1
|
Perera GS, Huang X, Bagherjeri FA, Joglekar CM, Leo P, Duijf P, Bhaskaran M, Sriram S, Punyadeera C. Rapid and selective detection of TP53 mutations in cancer using a novel conductometric biosensor. Biosens Bioelectron 2025; 276:117252. [PMID: 39978233 DOI: 10.1016/j.bios.2025.117252] [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: 11/19/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
Tumour protein p53 (TP53) is a tumour suppressor gene that is frequently mutated in cancers. Traditional TP53 detection methods, such as polymerase chain reactions, are time-consuming and demand skilled laboratory personnel. As an alternative, in the current study, we have demonstrated a high resistivity silicon (HR-Si) based conductometric biosensor designed for the rapid and specific identification of TP53 point mutations directly at the point-of-need. This biosensor accurately detected R248Q and R248W point mutant single strand DNA (ssDNA) as models, in real-time. Both R248Q and R248W mutant ssDNA exhibited a limit of detection (LOD) of 0.5 ng/mL in human plasma. The selectivity studies revealed that both R248Q and R248W mutant ssDNA can be detected 10 × lower molar content against their wild-type ssDNA. Validation of the sensor using clinical samples harbouring known TP53 mutations demonstrated a sensitivity of 100%, a specificity of 100%, and a LOD of 2.5 ng/mL. This precision biosensing platform at the point-of-need has the potential to revolutionise cancer diagnostics.
Collapse
Affiliation(s)
- Ganganath S Perera
- Functional Materials and Microsystems Research Group and the Micro Nano Research Facility, RMIT University, Melbourne, VIC, 3001, Australia.
| | - Xiaomin Huang
- Institute for Biomedicine and Glycomics (IBG), Griffith University, Nathan, QLD 4111, Australia.
| | - Fateme Akhlaghi Bagherjeri
- Functional Materials and Microsystems Research Group and the Micro Nano Research Facility, RMIT University, Melbourne, VIC, 3001, Australia
| | - Chinmayee Manesh Joglekar
- Functional Materials and Microsystems Research Group and the Micro Nano Research Facility, RMIT University, Melbourne, VIC, 3001, Australia
| | - Paul Leo
- Australian Translational Genomic Centre (ATGC), Queensland University of Technology, Woolloongabba, QLD, 4102, Australia
| | - Pascal Duijf
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, SA 5000, Australia
| | - Madhu Bhaskaran
- Functional Materials and Microsystems Research Group and the Micro Nano Research Facility, RMIT University, Melbourne, VIC, 3001, Australia
| | - Sharath Sriram
- Functional Materials and Microsystems Research Group and the Micro Nano Research Facility, RMIT University, Melbourne, VIC, 3001, Australia.
| | - Chamindie Punyadeera
- Institute for Biomedicine and Glycomics (IBG), Griffith University, Nathan, QLD 4111, Australia.
| |
Collapse
|
2
|
Jiang M, Wang S, Ji J, Baral S, Sun Q, Wang Y, Liu B, Ren J, Wang W, Wang D. PWP1 transcriptionally regulates p53, modulating apoptosis and cell cycle to promote gastric cancer progression. Apoptosis 2025; 30:693-709. [PMID: 39720977 PMCID: PMC11947051 DOI: 10.1007/s10495-024-02049-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] [Accepted: 11/18/2024] [Indexed: 12/26/2024]
Abstract
Gastric cancer remains a leading cause of cancer-related mortality worldwide. The prognosis often depends on early detection and understanding the molecular mechanisms involved in its progression. Periodic tryptophan protein 1 (PWP1) has emerged as a novel diagnostic marker, potentially linked to gastric cancer progression. This study aims to elucidate the impact of PWP1 on gastric cancer development, focusing on apoptosis, cell cycle regulation, and the role of p53. This study utilized gastric cancer cell lines to investigate the expression and functional role of Pwp1. Quantitative PCR and Western blot analyses were conducted to measure PWP1 expression levels. Apoptosis was assessed by using flow cytometry and TUNEL assays, and cell cycle analysis was performed to evaluate the impact of PWP1 modulation. Additionally, animal experiments were conducted using mouse models injected with gastric cancer cells, with PWP1 knockdown or overexpression, to observe tumor growth and progression. Statistical significance was evaluated using t-tests and ANOVA where appropriate. Elevated PWP1 expression was observed in gastric cancer tissues compared to normal tissues. PWP1's knockdown resulted in increased apoptosis and cell cycle arrest at the G1 phase, suggesting its role in promoting invasion and proliferation. Furthermore, animal experiments demonstrated reduced tumor growth in mice with PWP1 knockdown. PWP1 was found to transcriptionally regulate p53, affecting its expression and thereby influencing apoptosis and cell cycle pathways in gastric cancer. Our study identifies PWP1 as a novel oncogene frequently overexpressed in gastric cancer (GC). Through transcriptional regulation of p53, PWP1 enhances cell growth by influencing apoptosis and inducing G1 phase cell cycle arrest. These findings underscore PWP1 as a promising therapeutic target for treating GC, suggesting its potential for future clinical applications.
Collapse
Affiliation(s)
- Mingrui Jiang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- Medical College of Yangzhou University, Yangzhou, 225001, China
| | - Sen Wang
- The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Jin Ji
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- Medical College of Yangzhou University, Yangzhou, 225001, China
| | - Shantanu Baral
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- Medical College of Yangzhou University, Yangzhou, 225001, China
| | - Qiannan Sun
- Northern Jiangsu People's Hospital, Yangzhou, China
| | - Yong Wang
- Northern Jiangsu People's Hospital, Yangzhou, China
| | - Bin Liu
- Northern Jiangsu People's Hospital, Yangzhou, China
| | - Jun Ren
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- Northern Jiangsu People's Hospital, Yangzhou, China
| | - Wei Wang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
- Northern Jiangsu People's Hospital, Yangzhou, China
| | - Daorong Wang
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China.
- Northern Jiangsu People's Hospital, Yangzhou, China.
- General Surgery Institute of Yangzhou, Yangzhou University, Yangzhou, 225001, China.
| |
Collapse
|
3
|
Wu Q, Zhong L, Zhang G, Han L, Xie J, Xu Y. Complementing therapeutic strategies for acute myeloid leukemia: Signaling pathways and targets of traditional Chinese medicine. Leuk Res 2025; 151:107672. [PMID: 40022774 DOI: 10.1016/j.leukres.2025.107672] [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: 12/17/2024] [Revised: 02/16/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Leukemia is a heterogeneous malignant tumor of the hematopoietic system and is characterized by the blockage of differentiation and uncontrolled proliferation of myeloid or lymphoid progenitor cells in the bone marrow and peripheral blood. Currently, intensified chemotherapy regimens and hematopoietic stem cell transplantation (HSCT) are the most common treatment methods for various types of leukemia. However, they are associated with severe side effects and multidrug resistance. Therefore, developing new treatment approaches with sufficient therapeutic effects to eliminate leukemia cells and improve leukemia outcomes selectively is essential. Traditional Chinese Medicine (TCM) has received widespread attention as an alternative treatment for acute myeloid leukemia (AML) because of its multi-component and multi-target characteristics. Increasing evidence suggests that TCM blocks AML progression by regulating various biological processes. Herein, we review the effects of TCM therapies for AML and its potential mechanisms and targets. Our findings will promote further research and improve the clinical application of TCM in treating AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Medicine, Chinese Traditional/methods
- Signal Transduction/drug effects
- Drugs, Chinese Herbal/therapeutic use
- Animals
Collapse
Affiliation(s)
- Qiaoliang Wu
- Department of Hematology, The First People's Hospital of Jiashan, China
| | - Lei Zhong
- Department of Laboratory Medicine, Tongxiang Traditional Chinese Medicine Hospital, China
| | - Guibing Zhang
- Department of Hematology, The First People's Hospital of Fuyang, Hangzhou, China
| | - Liying Han
- Department of Laboratory Medicine, Tongxiang Traditional Chinese Medicine Hospital, China
| | - Jing Xie
- Department of Laboratory Medicine, Taizhou First People's Hospital, China
| | - Yao Xu
- Department of Pediatric Medicine, The First People's Hospital of Jiashan, China.
| |
Collapse
|
4
|
Gupta H, Singh A, Gupta A. Cancer-associated mutation at glycine 400 in TIP60 disrupt its phase separation property and catalytic activity resulting in compromised DNA damage repair function of the cell. Biochem Biophys Res Commun 2025; 753:151457. [PMID: 39965267 DOI: 10.1016/j.bbrc.2025.151457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/22/2025] [Accepted: 02/04/2025] [Indexed: 02/20/2025]
Abstract
TIP60 is a tumor suppressor with histone acetyltransferase (HAT) activity, playing a crucial role in regulating chromatin architecture by acetylating histones to enhance accessibility for other regulatory factors. Its function is vital for several key cellular processes, including DNA damage repair, apoptosis, and autophagy. While the downregulation of TIP60 has been associated with various cancers, the effects of naturally occurring mutations in TIP60 on its function in malignancies remain poorly understood. In this study, we explored how cancer-related mutations in TIP60 impact its structure and function. Several deleterious and destabilizing missense mutations were identified and analyzed for structural changes. Molecular dynamics simulations revealed alterations in protein conformational stability and radius of gyration due to these mutations, supported by significant changes in TIP60's solvent accessibility and intramolecular hydrogen bonding. Biochemical assays with recombinant proteins showed a loss of catalytic activity in the G400W mutant. Live cell imaging indicated abnormal localization of the G400W mutant within the nucleus. Additionally, we observed aberrant phase separation of TIP60 caused by the G400W mutation. Notably, the G400W mutation impairs TIP60's catalytic function, preventing effective DNA repair and leaving the genome vulnerable to further mutations. Our findings highlight cancer-associated mutations in TIP60 that may contribute to the molecular mechanisms underlying cancer initiation and progression.
Collapse
Affiliation(s)
- Himanshu Gupta
- Epigenetics and Human Disease Laboratory, Centre of Excellence in Epigenetics, Department of Life Sciences, Shiv Nadar Institution of Eminence, Deemed to be University, Delhi-NCR, Uttar Pradesh, India, 201314
| | - Ashutosh Singh
- Department of Life Sciences, Shiv Nadar Institution of Eminence, Deemed to be University, Delhi-NCR, Uttar Pradesh, India, 201314
| | - Ashish Gupta
- Epigenetics and Human Disease Laboratory, Centre of Excellence in Epigenetics, Department of Life Sciences, Shiv Nadar Institution of Eminence, Deemed to be University, Delhi-NCR, Uttar Pradesh, India, 201314.
| |
Collapse
|
5
|
Ozturk H, Seker-Polat F, Abbaszadeh N, Kingham Y, Orsulic S, Adli M. High PRMT5 levels, maintained by KEAP1 inhibition, drive chemoresistance in high-grade serous ovarian cancer. J Clin Invest 2025; 135:e184283. [PMID: 40091834 PMCID: PMC11910213 DOI: 10.1172/jci184283] [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: 06/28/2024] [Accepted: 01/16/2025] [Indexed: 03/19/2025] Open
Abstract
Protein arginine methyl transferases (PRMTs) are generally upregulated in cancers. However, the mechanisms leading to this upregulation and its biological consequences are poorly understood. Here, we identify PRMT5, the main symmetric arginine methyltransferase, as a critical driver of chemoresistance in high-grade serous ovarian cancer (HGSOC). PRMT5 levels and its enzymatic activity are induced in a platinum-resistant (Pt-resistant) state at the protein level. To reveal potential regulators of high PRMT5 protein levels, we optimized intracellular immunostaining conditions and performed unbiased CRISPR screening. We identified Kelch-like ECH-associated protein 1 (KEAP1) as a top-scoring negative regulator of PRMT5. Our mechanistic studies show that KEAP1 directly interacted with PRMT5, leading to its ubiquitin-dependent degradation under normal physiological conditions. At the genomic level, ChIP studies showed that elevated PRMT5 directly interacted with the promoters of stress response genes and positively regulated their transcription. Combined PRMT5 inhibition with Pt resulted in synergistic cellular cytotoxicity in vitro and reduced tumor growth in vivo in Pt-resistant patient-derived xenograft tumors. Overall, the findings from this study identify PRMT5 as a critical therapeutic target in Pt-resistant HGSOC cells and reveal the molecular mechanisms that lead to high PRMT5 levels in Pt-treated and chemo-resistant tumors.
Collapse
Affiliation(s)
- Harun Ozturk
- Robert Lurie Comprehensive Cancer Center, Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois, USA
| | - Fidan Seker-Polat
- Robert Lurie Comprehensive Cancer Center, Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois, USA
| | - Neda Abbaszadeh
- Robert Lurie Comprehensive Cancer Center, Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois, USA
| | - Yasemin Kingham
- Robert Lurie Comprehensive Cancer Center, Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois, USA
| | - Sandra Orsulic
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Mazhar Adli
- Robert Lurie Comprehensive Cancer Center, Department of Obstetrics and Gynecology, Feinberg School of Medicine at Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
6
|
Gesztes WR, Lap CJ, Rajendran R, Dalivand MM, Diao G, Liu S, Jain M, Nava VE. Investigating Intensity and Percentage of p53 Nuclear Expression in Prostate Cancer: Findings from a Cohort of U.S. Military Veterans. Cancers (Basel) 2025; 17:1004. [PMID: 40149338 DOI: 10.3390/cancers17061004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Next-generation sequencing has revealed TP53 alterations in localized prostate cancer (PCa), suggesting growing clinical potential for p53 immunohistochemistry (IHC). Prior research supports the use of IHC for the detection of p53 overexpression to predict the presence of TP53 alterations known to be associated with adverse outcomes. However, to reach a consensus definition of p53 overexpression in PCa, further insights are needed. This study aimed to compare two fundamental approaches of evaluating p53 expression across a variety of specimens regarding PCa progression. METHODS This study included 84 patients (75% self-identified as African American) diagnosed with PCa between 1996 and 2021 at the DC VA Medical Center. Representative sections of core biopsies, radical prostatectomies, transurethral prostate resections, and metastatic deposits were examined. p53 nuclear expression was scored according to the highest intensity observed (0, 1+, 2+, 3+) and the percentage (0%, <1%, 1-5%, >5%) of tumor cells expressing any level of intensity in the aggregate tumor area. All slides were reviewed by two independent pathologists. Pertinent clinical data were collected. RESULTS A total of 34 patients (40%) exhibited p53 nuclear expression, of which 18 (21%) showed the maximum (3+) intensity. The presence of maximum intensity, regardless of percentage, was found to be associated with Grade Group (p < 0.001), higher PSA at biopsy (p < 0.001), BCR (p < 0.001) and metastasis (p < 0.001). Importantly, maximum p53 intensity was identified only in patients who developed metastatic disease. CONCLUSIONS Maximum (3+) p53 nuclear intensity of any percentage is highly associated with disease progression in PCa, suggesting that optimal determination of p53 overexpression should incorporate intensity.
Collapse
Affiliation(s)
- William R Gesztes
- Department of Pathology, Washington DC VA Medical Center, Washington, DC 20422, USA
- Department of Pathology, The George Washington University Hospital, Washington, DC 20037, USA
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Coen J Lap
- The Edward P. Evans Precision Oncology Center of Excellence, Washington DC VA Medical Center, Washington, DC 20422, USA
- Department of Hematology and Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC 20007, USA
| | - Rithika Rajendran
- Department of Pathology, Washington DC VA Medical Center, Washington, DC 20422, USA
| | - Maryam M Dalivand
- Department of Pathology, Washington DC VA Medical Center, Washington, DC 20422, USA
- Department of Pathology, The George Washington University Hospital, Washington, DC 20037, USA
| | - Guoqing Diao
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20037, USA
| | - Shanshan Liu
- Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20037, USA
| | - Maneesh Jain
- The Edward P. Evans Precision Oncology Center of Excellence, Washington DC VA Medical Center, Washington, DC 20422, USA
| | - Victor E Nava
- Department of Pathology, Washington DC VA Medical Center, Washington, DC 20422, USA
- Department of Pathology, The George Washington University Hospital, Washington, DC 20037, USA
| |
Collapse
|
7
|
Shi L, Wang H, Sun Y, Xu N, Pei A, Zhang N. Development and validation of a disulfidptosis-related prognostic model for colorectal cancer using multi-omics analysis. Discov Oncol 2025; 16:338. [PMID: 40095116 PMCID: PMC11914417 DOI: 10.1007/s12672-025-02055-8] [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: 09/14/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
This study aims to integrate multi-omic and clinical data concerning disulfidptosis-related genes (DRGs) to facilitate molecular typing and prognosis in colorectal cancer (CRC). Public databases provided CRC transcriptome and clinical data, enabling differential expression, genomic analyses, pathway enrichment, survival analysis, and subtyping based on the expression levels of 15 DRGs identified in published studies. Differentially expressed genes (DEGs) between subtypes were identified to create a disulfidptosis prognostic model using LASSO and Cox regression analyses. This model was evaluated by comparing risk scores, survival curves, cellular infiltration, and drug sensitivity between high- and low-risk groups. Analyses revealed differential expression, mutations, and copy number variations (CNV) in DRGs in CRC. Survival analysis demonstrated significant prognostic differences among DRG expression subtypes. GSVA and ssGSEA highlighted DRGs' regulatory roles in CRC. DEGs identified between DRG expression subtypes led to the classification into subtypes A and B. A disulfidptosis prognostic model, including genes VSIG4, SCG2, INHBB, DDC, CXCL13, KLK10, CXCL10, and CCL11A, was developed to stratify patients into high- and low-risk groups. This model displayed strong predictive capability (AUC = 0.700) and calibration. The risk score was also strongly associated with immune cell infiltration, stromal cell score, and stem cell index in the CRC tumor microenvironment. Drug sensitivity analysis indicated that high-risk samples were more responsive to most medications. We established a robust disulfidptosis prognostic model for CRC through comprehensive multi-omics analysis. Our findings provide valuable insights into the role of DRGs in CRC progression and disease management, presenting an important resource for further research.
Collapse
Affiliation(s)
- Lei Shi
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China
| | - Huimei Wang
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China
| | - Yongxiao Sun
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China
| | - Na Xu
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China
| | - Aiyue Pei
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China.
| | - Nan Zhang
- Gastroenterology Department & Endoscopic Center, The First Bethune Hospital of Jilin University, 1 Xinmin Street, Changchun City, 130021, China.
| |
Collapse
|
8
|
Bao ZC, Zhang Y, Liu ZD, Dai HJ, Ren F, Li N, Lv SY, Zhang Y. Tetrahydrocurcumin-induced apoptosis of hepatocellular carcinoma cells involves the TP53 signaling pathway, as determined by network pharmacology. World J Gastrointest Oncol 2025; 17:101174. [PMID: 40092919 PMCID: PMC11866214 DOI: 10.4251/wjgo.v17.i3.101174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/04/2024] [Accepted: 01/16/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a malignant disease with high incidence and mortality worldwide. This study focuses on the TP53 target protein to investigate the potential therapeutic effect of tetrahydrocurcumin (THC) on HCC and its mechanism of action. The research hypothesis is that THC can inhibit the proliferation, migration, and invasion of HCC cells, and promote their apoptosis by regulating the TP53 target protein. AIM To explore the mechanism by which THC inhibits HCC cell proliferation via the TP53 signaling pathway. METHODS Potential targets of THC and HCC were identified from multiple databases. The core targets were subjected to analyses using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes databases, and visualization processing, using the online platform Metascape to identify the key molecules and signaling pathways involved in the action of THC against HCC. The molecular mechanisms of action of THC against TP53 in the inhibition of HCC cells were verified using cell counting kit-8, Transwell, apoptosis, and western blotting assays. RESULTS Molecular docking results showed that THC had a high score for the TP53 target protein. In vitro experiments indicated that THC effectively inhibited the proliferation and migration of HCC cells, and affected the expression levels of TP53, MDM2, cyclin B, Bax, Bcl-2, caspase-9, and caspase-3. CONCLUSION THC induces the apoptosis of HCC cells through the TP53 signaling pathway, thereby inhibiting their proliferation and migration.
Collapse
Affiliation(s)
- Zhuo-Cong Bao
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Ye Zhang
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Zhao-Dong Liu
- Graduate School, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Hui-Jun Dai
- Guangxi Medical University Cancer Hospital, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Fu Ren
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Ning Li
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Department of Biochemistry, School of Basic Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Shang-Yu Lv
- Department of Clinical Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| | - Yan Zhang
- Key Laboratory of Human Ethnic Specificity and Phenomics of Critical Illness in Liaoning Province, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Key Laboratory of Phenomics in Shenyang, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- Department of Biochemistry, School of Basic Medicine, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
- International Education School, International Exchange and Cooperation Office, Shenyang Medical College, Shenyang 110034, Liaoning Province, China
| |
Collapse
|
9
|
García-Molina J, Saiz-Vázquez O, Santamaría-Vázquez M, Ortiz-Huerta JH. Efficacy of a Supervised Exercise Program on Pain, Physical Function, and Quality of Life in Patients With Breast Cancer: Protocol for a Randomized Clinical Trial. JMIR Res Protoc 2025; 14:e63891. [PMID: 40073395 PMCID: PMC11947629 DOI: 10.2196/63891] [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: 07/05/2024] [Revised: 09/30/2024] [Accepted: 01/10/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Breast cancer is the second most common cancer in women worldwide. Treatments for this disease often result in side effects such as pain, fatigue, loss of muscle mass, and reduced quality of life. Physical exercise has been shown to effectively mitigate these side effects and improve the quality of life in patients with breast cancer. OBJECTIVE This randomized clinical trial aims to evaluate the efficacy of a 12-week supervised exercise program on pain, physical function, and quality of life in female patients with cancer. METHODS This randomized, double-blind clinical trial will recruit 325 participants, divided into an intervention group receiving the exercise program and a control group receiving standard care recommendations. Outcome measures, including pain (assessed via the Brief Pain Inventory), physical function (Disability of the Arm, Shoulder, and Hand Questionnaire), and quality of life (European Organization for Research and Treatment of Cancer QLQ-C30 and European Organization for Research and Treatment of Cancer QLQ-BR23), will be evaluated at baseline, immediately post intervention, and 12 weeks post intervention. Statistical analysis will involve repeated measures of ANOVA and MANOVA to determine the significance of the intervention's effects across time points. RESULTS Recruitment and data collection will commence in February of 2025, and data analysis is scheduled for completion at the end of 2025. No results are currently available. CONCLUSIONS Physical exercise is anticipated to play a significant role in alleviating pain, enhancing physical function, and improving the quality of life in female patients with cancer. This study will provide robust evidence to support the integration of supervised exercise into standard care protocols for this population. TRIAL REGISTRATION ClinicalTrials.gov NCT06618690; https://clinicaltrials.gov/ct2/show/NCT06618690. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) PRR1-10.2196/63891.
Collapse
Affiliation(s)
- Jennifer García-Molina
- Paseo de los Encomendadores, Faculty of Health Sciences, University of Burgos, Burgos, Spain
| | - Olalla Saiz-Vázquez
- Paseo de los Encomendadores, Faculty of Health Sciences, University of Burgos, Burgos, Spain
| | | | | |
Collapse
|
10
|
Lock IC, Leisenring NH, Floyd W, Xu ES, Luo L, Ma Y, Mansell EC, Cardona DM, Lee CL, Kirsch DG. Mis-splicing drives loss of function of p53E224D point mutation. PLoS One 2025; 20:e0318856. [PMID: 40043089 PMCID: PMC11882087 DOI: 10.1371/journal.pone.0318856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/22/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND The tumor suppressor p53 (Trp53), also known as p53, is the most commonly mutated gene in cancer. Canonical p53 DNA damage response pathways are well characterized and classically thought to underlie the tumor suppressive effect of p53. Challenging this dogma, mouse models have revealed that p53-driven apoptosis and cell cycle arrest are dispensable for tumor suppression. Here, we investigated the inverse context of a p53 mutation predicted to drive the expression of canonical targets but is detected in human cancer. METHODS We established a novel mouse model with a single base pair mutation (GAG>GAT, p53E221D) in the DNA-Binding domain that has wild-type function in screening assays, but is paradoxically found in human cancer in Li-Fraumeni syndrome. Using mouse p53E221D and the analogous human p53E224D mutants, we evaluated expression, transcriptional activation, and tumor suppression in vitro and in vivo. RESULTS Expression of human p53E224D from cDNA translated to a fully functional p53 protein. However, p53E221D/E221D RNA transcribed from the endogenous locus is mis-spliced resulting in nonsense-mediated decay. Moreover, fibroblasts derived from p53E221D/E221D mice do not express a detectable protein product. Mice homozygous for p53E221D exhibited increased tumor penetrance and decreased life expectancy compared to p53WT/WT animals. CONCLUSIONS Mouse p53E221D and human p53E224D mutations lead to splice variation and a biologically relevant p53 loss of function in vitro and in vivo.
Collapse
Affiliation(s)
- Ian C. Lock
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Nathan H. Leisenring
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Dermatology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Warren Floyd
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Radiation Oncology, Maryland Anderson Cancer Center, Houston, Texas, United States of America
| | - Eric S. Xu
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Lixia Luo
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Yan Ma
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Erin C. Mansell
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Diana M. Cardona
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Chang-Lung Lee
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - David G. Kirsch
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Radiation Oncology, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
11
|
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
|
12
|
Chen YJ, Liao SW, Lai YL, Li YF, Lu YC, Tai CK. Epigenetic downregulation of the proapoptotic gene HOXA5 in oral squamous cell carcinoma. Mol Med Rep 2025; 31:56. [PMID: 39704209 DOI: 10.3892/mmr.2024.13421] [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: 07/16/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
Homeobox A5 (HOXA5) has been identified as a tumor suppressor gene in breast cancers, but its role in oral squamous cell carcinoma (OSCC) has not been confirmed. The Illumina GoldenGate Assay for methylation identified that DNA methylation patterns differ between tumorous and normal tissues in the oral cavity and that HOXA5 is one of the genes that are hypermethylated in oral tumor tissues. The present study obtained more‑complete information on the methylation status of HOXA5 by using the Illumina Infinium MethylationEPIC BeadChip and bisulfite sequencing assays. The results indicated that HOXA5 hypermethylation has great potential as a biomarker for detecting OSCC. Comparing HOXA5 RNA expression between normal oral tissue and OSCC tissue samples indicated that its median level was 2.06‑fold higher in normal tissues that in OSCC tissues. Moreover, treatment using the demethylating agent 5‑aza‑2'‑deoxycytidine can upregulate HOXA5 expression in OSCC cell lines, verifying that the silencing of HOXA5 is primarily regulated by its hypermethylation. It was also found that upregulation of HOXA5 expression can not only increase OSCC cell death but that it can also enhance the therapeutic effect of cisplatin both in vitro and in vivo, suggesting that HOXA5 is an epigenetically downregulated proapoptotic gene in OSCC.
Collapse
Affiliation(s)
- Ying-Ju Chen
- Department of Biomedical Sciences, National Chung Cheng University, Chia‑Yi 62102, Taiwan, R.O.C
| | - Shin-Wei Liao
- Department of Biomedical Sciences, National Chung Cheng University, Chia‑Yi 62102, Taiwan, R.O.C
| | - Yen-Ling Lai
- Department of Biomedical Sciences, National Chung Cheng University, Chia‑Yi 62102, Taiwan, R.O.C
| | - Yu-Fen Li
- Department of Public Health, China Medical University, Taichung 40402, Taiwan, R.O.C
| | - Yin-Che Lu
- Division of Hematology‑Oncology, Ditmanson Medical Foundation Chia‑Yi Christian Hospital, Chia‑Yi 60002, Taiwan, R.O.C
| | - Chien-Kuo Tai
- Department of Biomedical Sciences, National Chung Cheng University, Chia‑Yi 62102, Taiwan, R.O.C
| |
Collapse
|
13
|
Kim HY, Shin S, Lee JM, Kim IS, Kim B, Kim HJ, Choi YJ, Bae B, Kim Y, Ji E, Kim H, Kim H, Lee JS, Chang YH, Kim HK, Lee JY, Yu S, Kim M, Cho YU, Jang S, Kim M. TP53 Mutation Status in Myelodysplastic Neoplasm and Acute Myeloid Leukemia: Impact of Reclassification Based on the 5th WHO and International Consensus Classification Criteria: A Korean Multicenter Study. Ann Lab Med 2025; 45:160-169. [PMID: 39497415 PMCID: PMC11788706 DOI: 10.3343/alm.2024.0351] [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: 07/07/2024] [Revised: 08/05/2024] [Accepted: 10/24/2024] [Indexed: 01/24/2025] Open
Abstract
Background TP53 mutations are associated with poor prognosis in myelodysplastic neoplasm (MDS) and AML. The updated 5th WHO classification and International Consensus Classification (ICC) categorize TP53-mutated MDS and AML as unique entities. We conducted a multicenter study in Korea to investigate the characteristics of TP53-mutated MDS and AML, focusing on diagnostic aspects based on updated classifications. Methods This study included patients aged ≥ 18 yrs who were diagnosed as having MDS (N=1,244) or AML (N=2,115) at six institutions. The results of bone marrow examination, cytogenetic studies, and targeted next-generation sequencing, including TP53, were collected and analyzed. Results TP53 mutations were detected in 9.3% and 9.2% of patients with MDS and AML, respectively. Missense mutation was the most common, with hotspot codons R248/R273/G245/Y220/R175/C238 accounting for 25.4% of TP53 mutations. Ten percent of patients had multiple TP53 mutations, and 78.4% had a complex karyotype. The median variant allele frequency (VAF) of TP53 mutations was 41.5%, with a notable difference according to the presence of a complex karyotype. According to the 5th WHO classification and ICC, the multi-hit TP53 mutation criteria were met in 58.6% and 75% of MDS patients, respectively, and the primary determinants were a TP53 VAF >50% for the 5th WHO classification and the presence of a complex karyotype for the ICC. Conclusions Collectively, we elucidated the molecular genetic characteristics of patients with TP53-mutated MDS and AML, highlighting key factors in applying TP53 mutation-related criteria in updated classifications, which will aid in establishing diagnostic strategies.
Collapse
Affiliation(s)
- Hyun-Young Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Saeam Shin
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jong-Mi Lee
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - In-Suk Kim
- Department of Laboratory Medicine, Pusan National University School of Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Boram Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee-Jin Kim
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yu Jeong Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Byunggyu Bae
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yonggoo Kim
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eunhui Ji
- Department of Laboratory Medicine, Pusan National University School of Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Hyerin Kim
- Department of Laboratory Medicine, Pusan National University School of Medicine, Pusan National University Hospital, Busan, Korea
| | - Hyerim Kim
- Department of Laboratory Medicine, Pusan National University School of Medicine, Pusan National University Hospital, Busan, Korea
| | - Jee-Soo Lee
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Yoon Hwan Chang
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Kyung Kim
- Department of Laboratory Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Ja Young Lee
- Department of Laboratory Medicine, Inje University Busan Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Shinae Yu
- Department of Laboratory Medicine, Inje University Haeundae Paik Hospital, Inje University College of Medicine, Busan, Korea
| | - Miyoung Kim
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Young-Uk Cho
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seongsoo Jang
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Myungshin Kim
- Department of Laboratory Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
14
|
Ma X, Xu J, Wang Y, Fleishman JS, Bing H, Yu B, Li Y, Bo L, Zhang S, Chen ZS, Zhao L. Research progress on gene mutations and drug resistance in leukemia. Drug Resist Updat 2025; 79:101195. [PMID: 39740374 DOI: 10.1016/j.drup.2024.101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/05/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025]
Abstract
Leukemia is a type of blood cancer characterized by the uncontrolled growth of abnormal cells in the bone marrow, which replace normal blood cells and disrupt normal blood cell function. Timely and personalized interventions are crucial for disease management and improving survival rates. However, many patients experience relapse following conventional chemotherapy, and increasing treatment intensity often fails to improve outcomes due to mutated gene-induced drug resistance in leukemia cells. This article analyzes the association of gene mutations and drug resistance in leukemia. It explores genetic abnormalities in leukemia, highlighting recently identified mutations affecting signaling pathways, cell apoptosis, epigenetic regulation, histone modification, and splicing mechanisms. Additionally, the article discusses therapeutic strategies such as molecular targeting of gene mutations, alternative pathway targeting, and immunotherapy in leukemia. These approaches aim to combat specific drug-resistant mutations, providing potential avenues to mitigate leukemia relapse. Future research with these strategies holds promise for advancing leukemia treatment and addressing the challenges of drug-resistant mutations to improve patient outcomes.
Collapse
Affiliation(s)
- Xiangyu Ma
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Jiamin Xu
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Yanan Wang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA
| | - Hao Bing
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Boran Yu
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Yanming Li
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Letao Bo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA
| | - Shaolong Zhang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA.
| | - Libo Zhao
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
15
|
Rahmé R, Resnick-Silverman L, Anguiano V, Campbell MJ, Fenaux P, Manfredi JJ. Mutant p53 regulates a distinct gene set by a mode of genome occupancy that is shared with wild type. EMBO Rep 2025; 26:1315-1343. [PMID: 39875582 PMCID: PMC11893899 DOI: 10.1038/s44319-025-00375-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 01/05/2025] [Accepted: 01/14/2025] [Indexed: 01/30/2025] Open
Abstract
To directly examine the interplay between mutant p53 or Mdm2 and wild type p53 in gene occupancy and expression, an integrated RNA-seq and ChIP-seq analysis was performed in vivo using isogenically matched mouse strains. Response to radiation was used as an endpoint to place findings in a biologically relevant context. Unexpectedly, mutant p53 and Mdm2 only inhibit a subset of wild type p53-mediated gene expression. In contrast to a dominant-negative or inhibitory role, the presence of either mutant p53 or Mdm2 actually enhances the occupancy of wild type p53 on many canonical targets. The C-terminal 19 amino acids of wild type p53 suppress the p53 response allowing for survival at sublethal doses of radiation. Further, the p53 mutant 172H is shown to occupy genes and regulate their expression via non-canonical means that are shared with wild type p53. This results in the heterozygous 172H/+ genotype having an expanded transcriptome compared to wild type p53 + /+.
Collapse
Affiliation(s)
- Ramy Rahmé
- Department of Oncological Sciences and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Institut de Recherche Saint Louis (IRSL), INSERM U1131, Université de Paris, Paris, France
- Ecole Doctorale Hématologie-Oncogenèse-Biothérapies, Université de Paris, Paris, France
| | - Lois Resnick-Silverman
- Department of Oncological Sciences and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Vincent Anguiano
- Department of Oncological Sciences and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Pierre Fenaux
- Institut de Recherche Saint Louis (IRSL), INSERM U1131, Université de Paris, Paris, France
- Service Hématologie Seniors, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université de Paris, Paris, France
| | - James J Manfredi
- Department of Oncological Sciences and Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- The Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
16
|
Saito Y, Hoshi Y, Sato M, Seino M, Watanabe N, Kawai M, Suzuki S. Clinical Characteristics and Chemosensitivity in Germline TP53 Pathogenic Variant Cases Identified by Cancer Genomic Testing. Cancer Genomics Proteomics 2025; 22:354-362. [PMID: 39993798 PMCID: PMC11880932 DOI: 10.21873/cgp.20506] [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: 11/18/2024] [Revised: 12/12/2024] [Accepted: 01/08/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND/AIM The widespread implementation of cancer genomic profiling (CGP) has led to an increase in the detection of germline TP53 pathogenic variants (gTP53v) in patients who do not meet the classical Li-Fraumeni syndrome (LFS) criteria. The present study aimed to characterize the clinical features and treatment outcomes of gTP53v cases identified through routine CGP testing. PATIENTS AND METHODS We conducted a retrospective analysis of 43 patients with gTP53v identified through CGP testing between June 2019 and August 2024. Clinical characteristics, molecular features, and treatment outcomes were analyzed and compared with TP53 wild-type cases from the same database (n=6,515). RESULTS The median age at diagnosis was 38 years (range=1-83 years), with 58.1% of cases presenting with non-core LFS tumors. A genomic analysis revealed diverse variant types (missense: 32, frameshift: 8, and nonsense: 3) with variant allele frequencies ranging between 0.10 and 0.696. Among 37 patients who received first-line chemotherapy, the objective response rate was 62%, which was significantly higher than in TP53 wild-type cases (32%, p=0.02). Complete responses were observed in six patients and partial responses in 14. CONCLUSION The present results suggest that gTP53v carriers identified through CGP represent a broader clinical spectrum than classical LFS, while demonstrating potentially favorable treatment outcomes. These results challenge traditional paradigms and emphasize the need for individualized approaches to patient care, particularly in cases with atypical presentations requiring the careful interpretation of mosaicism, de novo mutations, and clonal hematopoiesis.
Collapse
Affiliation(s)
- Yosuke Saito
- Department of Gastroenterology, Yamagata City Hospital Saiseikan, Yamagata, Japan
| | - Yuki Hoshi
- Department of Genetic Counseling, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Masamichi Sato
- Department of Respiratory Medicine, Okitama General Hospital, Kawanishi, Japan
- Yamagata Hereditary Tumor Research Center, Yamagata University School of Medicine, Yamagata, Japan
| | - Manabu Seino
- Yamagata Hereditary Tumor Research Center, Yamagata University School of Medicine, Yamagata, Japan
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Norikazu Watanabe
- Yamagata Hereditary Tumor Research Center, Yamagata University School of Medicine, Yamagata, Japan
- Department of Obstetrics and Gynecology, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Masaaki Kawai
- Yamagata Hereditary Tumor Research Center, Yamagata University School of Medicine, Yamagata, Japan
- Department of Surgery I, Yamagata University Faculty of Medicine, Yamagata, Japan
| | - Shuhei Suzuki
- Yamagata Hereditary Tumor Research Center, Yamagata University School of Medicine, Yamagata, Japan;
- Department of Clinical Oncology, Yamagata Prefectural Shinjo Hospital, Shinjo, Japan
| |
Collapse
|
17
|
Cai BH, Wang YT, Chen CC, Yeh FY, Lin YR, Lin YC, Wu TY, Wu KY, Lien CF, Shih YC, Shaw JF. Chlorophyllides repress gain-of-function p53 mutated HNSCC cell proliferation via activation of p73 and repression of p53 aggregation in vitro and in vivo. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167662. [PMID: 39788216 DOI: 10.1016/j.bbadis.2025.167662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 12/14/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC) cells have a high p53 mutation rate, but there were rare reported about the p53 gain of function through the prion-like aggregated form in p53 mutated HNSCC cells. Thioflavin T (ThT) is used to stain prion-like proteins in cells. Previously, we found that ThT and p53 staining were co-localized in HNSCC cells (Detroit 562 cells) with homozygous p53 R175H mutation. NAMPT inhibitor can repress ThT staining in Detroit 562 cells. In our previous study, co-treatment with p73 activator NSC59984 and NAMPT inhibitor FK886 synergistically repressed Detroit 562 cell proliferation. In this study, we found that two heterozygous p53-R280T mutation HNSCC cell lines, TW01 and HONE-1, also have the ThT staining signal. Treatment with chlorophyllides and p73 activator or NAMPT inhibitor did not synergistically repress cell proliferation in either Detroit 562 or HONE-1 cells. Chlorophyllides reduced the ThT aggregation signal in both Detroit 562 and HONE-1 cells. Chlorophyllides also induced p73 and caspase 3/7 expression and repressed NAMPT expression in both Detroit 562 and HONE-1 cells. Chlorophyllides reduced tumor size in vivo in Detroit 562 cells injected into a xenograft nude mice model, but this in vivo tumor repression effect was not found in p73 knockdown Detroit 562 cells. Moreover, NAMPT was repressed by chlorophyllides independent of p73 status in vivo. We thus concluded that chlorophyllides have a dual anticancer function when applied to HNSCC cells with p53 gain-of-function mutation, via activation of p73 and repression of p53 aggregation.
Collapse
Affiliation(s)
- Bi-He Cai
- School of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan.
| | - Yi-Ting Wang
- Department of Medical Science and Biotechnology, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Chia-Chi Chen
- School of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan; Department of Pathology, E-Da Hospital, Kaohsiung City 82445, Taiwan
| | - Fang-Yu Yeh
- School of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Yu-Rou Lin
- School of Medicine, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Ying-Chen Lin
- Department of Medical Laboratory Science, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Tze-You Wu
- Department of Biomedical Engineering, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Kuan-Yo Wu
- Department of Medical Science and Biotechnology, I-Shou University, Kaohsiung City 82445, Taiwan
| | - Ching-Feng Lien
- Department of Otolaryngology-Head and Neck Surgery, E-Da Hospital, Kaohsiung City 82445, Taiwan
| | - Yu-Chen Shih
- Department of Otolaryngology-Head and Neck Surgery, E-Da Hospital, Kaohsiung City 82445, Taiwan.
| | - Jei-Fu Shaw
- Department of Medical Science and Biotechnology, I-Shou University, Kaohsiung City 82445, Taiwan.
| |
Collapse
|
18
|
Narasimhamurthy M, Le A, Boruah N, Moses R, Kelly G, Bleiweiss I, Maxwell KN, Nayak A. Clinicopathologic Features of Breast Tumors in Germline TP53 Variant-Associated Li-Fraumeni Syndrome. Am J Surg Pathol 2025; 49:195-205. [PMID: 39629784 DOI: 10.1097/pas.0000000000002338] [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: 02/20/2025]
Abstract
We present one of the largest cohorts of TP53 -pathogenic germline variants (PGVs) associated with patients with Li-Fraumeni syndrome (n = 82) with breast tumors (19 to 76 y; median age: 35). Most had missense variants (77%), followed by large gene rearrangements (LGRs; 12%), truncating (6%), and splice-site (5%) variants. Twenty-one unique germline missense variants were found, with hotspots at codons 175, 181, 245, 248, 273, 334, and 337. Of 100 total breast tumors, 63% were invasive (mostly ductal), 30% pure ductal carcinoma in situ, 4% fibroepithelial lesions, and 3% with unknown histology. Unlike BRCA -associated tumors, approximately half of the breast cancers exhibited HER2 -positivity, of which ~50% showed estrogen receptor coexpression. Pathology slides were available for review for 61 tumors (44 patients), and no significant correlation between the type of TP53 PGVs and histologic features was noted. High p53 immunohistochemistry expression (>50%) was seen in 67% of tumors tested (mostly missense variant). Null pattern (<1% cells) was seen in 2 (LGR and splicing variants carriers). Surprisingly, 2 tumors from patients with an LGR and 1 tumor from a patient with a truncating variant showed p53 overexpression (>50%). The subset of patients with the Brazilian p.R337H variant presented at a higher age than those with non-p.R337H variant (46 vs 35 y) though statistically insignificant ( P = 0.071) due to an imbalance in the sample size, and were uniquely negative for HER2 -overexpressing tumors. To conclude, breast cancer in carriers of TP53 PGVs has some unique clinicopathological features that suggest differential mechanisms of tumor formation. p53 immunohistochemistry cannot be used as a surrogate marker to identify germline TP53 -mutated breast cancers.
Collapse
Affiliation(s)
| | - Anh Le
- Department of Medicine, Division of Hematology/Oncology
| | | | - Renyta Moses
- Department of Medicine, Division of Hematology/Oncology
| | - Gregory Kelly
- Department of Medicine, Division of Hematology/Oncology
| | | | - Kara N Maxwell
- Department of Medicine, Division of Hematology/Oncology, University of Pennsylvania Health System
| | - Anupma Nayak
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Health System, Philadelphia, PA
| |
Collapse
|
19
|
Standing S, Malkin D, Johnston DL. A Unique Case of a Pediatric Patient with Six Childhood Cancers in Association with a Germline TP53 Gene Pathogenic Variant. Pediatr Blood Cancer 2025; 72:e31487. [PMID: 39702904 DOI: 10.1002/pbc.31487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 11/29/2024] [Indexed: 12/21/2024]
Affiliation(s)
| | - David Malkin
- Division of Hematology/Oncology, Department of Pediatircs, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Donna L Johnston
- Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Division of Hematology/Oncology, Children's Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| |
Collapse
|
20
|
Hariri D, Pavlick D, Spiess P, Li R, Kamat A, Grivas P, Agarwal N, Gupta S, Necchi A, Bratslavsky G, Basnet A, Jacob J, Ross J, Kravtsov O. Primary sarcomas of the bladder and prostate: A genomic landscape study. Pathol Res Pract 2025; 267:155840. [PMID: 39965403 DOI: 10.1016/j.prp.2025.155840] [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: 12/20/2024] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
Primary sarcomas of the bladder and prostate are exceedingly rare, often highly aggressive and account for less than 1 % of all malignant tumors of these organs. In this landscape study, we searched for genomic biomarkers that could aid in either treatment selection or further classification of these tumors. 18 (0.2 %) bladder sarcomas were identified from 11,193 bladder cancers and 11 (0.6 %) prostate sarcomas were identified from 19,057 prostate cancers that underwent hybrid capture-based comprehensive genomic profiling (CGP) to assess all classes of genomic alterations (GA). The bladder sarcomas included 12 leiomyosarcomas, 3 rhabdomyosarcomas and 3 high-grade undifferentiated sarcomas. The sarcomas of the prostate featured 9 primary prostatic stromal sarcomas, 1 leiomyosarcoma and 1 rhabdomyosarcoma. The most frequent gene alterations were in TP53, RB1 and ATRX. Bladder sarcomas were also found to have more than 2-fold more genetic alterations than compared to prostatic sarcomas.
Collapse
Affiliation(s)
- Dana Hariri
- Department of Pathology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA.
| | - Dean Pavlick
- Foundation One, 150 Second Street, Cambridge, MA 02141, USA
| | | | - Roger Li
- Foundation One, 150 Second Street, Cambridge, MA 02141, USA
| | - Ashish Kamat
- Foundation One, 150 Second Street, Cambridge, MA 02141, USA
| | - Petros Grivas
- Foundation One, 150 Second Street, Cambridge, MA 02141, USA
| | - Neeraj Agarwal
- Foundation One, 150 Second Street, Cambridge, MA 02141, USA
| | - Shilpa Gupta
- Foundation One, 150 Second Street, Cambridge, MA 02141, USA
| | - Andrea Necchi
- Foundation One, 150 Second Street, Cambridge, MA 02141, USA
| | - Gennady Bratslavsky
- Department of Pathology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Alina Basnet
- Department of Pathology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Joseph Jacob
- Department of Pathology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Jeffrey Ross
- Department of Pathology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA; Foundation One, 150 Second Street, Cambridge, MA 02141, USA
| | - Oleksandr Kravtsov
- Department of Pathology, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| |
Collapse
|
21
|
Yokoi K, Wang J, Yoshioka Y, Fujisawa Y, Fujimoto M, Ochiya T, Tanemura A. Novel Detection and Clinical Utility of Serum-Derived Extracellular Vesicle in Angiosarcoma. Acta Derm Venereol 2025; 105:adv40902. [PMID: 40001341 DOI: 10.2340/actadv.v105.40902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Cutaneous angiosarcoma is a rare and highly aggressive skin malignancy. The aim of this study is to explore the alteration of serum-derived extracellular vesicle (EV) in angiosarcoma patients and to evaluate its clinical utility as a novel circulating biomarker. In a microarray analysis to examine the differential expression of specific EV-associated microRNAs in sera between cutaneous angiosarcoma patients and healthy controls, 73 microRNAs with significant upregulation and 100 microRNAs with significant downregulation, respectively, were identified in patients with angio-sarcoma. Among them, quantitative PCR confirmed that miR-184, miR-3925-5p, miR-3926, and miR-5703 were upregulated in sera of cutaneous angiosarcoma patients compared with those of healthy controls and melanoma patients. Additionally, these 4 microRNAs were expressed more highly in angiosarcoma cell lines compared with normal human endothelial cell lines and were prone to elevate along with disease progression. Furthermore, a gene analysis predicted that the target gene set of microRNAs might affect the regulation of TP53 via the epigenetic regulation of MECP2. Taken together, these 4 extracellular vesicle-associated microRNAs in circulation serve as a promising liquid biomarker to identify angiosarcoma patients and trace disease progression.
Collapse
Affiliation(s)
- Kazunori Yokoi
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Jing Wang
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Yoshioka
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Yasuhiro Fujisawa
- Department of Dermatology, Tsukuba University, Tsukuba, Ibaraki, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takahiro Ochiya
- Department of Molecular and Cellular Medicine, Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Osaka University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
22
|
Wang T, Wang S, Li Z, Xie J, Jia Q, Hou J. Integrative machine learning model of RNA modifications predict prognosis and treatment response in patients with breast cancer. Cancer Cell Int 2025; 25:43. [PMID: 39948551 PMCID: PMC11827143 DOI: 10.1186/s12935-025-03651-y] [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: 08/08/2024] [Accepted: 01/10/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Breast cancer, a highly heterogeneous and complex disease, remains the leading cause of cancer-related death among women worldwide. Despite advances in treatment modalities, effective prognostic models and therapeutic strategies are still urgently needed. METHODS We retrospectively analyzed 15 independent breast cancer cohorts to explore the role of RNA modifications in the prognosis of patients with breast cancer. By integrating nine types of RNA modifications, we developed a comprehensive machine learning-based RNA modification signature (CMRS). Furthermore, single-cell RNA sequencing data were analyzed to understand the biological mechanisms underlying CMRS. In addition, immune infiltration levels were evaluated via six different algorithms, and immune checkpoint inhibitor responsiveness was predicted. Moreover, the response of high-CMIS patients to chemotherapy was predicted via multiple datasets. Finally, immunohistochemistry was performed on tissue samples from breast cancer patients to validate protein expression levels. RESULTS Our analysis revealed five key RNA modification-related genes (ENO1, ARAF, WT1, GADD45A, and BIRC3) associated with breast cancer prognosis. The CMRS model demonstrated high predictive accuracy across multiple cohorts and was significantly correlated with patient survival outcomes. Multiomics analysis revealed that high CMRS was associated with increased tumor mutational burden and distinct mutational signatures, particularly in pathways related to TP53, MYC, and cell proliferation. Single-cell analysis highlighted the involvement of epithelial cells and MYC signaling in high CMRS activity. Cell‒cell communication analysis revealed reduced interaction strength in hig CMRS patients, indicating poor prognosis. Furthermore, low CMRS patients presented increased immune cell infiltration and improved responsiveness to immune checkpoint inhibitors, whereas high CMRS patients were identified as potential candidates for treatment with panobinostat and vincristine. CONCLUSION Our study elucidates the significant role of RNA modifications in breast cancer prognosis and treatment. The CMRS model serves as a sensitive biomarker for predicting patient survival and treatment responsiveness, offering a new avenue for personalized therapy in patients with breast cancer.
Collapse
Affiliation(s)
- Tao Wang
- Research Laboratory Center, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Shu Wang
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Zhuolin Li
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Jie Xie
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, China
| | - Qi Jia
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
| | - Jing Hou
- Department of Breast Surgery, Guizhou Provincial People's Hospital, Guiyang, 550002, China.
| |
Collapse
|
23
|
R HC, C GPD. Investigation of the impact of R273H and R273C mutations on the DNA binding domain of P53 protein through molecular dynamic simulation. J Biomol Struct Dyn 2025; 43:798-812. [PMID: 39737749 DOI: 10.1080/07391102.2023.2283793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/09/2023] [Indexed: 01/01/2025]
Abstract
The P53 protein, a cancer-associated transcriptional factor and tumor suppressor, houses a Zn2+ ion in its DNA-binding domain (DBD), essential for sequence-specific DNA binding. However, common mutations at position 273, specifically from Arginine to Histidine and Cysteine, lead to a loss of function as a tumor suppressor, also called DNA contact mutations. The mutant (MT) P53 structure cannot stabilize DNA due to inadequate interaction. To investigate the conformational changes, we performed a comparative molecular dynamic simulation (MDS) to study the effect of the P53-Wildtype (P53-WT) and the DNA contact mutations (R273H and R273C) on the DBD. Our research indicated that the DNA binding bases lose Hydrogen bonds (H bonds) when mutated to P53-R273H and P53-R273C during the simulation. We employed tools, such as PDIviz to highlight the contacts with DNA bases and backbone, major and minor grooves, and various pharmacophore forms of atoms. The contact maps for R273H and R273C were generated using the COZOID tool, which displayed changes in the frequency of the amino acids and DNA bases interaction in the DNA binding domain. These residues have diminished interactions, and the zinc-binding domain shows significant movements by Zn2+ ion binding to the phosphate group of the DNA, moving away from its binding sites. In conclusion, our research suggests that R273H and R273C each have unique stability and self-assembly properties. This understanding might assist researchers in better comprehending the function of the p53 protein and its importance in cancer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Hephzibah Cathryn R
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, India
| | - George Priya Doss C
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of Bio Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, India
| |
Collapse
|
24
|
Zhao R, Cui Y, Li D, Guo X, Cheng C, He R, Hu C, Wei X. Anlotinib enhances the pro-apoptotic effect of APG-115 on acute myeloid leukemia cell lines by inhibiting the P13K/AKT signaling pathway. Leuk Res 2025; 149:107637. [PMID: 39732045 DOI: 10.1016/j.leukres.2024.107637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND APG-115 is a novel small-molecule selective inhibitor that destabilizes the p53-MDM2 complex and activates p53-mediated apoptosis in tumor cells. Anlotinib inhibits tumor angiogenesis and promotes apoptosis. In this study, we investigated the apoptotic effect and potential mechanism of APG-115 and anlotinib combination on AML cell lines with different p53 backgrounds. MATERIAL AND METHODS The IC50 values of APG-115 and anlotinib were detected by CCK-8 assay. The apoptosis rate of AML cells was evaluated by Annexin-V and PI double staining. Transcriptome sequencing was performed on the MOLM16 cell line treated with APG-115 and anlotinib, and differential analysis and enrichment analysis were performed. Real-time quantitative PCR and Western blot were used to detect the changes in cell cycle and pathway-related genes and proteins in AML cell lines after drug treatment. In vivo experiments, the anti-leukemia effects of APG-115 and anlotinib on AML xenograft mouse models were evaluated. RESULTS APG-115 and anlotinib could independently promote AML cell apoptosis, and the combination of the two drugs could produce a synergistic effect. Transcriptome sequencing showed that compared with the APG-115 monotherapy group, the differentially expressed genes were mainly enriched in the MDM2-p53 and PI3K/AKT pathways. In vivo experiments showed that compared with AML xenograft mice treated with either drug alone, AML progression was slowed in AML xenograft mice treated with APG-115 and anlotinib. CONCLUSION In vivo and in vitro experimental have shown that APG-115 combined with anlotinib can promote AML cells apoptosis and inhibit the progression of disease is independent of the p53 status.
Collapse
Affiliation(s)
- Rui Zhao
- Department of Hematopathy, Henan Institute of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.
| | - Yu Cui
- Department of Hematopathy, Henan Institute of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.
| | - Dongbei Li
- Central Laboratory, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Xiaoli Guo
- Department of Hematopathy, Henan Institute of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Cheng Cheng
- Department of Hematopathy, Henan Institute of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Rongheng He
- Department of Hematopathy, Henan Institute of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Chenxi Hu
- Central Laboratory, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China
| | - Xudong Wei
- Department of Hematopathy, Henan Institute of Hematology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China; The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou 450008, China.
| |
Collapse
|
25
|
Hsu CL, Chang YS, Li HP. Molecular diagnosis of nasopharyngeal carcinoma: Past and future. Biomed J 2025; 48:100748. [PMID: 38796105 PMCID: PMC11772973 DOI: 10.1016/j.bj.2024.100748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/01/2024] [Accepted: 05/12/2024] [Indexed: 05/28/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a malignant tumor originated from the nasopharynx epithelial cells and has been linked with Epstein-Barr virus (EBV) infection, dietary habits, environmental and genetic factors. It is a common malignancy in Southeast Asia, especially with gender preference among men. Due to its non-specific symptoms, NPC is often diagnosed at a late stage. Thus, the molecular diagnosis of NPC plays a crucial role in early detection, treatment selection, disease monitoring, and prognosis prediction. This review aims to provide a summary of the current state and the latest emerging molecular diagnostic techniques for NPC, including EBV-related biomarkers, gene mutations, liquid biopsy, and DNA methylation. Challenges and potential future directions of NPC molecular diagnosis will be discussed.
Collapse
Affiliation(s)
- Cheng-Lung Hsu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Sun Chang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
- Department of Otolaryngology-Head and Neck Surgery, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Hsin-Pai Li
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
26
|
Galante PA, Guardia GD, Pisani J, Sandoval RL, Barros-Filho MC, Gifoni ACLVC, Patrão DF, Ashton-Prolla P, de Vasconcellos VF, Freycon C, Levine A, Hainaut P, Achatz MI. Personalized screening strategies for TP53 R337H carriers: a retrospective cohort study of tumor spectrum in Li-Fraumeni syndrome adult carriers. LANCET REGIONAL HEALTH. AMERICAS 2025; 42:100982. [PMID: 39895904 PMCID: PMC11787607 DOI: 10.1016/j.lana.2024.100982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 12/13/2024] [Accepted: 12/20/2024] [Indexed: 02/04/2025]
Abstract
Background Li-Fraumeni Syndrome (LFS) is a predisposition associated with early onset malignant tumors caused by germline pathogenic variants in the TP53 gene. Although rare worldwide, LFS is prevalent in Southern Brazil due to the founder pathogenic variant R337H. Here, we assessed tumor patterns and temporal trends, cancer risk, and sex differences of adult R337H carriers and carriers of other LFS-associated variants. Methods We retrospectively analyzed 708 adults, combining data from two sources: the Brazilian Li-Fraumeni Syndrome Study cohort and the NCI TP53 database. We assessed the clinical characteristics of 303 adults with R337H and compared them with those associated with 405 carriers of other TP53 variants. Findings R337H carriers, compared to adult carriers of other TP53 variants typical of LFS, had a lower cumulative risk of developing cancer (54% vs 78%). Female R337H carriers were at a higher risk than males (65% vs 30%) and had a higher risk of developing a second primary cancer, underscoring a strong sex bias not observed in carriers of other variants. The most common cancers were breast cancer and soft tissue sarcoma in females, and soft tissue sarcoma and prostate cancer in males. Common second malignancies were breast cancer in females and lung cancer in males. Interpretation This study shows that R337H is associated with a lifetime risk of multiple LFS-spectrum cancers but with incomplete penetrance, particularly in males. Our findings suggest that R337H carriers would benefit from tailored surveillance and risk reduction strategies. Funding São Paulo Research Foundation, Conselho Nacional de Pesquisa, and Hospital Sírio-Libanês.
Collapse
Affiliation(s)
- Pedro A.F. Galante
- Centro de Oncologia Molecular, Hospital Sirio-Libanes, São Paulo, Brazil
| | | | - Janina Pisani
- Department of Oncology, Hospital Sírio-Libanês, São Paulo, Brazil
| | | | | | | | | | | | | | - Claire Freycon
- University Grenoble Alpes, Inserm 1209 CNRS 5309, Institute for Advanced Biosciences, Grenoble, France
| | - Arnold Levine
- Institute for Advanced Study, Princeton, New Jersey, USA
| | - Pierre Hainaut
- University Grenoble Alpes, Inserm 1209 CNRS 5309, Institute for Advanced Biosciences, Grenoble, France
| | | |
Collapse
|
27
|
Gebrael G, Sayegh N, Hage Chehade C, Jo Y, Narang A, Chigarira B, Tripathi N, Srivastava A, Tandar C, Williams JF, Garg D, Ji R, Maughan BL, Swami U, Agarwal N. Genomic biomarkers of survival in patients with metastatic hormone-sensitive prostate cancer undergoing intensified androgen deprivation therapy. Prostate Cancer Prostatic Dis 2025:10.1038/s41391-025-00936-1. [PMID: 39885371 DOI: 10.1038/s41391-025-00936-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 12/04/2024] [Accepted: 01/03/2025] [Indexed: 02/01/2025]
Abstract
INTRODUCTION Androgen deprivation therapy intensification (ADTi) with androgen receptor pathway inhibitors (ARPI), docetaxel or both has been shown to improve survival outcomes in patients with metastatic hormone-sensitive prostate cancer (mHSPC). Currently, baseline tumor genomic markers have no role in clinical decision-making in patients with mHSPC. METHODS In this IRB-approved retrospective study, patients diagnosed with mHSPC who underwent comprehensive genomic profiling from primary tissue or metastatic sites and treated with ADTi were included. Genomic alterations with an incidence ≥5% were included in the analysis. RESULTS A total of 276 patients were eligible and included in the study. In the multivariable analysis, TP53 (HR 1.71, 95% CI 1.17-2.49, p = 0.006), RB1 (HR 2.32, 95% CI 1.28-4.18, p = 0.006), PTEN (HR 1.74, 95% CI 1.12-2.7, p = 0.014), and BRCA2 (HR 2.64, 95% CI 1.42-4.92, p = 0.003) were associated with significantly shorter PFS, while TP53 (HR 1.63, 95% CI 1.00-2.64, p = 0.049), RB1 (HR 4.5, 95% CI 2.32-8.70, p < 0.001), and PTEN (HR 2.4, 95% CI 1.38-4.2, p = 0.003) were associated with significantly worse OS. CONCLUSIONS This is one of the largest studies to show the association of baseline tumor genomic markers with survival in patients with mHSPC treated with ADTi. Upon external validation, these results may aid in developing a clinical-genomic risk stratification model, patient counseling, and prognostication.
Collapse
Affiliation(s)
- Georges Gebrael
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nicolas Sayegh
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chadi Hage Chehade
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Yeonjung Jo
- Division of Biostatistics, Department of Population Health Sciences, School of Medicine, University of Utah, Salt Lake City, UT, USA
- Cancer Biostatistics, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Arshit Narang
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Beverly Chigarira
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Nishita Tripathi
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Ayana Srivastava
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Clara Tandar
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | | | - Diya Garg
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Richard Ji
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Benjamin L Maughan
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Umang Swami
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| | - Neeraj Agarwal
- Division of Medical Oncology, Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
28
|
Zhang G, Pannucci A, Ivanov AA, Switchenko J, Sun SY, Sica GL, Liu Z, Huang Y, Schmitz JC, Owonikoko TK. Polo-like Kinase 1 Inhibitors Demonstrate In Vitro and In Vivo Efficacy in Preclinical Models of Small Cell Lung Cancer. Cancers (Basel) 2025; 17:446. [PMID: 39941812 PMCID: PMC11815996 DOI: 10.3390/cancers17030446] [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: 12/18/2024] [Revised: 01/19/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Objective: To investigate the preclinical efficacy and identify predictive biomarkers of polo-like kinase 1 (PLK1) inhibitors in small cell lung cancer (SCLC) models. Methods: We tested the cytotoxicity of selective PLK1 inhibitors (rigosertib, volasertib, and onvansertib) in a panel of SCLC cell lines. We confirmed the therapeutic efficacy of subcutaneous xenografts of representative cell lines and in four patient-derived xenograft models generated from patients with platinum-sensitive and platinum-resistant SCLC. We employed an integrated analysis of genomic and transcriptomic sequencing data to identify potential biomarkers of the activity and mechanisms of resistance in laboratory-derived resistance models. Results: Volasertib, rigosertib, and onvansertib showed strong in vitro cytotoxicity at nanomolar concentrations in human SCLC cell lines. Rigosertib, volasertib, and onvansertib showed equivalent efficacy to that of standard care agents (irinotecan and cisplatin) in vivo with significant growth inhibition superior to cisplatin in PDX models of platinum-sensitive and platinum-resistant SCLC. There was an association between YAP1 expression and disruptive or inactivation TP53 gene mutations, with greater efficacy of PLK1 inhibitors. Comparison of lab-derived onvansertib-resistant H526 cells to parental cells revealed differential gene expression with upregulation of NAP1L3, CYP7B1, AKAP7, and FOXG1 and downregulation of RPS4Y1, KDM5D, USP9Y, and EIF1AY highlighting the potential mechanisms of resistance in the clinical setting. Conclusions: We established the efficacy of PLK1 inhibitors in vitro and in vivo using PDX models of platinum-sensitive and resistant relapsed SCLC. An ongoing phase II trial is currently testing the efficacy of onvansertib in patients with SCLC (NCT05450965).
Collapse
Affiliation(s)
- Guojing Zhang
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (A.P.); (Z.L.); (Y.H.)
- University of Maryland Greenebaum Comprehensive Cancer Center (UMGCCC), 22 South Green Street N9E17, Baltimore, MD 21201, USA; (G.Z.); (J.C.S.)
| | - Abbe Pannucci
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (A.P.); (Z.L.); (Y.H.)
| | - Andrey A. Ivanov
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine and Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA;
| | - Jeffrey Switchenko
- Biostatistics Shared Resource of Winship Cancer Institute, Atlanta, GA 30322, USA;
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA 30322, USA;
| | - Gabriel L. Sica
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA;
| | - Zhentao Liu
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (A.P.); (Z.L.); (Y.H.)
- Cancer Virology Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Yufei Huang
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (A.P.); (Z.L.); (Y.H.)
- Cancer Virology Program, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - John C. Schmitz
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (A.P.); (Z.L.); (Y.H.)
- University of Maryland Greenebaum Comprehensive Cancer Center (UMGCCC), 22 South Green Street N9E17, Baltimore, MD 21201, USA; (G.Z.); (J.C.S.)
| | - Taofeek K. Owonikoko
- Division of Hematology and Oncology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA; (A.P.); (Z.L.); (Y.H.)
- University of Maryland Greenebaum Comprehensive Cancer Center (UMGCCC), 22 South Green Street N9E17, Baltimore, MD 21201, USA; (G.Z.); (J.C.S.)
| |
Collapse
|
29
|
Tseng YH, Tran TTM, Tsai Chang J, Huang YT, Nguyen AT, Chang IYF, Chen YT, Hsieh HW, Juang YL, Chang PMH, Huang TY, Chang YC, Chen YM, Liu H, Huang CYF. Utilizing TP53 hotspot mutations as effective predictors of gemcitabine treatment outcome in non-small-cell lung cancer. Cell Death Discov 2025; 11:26. [PMID: 39870629 PMCID: PMC11772833 DOI: 10.1038/s41420-025-02300-7] [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: 03/27/2024] [Revised: 12/11/2024] [Accepted: 01/14/2025] [Indexed: 01/29/2025] Open
Abstract
TP53 mutations are recognized to correlate with a worse prognosis in individuals with non-small cell lung cancer (NSCLC). There exists an immediate necessity to pinpoint selective treatment for patients carrying TP53 mutations. Potential drugs were identified by comparing drug sensitivity differences, represented by the half-maximal inhibitory concentration (IC50), between TP53 mutant and wild-type NSCLC cell lines using database analysis. In addition, clinical data from NSCLC patients were collected to evaluate both their TP53 status and their response to gemcitabine, thereby facilitating further validation. Subsequently, NSCLC cell lines with different TP53 status (A549 and H1299) were subjected to gemcitabine treatment to investigate the association between TP53 mutations and gemcitabine response. According to the dataset, NSCLC cell lines carrying TP53 mutations displayed heightened sensitivity to gemcitabine. From a clinical standpoint, patients exhibiting TP53 hotspot mutations demonstrated prolonged overall survival upon gemcitabine treatment. In vitro, overexpressing various hotspot TP53 mutations significantly sensitized H1299 cells to gemcitabine. Moreover, the knockdown of TP53 in A549 cells notably augmented sensitivity to gemcitabine treatment, as evidenced by cell viability and reproductive cell death assays. Conversely, the overexpression of wild-type TP53 in H1299 cells led to an increased resistance against gemcitabine. Gemcitabine is a treatment option for patients with non-small cell lung cancer (NSCLC) who carry TP53 hotspot mutations. This potential effectiveness might arise from its ability to disrupt DNA damage repair processes, leading to G2/M phase cell cycle arrest or an augmentation of mitotic abnormalities, eventually cause cell death. As a result, when planning treatment strategies for NSCLC patients possessing TP53 hotspot mutations, gemcitabine should be considered to incorporate into the indication.
Collapse
Affiliation(s)
- Yen-Han Tseng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Program in Molecular Medicine, School of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Trieu Thi My Tran
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Jinghua Tsai Chang
- Institute of Medicine, Chung Shan Medical University, Taichung City, Taiwan
| | - Yu-Tang Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Biomedical Industry Ph.D. Program, National Yang Ming Chiao Tung University, Hsinchu City, Taiwan
| | - Anh Thuc Nguyen
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Taiwan National Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Ian Yi-Feng Chang
- Taiwan National Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Tung Chen
- Taiwan National Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan
| | - Hao-Wen Hsieh
- Department of Neurosurgery, Lin-Kou Medical Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yue-Li Juang
- Institute of Clinical Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Peter Mu-Hsin Chang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City, Taiwan
- Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzu-Yi Huang
- Program in Molecular Medicine, School of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ying-Chih Chang
- Genomics Research Center, Academia Sinica, Taipei, 115, Taiwan
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| | - Hsuan Liu
- Biomedical Industry Ph.D. Program, National Yang Ming Chiao Tung University, Hsinchu City, Taiwan.
- Department of Cell and Molecular Biology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, College of Pharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan.
- Department of Biochemistry, School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Chong Hin Loon Memorial Cancer and Biotherapy Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
30
|
Heo Y, Kim WJ, Cho YJ, Jung JW, Kim NS, Choi IY. Advances in cancer genomics and precision oncology. Genes Genomics 2025:10.1007/s13258-024-01614-7. [PMID: 39849190 DOI: 10.1007/s13258-024-01614-7] [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: 11/07/2024] [Accepted: 12/27/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Next-generation sequencing has revolutionized genome science over the last two decades. Indeed, the wealth of sequence information on our genome has deepened our understanding on cancer. Cancer is a genetic disease caused by genetic or epigenetic alternations that affect the expression of genes that control cell functions, particularly cell growth and division. Utilization of next-generation sequencing in cancer gene panels has enabled the identification of actionable gene alterations in cancer patients to guide personalized precision medicine. OBJECTIVE The aim is to provide information that can identify actionable gene alterations, enabling personalized precision medicine for cancer patients. RESULTS & DISCUSSION Equipped with next-generation sequencing techniques, international collaboration programs on cancer genomics have identified numerous mutations, gene fusions, microsatellite variations, copy number variations, and epigenetics changes that promote the transformation of normal cells into tumors. Cancer classification has traditionally been based on cell type or tissue-of-origin and the morphological characteristics of the cancer. However, interactive genomic analyses have currently reclassified cancers based on systemic molecular-based taxonomy. Although all cancer-causing genes and mechanisms have yet to be completely understood or identified, personalized or precision medicine is now currently possible for some forms of cancer. Unlike the "one-size-fits-all" approach of traditional medicine, precision medicine allows for customized or personalized treatment based on genomic information. CONCLUSION Despite the availability of numerous cancer gene panels, technological innovation in genomics and expansion of knowledge on the cancer genome will allow precision oncology to manage even more types of cancers.
Collapse
Affiliation(s)
- Yonjong Heo
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, Gangwon, Republic of Korea
| | - Woo-Jin Kim
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, Gangwon, Republic of Korea
| | - Yong-Joon Cho
- Department of Molecular Bioscience, Kangwon National University, Chuncheon, 24341, Republic of Korea
- Multidimensional Genomics Research Center, Kangwon National University, Chuncheon, 24341, Republic of Korea
| | - Jae-Won Jung
- Genetic Sciences Group, Thermo Fisher Scientific Solutions Korea Co., Ltd., Seoul, 06349, Republic of Korea
| | - Nam-Soo Kim
- Department of Molecular Bioscience, Kangwon National University, Chuncheon, 24341, Republic of Korea.
- NBIT Co., Ltd., Chuncheon, 24341, Republic of Korea.
| | - Ik-Young Choi
- Department of Smart Farm and Agricultural Industry, Kangwon National University, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
31
|
Wang Y, Armendariz DA, Wang L, Zhao H, Xie S, Hon GC. Enhancer regulatory networks globally connect non-coding breast cancer loci to cancer genes. Genome Biol 2025; 26:10. [PMID: 39825430 PMCID: PMC11740497 DOI: 10.1186/s13059-025-03474-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 01/02/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Genetic studies have associated thousands of enhancers with breast cancer (BC). However, the vast majority have not been functionally characterized. Thus, it remains unclear how BC-associated enhancers contribute to cancer. RESULTS Here, we perform single-cell CRISPRi screens of 3513 regulatory elements associated with breast cancer to measure the impact of these regions on transcriptional phenotypes. Analysis of > 500,000 single-cell transcriptomes in two breast cancer cell lines shows that perturbation of BC-associated enhancers disrupts breast cancer gene programs. We observe BC-associated enhancers that directly or indirectly regulate the expression of cancer genes. We also find one-to-multiple and multiple-to-one network motifs where enhancers indirectly regulate cancer genes. Notably, multiple BC-associated enhancers indirectly regulate TP53. Comparative studies illustrate subtype specific functions between enhancers in ER + and ER - cells. Finally, we develop the pySpade package to facilitate analysis of single-cell enhancer screens. CONCLUSIONS Overall, we demonstrate that enhancers form regulatory networks that link cancer genes in the genome, providing a more comprehensive understanding of the contribution of enhancers to breast cancer development.
Collapse
Affiliation(s)
- Yihan Wang
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Daniel A Armendariz
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lei Wang
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Huan Zhao
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Shiqi Xie
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Present Address: Genentech, 1 DNA Way, South San Francisco, CA, 94080, USA
| | - Gary C Hon
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Division of Basic Reproductive Biology Research, Department of Obstetrics and Gynecology, Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
32
|
Aguilar D, Garza-Rodríguez ML, Muñiz-Garza CE, Nuñez FA, Villarreal-Garza CM, Vidal-Gutiérrez O, Pérez-Ibave DC, Burciaga-Flores CH. Los olvidados: Non-BRCA variants associated with Hereditary breast cancer in Mexican population. Breast Cancer Res 2025; 27:7. [PMID: 39815370 PMCID: PMC11737022 DOI: 10.1186/s13058-024-01957-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 12/20/2024] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Hereditary predisposition to breast and ovarian cancer syndrome (HBOC) is a pathological condition with increased cancer risk, including breast (BC), ovarian cancer (OC), and others. HBOC pathogenesis is caused mainly by germline pathogenic variants (GPV) in BRCA1 and BRCA2 genes. However, other relevant genes are related to this syndrome diagnosis, prognosis, and treatment, including TP53, PALB2, CHEK2, ATM, etc. This study aimed to identify the prevalence of non-BRCA genes in HBOC patients of Northeast Mexico. METHODS This multicentric study included 1285 patients with HBOC diagnosis from four oncologic centers in northeast Mexico from 2016 to 2023. Genomic and clinical data were analyzed based on multi-gene panel results and electronic records of the medical geneticist consultation. For the data analysis of qualitative and quantitative variants, JASP statistical software (version 0.18.1) was used, taking p < 0.05 as a significant result. RESULTS We found that 32.7% of the patients had at least one GPV in non-BRCA genes. The five most frequent non-BRCA genes were CHEK2, PALB2, MUTYH, CDKN2A, and ATM. Among the group of non-BRCA genes, six are involved in the homologous repair pathway (HR), and three are related to DNA damage repair (DDR) pathways. In analyzing GPVs in molecular pathways, both have similar frequencies with no statistical difference for BC. CONCLUSION Multi-gene testing implementation improves the detection of often overlooked genes related to HBOC pathogenesis and treatment. Non-BRCA GPVs in Northern Mexico correspond to one-third of the HBOC cases, including HR and DDR pathways genes that would be misdiagnosed if not tested. HR patient carriers are potential targets of iPARP therapies. The optimal approach to cancer treatment for non-BRCA mutation carriers warrants further investigation to develop newer therapies.
Collapse
Affiliation(s)
- Dione Aguilar
- Breast Cancer Center, Hospital Zambrano Hellion TecSalud, San Pedro Garza García, 64710, México, México
| | - María Lourdes Garza-Rodríguez
- Servicio de Oncología, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, 66451, Monterrey, Nuevo León, México
| | - Carolina Elizabeth Muñiz-Garza
- Facultad de Medicina, Universidad Autónoma de Nuevo León, Av. Francisco I. Madero S/N, Mitras Centro Monterrey, 64460, Monterrey, México
| | - Fernando Alcorta Nuñez
- Breast Cancer Center, Hospital Zambrano Hellion TecSalud, San Pedro Garza García, 64710, México, México
| | | | - Oscar Vidal-Gutiérrez
- Servicio de Oncología, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, 66451, Monterrey, Nuevo León, México
| | - Diana Cristina Pérez-Ibave
- Servicio de Oncología, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, 66451, Monterrey, Nuevo León, México.
| | - Carlos Horacio Burciaga-Flores
- Servicio de Oncología, Centro Universitario Contra el Cáncer (CUCC), Hospital Universitario "Dr. José Eleuterio González", Universidad Autónoma de Nuevo León, 66451, Monterrey, Nuevo León, México.
- Instituto Mexicano del Seguro Social (IMSS), Unidad Médica de Alta Especialidad, Hospital de Gineco Obstetricia (HGO), No. 23, 64000, Monterrey, Nuevo León, México.
| |
Collapse
|
33
|
Ghosh P, Kundu A, Ganguly D. From experimental studies to computational approaches: recent trends in designing novel therapeutics for amyloidogenesis. J Mater Chem B 2025; 13:858-881. [PMID: 39664012 DOI: 10.1039/d4tb01890g] [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: 12/13/2024]
Abstract
Amyloidosis is a condition marked by misfolded proteins that build up in tissues and eventually destroy organs. It has been connected to a number of fatal illnesses, including non-neuropathic and neurodegenerative conditions, which in turn have a significant influence on the worldwide health sector. The inability to identify the underlying etiology of amyloidosis has hampered efforts to find a treatment for the condition. Despite the identification of a multitude of putative pathogenic variables that may operate independently or in combination, the molecular mechanisms responsible for the development and progression of the disease remain unclear. A thorough investigation into protein aggregation and the impacts of toxic aggregated species will help to clarify the cytotoxicity of aggregation-mediated cellular apoptosis and lay the groundwork for future studies aimed at creating effective treatments and medications. This review article provides a thorough summary of the combination of various experimental and computational approaches to modulate amyloid aggregation. Further, an overview of the latest developments of novel therapeutic agents is given, along with a discussion of the possible obstacles and viewpoints on this developing field. We believe that the information provided by this review will help scientists create innovative treatment strategies that affect the way proteins aggregate.
Collapse
Affiliation(s)
- Pooja Ghosh
- Centre for Interdisciplinary Sciences, JIS Institute of Advanced Studies & Research (JISIASR) Kolkata, JIS University, GP Block, Sector-5, Salt Lake, Kolkata 700091, West Bengal, India.
| | - Agnibin Kundu
- Department of Medicine, District Hospital Howrah, 10, Biplabi Haren Ghosh Sarani Lane, Howrah 711101, West Bengal, India
| | - Debabani Ganguly
- Centre for Health Science & Technology, JIS Institute of Advanced Studies & Research (JISIASR) Kolkata, JIS University, GP Block, Sector-5, Salt Lake, Kolkata 700091, West Bengal, India.
| |
Collapse
|
34
|
Ding C, Cao L, Wang R, Wu Q, Li M, Zhang J, Thorne RF, Li J, Ma J, Wu M, Cang S. OTUD7B is a new deubiquitinase targeting p53. Theranostics 2025; 15:2121-2138. [PMID: 39990225 PMCID: PMC11840744 DOI: 10.7150/thno.103012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/04/2025] [Indexed: 02/25/2025] Open
Abstract
Rationale: The tumor suppressor p53 safeguards against cellular transformation, with its expression regulated by diverse post-translational modifications (PTMs). While polyubiquitination by Mdm2 principally drives its proteasomal degradation, the identity of p53 deubiquitinases (DUBs) remains less well defined. This study investigates the role of the deubiquitinase enzyme OTUD7B in hepatocellular carcinoma (HCC), where it is notably downregulated and proposed to function as a tumor suppressor. Methods: Mass spectrometry screening of immunoprecipitates from HCC cells was used to identify OTUD7B-binding proteins. Co-immunoprecipitation assays with endogenous, ectopic, and mutant forms of OTUD7B and p53 assessed binding interactions and p53 polyubiquitination levels, respectively. Regulatory mechanisms were explored via luciferase reporter and chromatin immunoprecipitation (ChIP) assays. OTUD7B function was evaluated in vitro and in xenograft models using shRNA knockdown, overexpression, and CRISPR-Cas9 knockout. OTUD7B expression in normal and HCC tissues was analyzed by immunohistochemistry and immunoblotting. Results: We identified p53 as a binding partner of OTUD7B, confirming interactions with both wild-type and mutant p53 in HCC cells. OTUD7B was shown to remove lysine-linked polyubiquitin chains in p53, including those mediated by Mdm2, thereby stabilizing p53 by inhibiting its proteasomal degradation. Overexpression of OTUD7B suppressed growth in HCC cultures and xenografts through p53-dependent mitochondrial apoptosis, marked by PUMA and BAX induction. Conversely, OTUD7B knockdown promoted tumor growth. These effects were absent in p53-null or CRISPR-knockout cells, underscoring p53 as a key OTUD7B substrate. Additionally, OTUD7B expression was found to be transcriptionally repressed via p53-dependent mechanisms. Bioinformatics and ex vivo analysis revealed a positive correlation between OTUD7B and p53 protein levels in HCC tissues. Conclusion: OTUD7B plays a critical role in stabilizing both wild-type and mutant p53 in HCC cells, with its expression regulated through a mutual feedback loop involving p53. By inhibiting cell growth, OTUD7B exhibits tumor-suppressive properties, underscored by its atypical downregulation in patient tissues and its positive correlation with p53 expression. These findings highlight the clinical significance of OTUD7B and position it as a promising therapeutic target for modulating the p53 pathway in HCC.
Collapse
Affiliation(s)
- Caoyuan Ding
- Translational Research Institute, People's Hospital of Zhengzhou University, 450003 Zhengzhou, Henan, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, 450001 Zhengzhou, Henan, China
| | - Leixi Cao
- Translational Research Institute, People's Hospital of Zhengzhou University, 450003 Zhengzhou, Henan, China
| | - Ruijie Wang
- Translational Research Institute, People's Hospital of Zhengzhou University, 450003 Zhengzhou, Henan, China
| | - Qichen Wu
- Translational Research Institute, People's Hospital of Zhengzhou University, 450003 Zhengzhou, Henan, China
| | - Mengfan Li
- Translational Research Institute, People's Hospital of Zhengzhou University, 450003 Zhengzhou, Henan, China
| | - Jinjing Zhang
- Translational Research Institute, People's Hospital of Zhengzhou University, 450003 Zhengzhou, Henan, China
| | - Rick F. Thorne
- Translational Research Institute, People's Hospital of Zhengzhou University, 450003 Zhengzhou, Henan, China
| | - Jinming Li
- Translational Research Institute, People's Hospital of Zhengzhou University, 450003 Zhengzhou, Henan, China
| | - Jianli Ma
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, 150081 Harbin, Heilongjiang, China
| | - Mian Wu
- Translational Research Institute, People's Hospital of Zhengzhou University, 450003 Zhengzhou, Henan, China
| | - Shundong Cang
- Translational Research Institute, People's Hospital of Zhengzhou University, 450003 Zhengzhou, Henan, China
| |
Collapse
|
35
|
Chakraborty R, Dutta A, Mukhopadhyay R. TP53 mutations and MDM2 polymorphisms in breast and ovarian cancers: amelioration by drugs and natural compounds. Clin Transl Oncol 2025:10.1007/s12094-024-03841-6. [PMID: 39797946 DOI: 10.1007/s12094-024-03841-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025]
Abstract
Globally, breast and ovarian cancers are major health concerns in women and account for significantly high cancer-related mortality rates. Dysregulations and mutations in genes like TP53, BRCA1/2, KRAS and PTEN increase susceptibility towards cancer. Here, we discuss the impact of mutations in the key regulatory gene, TP53 and polymorphisms in its negative regulator MDM2 which are reported to accelerate cancer progression. Missense mutations, null mutations, transversions, transitions, and point mutations occurring in the TP53 gene can cause an increase in metastatic activity. This review discusses mutations occurring in exon regions of TP53, polymorphisms in MDM2 and their interaction with large ribosomal subunit protein (RPL) leading to cancer development. We also highlight the potential of small molecules e.g. p53 activators like XI-011, Tenovin-1, and Nutlin-3a for the treatment of breast and ovarian cancers. The therapeutic efficacy of natural compounds in amelioration of these two types of cancers is also discussed.
Collapse
Affiliation(s)
- Rituraj Chakraborty
- Inflammation and Cancer Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, 784028, India
| | - Anupam Dutta
- Inflammation and Cancer Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, 784028, India
| | - Rupak Mukhopadhyay
- Inflammation and Cancer Biology Laboratory, Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, Assam, 784028, India.
| |
Collapse
|
36
|
Zhang Q, Han L, Luo X, Bao Y, Wang S, Li T, Huo J, Meng X. Enhancing inhibitory effect in SMMC-7721 hepatoma cells through combined treatment of gallic acid and hUC-MSCs-Exos. Int Immunopharmacol 2025; 144:113704. [PMID: 39608175 DOI: 10.1016/j.intimp.2024.113704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 09/23/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024]
Abstract
BACKGROUND Clinically, hepatoma patients are more frequently encountered in the intermediate and advanced stages. Consequently, the majority of patients miss out on the chance to undergo liver transplantation or radical surgery. Radiotherapy and chemotherapy often fall short of delivering satisfactory outcomes. The incidence and mortality rates for liver cancer approach nearly 100%. In recent years, both exosomes (Exos) and natural chemical compounds have demonstrated robust anti-cancer properties; however, the synergistic effect of their combination remains unexplored. METHODS Exos were extracted from human umbilical cord mesenchymal stem cells (hUC-MSCs). The impact of gallic acid (GA), hUC-MSCs-Exos, and their combined administration on the proliferation inhibition rate and apoptosis of SMMC-7721 hepatoma cells was assessed to ascertain the efficacy differences before and after the combined treatment. A combination of cells metabolomics and network pharmacology techniques was employed to investigate the underlying mechanisms of action. The pivotal targets associated with glycolysis, inflammation, and oxidative stress pathways were confirmed through ELISA assays. RESULTS The findings elucidate that GA profoundly impedes the proliferation of SMMC-7721 hepatoma cells and instigates apoptotic processes therein. While the impact of hUC-MSCs-Exos alone was inconspicuous, a notable augmentation in effect ensued upon their combined application. Concomitantly, a marked reduction was observed in the expressionlevels of key enzymes including HK, PFK, PK, LDH, TNF-α, IL-1β, CAT, SOD and GSH-Px in the malignant hepatocytes, while IL-6 and MDA exhibited heightened expression. Pathway enrichment analysis underscored selenocompound metabolism and cysteine and methionine metabolism as pivotal pathways. CONCLUSION The potentiated efficacy of GA conjunction with hUC-MSCs-Exos may be attributed to their synergistic modulation of anti-inflammatory, antioxidant, and glycolytic functions, thereby influencing selenocompound metabolism and cysteine and methionine metabolism. This study reveals the efficacy and mechanism of Exos and GA combined therapy for hepatoma, providing new methods and ideas for the clinical treatment of hepatoma.
Collapse
Affiliation(s)
- Qiang Zhang
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Liaoning Dalian China
| | - Liying Han
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Liaoning Dalian China
| | - Xi Luo
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Liaoning Dalian China
| | - Yongrui Bao
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Liaoning Dalian China; Professional Innovation Technology Center for Multidimensional Analysis of Traditional Chinese Medicine, Liaoning Dalian China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Liaoning Dalian China
| | - Shuai Wang
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Liaoning Dalian China; Professional Innovation Technology Center for Multidimensional Analysis of Traditional Chinese Medicine, Liaoning Dalian China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Liaoning Dalian China
| | - Tianjiao Li
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Liaoning Dalian China; Professional Innovation Technology Center for Multidimensional Analysis of Traditional Chinese Medicine, Liaoning Dalian China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Liaoning Dalian China
| | - Jinnan Huo
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Liaoning Dalian China
| | - Xiansheng Meng
- College of Pharmacy, Liaoning University of Traditional Chinese Medicine, Liaoning Dalian China; Professional Innovation Technology Center for Multidimensional Analysis of Traditional Chinese Medicine, Liaoning Dalian China; Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Liaoning Dalian China.
| |
Collapse
|
37
|
Pikul J, Machnicki MM, Rzepakowska A, Winiarska N, Chudy A, Moskowicz A, Król K, Fus Ł, Kostrzewa G, Stokłosa T. Potentially actionable molecular alterations in particular related to poor oncologic outcomes in salivary gland carcinomas. BMC Cancer 2025; 25:42. [PMID: 39780157 PMCID: PMC11708168 DOI: 10.1186/s12885-024-13421-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 12/31/2024] [Indexed: 01/11/2025] Open
Abstract
AIM The study was designed to evaluate molecular alterations, relevant to the prognosis and personalized therapy of salivary gland cancers (SGCs). MATERIALS AND METHODS DNA was extracted from archival tissue of 40 patients with various SGCs subtypes. A targeted next-generation sequencing (NGS) panel was used for the identification of small-scale mutations, focal and chromosomal arm-level copy number changes. The final analysis included selected genes with potential actionable aberrations for targeted therapies and outcome predictions in 37 tumours' samples. RESULTS The follow-up of the SGCs study cohort revealed disease recurrence or metastasis in 19 patients and indicated poor individual outcomes. The mean disease-free survival (DFS) within the poor outcome group was 2.4 years, and the overall survival (OS) was 5.4 years. The DFS and OS of the remaining 18 patients with favourable outcomes were 8.3 years. The genes most frequently affected with aberrations were NF1 (n = 9, 24%) and TP53 (n = 8, 22%), with increased occurrence observed in the poor outcome group: NF1 (n = 6, 32%) and TP53 (n = 6, 32%). CDKN2A biallelic deletion was the most common copy number variation (n = 5), and was detected in 4 cases with identified disease relapse. TERT promoter mutation and amplification were found in myoepithelial carcinoma. A p.Ile35Thr mutation was discovered in CTNNB1 in two cases of adenoid cystic carcinoma. ERBB2 alterations were remarkable for SDC ex PA. Furthermore, TP53 mutation was established as a relevant negative prognostic factor for overall survival (p = 0,04). The analysis revealed potentially actionable genes in detected alterations in: MECA 100% (1/1), SDC 100% (7/7), AD 92% (11/12), Ca ex PA 82% (18/22), MECA 65% (20/31), AdCC 64% (9/14) and AcCC 0% (0/1). CONCLUSIONS SGCs are a heterogeneous group of malignancies with distinct molecular landscape that characterized by poor prognosis and inadequate treatment options. Nonstandard strategies might be beneficial for patients who suffer from salivary gland cancers. Wider utilization of NGS analysis may increase the opportunity for patients with those rare cancers to receive more precise, personalized therapy.
Collapse
Affiliation(s)
- Julia Pikul
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Marcin M Machnicki
- Department of Tumor Biology and Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Anna Rzepakowska
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical University of Warsaw, Warsaw, Poland.
| | - Natalia Winiarska
- Student Scientific Research Group at Otorhinolaryngology Department, Head and Neck Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Chudy
- Laboratory of Genetics, University Clinical Hospital, Medical University of Warsaw, Warsaw, Poland
| | - Albert Moskowicz
- Laboratory of Genetics, University Clinical Hospital, Medical University of Warsaw, Warsaw, Poland
| | - Kacper Król
- Student Scientific Research Group at Otorhinolaryngology Department, Head and Neck Surgery, Medical University of Warsaw, Warsaw, Poland
| | - Łukasz Fus
- Department of Pathology Department, Medical University of Warsaw, Warsaw, Poland
| | - Grażyna Kostrzewa
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Tomasz Stokłosa
- Department of Tumor Biology and Genetics, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
38
|
Trendowski MR, Lusk CM, Wenzlaff AS, Neslund-Dudas C, Purrington KS, Beebe-Dimmer JL, Schwartz AG. Association of Germline Pathogenic Variants in MUTYH and Other DNA Damage Response Genes With Lung Cancer Risk Among Non-Hispanic Whites and African Americans. JCO Precis Oncol 2025; 9:e2400558. [PMID: 39854657 PMCID: PMC11771983 DOI: 10.1200/po-24-00558] [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: 08/05/2024] [Revised: 11/19/2024] [Accepted: 12/17/2024] [Indexed: 01/26/2025] Open
Abstract
PURPOSE Although lung cancer is one of the most common malignancies, the underlying genetics regarding susceptibility remain poorly understood. We characterized the spectrum of pathogenic/likely pathogenic (P/LP) germline variants within DNA damage response (DDR) genes among lung cancer cases and controls in non-Hispanic Whites (NHWs) and African Americans (AAs). MATERIALS AND METHODS Rare, germline variants in 67 DDR genes with evidence of pathogenicity were identified using the ClinVar database. These P/LP variants were genotyped in a sample of 3,040 lung cancer cases and controls from the Inflammation, Health, Ancestry, and Lung Epidemiology study (NHW: n = 1,915; AA: n = 1,125) and were tested for their association with lung cancer using multivariate logistic regression adjusting for age, sex, pack-years, and race. RESULTS We identified 49 unique rare P/LP variants in 21 genes among 156 carriers. Approximately 5.9% of lung cancer cases and 4.2% of controls carried at least one P/LP variant. P/LP variants in DDR genes were more common in lung cancer cases, particularly those diagnosed with adenocarcinoma (odds ratio [OR], 1.46 [95% CI, 1.00 to 2.14]). MUTYH variants were associated with lung cancer overall (OR, 1.82 [95% CI, 1.10 to 3.12]), with the strongest associations among never smokers (OR, 3.37 [95% CI, 1.08 to 10.26]), and in individuals who do not meet current USPSTF screening criteria (OR, 2.85 [95% CI, 1.20 to 7.53]). CONCLUSION Germline variants in DDR genes appear to be associated with lung cancer, particularly when examined by gene subtype and morphologic subtype. MUTYH, a gene historically associated with colorectal and other GI malignancies, emerged as a candidate gene that should be examined in individuals who do not have a significant smoking history.
Collapse
Affiliation(s)
- Matthew R. Trendowski
- Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Christine M. Lusk
- Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Angela S. Wenzlaff
- Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | | | - Kristen S. Purrington
- Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jennifer L. Beebe-Dimmer
- Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ann G. Schwartz
- Karmanos Cancer Institute and Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
39
|
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
|
40
|
Gu L, Zhu Y, Nandi SP, Lee M, Watari K, Bareng B, Ohira M, Liu Y, Sakane S, Carlessi R, Sauceda C, Dhar D, Ganguly S, Hosseini M, Teneche MG, Adams PD, Gonzalez DJ, Kisseleva T, Tirnitz-Parker JEE, Simon MC, Alexandrov LB, Karin M. FBP1 controls liver cancer evolution from senescent MASH hepatocytes. Nature 2025; 637:461-469. [PMID: 39743585 DOI: 10.1038/s41586-024-08317-9] [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: 07/22/2023] [Accepted: 10/30/2024] [Indexed: 01/04/2025]
Abstract
Hepatocellular carcinoma (HCC) originates from differentiated hepatocytes undergoing compensatory proliferation in livers damaged by viruses or metabolic-dysfunction-associated steatohepatitis (MASH)1. While increasing HCC risk2, MASH triggers p53-dependent hepatocyte senescence3, which we found to parallel hypernutrition-induced DNA breaks. How this tumour-suppressive response is bypassed to license oncogenic mutagenesis and enable HCC evolution was previously unclear. Here we identified the gluconeogenic enzyme fructose-1,6-bisphosphatase 1 (FBP1) as a p53 target that is elevated in senescent-like MASH hepatocytes but suppressed through promoter hypermethylation and proteasomal degradation in most human HCCs. FBP1 first declines in metabolically stressed premalignant disease-associated hepatocytes and HCC progenitor cells4,5, paralleling the protumorigenic activation of AKT and NRF2. By accelerating FBP1 and p53 degradation, AKT and NRF2 enhance the proliferation and metabolic activity of previously senescent HCC progenitors. The senescence-reversing and proliferation-supportive NRF2-FBP1-AKT-p53 metabolic switch, operative in mice and humans, also enhances the accumulation of DNA-damage-induced somatic mutations needed for MASH-to-HCC progression.
Collapse
Affiliation(s)
- Li Gu
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA.
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, China.
- Clinical Laboratory Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.
| | - Yahui Zhu
- School of Medicine, Chongqing University, Chongqing, China.
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Shuvro P Nandi
- Department of Cellular and Molecular Medicine, UCSD, La Jolla, CA, USA
- Department of Bioengineering, UCSD, La Jolla, CA, USA
- Moores Cancer Center, UCSD, La Jolla, CA, USA
| | - Maiya Lee
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Kosuke Watari
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Breanna Bareng
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Masafumi Ohira
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | - Yuxiao Liu
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA
| | | | - Rodrigo Carlessi
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Consuelo Sauceda
- Department of Pharmacology, UCSD, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, CA, USA
| | | | | | | | - Marcos G Teneche
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Peter D Adams
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - David J Gonzalez
- Department of Pharmacology, UCSD, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, UCSD, La Jolla, CA, USA
| | | | - Janina E E Tirnitz-Parker
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Bentley, Western Australia, Australia
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | - M Celeste Simon
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine, UCSD, La Jolla, CA, USA
- Department of Bioengineering, UCSD, La Jolla, CA, USA
- Moores Cancer Center, UCSD, La Jolla, CA, USA
| | - Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of Medicine, University of California San Diego (UCSD), La Jolla, CA, USA.
| |
Collapse
|
41
|
Zhang X, Zhang M, Sun H, Wang X, Wang X, Sheng W, Xu M. The role of transcription factors in the crosstalk between cancer-associated fibroblasts and tumor cells. J Adv Res 2025; 67:121-132. [PMID: 38309692 PMCID: PMC11725164 DOI: 10.1016/j.jare.2024.01.033] [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: 11/29/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024] Open
Abstract
BACKGROUND Transcription factors (TFs) fulfill a critical role in the formation and maintenance of different cell types during the developmental process as well as disease. It is believed that cancer-associated fibroblasts (CAFs) are activation status of tissue-resident fibroblasts or derived from form other cell types via transdifferentiation or dedifferentiation. Despite a subgroup of CAFs exhibit anti-cancer effects, most of them are reported to exert effects on tumor progression, further indicating their heterogeneous origin. AIM OF REVIEW This review aimed to summarize and review the roles of TFs in the reciprocal crosstalk between CAFs and tumor cells, discuss the emerging mechanisms, and their roles in cell-fate decision, cellular reprogramming and advancing our understanding of the gene regulatory networks over the period of cancer initiation and progression. KEY SCIENTIFIC CONCEPTS OF REVIEW This manuscript delves into the key contributory factors of TFs that are involved in activating CAFs and maintaining their unique states. Additionally, it explores how TFs play a pivotal and multifaceted role in the reciprocal crosstalk between CAFs and tumor cells. This includes their involvement in processes such as epithelial-mesenchymal transition (EMT), proliferation, invasion, and metastasis, as well as metabolic reprogramming. TFs also have a role in constructing an immunosuppressive microenvironment, inducing resistance to radiation and chemotherapy, facilitating angiogenesis, and even 'educating' CAFs to support the malignancies of tumor cells. Furthermore, this manuscript delves into the current status of TF-targeted therapy and considers the future directions of TFs in conjunction with anti-CAFs therapies to address the challenges in clinical cancer treatment.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Meng Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Hui Sun
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xu Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Xin Wang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| | - Midie Xu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China.
| |
Collapse
|
42
|
Tian R, Zhang H, Wang C, Zhou S, Zhang L, Wang H. Research on Prediction of Multiple Degenerative Diseases and Biomarker Screening Based on DNA Methylation. Int J Mol Sci 2025; 26:313. [PMID: 39796173 PMCID: PMC11719970 DOI: 10.3390/ijms26010313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/21/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
The aging process will lead to a gradual functional decline in the human body, and even accelerate a significantly increased risk of degenerative diseases. DNA methylation patterns change markedly with one's age, serving as a biomarker of biological age and closely linked to the occurrence and progression of age-related diseases. Currently, diagnostic methods for individual degenerative diseases are relatively mature. However, aging often accompanies the onset of multiple degenerative diseases, presenting certain limitations in existing diagnostic models. Additionally, some identified DNA methylation biomarkers are typically applicable to only one or a few types of cancer or diseases, further restricting their utility. We endeavor to screen for biomarkers associated with multiple degenerative diseases from the perspective of aging-related co-morbid mechanisms and to perform multiple degenerative disease diagnoses. In this study, we explored research based on methylation correlations and patterns to investigate shared mechanisms across multiple degenerative diseases, identifying a set of biomarkers associated with them. We validated these biomarkers with biological omics analysis and the prediction of multiple classes of degenerative diseases, screened the biomarkers from 600 to 110 by biological omics analysis, and demonstrated the validity and predictive ability of the screened 110 biomarkers. We propose a disease diagnostic model based on a multi-scale one-dimensional convolutional neural network (MSDCNN) and a multi-class degenerative disease prediction model (ResDegNet). The two models are well trained and tested to accurately diagnose diseases and categorize four types of degenerative diseases. The research identified 110 biomarkers associated with degenerative diseases, providing a foundation for further exploration of age-related degenerative conditions. This work aims to facilitate early diagnosis, the identification of biomarkers, and the development of therapeutic targets for drug interventions.
Collapse
Affiliation(s)
- Ruoting Tian
- College of Computer Science and Engineering, Changchun University of Technology, Changchun 130051, China; (R.T.); (C.W.); (S.Z.)
| | - Hao Zhang
- School of Information Science and Engineering (School of Software), Yanshan University, Qinhuangdao 066000, China;
| | - Chencai Wang
- College of Computer Science and Engineering, Changchun University of Technology, Changchun 130051, China; (R.T.); (C.W.); (S.Z.)
| | - Shengyang Zhou
- College of Computer Science and Engineering, Changchun University of Technology, Changchun 130051, China; (R.T.); (C.W.); (S.Z.)
| | - Li Zhang
- College of Computer Science and Engineering, Changchun University of Technology, Changchun 130051, China; (R.T.); (C.W.); (S.Z.)
| | - Han Wang
- School of Information Science and Technology, Institute of Computational Biology, Northeast Normal University, Changchun 130117, China;
| |
Collapse
|
43
|
Casalini R, Romei C, Ciampi R, Ramone T, Prete A, Gambale C, Matrone A, Torregrossa L, Ugolini C, Elisei R. Minor role of TP53 and TERT promoter mutations in medullary thyroid carcinoma: report of new cases and revision of the literature. Endocrine 2025; 87:243-251. [PMID: 39179735 DOI: 10.1007/s12020-024-03990-2] [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: 07/03/2024] [Accepted: 08/02/2024] [Indexed: 08/26/2024]
Abstract
PURPOSE Aims of this study were to investigate the prevalence of TP53 and TERT mutations in Medullary Thyroid carcinoma (MTC) and their role in inducing aggressiveness in positive cases. METHODS We performed a literature search in PubMed to identify studies investigating the prevalence of TERT and TP53 mutations in MTC. We also included data on MTC cases (n = 193) obtained at our center and unpublished. The in-silico pathogenicity of the TP53 mutations has been evaluated by predictor tools. RESULTS We identified a total of 25 and 11 published papers: all together 1280 cases have been investigated for the presence of TP53 mutations and 974 for TERT promoter mutation. Twenty-five out of 1280 (2%) cases had a TP53 mutation while only 3/974 MTC cases (0.3%) have been found to be positive for TERT promoter mutations. Among all, we identified 19 different TP53 mutations that in 12 cases were demonstrated to have an in silico predicted high pathogenic role and a high impact on protein function. Three non-sense and 4 probably not damaging mutations were also reported. The pathogenic role of the TERT promoter mutations has been previously in vitro determined. No correlation between TP53 and/or TERT mutations and aggressiveness of MTC has been demonstrated. CONCLUSION The prevalence of TP53 and TERT promoter mutations is very low in MTC. The reported mutations are pathogenic in the majority of cases. Because of their rarity it is not possible to clarify if they play or not a role in the pathogenesis and/or aggressiveness of this specific thyroid tumor.
Collapse
Affiliation(s)
- Roberta Casalini
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Cristina Romei
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Raffaele Ciampi
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Teresa Ramone
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Alessandro Prete
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Carla Gambale
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Antonio Matrone
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Liborio Torregrossa
- Department of Surgical, Medical, Molecular Pathology and Critical Area, Unit of Pathology, University Hospital of Pisa, Pisa, Italy
| | - Clara Ugolini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, Unit of Pathology, University Hospital of Pisa, Pisa, Italy
| | - Rossella Elisei
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy.
| |
Collapse
|
44
|
Alfaraidi M, Gilks CB, Hoang L. Typing of Vulvar Squamous Cell Carcinoma: Why it is Important? Adv Anat Pathol 2025; 32:20-29. [PMID: 39318249 DOI: 10.1097/pap.0000000000000466] [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: 09/26/2024]
Abstract
The classification of vulvar squamous cell carcinoma (VSCC), as in endometrial cancer, has shifted from the histology-based descriptors toward molecular-based identifiers. Recently, it has been reported that there are 3 genetically distinct and clinically significant subtypes of VSCC: HPV-associated VSCC, HPV-independent/p53 wild-type VSCC, and HPV-independent/p53-mutated VSCC. Each group has different prognostic implications as well as response to treatment, thus reinforcing the need for this 3-tier molecular classification. This molecular subtyping can easily be done on vulvar biopsies using p16 and p53 immunohistochemistry stains to further improve risk prediction and individualized treatment decisions, leading to better patient outcomes.
Collapse
Affiliation(s)
- Mona Alfaraidi
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, Canada
| | - C Blake Gilks
- Department of Pathology, Prince Sultan Military Medical Hospital, Riyadh, Saudi Arabia
| | - Lynn Hoang
- Department of Pathology, Prince Sultan Military Medical Hospital, Riyadh, Saudi Arabia
| |
Collapse
|
45
|
Wu S, Zuo Y, Ye M, Wang K, Wang X, Yang X, Wang C. Co‑occurrence of clear cell renal cell carcinoma and bladder urothelial carcinoma: A case report and literature review. Oncol Lett 2025; 29:21. [PMID: 39492932 PMCID: PMC11526436 DOI: 10.3892/ol.2024.14768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/25/2024] [Indexed: 11/05/2024] Open
Abstract
The co-occurrence of clear cell renal cell carcinoma (ccRCC) and bladder urothelial carcinoma (bUC) is rare, and owing to the lack of a unified treatment plan, the prognosis is poor. The present report describes the case of a 65-year-old male patient with a history of smoking and no history of malignant tumors who presented with hematuria at the Huanghe Sanmenxia Hospital Affiliated to Henan University of Science and Technology (Sanmenxia, China) in July 2021. Urinary system computed tomography urography revealed a right renal tumor, and cystoscopy revealed intravesical lesions. The patient underwent transurethral resection of a bladder tumor + laparoscopic partial nephrectomy + laparoscopic radical cystectomy and bilateral ureterostomy. Pathological examination revealed right-sided ccRCC (pT1aN0M0) and high-grade invasive bUC (pT2N0M0). After surgery, the patient underwent bilateral ureteral single J tube replacement in the outpatient clinic every 3 months. In September 2022, the patient presented with a mass on the right side of the neck. Further examination revealed a space-occupying lesion in the lower part of the left kidney and space-occupying lesions in the neck, axilla, mediastinal lymph nodes and liver. A neck lymph node puncture biopsy suggested UC, and the patient was diagnosed with metastatic UC (T4N0M1). The patient received tislelizumab (200 mg once every 3 weeks) + sunitinib (50 mg/day, administered for 4 weeks with a 2-week interval) for a total of 2 months and died of an advanced tumor in January 2023. In addition, the data of 36 patients with ccRCC and bUC from the literature were analyzed for the present report. The results showed that the median age at first onset was 56.5 years (range, 31-82 years) and the male-to-female ratio was 6:1. Smoking and male sex may be risk factors for this disease, which has a median survival time of 47.5 months. The survival analysis results showed that the pathological stage of bladder cancer may be associated with its prognosis. The present study reviews the potential risks, clinicopathological characteristics and treatment methods of co-occurrence of clear ccRCC and bUC. In conclusion, the high-risk factors for the co-occurrence of ccRCC and bUC were smoking and male sex, and the median survival time was 47.5 months. The pathological stage of bladder cancer may be related to the prognosis.
Collapse
Affiliation(s)
- Shuo Wu
- Department of Urology, Huanghe Sanmenxia Hospital Affiliated to Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| | - Yuliang Zuo
- Department of Urology, Huanghe Sanmenxia Hospital Affiliated to Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| | - Meihong Ye
- Department of Pathology, Huanghe Sanmenxia Hospital Affiliated to Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| | - Kuan Wang
- Department of Urology, Huanghe Sanmenxia Hospital Affiliated to Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| | - Xiaolong Wang
- Department of Urology, Huanghe Sanmenxia Hospital Affiliated to Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| | - Xudong Yang
- Department of Urology, Huanghe Sanmenxia Hospital Affiliated to Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| | - Chaoming Wang
- Department of Urology, Huanghe Sanmenxia Hospital Affiliated to Henan University of Science and Technology, Sanmenxia, Henan 472000, P.R. China
| |
Collapse
|
46
|
Guo L, Chen W, Yue J, Gao M, Zhang J, Huang Y, Xiong H, Li X, Wang Y, Yuan Y, Chen L, Fei F, Xu R. Unlocking the potential of LHPP: Inhibiting glioma growth and cell cycle via the MDM2/p53 pathway. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167509. [PMID: 39277057 DOI: 10.1016/j.bbadis.2024.167509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/31/2024] [Accepted: 09/07/2024] [Indexed: 09/17/2024]
Abstract
The recurrence of glioma after treatment has remained an intractable problem for many years. Recently, numerous studies have explored the pivotal role of the mouse double minute 2 (MDM2)/p53 pathway in cancer treatment. Lysine phosphate phosphohistidine inorganic pyrophosphate phosphatase (LHPP), a newly discovered tumor suppressor, has been confirmed in numerous studies on tumors, but its role in glioma remains poorly understood. Expression matrices in The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) databases were analyzed using gene set enrichment analysis (GSEA), revealing significant alterations in the p53 pathway among glioma patients with high LHPP expression. The overexpression of LHPP in glioma cells resulted in a reduction in cell proliferation, migration, and invasive ability, as well as an increase in apoptosis and alterations to the cell cycle. The present study has identified a novel inhibitory mechanism of LHPP against glioma, both in vivo and in vitro. The results demonstrate that LHPP exerts anti-glioma effects via the MDM2/p53 pathway. These findings may offer a new perspective for the treatment of glioma in the clinic.
Collapse
Affiliation(s)
- Lili Guo
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenjin Chen
- Department of Neurosurgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Jiong Yue
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Mingjun Gao
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jin Zhang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yukai Huang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Huan Xiong
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinda Li
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yangyang Wang
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ying Yuan
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Longyi Chen
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Fan Fei
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Ruxiang Xu
- Department of Neurosurgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
47
|
Xiong B, Zhang X, Sangji D, Ni L, Fan M, Fan B. Mechanisms of breast cancer treatment using Gentiana robusta: evidence from comprehensive bioinformatics investigation. Sci Rep 2024; 14:31567. [PMID: 39738201 PMCID: PMC11686125 DOI: 10.1038/s41598-024-76063-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/10/2024] [Indexed: 01/01/2025] Open
Abstract
This study investigates the potential treatment of breast cancer utilizing Gentiana robusta King ex Hook. f. (QJ) through an integrated approach involving network pharmacology, molecular docking, and molecular dynamics simulation. Building upon prior research on QJ's chemical constituents, we conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis using the DAVID database. Network interactions and core genes were identified using Cytoscape 3.9.1. Key target genes, including Interleukin-6 (IL-6), tumour suppressor gene P53 (TP53), and epidermal growth factor receptor (EGFR), were selected for molecular docking with QJ's active components, 2'-O-β-D-glucopyranosyl-gentiopicroside and macrophylloside D, employing Schrodinger Maestro 13.5. Molecular dynamics (MD) simulations were performed using the Desmond program. A total of 270 intersection targets of active ingredients and diseases were identified, with three core targets determined through network topology screening. Enrichment analysis highlighted the involvement of QJ in breast cancer treatment, primarily through the hsa05200 cancer signaling pathway and the hsa04066 HIF-1 signaling pathway. Molecular docking and dynamics simulations demonstrated the close interaction of 2'-O-β-D-glucopyranosyl-gentiopicroside (QJ17) and macrophylloside D (QJ25) with IL6, TP53, and EGFR, and other target genes, showcasing a stabilizing effect. In conclusion, this study unveils the effective components and potential mechanisms of 2'-O-β-D-glucopyranosyl-gentiopicroside and macrophylloside D in breast cancer prevention and treatment. The identified components act on target genes such as IL6, TP53, and EGFR, regulating crucial pathways including the cancer signaling and Hypoxia-inducible factor 1 (HIF-1) signaling pathways. These findings provide valuable insights into the therapeutic potential of QJ in breast cancer management. However, further experimental research are needed to validate the computational findings of QJ.
Collapse
Affiliation(s)
- Bo Xiong
- Department of Clinical Pharmacy, Baoshan Hospital Affiliated to, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinxin Zhang
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Dongzhi Sangji
- Tibetan Medical Hospital of Xizang Autonomous Region, Lhasa, China
| | - Lianghong Ni
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Mingjie Fan
- Department of Pharmacy, Shanghai Fourth Rehabilitation Hospital, Shanghai, China.
| | - Beibei Fan
- Department of Clinical Pharmacy, Baoshan Hospital Affiliated to, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
48
|
Chai D, Wang X, Fan C, Wang J, Lim JM, Yu X, Young KH, Li Y. Vaccines targeting p53 mutants elicit anti-tumor immunity. Cancer Lett 2024; 611:217421. [PMID: 39740750 DOI: 10.1016/j.canlet.2024.217421] [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: 10/14/2024] [Revised: 12/09/2024] [Accepted: 12/22/2024] [Indexed: 01/02/2025]
Abstract
The p53 tumor suppressor is commonly mutated in cancer; however, there are no effective treatments targeting p53 mutants. A DNA vaccine gWIZ-S237G targeting the p53 S237G mutant, which is highly expressed in A20 murine tumor cells, was developed and administered intramuscularly via electroporation, either alone or in combination with PD-1 blockade. The anti-p53-S237G immunization elicited a robust protective response against subcutaneous A20 tumors and facilitated the infiltration of immune cells including CD8+ T cells, NK cells, and DCs. The vaccine enhanced the induction and maturation of CD11c+, CD103+CD11c+, and CD8+CD11c+ cells, which in turn promoted tumor-specific antibody production, as well as Th1 and CD8+ T cell-mediated immune responses. Several antigenic epitopes of p53-S237G effectively stimulated multifunctional CD8+ T cells to secrete IFN-γ and TNF-α. The vaccine showed long-term anti-tumor effects that were dependent on memory CD8+ T cells. Furthermore, the anti-p53-S237G vaccine exhibited significant protective efficacy in the A20 liver metastasis models. When combined with PD-1 inhibition, the vaccine showed superior inhibition of tumor growth and liver metastasis. Targeting p53 mutants by vaccination represents a potential precision medicine strategy against cancers harboring p53 mutations.
Collapse
Affiliation(s)
- Dafei Chai
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Xu Wang
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Chunmei Fan
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Junhao Wang
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jing Ming Lim
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xinfang Yu
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ken H Young
- Department of Pathology, Division of Hematopathology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Yong Li
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
49
|
Zhong C, Wang J, Peng H, Lu J, Long Z, Lin Z, Chen G, Cai C, Cheng S, Chen Z, Zhang L, Zhong W, Mo R, Mao X. GG-NER's role in androgen receptor signaling inhibitor response for advanced prostate cancer. Cell Commun Signal 2024; 22:600. [PMID: 39696559 DOI: 10.1186/s12964-024-01977-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Advanced prostate cancer (PCa) often initially responds to androgen receptor signaling inhibitors (ARSI) but frequently develops resistance, driven by tumor heterogeneity and therapeutic pressure. Addressing the clinical challenge of identifying non-responsive patients and discovering new therapeutic targets is urgently needed. METHODS We utilized single-sample gene set enrichment analysis (ssGSEA) to elucidate the influence of the GG-NER pathway on ARSI response in PCa. We then constructed and validated a prognostic model based on this pathway using LASSO regression, Kaplan-Meier analysis, Cox regression, and ROC analysis. Additionally, we mapped tumor mutations to delineate the mutational landscapes across different risk groups and explored functional pathways through GO, KEGG, and GSEA analyses. The impact of the GG-NER pathway on enzalutamide sensitivity and DNA repair in PCa was further validated through CCK-8 assays, colony formation assays, in vivo experiments, and immunofluorescence. RESULTS ssGSEA indicated a trend of GG-NER pathway upregulation in patients with poor ARSI response. The GG-NER characteristic gene score (NECGS) identified a high-risk group with diminished ARSI response, serving as an independent prognostic indicator with strong predictive power. This high-risk group exhibited elevated TP53 mutation frequencies and significant enrichment in key pathways such as ribosome and mitochondrial functions, as well as MYC and E2F signaling. Experimental validation confirmed that targeting the GG-NER pathway or its key gene, ACTL6A, significantly reduces enzalutamide resistance in resistant cell lines and increases γH2AX expression. CONCLUSION NECGS effectively predicts ARSI response in PCa, and our comprehensive analysis underscores the critical role of the GG-NER pathway in enzalutamide resistance, positioning ACTL6A as a potential therapeutic target for PCa.
Collapse
Affiliation(s)
- Chuanfan Zhong
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China
- Department of Urology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), 523059, Dongguan, Guangdong, China
| | - Jiaxing Wang
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China
| | - Hangyang Peng
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China
| | - Jianming Lu
- Department of Andrology, Guangzhou First People's Hospital, Guangzhou Medical University, 510180, Guangzhou, Guangdong, China.
| | - Zining Long
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China
| | - Zhuoyuan Lin
- Department of Urology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Guo Chen
- Department of Urology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Chao Cai
- Department of Urology, Minimally Invasive Surgery Center, Guangdong Key Laboratory of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shilong Cheng
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China
| | - Zhongjie Chen
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China
| | - Le Zhang
- Institute for Integrative Genome Biology, University of California, Riverside, California, United States
| | - Weibo Zhong
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China.
| | - Rujun Mo
- Department of Urology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan people's hospital), 523059, Dongguan, Guangdong, China.
| | - Xiangming Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, 510282, Guangzhou, Guangdong, China.
| |
Collapse
|
50
|
Sun JX, An Y, Xu MY, Ma SY, Liu CQ, Xu JZ, Xia QD, Wang SG. Analysis of transcriptomic data reveals the landscape of cholesterol metabolism in prostate cancer and impact of related signature on survival. Discov Oncol 2024; 15:777. [PMID: 39692951 DOI: 10.1007/s12672-024-01658-x] [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: 08/09/2024] [Accepted: 12/02/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Cholesterol metabolism is essential for the development and progression of prostate cancer (PCa). Our previous study provided a new insight of cholesterol metabolism in the systematic management of PCa. However, the comprehensive role of cholesterol metabolism in PCa remains unclear. METHODS Using the cholesterol metabolism related genes (CMRGs) downloaded from the MSigDB database, and gene expression data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), we constructed a cholesterol risk index by the least absolute shrinkage and selection operator (LASSO) model, and correlated the risk index with prognosis, tumor mutation burden (TMB), tumor microenvironment (TME) infiltration and response to chemotherapy and immunotherapy. RT-qPCR, western blot, immunohistochemistry, cell proliferation assays by CCK-8 and EdU assays, and cell apoptosis assays by flow cytometry analysis were also performed. RESULTS We found PCa was tightly correlated with the cholesterol metabolism pathways. The cholesterol risk index was an excellent and independent predictor of prognosis for PCa. A nomogram involving the risk index and other clinical factors (age, T stage) was established to explore the clinical value of risk index. We found high-risk index group was associated with worse prognosis, higher TMB, lower infiltration level of CD8+ T cells and a worse response to chemotherapy and immunotherapy. RT-qPCR, western blot and immunohistochemical staining validated the expression level of important CMRGs in PCa. In vitro experiments revealed downregulation of cholesterol metabolism could inhibit the proliferation of PCa cells and promoted their apoptosis. CONCLUSIONS We demonstrated the comprehensive role of cholesterol metabolism in PCa. Using the risk index, we could predict the prognosis, TME infiltration and response to chemotherapy/immunotherapy of PCa. Better understanding and evaluating the cholesterol metabolism could aid in precision medicine and promoting prognosis of PCa.
Collapse
Affiliation(s)
- Jian-Xuan Sun
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095Jiefang Avenue, Wuhan, 430030, Wuhan, P.R. China
| | - Ye An
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095Jiefang Avenue, Wuhan, 430030, Wuhan, P.R. China
| | - Meng-Yao Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095Jiefang Avenue, Wuhan, 430030, Wuhan, P.R. China
| | - Si-Yang Ma
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095Jiefang Avenue, Wuhan, 430030, Wuhan, P.R. China
| | - Chen-Qian Liu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095Jiefang Avenue, Wuhan, 430030, Wuhan, P.R. China
- Laboratory of Signaling and Gene Regulation, Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Jin-Zhou Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095Jiefang Avenue, Wuhan, 430030, Wuhan, P.R. China
| | - Qi-Dong Xia
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095Jiefang Avenue, Wuhan, 430030, Wuhan, P.R. China.
| | - Shao-Gang Wang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095Jiefang Avenue, Wuhan, 430030, Wuhan, P.R. China.
| |
Collapse
|