivo experiments demonstrated that pyrogallol exerted growth inhibition on NSCLC with low toxicity through the same molecular mechanism as observed in vitro. Our findings could contribute to the understanding of the mechanism by which pyrogallol negatively regulates NSCLC growth, which could be effective in treating NSCLC.
Collapse
Affiliation(s)
- Beixian Zhou
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
- The People's Hospital of Gaozhou, Gaozhou, China
| | - Linxin Wang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zhixian Ren
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicine Utilization, Yunnan University of Chinese Medicine, Kunming, China
| | - Yueyun Liang
- The People's Hospital of Gaozhou, Gaozhou, China
| | - Sushan Yang
- The People's Hospital of Gaozhou, Gaozhou, China
| | - Yuehan Zhang
- The People's Hospital of Gaozhou, Gaozhou, China
| | - Siyao Che
- The People's Hospital of Gaozhou, Gaozhou, China
| | - Weiyi Fang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
43
|
Dixon-Zegeye M, Shaw R, Collins L, Perez-Smith K, Ooms A, Qiao M, Pantziarka P, Izatt L, Tischkowitz M, Harrison RE, George A, Woodward ER, Lord S, Hawkes L, Evans DG, Franklin J, Hanson H, Blagden SP. Cancer Precision-Prevention trial of Metformin in adults with Li Fraumeni syndrome (MILI) undergoing yearly MRI surveillance: a randomised controlled trial protocol. Trials 2024; 25:103. [PMID: 38308321 PMCID: PMC10837926 DOI: 10.1186/s13063-024-07929-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/16/2024] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Li-Fraumeni syndrome (LFS) is a rare autosomal dominant disease caused by inherited or de novo germline pathogenic variants in TP53. Individuals with LFS have a 70-100% lifetime risk of developing cancer. The current standard of care involves annual surveillance with whole-body and brain MRI (WB-MRI) and clinical review; however, there are no chemoprevention agents licensed for individuals with LFS. Preclinical studies in LFS murine models show that the anti-diabetic drug metformin is chemopreventive and, in a pilot intervention trial, short-term use of metformin was well-tolerated in adults with LFS. However, metformin's mechanism of anticancer activity in this context is unclear. METHODS Metformin in adults with Li-Fraumeni syndrome (MILI) is a Precision-Prevention phase II open-labelled unblinded randomised clinical trial in which 224 adults aged ≥ 16 years with LFS are randomised 1:1 to oral metformin (up to 2 mg daily) plus annual MRI surveillance or annual MRI surveillance alone for up to 5 years. The primary endpoint is to compare cumulative cancer-free survival up to 5 years (60 months) from randomisation between the intervention (metformin) and control (no metformin) arms. Secondary endpoints include a comparison of cumulative tumour-free survival at 5 years, overall survival at 5 years and clinical characteristics of emerging cancers between trial arms. Safety, toxicity and acceptability of metformin; impact of metformin on quality of life; and impact of baseline lifestyle risk factors on cancer incidence will be assessed. Exploratory end-points will evaluate the mechanism of action of metformin as a cancer preventative, identify biomarkers of response or carcinogenesis and assess WB-MRI performance as a diagnostic tool for detecting cancers in participants with LFS by assessing yield and diagnostic accuracy of WB-MRI. DISCUSSION Alongside a parallel MILI study being conducted by collaborators at the National Cancer Institute (NCI), MILI is the first prevention trial to be conducted in this high-risk group. The MILI study provides a unique opportunity to evaluate the efficacy of metformin as a chemopreventive alongside exploring its mechanism of anticancer action and the biological process of mutated P53-driven tumourigenesis. TRIAL REGISTRATION ISRCTN16699730. Registered on 28 November 2022. URL: https://www.isrctn.com/ EudraCT/CTIS number 2022-000165-41.
Collapse
Affiliation(s)
- Miriam Dixon-Zegeye
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Rachel Shaw
- Oncology Clinical Trials Office, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Linda Collins
- Oncology Clinical Trials Office, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Kendra Perez-Smith
- Trial Support Unit, Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, UK
| | - Alexander Ooms
- Centre for Statistics in Medicine and Oxford Clinical Trials Research Unit (OCTRU), Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Headington, Oxford, UK
| | - Maggie Qiao
- Centre for Statistics in Medicine and Oxford Clinical Trials Research Unit (OCTRU), Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Headington, Oxford, UK
| | - Pan Pantziarka
- George Pantziarka TP53 Trust, 7 Surbiton Cres, Kingston upon Thames, UK
| | - Louise Izatt
- Guy's and St Thomas' NHS Foundation Trust, Great Maze Pond, London, UK
| | - Marc Tischkowitz
- Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Rachel E Harrison
- Department of Clinical Genetics, Nottingham University Hospitals NHS Trust, Hucknall Rd, Nottingham, UK
| | | | - Emma R Woodward
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Simon Lord
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Lara Hawkes
- Oxford Centre for Genomic Medicine, ACE building, Nuffield Orthopaedic Centre, Windmill Road, Headington, Oxford, UK
| | - D Gareth Evans
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - James Franklin
- Institute of Medical Imaging and Visualisation, Bournemouth University, St Pauls Lane, Bournemouth, UK
| | - Helen Hanson
- Peninsula Clinical Genetics Service, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
- Faculty of Health and Life Sciences, University of Exeter, Heavitree Road, Exeter, UK
| | - Sarah P Blagden
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.
- Oncology Clinical Trials Office, University of Oxford, Old Road Campus Research Building, Oxford, UK.
| |
Collapse
|
44
|
Arai H, Matsui H, Chi S, Utsu Y, Masuda S, Aotsuka N, Minami Y. Germline Variants and Characteristic Features of Hereditary Hematological Malignancy Syndrome. Int J Mol Sci 2024; 25:652. [PMID: 38203823 PMCID: PMC10779750 DOI: 10.3390/ijms25010652] [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: 11/07/2023] [Revised: 12/25/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Due to the proliferation of genetic testing, pathogenic germline variants predisposing to hereditary hematological malignancy syndrome (HHMS) have been identified in an increasing number of genes. Consequently, the field of HHMS is gaining recognition among clinicians and scientists worldwide. Patients with germline genetic abnormalities often have poor outcomes and are candidates for allogeneic hematopoietic stem cell transplantation (HSCT). However, HSCT using blood from a related donor should be carefully considered because of the risk that the patient may inherit a pathogenic variant. At present, we now face the challenge of incorporating these advances into clinical practice for patients with myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) and optimizing the management and surveillance of patients and asymptomatic carriers, with the limitation that evidence-based guidelines are often inadequate. The 2016 revision of the WHO classification added a new section on myeloid malignant neoplasms, including MDS and AML with germline predisposition. The main syndromes can be classified into three groups. Those without pre-existing disease or organ dysfunction; DDX41, TP53, CEBPA, those with pre-existing platelet disorders; ANKRD26, ETV6, RUNX1, and those with other organ dysfunctions; SAMD9/SAMD9L, GATA2, and inherited bone marrow failure syndromes. In this review, we will outline the role of the genes involved in HHMS in order to clarify our understanding of HHMS.
Collapse
Affiliation(s)
- Hironori Arai
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (H.A.); (S.C.)
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Iidacho, Narita 286-0041, Japan; (Y.U.); (S.M.); (N.A.)
| | - Hirotaka Matsui
- Department of Laboratory Medicine, National Cancer Center Hospital, Tsukiji, Chuoku 104-0045, Japan;
- Department of Medical Oncology and Translational Research, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-8665, Japan
| | - SungGi Chi
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (H.A.); (S.C.)
| | - Yoshikazu Utsu
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Iidacho, Narita 286-0041, Japan; (Y.U.); (S.M.); (N.A.)
| | - Shinichi Masuda
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Iidacho, Narita 286-0041, Japan; (Y.U.); (S.M.); (N.A.)
| | - Nobuyuki Aotsuka
- Department of Hematology and Oncology, Japanese Red Cross Narita Hospital, Iidacho, Narita 286-0041, Japan; (Y.U.); (S.M.); (N.A.)
| | - Yosuke Minami
- Department of Hematology, National Cancer Center Hospital East, Kashiwa 277-8577, Japan; (H.A.); (S.C.)
| |
Collapse
|
45
|
Alvarado-Ortiz E, Ortiz-Sánchez E, Sarabia-Sánchez MA, de la Cruz-López KG, García-Carrancá A, Robles-Flores M. Mutant p53 gain-of-function stimulates canonical Wnt signaling via PI3K/AKT pathway in colon cancer. J Cell Commun Signal 2023; 17:1389-1403. [PMID: 37982965 PMCID: PMC10713888 DOI: 10.1007/s12079-023-00793-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/05/2023] [Indexed: 11/21/2023] Open
Abstract
Aberrant canonical Wnt signaling is a hallmark of colon cancer. The TP53 tumor suppressor gene is altered in many solid tumors, including colorectal cancer, resulting in mutant versions of p53 (mut-p53) that lose their tumor suppressor capacities and acquire new-oncogenic functions (GOFs) critical for disease progression. Although the mechanisms related to mut-p53 GOF have been explored extensively, the relevance of mut-p53 in the canonical Wnt pathway is not well defined. This work investigated the influence of mut-p53 compared to wt-p53 in β-catenin-dependent Wnt signaling. Using the TCGA public data from Pan-Cancer and the GEPIA2 platform, an in silico analysis of wt-p53 versus mut-p53 genotyped colorectal cancer patients showed that TP53 (p53) and CTNNB1 (β-catenin) are significantly overexpressed in colorectal cancer, compared with normal tissue. Using p53 overexpression or p53 knockdown assays of wt-p53 or mut-p53, we found that while wt-p53 antagonizes canonical Wnt signaling, mut-p53 induces the opposite effect, improving the β-catenin-dependent transcriptional activity and colony formation ability of colon cancer cells, which were both decreased by mut-p53 knockdown expression. The mechanism involved in mut-p53-induced activation of canonical Wnt appears to be via AKT-mediated phosphorylation of Ser 552 of β-catenin, which is known to stabilize and enhance its transcriptional activity. We also found that while wt-p53 expression contributes to 5-FU sensitivity in colon cancer cells, the RITA p53 reactivating molecule counteracted the resistance against 5-FU in cells expressing mut-p53. Our results indicate that mut-p53 GOF acts as a positive regulator of canonical Wnt signaling and participates in the induction of resistance to 5-FU in colon cancer cells.
Collapse
Affiliation(s)
- Eduardo Alvarado-Ortiz
- Programa de Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Elizabeth Ortiz-Sánchez
- Subdirección de Investigación Básica, Instituto Nacional de Cancerología, Secretaría de Salud, Ciudad de México, Mexico City, Mexico
| | | | - Karen Griselda de la Cruz-López
- Laboratorio de Virus & Cáncer, Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México & Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Alejandro García-Carrancá
- Laboratorio de Virus & Cáncer, Unidad de Investigación Biomédica en Cáncer, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México & Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Martha Robles-Flores
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
46
|
Pereira M, Glogova A, Haagsma J, Stewart J, Shepherd TG, Petrik J. Mutant p53 murine oviductal epithelial cells induce progression of high-grade serous carcinoma and are most sensitive to simvastatin therapy in vitro and in vivo. J Ovarian Res 2023; 16:218. [PMID: 37986175 PMCID: PMC10662458 DOI: 10.1186/s13048-023-01307-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 11/02/2023] [Indexed: 11/22/2023] Open
Abstract
High-grade serous carcinoma (HGSC) is the most common and aggressive subtype of epithelial ovarian cancer, characterized by gain-of-function TP53 mutations originating in the fallopian tube epithelium. Therapeutic intervention occurs at advanced metastatic disease, due to challenges in early-stage diagnosis, with common disease recurrence and therapy resistance despite initial therapy success. The mevalonate pathway is exploited by many cancers and is potently inhibited by statin drugs. Statins have shown anti-cancer activity in many, but not all cancers. Here, we investigated the role of p53 status in relation to mevalonate pathway signaling in murine oviductal epithelial (OVE) cells and identified OVE cell sensitivity to statin inhibition. We found that p53R175H mutant and Trp53 knockout OVE cells have increased mevalonate pathway signaling compared to p53 wild-type OVE cells. Through orthotopic implantation to replicate the fallopian tube origin of HGSC, p53R175H mutant cells upregulated the mevalonate pathway to drive progression to advanced-stage ovarian cancer, and simvastatin treatment abrogated this effect. Additionally, simvastatin was more efficacious at inhibiting cell metabolic activity in OVE cells than atorvastatin, rosuvastatin and pravastatin. In vitro, simvastatin demonstrated potent effects on cell proliferation, apoptosis, invasion and migration in OVE cells regardless of p53 status. In vivo, simvastatin induced ovarian cancer disease regression through decreased primary ovarian tumor weight and increased apoptosis. Simvastatin also significantly increased cytoplasmic localization of HMG-CoA reductase in ovarian tumors. Downstream of the mevalonate pathway, simvastatin had no effect on YAP or small GTPase activity. This study suggests that simvastatin can induce anti-tumor effects and could be an important inhibitor of ovarian cancer progression.
Collapse
Affiliation(s)
- Madison Pereira
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Alice Glogova
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Jacob Haagsma
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Julia Stewart
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Trevor G Shepherd
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Obstetrics & Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Jim Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
47
|
Kashiwagi R, Funayama R, Aoki S, Matsui A, Klein S, Sato Y, Suzuki T, Murakami K, Inoue K, Iseki M, Masuda K, Mizuma M, Naito H, Duda DG, Unno M, Nakayama K. Collagen XVII regulates tumor growth in pancreatic cancer through interaction with the tumor microenvironment. Cancer Sci 2023; 114:4286-4298. [PMID: 37688308 PMCID: PMC10637054 DOI: 10.1111/cas.15952] [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: 04/01/2023] [Revised: 08/15/2023] [Accepted: 08/21/2023] [Indexed: 09/10/2023] Open
Abstract
Expression of the gene for collagen XVII (COL17A1) in tumor tissue is positively or negatively associated with patient survival depending on cancer type. High COL17A1 expression is thus a favorable prognostic marker for breast cancer but unfavorable for pancreatic cancer. This study explored the effects of COL17A1 expression on pancreatic tumor growth and their underlying mechanisms. Analysis of published single-cell RNA-sequencing data for human pancreatic cancer tissue revealed that COL17A1 was expressed predominantly in cancer cells rather than surrounding stromal cells. Forced expression of COL17A1 did not substantially affect the proliferation rate of the mouse pancreatic cancer cell lines KPC and AK4.4 in vitro. However, in mouse homograft tumor models in which KPC or AK4.4 cells were injected into syngeneic C57BL/6 or FVB mice, respectively, COL17A1 expression promoted or suppressed tumor growth, respectively, suggesting that the effect of COL17A1 on tumor growth was influenced by the tumor microenvironment. RNA-sequencing analysis of tumor tissue revealed effects of COL17A1 on gene expression profiles (including the expression of genes related to cell proliferation, the immune response, Wnt signaling, and Hippo signaling) that differed between C57BL/6-KPC and FVB-AK4.4 tumors. Our data thus suggest that COL17A1 promotes or suppresses cancer progression in a manner dependent on the interaction of tumor cells with the tumor microenvironment.
Collapse
Affiliation(s)
- Ryosuke Kashiwagi
- Department of Cell ProliferationART, Graduate School of Medicine, Tohoku UniversitySendaiJapan
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Ryo Funayama
- Department of Cell ProliferationART, Graduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Shuichi Aoki
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Aya Matsui
- Department of Vascular Physiology, Graduate School of Medical ScienceKanazawa UniversityKanazawaJapan
| | - Sebastian Klein
- PathologyUniversity Hospital CologneCologneGermany
- Radiation Oncology/Steele Laboratories for Tumor BiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Yukihiro Sato
- Department of Cell ProliferationART, Graduate School of Medicine, Tohoku UniversitySendaiJapan
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Tsubasa Suzuki
- Department of Cell ProliferationART, Graduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Keigo Murakami
- Department of Investigative Pathology, Graduate School of MedicineTohoku UniversitySendaiJapan
| | - Koetsu Inoue
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Masahiro Iseki
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Kunihiro Masuda
- Department of SurgerySouth Miyagi Medical CenterShibata‐gunJapan
| | - Masamichi Mizuma
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Hisamichi Naito
- Department of Vascular Physiology, Graduate School of Medical ScienceKanazawa UniversityKanazawaJapan
| | - Dan G. Duda
- Radiation Oncology/Steele Laboratories for Tumor BiologyMassachusetts General HospitalBostonMassachusettsUSA
| | - Michiaki Unno
- Department of SurgeryGraduate School of Medicine, Tohoku UniversitySendaiJapan
| | - Keiko Nakayama
- Department of Cell ProliferationART, Graduate School of Medicine, Tohoku UniversitySendaiJapan
| |
Collapse
|
48
|
Ratan C, Arian AM, Rajendran R, Jayakumar R, Masson M, Mangalathillam S. Nano-based formulations of curcumin: elucidating the potential benefits and future prospects in skin cancer. Biomed Mater 2023; 18:052008. [PMID: 37582394 DOI: 10.1088/1748-605x/acf0af] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/15/2023] [Indexed: 08/17/2023]
Abstract
Skin cancer refers to any malignant lesions that occur in the skin and are observed predominantly in populations of European descent. Conventional treatment modalities such as excision biopsy, chemotherapy, radiotherapy, immunotherapy, electrodesiccation, and photodynamic therapy (PDT) induce several unintended side effects which affect a patient's quality of life and physical well-being. Therefore, spice-derived nutraceuticals like curcumin, which are well tolerated, less expensive, and relatively safe, have been considered a promising agent for skin cancer treatment. Curcumin, a chemical constituent extracted from the Indian spice, turmeric, and its analogues has been used in various mammalian cancers including skin cancer. Curcumin has anti-neoplastic activity by triggering the process of apoptosis and preventing the multiplication and infiltration of the cancer cells by inhibiting some signaling pathways and thus subsequently preventing the process of carcinogenesis. Curcumin is also a photosensitizer and has been used in PDT. The major limitations associated with curcumin are poor bioavailability, instability, limited permeation into the skin, and lack of solubility in water. This will constrain the use of curcumin in clinical settings. Hence, developing a proper formulation that can ideally release curcumin to its targeted site is important. So, several nanoformulations based on curcumin have been established such as nanogels, nanoemulsions, nanofibers, nanopatterned films, nanoliposomes and nanoniosomes, nanodisks, and cyclodextrins. The present review mainly focuses on curcumin and its analogues as therapeutic agents for treating different types of skin cancers. The significance of using various nanoformulations as well non-nanoformulations loaded with curcumin as an effective treatment modality for skin cancer is also emphasized.
Collapse
Affiliation(s)
- Chameli Ratan
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041 Kerala, India
| | - Arya Mangalath Arian
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041 Kerala, India
| | - Rajalakshmi Rajendran
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041 Kerala, India
| | - Rangasamy Jayakumar
- Polymeric Biomaterials Lab, School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041 Kerala, India
| | - Mar Masson
- Faculty of Pharmaceutical Sciences, School of Health Sciences, University of Iceland, Hofsvallagata 53, IS-107, Reykjavík, Iceland
| | - Sabitha Mangalathillam
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences and Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041 Kerala, India
| |
Collapse
|
49
|
Wang J, Liu W, Zhang L, Zhang J. Targeting mutant p53 stabilization for cancer therapy. Front Pharmacol 2023; 14:1215995. [PMID: 37502209 PMCID: PMC10369794 DOI: 10.3389/fphar.2023.1215995] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023] Open
Abstract
Over 50% cancer bears TP53 mutation, the highly stabilized mutant p53 protein drives the tumorigenesis and progression. Mutation of p53 not only cause loss-of-function and dominant-negative effects (DNE), but also results in the abnormal stability by the regulation of the ubiquitin-proteasome system and molecular chaperones that promote tumorigenesis through gain-of-function effects. The accumulation of mutant p53 is mainly regulated by molecular chaperones, including Hsp40, Hsp70, Hsp90 and other biomolecules such as TRIM21, BAG2 and Stat3. In addition, mutant p53 forms prion-like aggregates or complexes with other protein molecules and result in the accumulation of mutant p53 in tumor cells. Depleting mutant p53 has become one of the strategies to target mutant p53. This review will focus on the mechanism of mutant p53 stabilization and discuss how the strategies to manipulate these interconnected processes for cancer therapy.
Collapse
Affiliation(s)
- Jiajian Wang
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Wenjun Liu
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Lanqing Zhang
- Medical School, Kunming University of Science and Technology, Kunming, China
| | - Jihong Zhang
- Medical School, Kunming University of Science and Technology, Kunming, China
- Yunnan Province Clinical Research Center for Hematologic Disease, Kunming, China
| |
Collapse
|
50
|
Alvarez-Rivera E, Ortiz-Hernández EJ, Lugo E, Lozada-Reyes LM, Boukli NM. Oncogenic Proteomics Approaches for Translational Research and HIV-Associated Malignancy Mechanisms. Proteomes 2023; 11:22. [PMID: 37489388 PMCID: PMC10366845 DOI: 10.3390/proteomes11030022] [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: 03/30/2023] [Revised: 06/09/2023] [Accepted: 06/29/2023] [Indexed: 07/26/2023] Open
Abstract
Recent advances in the field of proteomics have allowed extensive insights into the molecular regulations of the cell proteome. Specifically, this allows researchers to dissect a multitude of signaling arrays while targeting for the discovery of novel protein signatures. These approaches based on data mining are becoming increasingly powerful for identifying both potential disease mechanisms as well as indicators for disease progression and overall survival predictive and prognostic molecular markers for cancer. Furthermore, mass spectrometry (MS) integrations satisfy the ongoing demand for in-depth biomarker validation. For the purpose of this review, we will highlight the current developments based on MS sensitivity, to place quantitative proteomics into clinical settings and provide a perspective to integrate proteomics data for future applications in cancer precision medicine. We will also discuss malignancies associated with oncogenic viruses such as Acquire Immunodeficiency Syndrome (AIDS) and suggest novel mechanisms behind this phenomenon. Human Immunodeficiency Virus type-1 (HIV-1) proteins are known to be oncogenic per se, to induce oxidative and endoplasmic reticulum stresses, and to be released from the infected or expressing cells. HIV-1 proteins can act alone or in collaboration with other known oncoproteins, which cause the bulk of malignancies in people living with HIV-1 on ART.
Collapse
Affiliation(s)
- Eduardo Alvarez-Rivera
- Biomedical Proteomics Facility, Department of Microbiology and Immunology, Universidad Central del Caribe, School of Medicine, Bayamón, PR 00960, USA
| | - Emanuel J. Ortiz-Hernández
- Biomedical Proteomics Facility, Department of Microbiology and Immunology, Universidad Central del Caribe, School of Medicine, Bayamón, PR 00960, USA
| | - Elyette Lugo
- Biomedical Proteomics Facility, Department of Microbiology and Immunology, Universidad Central del Caribe, School of Medicine, Bayamón, PR 00960, USA
| | | | - Nawal M. Boukli
- Biomedical Proteomics Facility, Department of Microbiology and Immunology, Universidad Central del Caribe, School of Medicine, Bayamón, PR 00960, USA
| |
Collapse
|