1
|
Lee S, Lee B, Kwon SH, Park J, Kim SH. MCC in the spotlight: Its dual role in signal regulation and oncogenesis. Cell Signal 2025; 131:111756. [PMID: 40118128 DOI: 10.1016/j.cellsig.2025.111756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
The mutated in colorectal cancer (MCC) gene is closely associated with the onset and progression of colorectal cancer. MCC plays a critical role in regulating the cell cycle and various signaling pathways and is recognized to inhibit cancer cell proliferation via the β-catenin signaling pathway. β-catenin is a key component of the WNT signaling pathway that influences cell growth, differentiation, survival, and migration, thereby positioning MCC as an important tumor suppressor. Notably, MCC has also been implicated in other cancer types, including lung, liver, and brain cancers. However, the precise mechanisms by which MCC functions in these malignancies remain inadequately understood. Comprehensive investigations into the interactions among MCC, various signaling pathways, and metabolic processes are essential for uncovering the molecular mechanisms of cancer and the pathological features characteristic of different cancer stages. This review presents the structural characteristics of MCC and its cell growth regulation mechanisms and functional roles within tissues, with the aims of enhancing our understanding of the role of MCC in cancer biology and highlighting potential therapeutic strategies targeting this gene.
Collapse
Affiliation(s)
- Soohyeon Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - Beomwoo Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, South Korea
| | - So Hee Kwon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, South Korea.
| | - Jongsun Park
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, South Korea; Biomedical Research Institute, Chungnam National University Hospital, Daejeon 35015, Republic of Korea.
| | - Seon-Hwan Kim
- Biomedical Research Institute, Chungnam National University Hospital, Daejeon 35015, Republic of Korea; Department of Neurosurgery, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon 35015, South Korea.
| |
Collapse
|
2
|
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
|
3
|
Terzapulo X, Dyussupova A, Ilyas A, Boranova A, Shevchenko Y, Mergenbayeva S, Filchakova O, Gaipov A, Bukasov R. Detection of Cancer Biomarkers: Review of Methods and Applications Reported from Analytical Perspective. Crit Rev Anal Chem 2025:1-46. [PMID: 40367278 DOI: 10.1080/10408347.2025.2497868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
One in five deaths in developed countries is related to cancer. The cancer prevalence is likely to grow with aging population. The affordable and accurate early diagnostics of cancer based on detection of cancer biomarkers at low concentration during its early stages is one of the most efficient way to decrease mortality and human suffering from cancer. The data from 201 analytical papers are tabulated in 9 tables, illustrated in 8 figures and used for comparative analysis of methods applied for cancer biomarker detection, including polymerase chain reaction, Loop-mediated isothermal amplification (LAMP), mass spectrometry, enzyme-linked immunosorbent assay, electroanalytical methods, immunoassays, surface enhanced Raman scattering, Fourier Transform Infrared and others in terms of above-mentioned performance parameters. Median and/or average limit of detection (LOD) are calculated and compared between different analytical methods. We also described and compared LOD of the methods used for detection of three frequently detected cancer biomarkers: carcinoembryonic antigen, prostate-specific antigen and alpha-fetoprotein. Among those methods of detection, the reported electrochemical sensors often demonstrate relatively high sensitivity/low LOD while they often have a moderate instrumental cost and fast time to results. The review tabulates, compares and discusses analytical papers, which report LOD of cancer biomarkers and comprehensive quantitative comparison of various analytical methods is made. The discussion of those techniques applied for cancer biomarker detection included brief summary of pro and cons for each of those methods.
Collapse
Affiliation(s)
- Xeniya Terzapulo
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Aigerim Dyussupova
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Aisha Ilyas
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Aigerim Boranova
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Yegor Shevchenko
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Saule Mergenbayeva
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Olena Filchakova
- Biology Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Abduzhappar Gaipov
- Department of Medicine, Nazarbayev University School of Medicine, Astana, Republic of Kazakhstan
| | - Rostislav Bukasov
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| |
Collapse
|
4
|
Gori K, Baez-Ortega A, Strakova A, Stammnitz MR, Wang J, Chan J, Hughes K, Belkhir S, Hammel M, Moralli D, Bancroft J, Drydale E, Allum KM, Brignone MV, Corrigan AM, de Castro KF, Donelan EM, Faramade IA, Hayes A, Ignatenko N, Karmacharya R, Koenig D, Lanza-Perea M, Lopez Quintana AM, Meyer M, Neunzig W, Pedraza-Ordoñez F, Phuentshok Y, Phuntsho K, Ramirez-Ante JC, Reece JF, Schmeling SK, Singh S, Tapia Martinez LJ, Taulescu M, Thapa S, Thapa S, van der Wel MG, Wehrle-Martinez AS, Stratton MR, Murchison EP. Horizontal transfer of nuclear DNA in transmissible cancer. Proc Natl Acad Sci U S A 2025; 122:e2424634122. [PMID: 40261943 PMCID: PMC12067285 DOI: 10.1073/pnas.2424634122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/05/2025] [Indexed: 04/24/2025] Open
Abstract
Horizontal transfer of nuclear DNA between cells of host and cancer is a potential source of adaptive variation in cancer cells. An understanding of the frequency and significance of this process in naturally occurring tumors is, however, lacking. We screened for this phenomenon in the transmissible cancers of dogs and Tasmanian devils and found an instance in the canine transmissible venereal tumor (CTVT). This involved introduction of a 15-megabase dicentric genetic element, composed of 11 fragments of six chromosomes, to a CTVT sublineage occurring in Asia around 2,000 y ago. The element forms the short arm of a small submetacentric chromosome and derives from a dog with ancestry associated with the ancient Middle East. The introduced DNA fragment is transcriptionally active and has adopted the expression profile of CTVT. Its features suggest that it may derive from an engulfed apoptotic body. Our findings indicate that nuclear horizontal gene transfer, although likely a rare event in tumor evolution, provides a viable mechanism for the acquisition of genetic material in naturally occurring cancer genomes.
Collapse
Affiliation(s)
- Kevin Gori
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, CambridgeCB3 0ES, United Kingdom
| | - Adrian Baez-Ortega
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, CambridgeCB3 0ES, United Kingdom
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, HinxtonCB10 1SA, United Kingdom
| | - Andrea Strakova
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, CambridgeCB3 0ES, United Kingdom
| | - Maximilian R. Stammnitz
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, CambridgeCB3 0ES, United Kingdom
| | - Jinhong Wang
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, CambridgeCB3 0ES, United Kingdom
| | - Jonathan Chan
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, CambridgeCB3 0ES, United Kingdom
| | - Katherine Hughes
- Department of Veterinary Medicine, University of Cambridge, CambridgeCB3 0ES, United Kingdom
| | - Sophia Belkhir
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, CambridgeCB3 0ES, United Kingdom
| | - Maurine Hammel
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, CambridgeCB3 0ES, United Kingdom
| | - Daniela Moralli
- Pandemic Sciences Institute, University of Oxford, OxfordOX3 7DQ, United Kingdom
| | - James Bancroft
- Cellular Imaging Core Facility, Centre for Human Genetics, University of Oxford, OxfordOX3 7BM, United Kingdom
| | - Edward Drydale
- Cellular Imaging Core Facility, Centre for Human Genetics, University of Oxford, OxfordOX3 7BM, United Kingdom
| | | | - María Verónica Brignone
- Faculty of Veterinary Sciences, Universidad de Buenos Aires, Buenos AiresC1053ABJ, Argentina
| | - Anne M. Corrigan
- School of Veterinary Medicine, St. George’s University, True Blue, Grenada
| | - Karina F. de Castro
- Faculty of Agrarian and Veterinary Sciences, São Paulo State University, Jaboticabal14884-900, Brazil
| | - Edward M. Donelan
- Animal Management in Rural and Remote Indigenous Communities, Darwin, NT0820, Australia
| | | | - Alison Hayes
- Department of Veterinary Medicine, University of Cambridge, CambridgeCB3 0ES, United Kingdom
| | | | - Rockson Karmacharya
- Veterinary Diagnostic and Research Laboratory Pvt. Ltd., Kathmandu44600, Nepal
| | | | - Marta Lanza-Perea
- School of Veterinary Medicine, St. George’s University, True Blue, Grenada
| | | | | | | | | | | | | | - Juan C. Ramirez-Ante
- Facultad de Ciencias Pecuarias, Corporación Universitaria Santa Rosa de Cabal, Santa Rosa de Cabal661020, Colombia
| | | | | | - Sanjay Singh
- Help in Suffering, Jaipur302018, Rajasthan, India
| | | | - Marian Taulescu
- Department of Anatomic Pathology, Faculty of Veterinary Medicine, University of Agricultural Sciences and Veterinary Medicine, Cluj-Napoca400372, Romania
| | - Samir Thapa
- Kathmandu Animal Treatment Centre, Kathmandu44622, Nepal
| | - Sunil Thapa
- Animal Nepal, Dobighat, Kathmandu44600, Nepal
| | | | | | - Michael R. Stratton
- Cancer, Ageing and Somatic Mutation Programme, Wellcome Sanger Institute, HinxtonCB10 1SA, United Kingdom
| | - Elizabeth P. Murchison
- Transmissible Cancer Group, Department of Veterinary Medicine, University of Cambridge, CambridgeCB3 0ES, United Kingdom
| |
Collapse
|
5
|
Choudalakis S, Kastis GA, Dikaios N. Intra-clustering analysis reveals tissue-specific mutational patterns. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 263:108681. [PMID: 40050208 DOI: 10.1016/j.cmpb.2025.108681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 02/06/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND AND OBJECTIVE The identification of tissue-specific mutational patterns associated with cancer is challenging due to the low frequency of certain mutations and the high variability among tumors within the same cancer type. To address the inter-tumoral heterogeneity issue, our study aims to uncover infrequent mutational patterns by proposing a novel intra-clustering analysis. METHODS A Network Graph of 8303 patients and 198 genes was constructed using single-point-mutation data from The Cancer Genome Atlas (TCGA). Patient-gene groups were retrieved with the parallel use of two separate methodologies based on the: (a) Barber's modularity index, and (b) network dynamics. An intra-clustering analysis was employed to explore the patterns within smaller patient subgroups in two phases: i) to determine the significant presence of a gene with a cancer type using the Fisher's exact test and ii) to determine gene-to-gene patterns using multiple correspondence analysis and DISCOVER. The results are followed by a Benjamini-Hochberg false discovery rate of 5%. RESULTS This analysis was applied over 24 statistically meaningful groups of 2619 patients spanning 21 cancer types and it recovered 42 mutational patterns that are not reported in the TCGA consortium publications. Notably, our findings: (i) suggest that AMER1 mutations are a putative separative element between colon and rectal adenocarcinomas, (ii) highlight the significant presence of RAC1 in head and neck squamous cell carcinoma (iii) suggest that EP300 mutations in head and neck squamous cell carcinoma are irrelevant of the HPV status of the patients and (iv) show that mutational-based clusters can contain patients with contrasting genetic alterations. CONCLUSIONS The proposed intra-clustering analysis extracted statistically significant relationships within clusters, uncovering putative clinically relevant connections and disentangling mutational heterogeneity.
Collapse
Affiliation(s)
- Stamatis Choudalakis
- Mathematics Research Center, Academy of Athens, 4, Soranou Efesiou str., 11527 Athens, Greece; Medical School of Athens, National and Kapodistrian University of Athens, 75, Mikras Asias str., 11527 Athens, Greece.
| | - George A Kastis
- Mathematics Research Center, Academy of Athens, 4, Soranou Efesiou str., 11527 Athens, Greece.
| | - Nikolaos Dikaios
- Mathematics Research Center, Academy of Athens, 4, Soranou Efesiou str., 11527 Athens, Greece.
| |
Collapse
|
6
|
Pascoal LB, Jalalizadeh M, Barbosa G, da Silva ANMR, Queiroz MAF, Laukhtina E, Shariat SF, Gambero A, Reis LO. Viral infections and immune modulation in bladder cancer: implications for immunotherapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002311. [PMID: 40291982 PMCID: PMC12022759 DOI: 10.37349/etat.2025.1002311] [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: 01/29/2025] [Accepted: 03/27/2025] [Indexed: 04/30/2025] Open
Abstract
This review explores the intricate relationship between viral infections and Bacillus Calmette-Guerin (BCG) efficacy, emphasizing immune modulation mechanisms that may influence treatment outcomes. Since its introduction in 1976, intravesical BCG has been a cornerstone in managing non-muscle invasive bladder cancer (NMIBC) after transurethral resection of bladder tumors (TURBT). Despite its success, variability in response rates suggests that host immune status, influenced by persistent infections, immunosenescence, and antigenic overload, may play a crucial role in therapeutic effectiveness. Chronic viral infections can modulate T cell responses, leading to immune exhaustion and impaired antitumor immunity. This review discusses the interplay between viral antigenic load, immune dysfunction, and tumor microenvironment remodeling, highlighting their potential impact on immunotherapies. By integrating insights from virome analysis, immune profiling, and tumor characterization, this review proposes personalized strategies to enhance immunotherapy efficacy. A deeper understanding of viral-induced immune dysregulation may improve prognostic assessment, optimize treatment protocols, and reduce healthcare costs associated with bladder cancer. Future research should focus on targeted interventions to mitigate the immunosuppressive effects of chronic infections, ultimately improving patient outcomes in NMIBC management.
Collapse
Affiliation(s)
- Lívia Bitencourt Pascoal
- UroScience, State University of Campinas, Campinas 13083-970, Brazil
- ImmunOncology, Pontifical Catholic University of Campinas, Campinas 13060-904, Brazil
- INCT UroGen, National Institute of Science, Technology and Innovation in Genitourinary Cancer (INCT), Campinas 13087-571, Brazil
| | | | - Gabriela Barbosa
- UroScience, State University of Campinas, Campinas 13083-970, Brazil
- ImmunOncology, Pontifical Catholic University of Campinas, Campinas 13060-904, Brazil
- INCT UroGen, National Institute of Science, Technology and Innovation in Genitourinary Cancer (INCT), Campinas 13087-571, Brazil
| | | | - Maria Alice Freitas Queiroz
- Laboratory of Virology, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
| | - Ekaterina Laukhtina
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Shahrokh F. Shariat
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman 19328, Jordan
- Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Urology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Alessandra Gambero
- ImmunOncology, Pontifical Catholic University of Campinas, Campinas 13060-904, Brazil
- INCT UroGen, National Institute of Science, Technology and Innovation in Genitourinary Cancer (INCT), Campinas 13087-571, Brazil
| | - Leonardo O. Reis
- UroScience, State University of Campinas, Campinas 13083-970, Brazil
- ImmunOncology, Pontifical Catholic University of Campinas, Campinas 13060-904, Brazil
- INCT UroGen, National Institute of Science, Technology and Innovation in Genitourinary Cancer (INCT), Campinas 13087-571, Brazil
| |
Collapse
|
7
|
Luo L, Yuan F, Palovcak A, Li F, Yuan Q, Calkins D, Manalo Z, Li Y, Wang D, Zhou M, Zhou C, Li M, Tan YD, Bai F, Ban Y, Mason C, Roberts E, Bilbao D, Liu ZJ, Briegel K, Welford SM, Pei XH, Daunert S, Liu W, Zhang Y. Oncogenic properties of wild-type DNA repair gene FANCA in breast cancer. Cell Rep 2025; 44:115480. [PMID: 40146775 DOI: 10.1016/j.celrep.2025.115480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/10/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
FANCA is one of the 23 genes whose deficiencies lead to defective DNA interstrand crosslink repair and cancer-prone Fanconi anemia disease. Beyond its functions in DNA repair and tumor suppression, we report that high FANCA expression is strongly associated with breast cancer development. Overexpression of WT-FANCA significantly promotes breast cancer cell proliferation and tumor growth both in vitro and in vivo, while FANCA deficiency severely compromises the proliferation of breast cancer cells, but not non-tumorigenic breast epithelial cells. Heterozygous knockout of FANCA in breast cancer mouse models is sufficient to cause significant reduction of breast tumor growth in vivo. Furthermore, we have shown that high FANCA expression in breast cancer correlates with promoter hypomethylation in a TET-dependent manner, and TET inhibition recapitulates the proliferation defects caused by FANCA deficiency. Our study identifies the oncogenic properties of WT-FANCA and shows that FANCA is a promising target for breast cancer intervention.
Collapse
Affiliation(s)
- Liang Luo
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Fenghua Yuan
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Anna Palovcak
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Fang Li
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Qingqi Yuan
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daniel Calkins
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Zoe Manalo
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yan Li
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Dazhi Wang
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Mike Zhou
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Catherine Zhou
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Matthew Li
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yuan-De Tan
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Feng Bai
- International Cancer Center, Shenzhen University Medical School, Shenzhen 518060, China
| | - Yuguang Ban
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Christian Mason
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Evan Roberts
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Daniel Bilbao
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Pathology & Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Zhao-Jun Liu
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Karoline Briegel
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Scott M Welford
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Xin-Hai Pei
- International Cancer Center, Shenzhen University Medical School, Shenzhen 518060, China
| | - Sylvia Daunert
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Wenjun Liu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310003, China.
| | - Yanbin Zhang
- Department of Biochemistry & Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
8
|
Zhang Y, Wang W. Advances in tumor subclone formation and mechanisms of growth and invasion. J Transl Med 2025; 23:461. [PMID: 40259385 PMCID: PMC12012948 DOI: 10.1186/s12967-025-06486-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/11/2025] [Indexed: 04/23/2025] Open
Abstract
Tumor subclones refer to distinct cell populations within the same tumor that possess different genetic characteristics. They play a crucial role in understanding tumor heterogeneity, evolution, and therapeutic resistance. The formation of tumor subclones is driven by several key mechanisms, including the inherent genetic instability of tumor cells, which facilitates the accumulation of novel mutations; selective pressures from the tumor microenvironment and therapeutic interventions, which promote the expansion of certain subclones; and epigenetic modifications, such as DNA methylation and histone modifications, which alter gene expression patterns. Major methodologies for studying tumor subclones include single-cell sequencing, liquid biopsy, and spatial transcriptomics, which provide insights into clonal architecture and dynamic evolution. Beyond their direct involvement in tumor growth and invasion, subclones significantly contribute to tumor heterogeneity, immune evasion, and treatment resistance. Thus, an in-depth investigation of tumor subclones not only aids in guiding personalized precision therapy, overcoming drug resistance, and identifying novel therapeutic targets, but also enhances our ability to predict recurrence and metastasis risks while elucidating the mechanisms underlying tumor heterogeneity. The integration of artificial intelligence, big data analytics, and multi-omics technologies is expected to further advance research in tumor subclones, paving the way for novel strategies in cancer diagnosis and treatment. This review aims to provide a comprehensive overview of tumor subclone formation mechanisms, evolutionary models, analytical methods, and clinical implications, offering insights into precision oncology and future translational research.
Collapse
Affiliation(s)
- Yuhong Zhang
- Department of Oncology, Clinical Medical College, Southwest Medical University, No. 319, Section 3, Zhongshan Road, Luzhou, 646099, Sichuan, China
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Weidong Wang
- Department of Oncology, Clinical Medical College, Southwest Medical University, No. 319, Section 3, Zhongshan Road, Luzhou, 646099, Sichuan, China.
- Department of Radiation Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China.
| |
Collapse
|
9
|
Shu C, Zhou H, Xu J, Tang J, Zhou S. Up-regulation of thrombospondin-1 inhibits the progression of gallbladder cancer. Med Oncol 2025; 42:170. [PMID: 40259006 PMCID: PMC12011972 DOI: 10.1007/s12032-025-02719-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/14/2025] [Indexed: 04/23/2025]
Abstract
Gallbladder cancer, the most prevalent malignant neoplasm of the biliary tract, has garnered significant attention due to its dismal prognosis and high degree of malignancy. Identifying key regulatory genes is crucial for the development of effective therapeutic strategies. The differential gene expression in biliary tract malignancies was identified using the Gene Expression Omnibus (GEO) database. Subsequently, the interactions among these differentially expressed genes were analyzed employing the STRING database, and the resultant regulatory network was visualized using Cytoscape software. Utilizing the Cytoscape plugin CytoHubba, the core genes within the network were identified, followed by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Ultimately, the overexpression of THBS1 in the gallbladder cancer cell line (NOZ) was achieved through lentiviral transfection and both in vivo and in vitro experiments were conducted to evaluate its effects. We found that thrombospondin-1 (THBS1) was the core gene of gallbladder cancer and its expression was low in gallbladder cancer. Experimental data, both in vivo and in vitro, indicate that the up-regulation of THBS1 exerts an inhibitory effect on the proliferation, migration, and invasion of gallbladder cancer cells. Furthermore, it facilitates the process of apoptosis and suppresses tumor growth and angiogenesis. The expression of THBS1 is low in gallbladder cancer. Up-regulation of THBS1 can effectively inhibit the occurrence and development of gallbladder cancer and can be used as a biomarker for the diagnosis and treatment of gallbladder cancer.
Collapse
Affiliation(s)
- Chang Shu
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214100, Jiangsu, China
- Wuxi Medical Center, Nanjing Medical University, Wuxi, 214100, Jiangsu, China
- Wuxi People's Hospital, Wuxi, 214100, Jiangsu, China
| | - Hanxu Zhou
- General Surgery, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China
| | - Jinyu Xu
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214100, Jiangsu, China.
- Wuxi Medical Center, Nanjing Medical University, Wuxi, 214100, Jiangsu, China.
- Wuxi People's Hospital, Wuxi, 214100, Jiangsu, China.
| | - Jie Tang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, 214100, Jiangsu, China.
- Wuxi Medical Center, Nanjing Medical University, Wuxi, 214100, Jiangsu, China.
- Wuxi People's Hospital, Wuxi, 214100, Jiangsu, China.
- General Surgery, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China.
| | - Shaobo Zhou
- General Surgery, The Second Affiliated Hospital of Bengbu Medical College, Bengbu, 233000, Anhui, China.
- General Surgery, Shenzhen Yantian District People's Hospital, Shenzhen, 518081, Guangdong, China.
| |
Collapse
|
10
|
M A Basher AR, Hallinan C, Lee K. Heterogeneity-preserving discriminative feature selection for disease-specific subtype discovery. Nat Commun 2025; 16:3593. [PMID: 40234411 PMCID: PMC12000357 DOI: 10.1038/s41467-025-58718-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Disease-specific subtype identification can deepen our understanding of disease progression and pave the way for personalized therapies, given the complexity of disease heterogeneity. Large-scale transcriptomic, proteomic, and imaging datasets create opportunities for discovering subtypes but also pose challenges due to their high dimensionality. To mitigate this, many feature selection methods focus on selecting features that distinguish known diseases or cell states, yet often miss features that preserve heterogeneity and reveal new subtypes. To overcome this gap, we develop Preserving Heterogeneity (PHet), a statistical methodology that employs iterative subsampling and differential analysis of interquartile range, in conjunction with Fisher's method, to identify a small set of features that enhance subtype clustering quality. Here, we show that this method can maintain sample heterogeneity while distinguishing known disease/cell states, with a tendency to outperform previous differential expression and outlier-based methods, indicating its potential to advance our understanding of disease mechanisms and cell differentiation.
Collapse
Affiliation(s)
- Abdur Rahman M A Basher
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Caleb Hallinan
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA
| | - Kwonmoo Lee
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Lu D, Zheng Y, Yi X, Hao J, Zeng X, Han L, Li Z, Jiao S, Jiang B, Ai J, Peng J. Identifying potential risk genes for clear cell renal cell carcinoma with deep reinforcement learning. Nat Commun 2025; 16:3591. [PMID: 40234405 PMCID: PMC12000451 DOI: 10.1038/s41467-025-58439-5] [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: 05/13/2024] [Accepted: 03/18/2025] [Indexed: 04/17/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent type of renal cell carcinoma. However, our understanding of ccRCC risk genes remains limited. This gap in knowledge poses challenges to the effective diagnosis and treatment of ccRCC. To address this problem, we propose a deep reinforcement learning-based computational approach named RL-GenRisk to identify ccRCC risk genes. Distinct from traditional supervised models, RL-GenRisk frames the identification of ccRCC risk genes as a Markov Decision Process, combining the graph convolutional network and Deep Q-Network for risk gene identification. Moreover, a well-designed data-driven reward is proposed for mitigating the limitation of scant known risk genes. The evaluation demonstrates that RL-GenRisk outperforms existing methods in ccRCC risk gene identification. Additionally, RL-GenRisk identifies eight potential ccRCC risk genes. We successfully validated epidermal growth factor receptor (EGFR) and piccolo presynaptic cytomatrix protein (PCLO), corroborated through independent datasets and biological experimentation. This approach may also be used for other diseases in the future.
Collapse
Affiliation(s)
- Dazhi Lu
- AI for Science Interdisciplinary Research Center, School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Yan Zheng
- College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Xianyanling Yi
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jianye Hao
- College of Intelligence and Computing, Tianjin University, Tianjin, China.
| | - Xi Zeng
- AI for Science Interdisciplinary Research Center, School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Lu Han
- AI for Science Interdisciplinary Research Center, School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Zhigang Li
- College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Shaoqing Jiao
- School of Software, Northwestern Polytechnical University, Xi'an, China
| | - Bei Jiang
- Tianjin Second People's Hospital, Tianjin, China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China.
| | - Jiajie Peng
- AI for Science Interdisciplinary Research Center, School of Computer Science, Northwestern Polytechnical University, Xi'an, China.
- Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an, China.
| |
Collapse
|
12
|
Huang CY, Huang WK, Yeh KY, Chang JWC, Lin YC, Chou WC. Integrating comprehensive genomic profiling in the management of oncology patients: applications and challenges in Taiwan. Biomed J 2025:100851. [PMID: 40185203 DOI: 10.1016/j.bj.2025.100851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025] Open
Abstract
Comprehensive genomic profiling (CGP) refers to the detailed genomic analysis of cancers for oncology patients. With the rapid development of next-generation sequencing (NGS) technologies, CGP has been widely applied to clinical practice and managing oncology patients. CGP can be performed on the tumor DNA and RNA, as well as non-tumor tissues (e.g., blood, pleural effusion, and ascites). In this article, we review the current evidence supporting the use of CGP in the management of oncology patients, both in real-world practice and the bridging to clinical trials. We also discuss the role of the molecular tumor board on the application of CGP in oncology patients. We provide an overview of the current scheme of CGP reimbursement in Taiwan and the precision oncology branch of the National Biobank Consortium of Taiwan. Finally, we discuss about the potential barriers and challenges of applying CGP in managing oncology patients and the future perspectives of CGP in precision oncology.
Collapse
Affiliation(s)
- Chen-Yang Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan
| | - Wen-Kuan Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kun-Yun Yeh
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Division of Hematology-Oncology, Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - John Wen-Cheng Chang
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan
| | - Yung-Chang Lin
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan
| | - Wen-Chi Chou
- Division of Hematology-Oncology, Department of Internal Medicine, Linkou Chang Gung Memorial Hospital and Chang Gung University, Taoyuan 333, Taiwan.
| |
Collapse
|
13
|
Wu X, Peng L, Zheng M, Mao Y, Li H, Sun S. The Role of TRPA1 as a Prognostic Marker in Colon Adenocarcinoma and Its Correlation with Mutations and Immunity. IRANIAN JOURNAL OF PUBLIC HEALTH 2025; 54:762-774. [PMID: 40321910 PMCID: PMC12045875 DOI: 10.18502/ijph.v54i4.18414] [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: 03/10/2024] [Accepted: 05/18/2024] [Indexed: 05/08/2025]
Abstract
Background This study aimed to investigate the prognostic value of TRP ion channel genes (TRPICGs) in colorectal adenocarcinoma (COAD) and explore its related mechanisms. Methods The COAD dataset was downloaded from the Cancer Genome Atlas (TCGA) database. The differential expression genes (DEGs) were screened between COAD and normal samples. The differentially expressed TRPICGs (DE-TRPICGs) were obtained via intersection of DEGs and 28 TRPICGs. The Kaplan-Meier (K-M) survival curve was used to screen DE-TRPICGs with survival differences as prognostic markers. Afterward, the correlation of prognostic marker with clinical, immune cell, copy number variation were explored. Finally, immunohistochemistry (IHC) was used to verify the expression of prognostic marker. Results Overall, 6003 DEGs were screened, and 6 DE-TRPICGs were obtained. Only TRPA1 was identified as prognostic biomarker. Survival and clinical correlation analyses implied that TRPA1 played an inhibitory role in colon adenocarcinoma pathogenesis and progression. Gene Set Enrichment Analysis (GSEA) indicated that TRPA1 was associated with cell cycle and immune-related pathways. Immune infiltration analysis showed that TRPA1 expression was significantly correlated with the infiltration of B cells, CD4+T cells, CD8+T cells, neutrophils and dendritic cells. Eventually, TRPA1 expression was down-regulated at the protein level in COAD samples, which presented consistent results with expression in the database. Conclusion TRPA1 was identified in COAD as a prognostic marker associated with TRP ion channels, which provided a powerful reference value and a new direction for the diagnosis and treatment of COAD.
Collapse
Affiliation(s)
- Xingxing Wu
- Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Lifang Peng
- Jiading District Central Hospital Affiliated Shanghai University of Medicine & Health Sciences, Shanghai, China
| | - Mingxu Zheng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuqing Mao
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Heng Li
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Shaopeng Sun
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
14
|
Lai Y, Xie B, Zhang W, He W. Pure drug nanomedicines - where we are? Chin J Nat Med 2025; 23:385-409. [PMID: 40274343 DOI: 10.1016/s1875-5364(25)60851-x] [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: 08/11/2024] [Revised: 10/26/2024] [Accepted: 11/03/2024] [Indexed: 04/26/2025]
Abstract
Pure drug nanomedicines (PDNs) encompass active pharmaceutical ingredients (APIs), including macromolecules, biological compounds, and functional components. They overcome research barriers and conversion thresholds associated with nanocarriers, offering advantages such as high drug loading capacity, synergistic treatment effects, and environmentally friendly production methods. This review provides a comprehensive overview of the latest advancements in PDNs, focusing on their essential components, design theories, and manufacturing techniques. The physicochemical properties and in vivo behaviors of PDNs are thoroughly analyzed to gain an in-depth understanding of their systematic characteristics. The review introduces currently approved PDN products and further explores the opportunities and challenges in expanding their depth and breadth of application. Drug nanocrystals, drug-drug cocrystals (DDCs), antibody-drug conjugates (ADCs), and nanobodies represent the successful commercialization and widespread utilization of PDNs across various disease domains. Self-assembled pure drug nanoparticles (SAPDNPs), a next-generation product, still require extensive translational research. Challenges persist in transitioning from laboratory-scale production to mass manufacturing and overcoming the conversion threshold from laboratory findings to clinical applications.
Collapse
Affiliation(s)
- Yaoyao Lai
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Bing Xie
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Wanting Zhang
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China.
| |
Collapse
|
15
|
Salcedo A, Tarabichi M, Buchanan A, Espiritu SMG, Zhang H, Zhu K, Ou Yang TH, Leshchiner I, Anastassiou D, Guan Y, Jang GH, Mootor MFE, Haase K, Deshwar AG, Zou W, Umar I, Dentro S, Wintersinger JA, Chiotti K, Demeulemeester J, Jolly C, Sycza L, Ko M, Wedge DC, Morris QD, Ellrott K, Van Loo P, Boutros PC. Crowd-sourced benchmarking of single-sample tumor subclonal reconstruction. Nat Biotechnol 2025; 43:581-592. [PMID: 38862616 PMCID: PMC11994449 DOI: 10.1038/s41587-024-02250-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: 04/11/2022] [Accepted: 04/17/2024] [Indexed: 06/13/2024]
Abstract
Subclonal reconstruction algorithms use bulk DNA sequencing data to quantify parameters of tumor evolution, allowing an assessment of how cancers initiate, progress and respond to selective pressures. We launched the ICGC-TCGA (International Cancer Genome Consortium-The Cancer Genome Atlas) DREAM Somatic Mutation Calling Tumor Heterogeneity and Evolution Challenge to benchmark existing subclonal reconstruction algorithms. This 7-year community effort used cloud computing to benchmark 31 subclonal reconstruction algorithms on 51 simulated tumors. Algorithms were scored on seven independent tasks, leading to 12,061 total runs. Algorithm choice influenced performance substantially more than tumor features but purity-adjusted read depth, copy-number state and read mappability were associated with the performance of most algorithms on most tasks. No single algorithm was a top performer for all seven tasks and existing ensemble strategies were unable to outperform the best individual methods, highlighting a key research need. All containerized methods, evaluation code and datasets are available to support further assessment of the determinants of subclonal reconstruction accuracy and development of improved methods to understand tumor evolution.
Collapse
Affiliation(s)
- Adriana Salcedo
- Department of Human Genetics, University of California, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA.
- Institute for Precision Health, University of California, Los Angeles, CA, USA.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada.
| | - Maxime Tarabichi
- The Francis Crick Institute, London, UK.
- Wellcome Sanger Institute, Hinxton, UK.
- Institute for Interdisciplinary Research, Université Libre de Bruxelles, Brussels, Belgium.
| | - Alex Buchanan
- Oregon Health and Sciences University, Portland, OR, USA
| | | | - Hongjiu Zhang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Kaiyi Zhu
- Department of Systems Biology, Columbia University, New York, NY, USA
- Center for Cancer Systems Therapeutics, Columbia University, New York, NY, USA
- Department of Electrical Engineering, Columbia University, New York, NY, USA
| | - Tai-Hsien Ou Yang
- Department of Systems Biology, Columbia University, New York, NY, USA
- Center for Cancer Systems Therapeutics, Columbia University, New York, NY, USA
- Department of Electrical Engineering, Columbia University, New York, NY, USA
| | | | - Dimitris Anastassiou
- Department of Systems Biology, Columbia University, New York, NY, USA
- Center for Cancer Systems Therapeutics, Columbia University, New York, NY, USA
- Department of Electrical Engineering, Columbia University, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Yuanfang Guan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Electronic Engineering and Computer Science, University of Michigan, Ann Arbor, MI, USA
| | - Gun Ho Jang
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Mohammed F E Mootor
- Department of Human Genetics, University of California, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
- Institute for Precision Health, University of California, Los Angeles, CA, USA
| | | | - Amit G Deshwar
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - William Zou
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Imaad Umar
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Stefan Dentro
- The Francis Crick Institute, London, UK
- Wellcome Sanger Institute, Hinxton, UK
| | - Jeff A Wintersinger
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Kami Chiotti
- Oregon Health and Sciences University, Portland, OR, USA
| | - Jonas Demeulemeester
- The Francis Crick Institute, London, UK
- VIB Center for Cancer Biology, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | | | - Lesia Sycza
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Minjeong Ko
- Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - David C Wedge
- Big Data Institute, University of Oxford, Oxford, UK
- Manchester Cancer Research Center, University of Manchester, Manchester, UK
| | - Quaid D Morris
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
- Computational and Systems Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kyle Ellrott
- Oregon Health and Sciences University, Portland, OR, USA.
| | - Peter Van Loo
- The Francis Crick Institute, London, UK.
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Paul C Boutros
- Department of Human Genetics, University of California, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA.
- Institute for Precision Health, University of California, Los Angeles, CA, USA.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada.
- Department of Urology, University of California, Los Angeles, CA, USA.
- Broad Stem Cell Research Center, University of California, Los Angeles, CA, USA.
- California NanoSystems Institute, University of California, Los Angeles, CA, USA.
| |
Collapse
|
16
|
Nourbakhsh M, Zheng Y, Noor H, Chen H, Akhuli S, Tiberti M, Gevaert O, Papaleo E. Revealing cancer driver genes through integrative transcriptomic and epigenomic analyses with Moonlight. PLoS Comput Biol 2025; 21:e1012999. [PMID: 40258059 PMCID: PMC12058160 DOI: 10.1371/journal.pcbi.1012999] [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: 11/13/2024] [Revised: 05/07/2025] [Accepted: 03/26/2025] [Indexed: 04/23/2025] Open
Abstract
Cancer involves dynamic changes caused by (epi)genetic alterations such as mutations or abnormal DNA methylation patterns which occur in cancer driver genes. These driver genes are divided into oncogenes and tumor suppressors depending on their function and mechanism of action. Discovering driver genes in different cancer (sub)types is important not only for increasing current understanding of carcinogenesis but also from prognostic and therapeutic perspectives. We have previously developed a framework called Moonlight which uses a systems biology multi-omics approach for prediction of driver genes. Here, we present an important development in Moonlight2 by incorporating a DNA methylation layer which provides epigenetic evidence for deregulated expression profiles of driver genes. To this end, we present a novel functionality called Gene Methylation Analysis (GMA) which investigates abnormal DNA methylation patterns to predict driver genes. This is achieved by integrating the tool EpiMix which is designed to detect such aberrant DNA methylation patterns in a cohort of patients and further couples these patterns with gene expression changes. To showcase GMA, we applied it to three cancer (sub)types (basal-like breast cancer, lung adenocarcinoma, and thyroid carcinoma) where we discovered 33, 190, and 263 epigenetically driven genes, respectively. A subset of these driver genes had prognostic effects with expression levels significantly affecting survival of the patients. Moreover, a subset of the driver genes demonstrated therapeutic potential as drug targets. This study provides a framework for exploring the driving forces behind cancer and provides novel insights into the landscape of three cancer sub(types) by integrating gene expression and methylation data.
Collapse
Affiliation(s)
- Mona Nourbakhsh
- Cancer Systems Biology, Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
- Cancer Structural Biology, Danish Cancer Institute, Copenhagen, Denmark
| | - Yuanning Zheng
- Department of Biomedical Data Science, Stanford Center for Biomedical Informatics Research, Palo Alto, California, United States of America
| | - Humaira Noor
- Department of Biomedical Data Science, Stanford Center for Biomedical Informatics Research, Palo Alto, California, United States of America
| | - Hongjin Chen
- Cancer Systems Biology, Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Subhayan Akhuli
- Cancer Systems Biology, Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
| | - Matteo Tiberti
- Cancer Structural Biology, Danish Cancer Institute, Copenhagen, Denmark
| | - Olivier Gevaert
- Department of Biomedical Data Science, Stanford Center for Biomedical Informatics Research, Palo Alto, California, United States of America
| | - Elena Papaleo
- Cancer Systems Biology, Section for Bioinformatics, Department of Health Technology, Technical University of Denmark, Lyngby, Denmark
- Cancer Structural Biology, Danish Cancer Institute, Copenhagen, Denmark
| |
Collapse
|
17
|
Koch Z, Li A, Evans DS, Cummings S, Ideker T. Somatic mutation as an explanation for epigenetic aging. NATURE AGING 2025; 5:709-719. [PMID: 39806003 DOI: 10.1038/s43587-024-00794-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 12/12/2024] [Indexed: 01/16/2025]
Abstract
DNA methylation marks have recently been used to build models known as epigenetic clocks, which predict calendar age. As methylation of cytosine promotes C-to-T mutations, we hypothesized that the methylation changes observed with age should reflect the accrual of somatic mutations, and the two should yield analogous aging estimates. In an analysis of multimodal data from 9,331 human individuals, we found that CpG mutations indeed coincide with changes in methylation, not only at the mutated site but with pervasive remodeling of the methylome out to ±10 kilobases. This one-to-many mapping allows mutation-based predictions of age that agree with epigenetic clocks, including which individuals are aging more rapidly or slowly than expected. Moreover, genomic loci where mutations accumulate with age also tend to have methylation patterns that are especially predictive of age. These results suggest a close coupling between the accumulation of sporadic somatic mutations and the widespread changes in methylation observed over the course of life.
Collapse
Affiliation(s)
- Zane Koch
- Program in Bioinformatics and Systems Biology, University of California, San Diego, La Jolla, CA, USA
| | - Adam Li
- Program in Bioinformatics and Systems Biology, University of California, San Diego, La Jolla, CA, USA
| | - Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Steven Cummings
- California Pacific Medical Center Research Institute, San Francisco, CA, USA.
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA.
| | - Trey Ideker
- Program in Bioinformatics and Systems Biology, University of California, San Diego, La Jolla, CA, USA.
- Department of Medicine, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
18
|
Hu M, Zheng L, Li A, Li X, Liang W, Zhu Y, Wang A, He L, Liu X, Sun Q. Discovery of 3-indolylbenzoquinone derivatives with therapeutic potential for breast cancer. Bioorg Med Chem 2025; 120:118094. [PMID: 39933277 DOI: 10.1016/j.bmc.2025.118094] [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: 11/23/2024] [Revised: 01/25/2025] [Accepted: 01/29/2025] [Indexed: 02/13/2025]
Abstract
Breast cancer is one of the most prevalent malignant tumors in women, but the side effects and drug resistance limit the long-term effectiveness of existing drugs. To address these issues, we designed and synthesized a series of novel mono- and bis-indole-substituted 3-indolylbenzoquinone derivatives and evaluated their inhibitory activity against breast cancer. Among them, compound 1b demonstrated the most potent inhibitory activity against the MDA-MB-231 breast cancer cell line (IC50 = 3.2 µM) as well as the drug-resistant variant, MCF-7/ADR (IC50 = 8.36 µM). It demonstrated minimal toxicity and superior tumor suppression in a Balb/c mouse model of 4 T1 breast cancer. Mechanistically, compound 1b induced apoptosis and cell cycle arrest at the G2/M phase. Through computational study and CESTA assay, we implicated phosphoinositide 3-kinase α (PI3Kα) as a potential target. Thus, we present compound 1b as a lead candidate for the development of novel, safe, and effective small-molecule therapies against breast cancer.
Collapse
Affiliation(s)
- Mingli Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lang Zheng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Ailing Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xiao Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wengxue Liang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yuanhao Zhu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Aoxue Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China
| | - Ling He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xiuxiu Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Department of Medicinal Chemistry, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Qiu Sun
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
19
|
Su X, Lin Q, Liu B, Zhou C, Lu L, Lin Z, Si J, Ding Y, Duan S. The promising role of nanopore sequencing in cancer diagnostics and treatment. CELL INSIGHT 2025; 4:100229. [PMID: 39995512 PMCID: PMC11849079 DOI: 10.1016/j.cellin.2025.100229] [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: 09/14/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 02/26/2025]
Abstract
Cancer arises from genetic alterations that impact both the genome and transcriptome. The utilization of nanopore sequencing offers a powerful means of detecting these alterations due to its unique capacity for long single-molecule sequencing. In the context of DNA analysis, nanopore sequencing excels in identifying structural variations (SVs), copy number variations (CNVs), gene fusions within SVs, and mutations in specific genes, including those involving DNA modifications and DNA adducts. In the field of RNA research, nanopore sequencing proves invaluable in discerning differentially expressed transcripts, uncovering novel elements linked to transcriptional regulation, and identifying alternative splicing events and RNA modifications at the single-molecule level. Furthermore, nanopore sequencing extends its reach to detecting microorganisms, encompassing bacteria and viruses, that are intricately associated with tumorigenesis and the development of cancer. Consequently, the application prospects of nanopore sequencing in tumor diagnosis and personalized treatment are expansive, encompassing tasks such as tumor identification and classification, the tailoring of treatment strategies, and the screening of prospective patients. In essence, this technology stands poised to unearth novel mechanisms underlying tumorigenesis while providing dependable support for the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Xinming Su
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Qingyuan Lin
- The Second Clinical Medical College, Zhejiang Chinese Medicine University BinJiang College, Hangzhou 310053, Zhejiang, China
| | - Bin Liu
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Chuntao Zhou
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Liuyi Lu
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Zihao Lin
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Jiahua Si
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Yuemin Ding
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
- Institute of Translational Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Shiwei Duan
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
- Institute of Translational Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| |
Collapse
|
20
|
Bahari F, Ahangari Cohan R, Montazeri H. Element-specific estimation of background mutation rates in whole cancer genomes through transfer learning. NPJ Precis Oncol 2025; 9:92. [PMID: 40155429 PMCID: PMC11953285 DOI: 10.1038/s41698-025-00871-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
Mutational burden tests are essential for detecting signals of positive selection in cancer driver discovery by comparing observed mutation rates with background mutation rates (BMRs). However, accurate BMR estimation is challenging due to the diversity of mutational processes across genomes, complicating driver discovery efforts. Existing methods rely on various genomic regions and features for BMR estimation but lack a model that integrates both intergenic intervals and functional genomic elements on a comprehensive set of genomic features. Here, we introduce eMET (element-specific Mutation Estimator with boosted Trees), which employs 1372 (epi)genomic features from intergenic data and fine-tunes it with element-specific data through transfer learning. Applied to PCAWG somatic mutations, eMET significantly improves BMR accuracy and has potential to enhance driver discovery. Additionally, we provide an extensive analysis of BMR estimation, examining different machine learning models, genomic interval strategies, feature categories, and dimensionality reduction techniques.
Collapse
Affiliation(s)
- Farideh Bahari
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran.
| | - Hesam Montazeri
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
21
|
Zhu Z, Shen J, Ho PCL, Hu Y, Ma Z, Wang L. Transforming cancer treatment: integrating patient-derived organoids and CRISPR screening for precision medicine. Front Pharmacol 2025; 16:1563198. [PMID: 40201690 PMCID: PMC11975957 DOI: 10.3389/fphar.2025.1563198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
The persistently high mortality rates associated with cancer underscore the imperative need for innovative, efficacious, and safer therapeutic agents, as well as a more nuanced understanding of tumor biology. Patient-derived organoids (PDOs) have emerged as innovative preclinical models with significant translational potential, capable of accurately recapitulating the structural, functional, and heterogeneous characteristics of primary tumors. When integrated with cutting-edge genomic tools such as CRISPR, PDOs provide a powerful platform for identifying cancer driver genes and novel therapeutic targets. This comprehensive review delves into recent advancements in CRISPR-mediated functional screens leveraging PDOs across diverse cancer types, highlighting their pivotal role in high-throughput functional genomics and tumor microenvironment (TME) modeling. Furthermore, this review highlights the synergistic potential of integrating PDOs with CRISPR screens in cancer immunotherapy, focusing on uncovering immune evasion mechanisms and improving the efficacy of immunotherapeutic approaches. Together, these cutting-edge technologies offer significant promise for advancing precision oncology.
Collapse
Affiliation(s)
- Ziyi Zhu
- The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, Hubei, China
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Jiayang Shen
- The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, Hubei, China
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Paul Chi-Lui Ho
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
| | - Ya Hu
- The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, Hubei, China
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Zhaowu Ma
- The First Affiliated Hospital of Yangtze University, Yangtze University, Jingzhou, Hubei, China
- School of Basic Medicine, Yangtze University, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- NUS Centre for Cancer Research (N2CR), National University of Singapore, Singapore, Singapore
| |
Collapse
|
22
|
Zarrella S, Miranda MR, Covelli V, Restivo I, Novi S, Pepe G, Tesoriere L, Rodriquez M, Bertamino A, Campiglia P, Tecce MF, Vestuto V. Endoplasmic Reticulum Stress and Its Role in Metabolic Reprogramming of Cancer. Metabolites 2025; 15:221. [PMID: 40278350 PMCID: PMC12029571 DOI: 10.3390/metabo15040221] [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: 02/17/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Endoplasmic reticulum (ER) stress occurs when ER homeostasis is disrupted, leading to the accumulation of misfolded or unfolded proteins. This condition activates the unfolded protein response (UPR), which aims to restore balance or trigger cell death if homeostasis cannot be achieved. In cancer, ER stress plays a key role due to the heightened metabolic demands of tumor cells. This review explores how metabolomics can provide insights into ER stress-related metabolic alterations and their implications for cancer therapy. Methods: A comprehensive literature review was conducted to analyze recent findings on ER stress, metabolomics, and cancer metabolism. Studies examining metabolic profiling of cancer cells under ER stress conditions were selected, with a focus on identifying potential biomarkers and therapeutic targets. Results: Metabolomic studies highlight significant shifts in lipid metabolism, protein synthesis, and oxidative stress management in response to ER stress. These metabolic alterations are crucial for tumor adaptation and survival. Additionally, targeting ER stress-related metabolic pathways has shown potential in preclinical models, suggesting new therapeutic strategies. Conclusions: Understanding the metabolic impact of ER stress in cancer provides valuable opportunities for drug development. Metabolomics-based approaches may help identify novel biomarkers and therapeutic targets, enhancing the effectiveness of antitumor therapies.
Collapse
Affiliation(s)
- Salvatore Zarrella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Maria Rosaria Miranda
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Verdiana Covelli
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (V.C.); (M.R.)
| | - Ignazio Restivo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (L.T.)
| | - Sara Novi
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
- NBFC, National Biodiversity Future Center, 90133 Palermo, Italy
| | - Luisa Tesoriere
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Via Archirafi 28, 90123 Palermo, Italy; (I.R.); (L.T.)
| | - Manuela Rodriquez
- Department of Pharmacy, University of Naples Federico II, Via Domenico Montesano, 49, 80131 Napoli, Italy; (V.C.); (M.R.)
| | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Mario Felice Tecce
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, Italy; (S.Z.); (M.R.M.); (S.N.); (G.P.); (A.B.); (P.C.); (M.F.T.)
| |
Collapse
|
23
|
Challagundla KB, Pathania AS, Chava H, Kantem NM, Dronadula VM, Coulter DW, Clarke M. FOXJ3, a novel tumor suppressor in neuroblastoma. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200914. [PMID: 39811681 PMCID: PMC11731479 DOI: 10.1016/j.omton.2024.200914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/20/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025]
Abstract
Neuroblastoma (NB) poses a significant challenge in pediatric cancer care due to its aggressive nature and poor prognosis. While advances have been made in clinical treatments, therapy resistance remains a tough hurdle in NB treatment. While much research has focused on identifying oncogenes in NB, there has been less emphasis on understanding tumor suppressors. This study aimed to discover a new transcription factor that could address patient stage, risk level, and MYCN amplification status while exhibiting tumor-suppressive properties in NB patients. Using advanced bioinformatics techniques, we identified unique transcription factor signature that corresponded to patient characteristics. By analyzing regulon specificity scores, we prioritized Forkhead Box J3 (FOXJ3) as a potential novel driver transcription factor with tumor-suppressive functions in NB. Validation experiments on NB patients and patient-derived xenograft (PDX) tumors confirmed higher FOXJ3 expression in low-risk versus high-risk patients and in PDXs from diagnostic tumors versus relapse-specific tumors. Notably, the overexpression of FOXJ3 was associated with reduced cell density, proliferation, cells in S phase, colony-formation ability, transwell migration, neurosphere formation, spheroid diameter, and inhibition of AKT signaling in NB cells. Overall, these findings suggest that FOXJ3 functions as a novel tumor suppressor in NB, holding promise for potential therapeutic interventions.
Collapse
Affiliation(s)
- Kishore B. Challagundla
- School of Interdisciplinary Informatics, University of Nebraska Omaha, 1110 South 67th Street, Omaha, NE 68182, USA
- The Child Health Research Institute, Department of Biochemistry and Molecular Biology & Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Basic Biomedical Sciences, Touro College of Osteopathic Medicine, Middletown, NY 10940, USA
| | - Anup S. Pathania
- The Child Health Research Institute, Department of Biochemistry and Molecular Biology & Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Haritha Chava
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Naveenkumar M. Kantem
- Department of Mathematical and Statistical Sciences, University of Nebraska Omaha, 1110 South 67th Street, Omaha, NE 68182, USA
| | - Veena M. Dronadula
- School of Interdisciplinary Informatics, University of Nebraska Omaha, 1110 South 67th Street, Omaha, NE 68182, USA
| | - Don W. Coulter
- Department of Pediatrics, Division of Hematology/Oncology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Martina Clarke
- School of Interdisciplinary Informatics, University of Nebraska Omaha, 1110 South 67th Street, Omaha, NE 68182, USA
- Department of Biomedical Informatics, University of Nebraska Medical Center, 42nd and Emile Street, Omaha, NE 68198, USA
| |
Collapse
|
24
|
Boga S, Bouzada D, Lopez-Blanco R, Sarmiento A, Salvadó I, Alvar Gil D, Brea J, Loza MI, Barreiro-Piñeiro N, Martínez-Costas J, Mena S, Guirado G, Santoro A, Faller P, Vázquez ME, Vázquez López M. Copper(II) Cyclopeptides with High ROS-Mediated Cytotoxicity. Bioconjug Chem 2025; 36:500-509. [PMID: 40059798 DOI: 10.1021/acs.bioconjchem.4c00561] [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: 03/20/2025]
Abstract
Cu(II) coordination complexes are emerging as promising anticancer agents due to their ability to induce oxidative stress through reactive oxygen species (ROS) generation. In this study, we synthesized and characterized two novel Cu(II) metallopeptide systems, 1/Cu(II) and 2/Cu(II), derived from the oligocationic bipyridyl cyclopeptides 1 and 2, and designed to enhance the transport of Cu(II) into cells and increase ROS levels. Spectroscopic and electrochemical analyses confirmed the formation of stable metallopeptide species in aqueous media. Inductively coupled plasma mass spectrometry (ICP-MS) studies demonstrated that both metallopeptides significantly increase intracellular Cu(II) accumulation in NCI/ADR-RES cancer cells, highlighting their role as efficient Cu(II) transporters. Additionally, ROS generation assays revealed that 1/Cu(II) induces a substantial increase in intracellular ROS levels, supporting the hypothesis of oxidative stress-induced cytotoxicity. Cell-viability assays further confirmed that both 1/Cu(II) and 2/Cu(II) exhibit strong anticancer activity in a number of cancer cell lines, with IC50 values significantly lower than those of their free cyclopeptide counterparts or Cu(II) alone, showing an order of activity higher than that of cisplatin. Finally, molecular modeling studies provided further insights into the structural stability and coordination environment of Cu(II) within the metallopeptide complexes. These findings suggest that these Cu(II) cyclometallopeptide systems hold potential as novel metal-based therapeutic agents, leveraging Cu(II) transport and ROS increase as key strategies for cancer treatment.
Collapse
Affiliation(s)
- Sonia Boga
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - David Bouzada
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Roi Lopez-Blanco
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Axel Sarmiento
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Iria Salvadó
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - David Alvar Gil
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - José Brea
- Innopharma Drug Screening and Pharmacogenomics Platform. Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - María Isabel Loza
- Innopharma Drug Screening and Pharmacogenomics Platform. Center for Research in Molecular Medicine and Chronic Diseases (CiMUS). Department of Pharmacology, Pharmacy and Pharmaceutical Technology, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Health Research Institute of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Natalia Barreiro-Piñeiro
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Bioquímica e Bioloxía Molecular, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - José Martínez-Costas
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Bioquímica e Bioloxía Molecular, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Silvia Mena
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Gonzalo Guirado
- Departament de Química, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Alice Santoro
- Institut de Chimie (UMR 7177), University of Strasbourg─CNRS, 67081 Strasbourg, France
| | - Peter Faller
- Institut de Chimie (UMR 7177), University of Strasbourg─CNRS, 67081 Strasbourg, France
- Institut Universitaire de France (IUF), 75231 Paris, France
| | - M Eugenio Vázquez
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Orgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Miguel Vázquez López
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Química Inorgánica, Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
25
|
Zhou Y, Tao Q, Luo C, Chen J, Chen G, Sun J. Epacadostat Overcomes Cetuximab Resistance in Colorectal Cancer by Targeting IDO-Mediated Tryptophan Metabolism. Cancer Sci 2025. [PMID: 40103010 DOI: 10.1111/cas.70057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025] Open
Abstract
Primary or acquired mutations in RAS/RAF genes resulting in cetuximab resistance have limited its clinical application in colorectal cancer (CRC) patients. The mechanism of this resistance remains unclear. RNA sequencing from cetuximab-sensitive and -resistant specimens revealed an activation of the tryptophan pathway and elevation of IDO1 and IDO2 in cetuximab-resistant CRC patients. In vitro, in vivo, and clinical specimens confirmed the upregulation of IDO1and IDO2 and the Kyn/Trp after cetuximab treatment. Additionally, the IDO inhibitor, epacadostat, could effectively inhibit the migration and proliferation of cetuximab-resistant CRC cells while promoting apoptosis. Compared to epacadostat monotherapy, the combination of cetuximab and epacadostat showed a stronger synergistic anti-tumor effect. Furthermore, in vivo experiments confirmed that combination therapy effectively suppressed tumor growth. Mechanistically, KEGG pathway analysis revealed the activation of the IFN-γ pathway in cetuximab-resistant CRC tissues. Luciferase reporter assays confirmed the transcriptional activity of IDO1 following cetuximab treatment. Silencing IFN-γ then suppressed the upregulation induced by cetuximab. Moreover, we observed that the combination reduced the concentration of the tryptophan metabolite kynurenine, promoted the infiltration of CD8+ T lymphocytes, and enhanced the polarization of M1 macrophages within the tumor microenvironment, thereby exerting potent anti-tumor immune effects. Overall, our results confirm the remarkable therapeutic efficacy of combining cetuximab with epacadostat in cetuximab-resistant CRC. Our findings may provide a novel target for overcoming cetuximab resistance in CRC.
Collapse
Affiliation(s)
- Yimin Zhou
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiongyan Tao
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chubin Luo
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Jinsong Chen
- Department of Clinical Medicine, Shaoguan University, Shaoguan, Guangdong, China
| | - Genwen Chen
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianyong Sun
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Xu Q, Cheng X, Li Q, Yu P, Zhou X, Chen Y, Lin L, Ni T, Zhao Z. 3' untranslated region somatic variants connect alternative polyadenylation dysregulation in human cancers. J Genet Genomics 2025:S1673-8527(25)00079-7. [PMID: 40107412 DOI: 10.1016/j.jgg.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Somatic variants in the cancer genome influence gene expression through diverse mechanisms depending on their specific locations. However, a systematic evaluation of the effects of somatic variants located in 3' untranslated regions (3' UTRs) on alternative polyadenylation (APA) of mRNA remains lacking. In this study, we analyze 10,199 tumor samples across 32 cancer types and identify 1,333 somatic single nucleotide variants (SNVs) associated with abnormal 3' UTR APA. Mechanistically, these 3' UTR SNVs can alter cis-regulatory elements, such as the poly(A) signal and UGUA motif, leading to changes in APA. Minigene assays confirm that 3' UTR SNVs in multiple genes, including RPS23 and CHTOP, induce aberrant APA. Among affected genes, 62 exhibit differential stability between tandem 3' UTR isoforms, including HSPA4 and UCK2, validated by experimental assays. Finally, we establish that SNV-related abnormal APA usage serves as an additional layer of expression regulation for tumor-suppressor gene HMGN2 in breast cancer. Collectively, this study reveals 3' UTR APA as a critical mechanism mediating the functional impact of somatic noncoding variants in human cancers.
Collapse
Affiliation(s)
- Qiushi Xu
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China; Center for Reproductive Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xiaomeng Cheng
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Qianru Li
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Peng Yu
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xiaolan Zhou
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yu Chen
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Limin Lin
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ting Ni
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China; State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot 010070, China.
| | - Zhaozhao Zhao
- State Key Laboratory of Genetic Engineering, National Clinical Research Center for Aging and Medicine, Huashan Hospital, Collaborative Innovation Center of Genetics and Development, Human Phenome Institute, Center for Evolutionary Biology, Shanghai Engineering Research Center of Industrial Microorganisms, School of Life Sciences, Fudan University, Shanghai 200438, China; MOE Key Laboratory of Contemporary Anthropology, School of Life Sciences, Fudan University, Shanghai 200438, China.
| |
Collapse
|
27
|
Tian L, Wang Y, Guan J, Zhang L, Fan J. The Prognostic Value and Immunomodulatory Role of Spsb2, a Novel Immune Checkpoint Molecule, in Hepatocellular Carcinoma. Genes (Basel) 2025; 16:346. [PMID: 40149497 PMCID: PMC11941779 DOI: 10.3390/genes16030346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Liver cancer, specifically hepatocellular carcinoma (LIHC), ranks as the second most common cause of cancer-related fatalities globally. Moreover, the occurrence rate of LIHC is steadily increasing. A recently identified gene, SPSB2, has been implicated in cell signaling, impacting the development and progression of non-small cell lung cancer. Nevertheless, studies on the role of SPSB2 in the pathogenesis of LIHC are lacking. METHODS Using the TCGA, GTEx, and GEO databases, we obtained differentially expressed genes that affect the prognosis of patients with LIHC. We utilized the Kruskal-Wallis test, along with univariate and multivariate COX regression analyses, to determine the correlation between SPSB2 and patient clinical indicators. Potential biological functions of SPSB2 in LIHC were explored by enrichment analysis, ssGSEA, and Spearman correlation analysis. Finally, LIHC cell lines Huh7 and SMMC-7721 were used to validate the biological function of SPSB2. RESULTS The results showed LIHC patients with higher SPSB2 expression had a poorer prognosis, and SPSB2 expression was significantly correlated with LIHC patients' Histologic grade, Pathologic T stage, Prothrombin time, Pathologic stage, BMI, weight, adjacent hepatic tissue inflammation, AFP level, and OS event (p < 0.05). SPSB2 shows notable enrichment in pathways linked to tumorigenesis and the immune system. Moreover, its expression is strongly connected to immune cells and immune checkpoints. Knockdown of SPSB2 expression in Huh7 cells and SMMC-7721 cells inhibits SPSB2's biological functions, including proliferation, invasion, metastasis, and other phenotypes. CONCLUSIONS SPSB2 plays a crucial role in the development of LIHC. It is related to the immune response and unfavorable outcomes. SPSB2 may function as a clinical biomarker for prognosis.
Collapse
Affiliation(s)
- Lv Tian
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Yiming Wang
- School of Nursing, Jilin University, Changchun 130021, China
| | - Jiexin Guan
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Lu Zhang
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Jun Fan
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 611731, China
| |
Collapse
|
28
|
Arnet L, Emilius L, Hamann A, Carmo-Fonseca M, Berking C, Dörrie J, Schaft N. The Influence of Indisulam on Human Immune Effector Cells: Is a Combination with Immunotherapy Feasible? Pharmaceutics 2025; 17:368. [PMID: 40143032 PMCID: PMC11945250 DOI: 10.3390/pharmaceutics17030368] [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: 07/31/2024] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Background: As a modulator of pre-mRNA splicing, the anti-cancer agent indisulam can induce aberrantly spliced neoantigens, enabling immunologic anti-tumor activity. Consequently, combining indisulam with immunotherapy is expected to be a promising novel approach in cancer therapy. However, a prerequisite for such a combination is that immune effector cells remain functional and unharmed by the chemical. Methods: To ensure the immunocompetence of human immune effector cells is maintained, we investigated the influence of indisulam on ex vivo-isolated T cells and monocyte-derived dendritic cells (moDCs) from healthy donors. We used indisulam concentrations from 0.625 µM to 160 µM and examined the impact on the following: (i) the activation of CD4+ and CD8+ T cells by CD3-crosslinking and via a high-affinity TCR, (ii) the cytotoxicity of CD8+ T cells, (iii) the maturation process of moDCs, and (iv) antigen-specific CD8+ T cell priming. Results: We observed dose-dependent inhibitory effects of indisulam, and substantial inhibition occurred at concentrations around 10 µM, but the various functions of the immune system exhibited different sensitivities. The weaker activation of T cells via CD3-crosslinking was more sensitive than the stronger activation via the high-affinity TCR. T cells remained capable of killing tumor cells after treatment with indisulam up to 40 µM, but T cell cytotoxicity was impaired at 160 µM indisulam. While moDC maturation was also rather resistant, T cell priming was almost completely abolished at a concentration of 10 µM. Conclusions: These effects should be considered in possible future combinations of immunotherapy with the mRNA splicing inhibitor indisulam.
Collapse
Affiliation(s)
- Lisa Arnet
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Lisabeth Emilius
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Annett Hamann
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Maria Carmo-Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisbon, Portugal;
| | - Carola Berking
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität ErlangenNürnberg, 91054 Erlangen, Germany; (L.A.); (L.E.); (A.H.); (C.B.); (J.D.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| |
Collapse
|
29
|
Choi J, Kim J, Jung YW, Park JH, Lee JH. Neurotrophic Receptor Tyrosine Kinase 3 as a Prognostic Biomarker in Breast Cancer Using Bioinformatic Analysis. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:474. [PMID: 40142285 PMCID: PMC11943874 DOI: 10.3390/medicina61030474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025]
Abstract
Background and Objectives: Neurotrophic receptor tyrosine kinase 3 (NTRK3) is a member of the tropomyosin receptor kinase family of receptor tyrosine kinases, which play a crucial role in neural development. However, owing to the limited number of studies about NTRK3 and cancer, we aimed to investigate NTRK3 as a potential prognostic marker for breast cancer (BC). Materials and Methods: We conducted a comprehensive analysis of NTRK3 expression in BC using the Tumor Immune Estimation Resource, Gene Expression Profiling Interactive Analysis 2, and Kaplan-Meier Plotter databases. We also explored the association between NTRK3 expression and tumor-infiltrating immune cells. Results: Low NTRK3 expression showed poorer prognosis in BC, as well as with T stage, pathology, and the Luminal subtype. In BC (BRCA), NTRK3 was positively correlated with CD4+ T cell, CD8+ T cell, macrophage, and neutrophil infiltration. Conclusions: These results suggest that NTRK3 may serve as a prognostic biomarker and provide novel insights into tumor immunology in BC. Therefore, NTRK3 represents a potential diagnostic and therapeutic target for BC treatment.
Collapse
Affiliation(s)
- Jeongmin Choi
- Medical Course, School of Medicine, Keimyung University, 1095 Dalgubeol-daero, Daegu 42601, Republic of Korea;
| | - Jongwan Kim
- Department of Anatomy, College of Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Yong Wook Jung
- Department of Anatomy, College of Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Jong Ho Park
- Department of Anatomy, School of Medicine, Keimyung University, 1095 Dalgubeol-daero, Daegu 42601, Republic of Korea;
| | - Jae-Ho Lee
- Department of Anatomy, School of Medicine, Keimyung University, 1095 Dalgubeol-daero, Daegu 42601, Republic of Korea;
| |
Collapse
|
30
|
Basher ARMA, Hallinan C, Lee K. Heterogeneity-Preserving Discriminative Feature Selection for Disease-Specific Subtype Discovery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.05.14.540686. [PMID: 38187596 PMCID: PMC10769187 DOI: 10.1101/2023.05.14.540686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
The identification of disease-specific subtypes can provide valuable insights into disease progression and potential individualized therapies, important aspects of precision medicine given the complex nature of disease heterogeneity. The advent of high-throughput technologies has enabled the generation and analysis of various molecular data types, such as single-cell RNA-seq, proteomic, and imaging datasets, on a large scale. While these datasets offer opportunities for subtype discovery, they also pose challenges in finding subtype signatures due to their high dimensionality. Feature selection, a key step in the machine learning pipeline, involves selecting signatures that reduce feature size for more efficient downstream computational analysis. Although many existing methods focus on selecting features that differentiate known diseases or cell states, they often struggle to identify features that both preserve heterogeneity and reveal subtypes. To address this, we utilized deep metric learning-based feature embedding to explore the statistical properties of features crucial for preserving heterogeneity. Our analysis indicated that features with a notable difference in interquartile range (IQR) between classes hold important subtype information. Guided by this insight, we developed a statistical method called PHet (Preserving Heterogeneity), which employs iterative subsampling and differential analysis of IQR combined with Fisher's method to identify a small set of features that preserve heterogeneity and enhance subtype clustering quality. Validation on public single-cell RNA-seq and microarray datasets demonstrated PHet's ability to maintain sample heterogeneity while distinguishing known disease/cell states, with a tendency to outperform previous differential expression and outlier-based methods. Furthermore, an analysis of a single-cell RNA-seq dataset from mouse tracheal epithelial cells identified two distinct basal cell subtypes differentiating towards a luminal secretory phenotype using PHet-based features, demonstrating promising results in a real-data application. These results highlight PHet's potential to enhance our understanding of disease mechanisms and cell differentiation, contributing significantly to the field of personalized medicine.
Collapse
Affiliation(s)
- Abdur Rahman M. A. Basher
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| | - Caleb Hallinan
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Kwonmoo Lee
- Vascular Biology Program, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
31
|
Dong G, Ma CC, Mao S, Naik SM, Brown KSM, McDonough GA, Kim J, Kirkham SL, Cherry JD, Uretsky M, Spurlock E, McKee AC, Huang AY, Miller MB, Lee EA, Walsh CA. Diverse somatic genomic alterations in single neurons in chronic traumatic encephalopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.03.641217. [PMID: 40093089 PMCID: PMC11908173 DOI: 10.1101/2025.03.03.641217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Chronic traumatic encephalopathy (CTE) is a neurodegenerative disease that is linked to exposure to repetitive head impacts (RHI), yet little is known about its pathogenesis. Applying two single-cell whole-genome sequencing methods to hundreds of neurons from prefrontal cortex of 15 individuals with CTE, and 4 with RHI without CTE, revealed increased somatic single-nucleotide variants in CTE, resembling a pattern previously reported in Alzheimer's disease (AD). Furthermore, we discovered remarkably high burdens of somatic small insertions and deletions in a subset of CTE individuals, resembling a known pattern, ID4, also found in AD. Our results suggest that neurons in CTE experience stereotyped mutational processes shared with AD; the absence of similar changes in RHI neurons without CTE suggests that CTE involves mechanisms beyond RHI alone.
Collapse
Affiliation(s)
- Guanlan Dong
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
- Bioinformatics and Integrative Genomics Program, Harvard Medical School; Boston, MA, USA
| | - Chanthia C. Ma
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
- Harvard-MIT MD-PhD Program, Harvard Medical School; Boston, MA, USA
| | - Shulin Mao
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School; Boston, MA, USA
| | - Samuel M. Naik
- Division of Neuropathology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School; Boston, MA, USA
| | - Katherine Sun-Mi Brown
- Division of Neuropathology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School; Boston, MA, USA
| | - Gannon A. McDonough
- Division of Neuropathology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School; Boston, MA, USA
| | - Junho Kim
- Department of Biological Sciences, Sungkyunkwan University; Suwon, South Korea
| | - Samantha L. Kirkham
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
| | - Jonathan D. Cherry
- Veterans Affairs (VA) Boston Healthcare System, US Department of Veteran Affairs; Boston, MA, USA
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University; Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Chobanian and Avedisian School of Medicine, Boston University; Boston, MA, USA
| | - Madeline Uretsky
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University; Boston, MA, USA
| | - Elizabeth Spurlock
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University; Boston, MA, USA
| | - Ann C. McKee
- Veterans Affairs (VA) Boston Healthcare System, US Department of Veteran Affairs; Boston, MA, USA
- Alzheimer’s Disease Research Center and Chronic Traumatic Encephalopathy Center, Chobanian and Avedisian School of Medicine, Boston University; Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Chobanian and Avedisian School of Medicine, Boston University; Boston, MA, USA
- Department of Neurology, Chobanian and Avedisian School of Medicine, Boston University; Boston, MA, USA
| | - August Yue Huang
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | - Michael B. Miller
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
- Division of Neuropathology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School; Boston, MA, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | - Eunjung Alice Lee
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, USA
| | - Christopher A. Walsh
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Boston Children’s Hospital; Boston, MA, USA
- Department of Pediatrics, Harvard Medical School; Boston, MA, USA
- Broad Institute of MIT and Harvard; Cambridge, MA, USA
- Howard Hughes Medical Institute; Boston, MA, USA
| |
Collapse
|
32
|
Pei Y, Liang H, Guo Y, Wang B, Wu H, Jin Z, Lin S, Zeng F, Wu Y, Shi Q, Xu J, Huang Y, Ren T, Liu J, Guo W. Liquid-liquid phase separation drives immune signaling transduction in cancer: a bibliometric and visualized study from 1992 to 2024. Front Oncol 2025; 15:1509457. [PMID: 40104511 PMCID: PMC11913689 DOI: 10.3389/fonc.2025.1509457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/28/2025] [Indexed: 03/20/2025] Open
Abstract
Background Liquid-liquid phase separation (LLPS) is a novel concept that could explain how living cells precisely modulate internal spatial and temporal functions. However, a comprehensive bibliometric analysis on LLPS and immune signaling processes in cancer is still scarce. This study aims to perform a bibliometric assessment of research to explore the landscape of LLPS research in immune signaling pathways for cancer. Methods Utilizing the Web of Science Core Collection database and multiple analysis software, we performed quantitative and qualitative analyses of the study situation between LLPS and immune signaling in cancer from 1992 to 2024. Results The corresponding authors were primarily from China and the USA. The most relevant references were the "International Journal of Molecular Sciences", "Proteomics". The annual number of publications exhibited a fast upward tendency from 2020 to 2024. The most frequent key terms included expression, separation, activation, immunotherapy, and mechanisms. Qualitative evaluation emphasized the TCR, BCR, cGAS-STING, RIG-1, NF-κB signaling pathways associated with LLPS processes. Conclusion This research is the first to integratively map out the knowledge structure and forward direction in the area of immune transduction linked with LLPS over the past 30 years. In summary, although this research area is still in its infancy, illustrating the coordinated structures and communications between cancer and immune signaling with LLPS within a spatial framework will offer deeper insights into the molecular mechanisms of cancer development and further enhance the effectiveness of existing immunotherapies.
Collapse
Affiliation(s)
- Yanhong Pei
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Haijie Liang
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Yu Guo
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Boyang Wang
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Han Wu
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Zhijian Jin
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Shanyi Lin
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Fanwei Zeng
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Yifan Wu
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Qianyu Shi
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Jiuhui Xu
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Yi Huang
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Tingting Ren
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Jiarui Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Wei Guo
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| |
Collapse
|
33
|
Butler G, Baker J, Amend SR, Pienta KJ, Venditti C. No evidence for Peto's paradox in terrestrial vertebrates. Proc Natl Acad Sci U S A 2025; 122:e2422861122. [PMID: 39993196 PMCID: PMC11892590 DOI: 10.1073/pnas.2422861122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/13/2025] [Indexed: 02/26/2025] Open
Abstract
Larger, longer-lived species are expected to have a higher cancer prevalence compared to smaller, shorter-lived species owing to the greater number of cell divisions that occur during their lifespan. Yet, to date, no evidence has been found to support this expectation, and no association has been found between cancer prevalence and body size across species-a phenomenon known as Peto's paradox. Specifically, while anticancer mechanisms have been identified for individual species, wider phylogenetic evidence has remained elusive. Here, we show that there is no evidence for Peto's paradox across amphibians, birds, mammals, and squamate reptiles: Larger species do in fact have a higher cancer prevalence compared to smaller species. Moreover, we demonstrate that the accumulation of repeated instances of accelerated body size evolution in mammals and birds is associated with a reduction in the prevalence of neoplasia and malignancy, suggesting that increased rates of body size evolution are associated with the evolution of improved cellular growth control. These results represent empirical evidence showing that larger body size is related to higher cancer prevalence, thus rejecting Peto's paradox, and demonstrating the importance of heterogenous routes of body size evolution in shaping anticancer defenses.
Collapse
Affiliation(s)
- George Butler
- University College London Cancer Institute, University College London, LondonWC1E 6DD, United Kingdom
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Joanna Baker
- School of Biological Sciences, University of Reading, ReadingRG6 6AS, United Kingdom
| | - Sarah R. Amend
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Kenneth J. Pienta
- Cancer Ecology Center, The Brady Urological Institute, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Chris Venditti
- School of Biological Sciences, University of Reading, ReadingRG6 6AS, United Kingdom
| |
Collapse
|
34
|
Sturgeon CM, Wagenblast E, Izzo F, Papapetrou EP. The Crossroads of Clonal Evolution, Differentiation Hierarchy, and Ontogeny in Leukemia Development. Blood Cancer Discov 2025; 6:94-109. [PMID: 39652739 PMCID: PMC11876951 DOI: 10.1158/2643-3230.bcd-24-0235] [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: 09/08/2024] [Revised: 11/19/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
SIGNIFICANCE In recent years, remarkable technological advances have illuminated aspects of the pathogenesis of myeloid malignancies-yet outcomes for patients with these devastating diseases have not significantly improved. We posit that a synthesized view of the three dimensions through which hematopoietic cells transit during their healthy and diseased life-clonal evolution, stem cell hierarchy, and ontogeny-promises high yields in new insights into disease pathogenesis and new therapeutic avenues.
Collapse
Affiliation(s)
- Christopher M. Sturgeon
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, New York
- Black Family Stem Cell Institute, Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Elvin Wagenblast
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, New York
- Black Family Stem Cell Institute, Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Pediatrics, Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Franco Izzo
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, New York
- Black Family Stem Cell Institute, Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Eirini P. Papapetrou
- Center for Advancement of Blood Cancer Therapies, Icahn School of Medicine at Mount Sinai, New York, New York
- Black Family Stem Cell Institute, Institute for Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Medicine, Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
35
|
Huang S, Soto AM, Sonnenschein C. The end of the genetic paradigm of cancer. PLoS Biol 2025; 23:e3003052. [PMID: 40100793 DOI: 10.1371/journal.pbio.3003052] [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: 03/20/2025] Open
Abstract
Genome sequencing of cancer and normal tissues, alongside single-cell transcriptomics, continues to produce findings that challenge the idea that cancer is a 'genetic disease', as posited by the somatic mutation theory (SMT). In this prevailing paradigm, tumorigenesis is caused by cancer-driving somatic mutations and clonal expansion. However, results from tumor sequencing, motivated by the genetic paradigm itself, create apparent 'paradoxes' that are not conducive to a pure SMT. But beyond genetic causation, the new results lend credence to old ideas from organismal biology. To resolve inconsistencies between the genetic paradigm of cancer and biological reality, we must complement deep sequencing with deep thinking: embrace formal theory and historicity of biological entities, and (re)consider non-genetic plasticity of cells and tissues. In this Essay, we discuss the concepts of cell state dynamics and tissue fields that emerge from the collective action of genes and of cells in their morphogenetic context, respectively, and how they help explain inconsistencies in the data in the context of SMT.
Collapse
Affiliation(s)
- Sui Huang
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Ana M Soto
- Tufts University School of Medicine, Immunology, Boston, Massachusetts, United States of America
| | - Carlos Sonnenschein
- Tufts University School of Medicine, Immunology, Boston, Massachusetts, United States of America
| |
Collapse
|
36
|
Ahn B, Chou C, Chou C, Chen J, Zug A, Baykara Y, Claus J, Hacking SM, Uzun A, Gamsiz Uzun E. The Atlas of Protein-Protein Interactions in Cancer (APPIC)-a webtool to visualize and analyze cancer subtypes. NAR Cancer 2025; 7:zcae047. [PMID: 39822275 PMCID: PMC11734624 DOI: 10.1093/narcan/zcae047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/27/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025] Open
Abstract
Cancer is a complex disease with heterogeneous mutational and gene expression patterns. Subgroups of patients who share a phenotype might share a specific genetic architecture including protein-protein interactions (PPIs). We developed the Atlas of Protein-Protein Interactions in Cancer (APPIC), an interactive webtool that provides PPI subnetworks of 10 cancer types and their subtypes shared by cohorts of patients. To achieve this, we analyzed publicly available RNA sequencing data from patients and identified PPIs specific to 26 distinct cancer subtypes. APPIC compiles biological and clinical information from various databases, including the Human Protein Atlas, Hugo Gene Nomenclature Committee, g:Profiler, cBioPortal and Clue.io. The user-friendly interface allows for both 2D and 3D PPI network visualizations, enhancing the usability and interpretability of complex data. For advanced users seeking greater customization, APPIC conveniently provides all output files for further analysis and visualization on other platforms or tools. By offering comprehensive insights into PPIs and their role in cancer, APPIC aims to support the discovery of tumor subtype-specific novel targeted therapeutics and drug repurposing. APPIC is freely available at https://appic.brown.edu.
Collapse
Affiliation(s)
- Benjamin Ahn
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - Charissa Chou
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| | - Caden Chou
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - Jennifer Chen
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - Amelia Zug
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
| | - Yigit Baykara
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| | - Jessica Claus
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
| | - Sean M Hacking
- Department of Pathology, NYU Grossman School of Medicine, 550 1st Ave., New York, NY 10016, USA
| | - Alper Uzun
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
- Legoretta Cancer Center, Brown University, 70 Ship Street, Providence, RI 02903, USA
- Brown Center for Clinical Cancer Informatics and Data Science (CCIDS), Brown University, 593 Eddy Street, Providence, RI 02903, USA
- Center for Computational Molecular Biology, Brown University, 164 Angell Street, Providence, RI 02906, USA
| | - Ece D Gamsiz Uzun
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, 593 Eddy Street, Providence, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, 593 Eddy Street, Providence, RI 02903, USA
- Legoretta Cancer Center, Brown University, 70 Ship Street, Providence, RI 02903, USA
- Brown Center for Clinical Cancer Informatics and Data Science (CCIDS), Brown University, 593 Eddy Street, Providence, RI 02903, USA
- Center for Computational Molecular Biology, Brown University, 164 Angell Street, Providence, RI 02906, USA
| |
Collapse
|
37
|
Wu S, Thawani R. Tumor-Agnostic Therapies in Practice: Challenges, Innovations, and Future Perspectives. Cancers (Basel) 2025; 17:801. [PMID: 40075649 PMCID: PMC11899253 DOI: 10.3390/cancers17050801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/22/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
This review comprehensively analyzes the current landscape of tumor-agnostic therapies in oncology. Tumor-agnostic therapies are designed to target specific molecular alterations rather than the primary site of the tumor, representing a shift in cancer treatment. We discuss recent approvals by regulatory agencies such as the FDA and EMA, highlighting therapies that have demonstrated efficacy across multiple cancer types sharing common alterations. We delve into the trial methodologies that underpin these approvals, emphasizing innovative designs such as basket trials and umbrella trials. These methodologies present unique advantages, including increased efficiency in patient recruitment and the ability to assess drug efficacy in diverse populations rapidly. However, they also entail certain challenges, including the need for robust biomarkers and the complexities of regulatory requirements. Moreover, we examine the promising prospects for developing therapies for rare cancers that exhibit common molecular targets typically associated with more prevalent malignancies. By synthesizing these insights, this review underscores the transformative potential of tumor-agnostic therapies in oncology. It offers a pathway for personalized cancer treatment that transcends conventional histology-based classification.
Collapse
Affiliation(s)
| | - Rajat Thawani
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, Chicago, IL 60637, USA;
| |
Collapse
|
38
|
Khranovska N, Gorbach O, Skachkova O, Klimnyuk G. Application of Next-Generation Sequencing to Realize Principles of Precision Therapy in Management of Cancer Patients. Exp Oncol 2025; 46:295-304. [PMID: 39985357 DOI: 10.15407/exp-oncology.2024.04.295] [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: 02/19/2025] [Indexed: 02/24/2025]
Abstract
All cancers are diseases of the genome, since the cancer cell genome typically consists of 10,000s of passenger alterations, 5-10 biologically relevant alterations, and 1-2 "actionable" alterations. Therefore, somatic mutations in cancer cells can have diagnostic, prognostic, and predictive value. Traditional methods are widely used for testing, such as immunohistochemistry, Sanger sequencing, and allele-specific PCR. However, due to the low throughput, these methods are focused exclusively on testing the most common mutations in target genes. The modern next generation sequencing (NGS) is a technology that enables precision oncology in its current form. ESCAT and ESMO Guidelines defined NGS for routine use in patients with advanced cancers such as non-squamous non-small cell lung cancer, prostate cancer, ovarian cancer, and cholangiocarcinoma. The high sensitivity of the NGS method allows it to be used to search for specific mutations in circulating tumor DNA in blood plasma and other body fluids. NGS testing has evolved from hotspot panels, actionable gene panels, and disease-specific panels to more comprehensive panels. The exome and whole genome sequencing approaches are just beginning to emerge, that is why panel-based testing remains most optimal in oncology practice. NGS is also widely used to identify new and rare mutations in cancer genes and detect inherited cancer mutations.
Collapse
Affiliation(s)
- N Khranovska
- Nonprofit organization "National Cancer Institute", Kyiv, Ukraine
| | - O Gorbach
- Nonprofit organization "National Cancer Institute", Kyiv, Ukraine
| | - O Skachkova
- Nonprofit organization "National Cancer Institute", Kyiv, Ukraine
| | - G Klimnyuk
- Nonprofit organization "National Cancer Institute", Kyiv, Ukraine
| |
Collapse
|
39
|
Yuan LX, Yue ZQ, Ma QR, Zhang P, Xiao F, Chen L. Identification of DAP3 as candidate prognosis marker and potential therapeutic target for hepatocellular carcinoma. Front Immunol 2025; 16:1528853. [PMID: 40051634 PMCID: PMC11882876 DOI: 10.3389/fimmu.2025.1528853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025] Open
Abstract
Background Among malignant tumors, hepatocellular carcinoma (HCC) is both prevalent and highly lethal. Most patients with advanced-stage liver cancer have a poor prognosis. Death-associated protein 3 (DAP3) is reportedly related to tumors and may hold great promise for the future. Methods DAP3 transcriptome data along with related clinical information were obtained from The Cancer Genome Atlas (TCGA), GEO, and ICGC databases. We assessed its prognostic value, clinical relevance, associated pathways, immune infiltration, gene mutations, and sensitivity to chemotherapeutics. A prognostic risk model was subsequently developed and evaluated using receiver operating characteristic (ROC) curves and Kaplan-Meier (KM) plots. Additionally, a nomogram was created and validated through calibration and decision curve analysis (DCA). Furthermore, quantitative real-time PCR (qRT-PCR), Western blot, and immunohistochemical (IHC) staining were performed to examine the expression of DAP3 in HCC. Finally, gene knockdown and overexpression experiments, along with cell counting kit-8 (CCK-8) assays, colony formation assays, and tests for cell apoptosis, migration, and invasion, were conducted to investigate the role of DAP3 in HCC. Results The study discovered that DAP3 expression was linked to HCC subtypes, and its high expression was linked to a poor prognosis. There were significant differences in immune infiltration level, mutation level, prognostic value and chemotherapeutic efficacy. Subsequently, we constructed a prognostic model and demonstrated that high risk score was significantly related to a poor survival rate. A predictive nomogram demonstrated that the nomogram model was effective prediction tool that can accurately predict the survival rate of patients with different clinical characteristics. Additionally, DAP3 expression significantly increased in both tissue samples and cell lines. Elevated levels of DAP3 were correlated with larger tumor size and higher alpha-fetoprotein (AFP) levels, and Cox analysis confirmed that DAP3 was a clinically independent prognostic marker. Finally, cell assays revealed that the knockdown of DAP3 significantly impeded cell proliferation and metabolic activity and induced apoptosis. Conversely, the overexpression of DAP3 had opposite effects on these cellular processes. Conclusions Our study on DAP3 can provide a reference for HCC diagnosis, treatment and prognosis assessment.
Collapse
Affiliation(s)
- Liu-Xia Yuan
- Institute of Liver Diseases, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Zhi-Qiang Yue
- Department of Hepatobiliary Surgery, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Qin-Rong Ma
- Department of Pathology, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Peng Zhang
- Department of Hepatobiliary Surgery, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Feng Xiao
- Department of Pathology, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| | - Lin Chen
- Institute of Liver Diseases, Nantong Third People’s Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
40
|
Niharika, Asthana S, Narayan Yadav H, Sharma N, Kumar Singh V. A compendium of methods: Searching allele specific expression via RNA sequencing. Gene 2025; 936:149102. [PMID: 39561903 DOI: 10.1016/j.gene.2024.149102] [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: 07/29/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 11/21/2024]
Abstract
Diploid mammalian genome has paired alleles for each gene; typically allowing for equal expression of the two alleles within the cell/tissue. However, genetic regulatory elements and epigenetic modifications can disrupt this equality, leading to preferential expression of one allele. Examining high-confidence allele-specific expression (ASE) is vital for understanding genetic variations and their impact on major diseases like cancers and diabetes. ASE analysis not only aids in disease prognosis and diagnosis but also helps to identify regulatory mechanisms operating within the genome. While advances in sequencing technologies have greatly improved our understanding of ASE, challenges remain in estimating it accurately. In this article, we reviewed methods for detecting ASE using both bulk RNASeq and single-cell RNASeq data to provide deeper insights beyond the mere prediction of ASE genes. Fundamentally, ASE detection methods are data-driven and can be classified according to type of data used. Some methods utilize both, DNA genotyping information and RNASeq while others rely solely on RNASeq data. This article offers a comparative analysis of these methods and compilation of repositories providing valuable insights.
Collapse
Affiliation(s)
- Niharika
- Department of Bioinformatics, Central University of South Bihar, Gaya, Bihar 824236, India
| | - Shailendra Asthana
- Computational and Mathematical Biology Centre, Translational Health Science and Technology Institute, NCR Biotech Science Cluster 3rd 15 Milestone, Faridabad-Gurugram 16 expressway, PO Box # 4. Faridabad, Haryana 121001, India
| | - Harlokesh Narayan Yadav
- Department of Pharmacology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Nanaocha Sharma
- Institute of Bioresources and Sustainable Development, Takyelpat, Manipur 795001 Imphal, India.
| | - Vijay Kumar Singh
- Department of Bioinformatics, Central University of South Bihar, Gaya, Bihar 824236, India.
| |
Collapse
|
41
|
Ren Y, Zhang T, Liu J, Ma F, Chen J, Li P, Xiao G, Sun C, Zhang Y. MONet: cancer driver gene identification algorithm based on integrated analysis of multi-omics data and network models. Exp Biol Med (Maywood) 2025; 250:10399. [PMID: 39968416 PMCID: PMC11834253 DOI: 10.3389/ebm.2025.10399] [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: 10/06/2024] [Accepted: 01/22/2025] [Indexed: 02/20/2025] Open
Abstract
Cancer progression is orchestrated by the accrual of mutations in driver genes, which endow malignant cells with a selective proliferative advantage. Identifying cancer driver genes is crucial for elucidating the molecular mechanisms of cancer, advancing targeted therapies, and uncovering novel biomarkers. Based on integrated analysis of Multi-Omics data and Network models, we present MONet, a novel cancer driver gene identification algorithm. Our method utilizes two graph neural network algorithms on protein-protein interaction (PPI) networks to extract feature vector representations for each gene. These feature vectors are subsequently concatenated and fed into a multi-layer perceptron model (MLP) to perform semi-supervised identification of cancer driver genes. For each mutated gene, MONet assigns the probability of being potential driver, with genes identified in at least two PPI networks selected as candidate driver genes. When applied to pan-cancer datasets, MONet demonstrated robustness across various PPI networks, outperforming baseline models in terms of both the area under the receiver operating characteristic curve and the area under the precision-recall curve. Notably, MONet identified 37 novel driver genes that were missed by other methods, including 29 genes such as APOBEC2, GDNF, and PRELP, which are corroborated by existing literature, underscoring their critical roles in cancer development and progression. Through the MONet framework, we successfully identified known and novel candidate cancer driver genes, providing biologically meaningful insights into cancer mechanisms.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yusen Zhang
- School of Mathematics and Statistics, Shandong University, Weihai, Shandong, China
| |
Collapse
|
42
|
Yang Y, Zhang X, Chen T, Wu F, Huang YS, Qu Y, Xu M, Ma L, Liu M, Zhai W. An Expanding Universe of Mutational Signatures and Its Rapid Evolution in Single-Stranded RNA Viruses. Mol Biol Evol 2025; 42:msaf009. [PMID: 39823310 PMCID: PMC11796089 DOI: 10.1093/molbev/msaf009] [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: 08/01/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/19/2025] Open
Abstract
The study of mutational processes in somatic genomes has gained recent momentum, uncovering a wide array of endogenous and exogenous factors associated with somatic changes. However, the overall landscape of mutational processes in germline mutations across the tree of life and associated evolutionary driving forces are rather unclear. In this study, we analyzed mutational processes in single-stranded RNA (ssRNA) viruses which are known to jump between different hosts with divergent exogenous environments. We found that mutational spectra in different ssRNA viruses differ significantly and are mainly associated with their genetic divergence. Surprisingly, host environments contribute much less significantly to the mutational spectrum, challenging the prevailing view that the exogenous cellular environment is a major determinant of the mutational spectrum in viruses. To dissect the evolutionary forces shaping viral spectra, we selected two important scenarios, namely the inter-host evolution between different viral strains as well as the intra-host evolution. In both scenarios, we found mutational spectra change significantly through space and time, strongly correlating with levels of natural selection. Combining the mutations across all ssRNA viruses, we identified a suite of mutational signatures with varying degrees of similarity to somatic signatures in humans, indicating universal and divergent mutational processes across the tree of life. Taken together, we unraveled an unprecedented dynamic landscape of mutational processes in ssRNA viruses, pinpointing important evolutionary forces shaping fast evolution of mutational spectra in different species.
Collapse
Affiliation(s)
- Yue Yang
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xinyi Zhang
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tingting Chen
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fengyuan Wu
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu S Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Genecast Biotechnology Co., Ltd., Wuxi 214105, China
| | - Yanhua Qu
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Miao Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Liang Ma
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mo Liu
- School of Basic Medical Sciences, Sino-French Hoffmann Institute, Guangzhou Medical University, Guangzhou 511436, China
| | - Weiwei Zhai
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| |
Collapse
|
43
|
Zhang G, Levin M. Bioelectricity is a universal multifaced signaling cue in living organisms. Mol Biol Cell 2025; 36:pe2. [PMID: 39873662 PMCID: PMC11809311 DOI: 10.1091/mbc.e23-08-0312] [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: 07/11/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/30/2025] Open
Abstract
The cellular electrical signals of living organisms were discovered more than a century ago and have been extensively investigated in the neuromuscular system. Neuronal depolarization and hyperpolarization are essential for our neuromuscular physiological and pathological functions. Bioelectricity is being recognized as an ancient, intrinsic, fundamental property of all living cells, and it is not limited to the neuromuscular system. Instead, emerging evidence supports a view of bioelectricity as an instructional signaling cue for fundamental cellular physiology, embryonic development, regeneration, and human diseases, including cancers. Here, we highlight the current understanding of bioelectricity and share our views on the challenges and perspectives.
Collapse
Affiliation(s)
- GuangJun Zhang
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN 47906
| | - Michael Levin
- Allen Discovery Center at Tufts University, Medford, MA 02155
| |
Collapse
|
44
|
Yan T, Yan Z, Chen G, Xu S, Wu C, Zhou Q, Wang G, Li Y, Jia M, Zhuang X, Yang J, Liu L, Wang L, Wu Q, Wang B, Yan T. Survival outcome prediction of esophageal squamous cell carcinoma patients based on radiomics and mutation signature. Cancer Imaging 2025; 25:9. [PMID: 39891186 PMCID: PMC11783911 DOI: 10.1186/s40644-024-00821-5] [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: 12/26/2023] [Accepted: 12/29/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND The present study aimed to develop a nomogram model for predicting overall survival (OS) in esophageal squamous cell carcinoma (ESCC) patients. METHODS A total of 205 patients with ESCC were enrolled and randomly divided into a training cohort (n = 153) and a test cohort (n = 52) at a ratio of 7:3. Multivariate Cox regression was used to construct the radiomics model based on CT data. The mutation signature was constructed based on whole genome sequencing data and found to be significantly associated with the prognosis of patients with ESCC. A nomogram model combining the Rad-score and mutation signature was constructed. An integrated nomogram model combining the Rad-score, mutation signature, and clinical factors was constructed. RESULTS A total of 8 CT features were selected for multivariate Cox regression analysis to determine whether the Rad-score was significantly correlated with OS. The area under the curve (AUC) of the radiomics model was 0.834 (95% CI, 0.767-0.900) for the training cohort and 0.733 (95% CI, 0.574-0.892) for the test cohort. The Rad-score, S3, and S6 were used to construct an integrated RM nomogram. The predictive performance of the RM nomogram model was better than that of the radiomics model, with an AUC of 0. 830 (95% CI, 0.761-0.899) in the training cohort and 0.793 (95% CI, 0.653-0.934) in the test cohort. The Rad-score, TNM stage, lymph node metastasis status, S3, and S6 were used to construct an integrated RMC nomogram. The predictive performance of the RMC nomogram model was better than that of the radiomics model and RM nomogram model, with an AUC of 0. 862 (95% CI, 0.795-0.928) in the training cohort and 0. 837 (95% CI, 0.705-0.969) in the test cohort. CONCLUSION An integrated nomogram model combining the Rad-score, mutation signature, and clinical factors can better predict the prognosis of patients with ESCC.
Collapse
Affiliation(s)
- Ting Yan
- Second Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Zhenpeng Yan
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Guohui Chen
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Songrui Xu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Chenxuan Wu
- School of Life Science, Beijing Institute of Technology, Beijing, People's Republic of China
| | - Qichao Zhou
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Guolan Wang
- School of Computer Information Engineering, Shanxi Technology and Business University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Ying Li
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, Shanxi, 030024, People's Republic of China
| | - Mengjiu Jia
- School of Computer Information Engineering, Shanxi Technology and Business University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Xiaofei Zhuang
- Department of Thoracic Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, 030013, People's Republic of China
| | - Jie Yang
- Department of Gastroenterology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Lili Liu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Lu Wang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Qinglu Wu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Bin Wang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, Shanxi, 030024, People's Republic of China.
| | - Tianyi Yan
- School of Life Science, Beijing Institute of Technology, Beijing, People's Republic of China.
| |
Collapse
|
45
|
Hoang Nguyen KH, Le NV, Nguyen PH, Nguyen HHT, Hoang DM, Huynh CD. Human immune system: Exploring diversity across individuals and populations. Heliyon 2025; 11:e41836. [PMID: 39911431 PMCID: PMC11795082 DOI: 10.1016/j.heliyon.2025.e41836] [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: 07/21/2024] [Revised: 12/23/2024] [Accepted: 01/08/2025] [Indexed: 02/07/2025] Open
Abstract
The immune response is an intricate system that involves the complex connection of cellular and molecular components, each with distinct functional specialisations. It has a distinct capacity to adjust and mould the immune response in accordance with specific stimuli, influenced by both genetic and environmental factors. The presence of genetic diversity, particularly across different ethnic and racial groups, significantly contributes to the impact of incidence of diseases, disease susceptibility, autoimmune disorders, and cancer risks in specific regions and certain populations. Environmental factors, including geography and socioeconomic status, further modulate the variety of the immune system responses. These, in turn, affect the susceptibility to infectious diseases and development of autoimmune disorders. Despite the complexity of the relationship, there remains a gap in understanding the specificity of immune indices across races, immune reference ranges among populations, highlighting the need for deeper understanding of immune diversity for personalized approaches in diagnostics and therapeutics. This review systematically organizes these findings, with the goal of emphasizing the potential of targeted interventions to address health disparities and advance translational research, enabling a more comprehensive strategy. This approach promises significant advancements in identifying specific immunological conditions, focusing on personalized interventions, through both genetic and environmental factors.
Collapse
Affiliation(s)
| | - Nghi Vinh Le
- College of Health Sciences, VinUniversity, Hanoi, Viet Nam
| | | | - Hien Hau Thi Nguyen
- College of Health Sciences, VinUniversity, Hanoi, Viet Nam
- Institute of Research and Development, Duy Tan University, Da Nang, Viet Nam
- School of Medicine and Pharmacy, Duy Tan University, Da Nang, Viet Nam
| | - Duy Mai Hoang
- College of Health Sciences, VinUniversity, Hanoi, Viet Nam
| | | |
Collapse
|
46
|
Dong S, Li A, Pan R, Hong J, Wang Z, Shen K. Carboplatin-resistance-related DNA damage repair prognostic gene signature and its association with immune infiltration in breast cancer. Front Immunol 2025; 16:1522149. [PMID: 39944694 PMCID: PMC11813922 DOI: 10.3389/fimmu.2025.1522149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/13/2025] [Indexed: 05/09/2025] Open
Abstract
Introduction Breast cancer is among the most prevalent malignant tumors globally, with carboplatin serving as a standard treatment option. However, resistance often compromises its efficacy. DNA damage repair (DDR) pathways are crucial in determining responses to treatment and are also associated with immune infiltration. This study aimed to identify the DDR genes involved in carboplatin resistance and to elucidate their effects on prognosis, immune infiltration, and drug sensitivity in breast cancer patients. Methods A 3D-culture model resistant to carboplatin was constructed and sequenced. Co-expressed DDR genes were analyzed to develop a predictive model. Immune infiltration analysis tools were employed to assess the immune microenvironment of patients with varying expression levels of these risk genes. Additionally, drug sensitivity predictions were made to evaluate the efficacy of other DNA damage-related drugs across different risk groups. Molecular assays were performed to investigate the role of the key gene TONSL in breast cancer. Results By integrating data from public database, we established a prognostic signature comprising thirteen DDR genes. Our analysis indicated that this model is associated with immune infiltration patterns in breast cancer patients, particularly concerning CD8+ T cells and NK cells. Additionally, it demonstrated a significant correlation with sensitivity to other DDR-related drugs, suggesting its potential as a biomarker for treatment efficacy. Compared to the control group, TONSL-knockdown cell lines exhibited a diminished response to DNA-damaging agents, marked by a notable increase in DNA damage levels and enhanced drug sensitivity. Furthermore, single-cell analysis revealed elevated TONSL expression in dendritic and epithelial cells, particularly in triple-negative breast cancers. Conclusions Carboplatin resistance-related DDR genes are associated with prognosis, immune infiltration, and drug sensitivity in breast cancer patients. TONSL may serve as a potential therapeutic target for breast cancer, particularly in triple-negative breast cancer, indicating new treatment strategies for these patients.
Collapse
Affiliation(s)
- Shuwen Dong
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Anqi Li
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruixin Pan
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jin Hong
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Wang
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kunwei Shen
- Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
47
|
Liu D, Liu L, Che X, Wu G. Discovery of paradoxical genes: reevaluating the prognostic impact of overexpressed genes in cancer. Front Cell Dev Biol 2025; 13:1525345. [PMID: 39911323 PMCID: PMC11794808 DOI: 10.3389/fcell.2025.1525345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 01/07/2025] [Indexed: 02/07/2025] Open
Abstract
Oncogenes are typically overexpressed in tumor tissues and often linked to poor prognosis. However, recent advancements in bioinformatics have revealed that many highly expressed genes in tumors are associated with better patient outcomes. These genes, which act as tumor suppressors, are referred to as "paradoxical genes." Analyzing The Cancer Genome Atlas (TCGA) confirmed the widespread presence of paradoxical genes, and KEGG analysis revealed their role in regulating tumor metabolism. Mechanistically, discrepancies between gene and protein expression-affected by pre- and post-transcriptional modifications-may drive this phenomenon. Mechanisms like upstream open reading frames and alternative splicing contribute to these inconsistencies. Many paradoxical genes modulate the tumor immune microenvironment, exerting tumor-suppressive effects. Further analysis shows that the stage- and tumor-specific expression of these genes, along with their environmental sensitivity, influence their dual roles in various signaling pathways. These findings highlight the importance of paradoxical genes in resisting tumor progression and maintaining cellular homeostasis, offering new avenues for targeted cancer therapy.
Collapse
Affiliation(s)
| | | | - Xiangyu Che
- *Correspondence: Guangzhen Wu, ; Xiangyu Che,
| | | |
Collapse
|
48
|
Ramos RH, Bardelotte YA, de Oliveira Lage Ferreira C, Simao A. Identifying key genes in cancer networks using persistent homology. Sci Rep 2025; 15:2751. [PMID: 39838168 PMCID: PMC11751331 DOI: 10.1038/s41598-025-87265-4] [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: 10/01/2024] [Accepted: 01/17/2025] [Indexed: 01/23/2025] Open
Abstract
Identifying driver genes is crucial for understanding oncogenesis and developing targeted cancer therapies. Driver discovery methods using protein or pathway networks rely on traditional network science measures, focusing on nodes, edges, or community metrics. These methods can overlook the high-dimensional interactions that cancer genes have within cancer networks. This study presents a novel method using Persistent Homology to analyze the role of driver genes in higher-order structures within Cancer Consensus Networks derived from main cellular pathways. We integrate mutation data from six cancer types and three biological functions: DNA Repair, Chromatin Organization, and Programmed Cell Death. We systematically evaluated the impact of gene removal on topological voids ([Formula: see text] structures) within the Cancer Consensus Networks. Our results reveal that only known driver genes and cancer-associated genes influence these structures, while passenger genes do not. Although centrality measures alone proved insufficient to fully characterize impact genes, combining higher-order topological analysis with traditional network metrics can improve the precision of distinguishing between drivers and passengers. This work shows that cancer genes play an important role in higher-order structures, going beyond pairwise measures, and provides an approach to distinguish drivers and cancer-associated genes from passenger genes.
Collapse
Affiliation(s)
- Rodrigo Henrique Ramos
- University of São Paulo, ICMC, São Carlos, 13566-590, Brazil.
- Federal Institute of São Paulo, São Carlos, 13565-820, Brazil.
| | | | | | - Adenilso Simao
- University of São Paulo, ICMC, São Carlos, 13566-590, Brazil
| |
Collapse
|
49
|
Zhang D, Zhang E, Cai Y, Sun Y, Zeng P, Jiang X, Lian Y. Deciphering the potential ability of DExD/H-box helicase 60 (DDX60) on the proliferation, diagnostic and prognostic biomarker in pancreatic cancer: a research based on silico, RNA-seq and molecular biology experiment. Hereditas 2025; 162:6. [PMID: 39844327 PMCID: PMC11753068 DOI: 10.1186/s41065-024-00361-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/22/2024] [Indexed: 01/24/2025] Open
Abstract
BACKGROUND Pancreatic cancer is one of the most malignant abdominal tumors. DDX60 has been shown to be associated with a variety of tumor biological processes. However, DDX60 in pancreatic cancer has not been reported. Our study confirmed that DDX60 can serve as a novel biomarker for diagnosis and treatment of pancreatic cancer. MATERIALS AND METHODS We downloaded pancreatic cancer datasets from GEO and TCGA databases, respectively. To investigate the relationship between DDX60 expression and prognosis in pancreatic cancer. GSEA analysis was performed on DDX60. We performed RNA-seq to further explore the downstream biological targets of DDX60 and the signaling pathways that may be involved in pancreatic cancer. Finally, we tested it through molecular biology experiments. First, we constructed the plasmid and tested the plasmid effect by WB. Then MTT assay was performed to explore the effect of DDX60 knockout on the proliferation of pancreatic cancer cells. LDH assay was performed to explore the effect of DDX60 on the release of lactate dehydrogenase from tumor cells. The effect of DDX60 on cell proliferation was further explored by clonal formation experiment. Continue to explore clinical therapeutic drugs sensitive to DDX60 targets. RESULTS By analyzing the GSE71729, GSE183795, GSE16515, GSE28735 and GSE62452 data sets, we found that DDX60 was highly expressed in pancreatic cancer. And is associated with poorer outcomes for pancreatic patients. The mRNA expression level of DDX60 was correlated with lymph node metastasis and grade in clinical pancreatic patients. Through the results of RNA-seq analysis, GO and KEGG analysis showed that DDX60 may be associated with cell cycle in pancreatic cancer. Through molecular biology experiments (MTT, LDH, and clonal formation experiment), we found that When DDX60 is knocked down in pancreatic cancer cells, the proliferation ability of tumor cells is significantly decreased. Several drugs targeting about DDX60 have been found, such as JW-7-52-1, this could provide direction for drug therapy against the DDX60 target. CONCLUSION In summary, DDX60 can be used as a novel biomarker related to the diagnosis and treatment of pancreatic cancer, participate in tumor proliferation, and is associated with poor prognosis in patients.
Collapse
Affiliation(s)
- Dongdong Zhang
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China
| | - Enze Zhang
- School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China
| | - Ying Cai
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China
| | - Yixin Sun
- School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China
- 3National Institute for Data Science in Health and Medicine, Xiamen UniversityXiamen, Fujian, 361000, China
| | - Peiji Zeng
- School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China
| | - Xiaohua Jiang
- Department of Orthopedics, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, People's Republic of China.
| | - Yifan Lian
- Department of Gastroenterology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
- School of Medicine, Xiamen University, Xiamen, 361000, Fujian, China.
| |
Collapse
|
50
|
Liu Z, Wang Y, Peng Z, Li H, Wang H, Wu Y, Jiang X, Fu P. Fusion of tumor cells and mesenchymal stem/stroma cells: a source of tumor heterogeneity, evolution and recurrence. Med Oncol 2025; 42:52. [PMID: 39838167 DOI: 10.1007/s12032-024-02595-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: 10/25/2024] [Accepted: 12/28/2024] [Indexed: 01/23/2025]
Abstract
The heterogeneity and evolution of tumors remain significant obstacles in cancer treatment, contributing to both therapy resistance and relapse. Mesenchymal stem/stromal cells (MSCs) are multipotent stromal cells within the tumor microenvironment that interact with tumor cells through various mechanisms, including cell fusion. While previous research has largely focused on the effects of MSC-tumor cell fusion on tumor proliferation, migration, and tumorigenicity, emerging evidence indicates that its role in tumor maintenance, evolution, and recurrence, particularly under stress conditions, may be even more pivotal. This review examines the connection between MSC-tumor cell fusion and several critical factors like tumor heterogeneity, cancer stem cells, and therapy resistance, highlighting the crucial role of cell fusion in tumor survival, evolution, and recurrence. Additionally, we explore potential therapeutic strategies aimed at targeting this process.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yihao Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zesheng Peng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Li
- Department of Cataract, Nanyang Eye Hospital, Nanyang, 473000, China
| | - Haofei Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuyi Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Peng Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|