1
|
Parra RG, Komives EA, Wolynes PG, Ferreiro DU. Frustration in physiology and molecular medicine. Mol Aspects Med 2025; 103:101362. [PMID: 40273505 DOI: 10.1016/j.mam.2025.101362] [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: 01/30/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/26/2025]
Abstract
Molecules provide the ultimate language in terms of which physiology and pathology must be understood. Myriads of proteins participate in elaborate networks of interactions and perform chemical activities coordinating the life of cells. To perform these often amazing tasks, proteins must move and we must think of them as dynamic ensembles of three dimensional structures formed first by folding the polypeptide chains so as to minimize the conflicts between the interactions of their constituent amino acids. It is apparent however that, even when completely folded, not all conflicting interactions have been resolved so the structure remains 'locally frustrated'. Over the last decades it has become clearer that this local frustration is not just a random accident but plays an essential part of the inner workings of protein molecules. We will review here the physical origins of the frustration concept and review evidence that local frustration is important for protein physiology, protein-protein recognition, catalysis and allostery. Also, we highlight examples showing how alterations in the local frustration patterns can be linked to distinct pathologies. Finally we explore the extensions of the impact of frustration in higher order levels of organization of systems including gene regulatory networks and the neural networks of the brain.
Collapse
Affiliation(s)
- R Gonzalo Parra
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Spain
| | | | - Peter G Wolynes
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA.
| | - Diego U Ferreiro
- Protein Physiology Lab, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina.
| |
Collapse
|
2
|
Shu QA, Huang XD, Wu J, Liang XR, Wang YL, Yu JM, Wang ZW, Liu N, Wang JH, Xie N. ARHGAP11A is a potential prognostic biomarker and therapeutic target for pancreatic adenocarcinoma. Int J Biochem Cell Biol 2025; 185:106804. [PMID: 40398712 DOI: 10.1016/j.biocel.2025.106804] [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: 02/27/2024] [Revised: 06/23/2024] [Accepted: 05/16/2025] [Indexed: 05/23/2025]
Abstract
The prognosis of pancreatic adenocarcinoma remains dismal up to now, partly owing to a lack of clinically feasible therapeutic targets. ARHGAP11A, a member of Rho GTPase-activating proteins family, has been reported as a prognostic biomarker and oncogene for multiple cancers. However, the specific effects of ARHGAP11A in pancreatic adenocarcinoma remain obscure. In this study, we explored and validated the oncogenic role of ARHGAP11A in pancreatic adenocarcinoma via thorough bioinformatics analyses of public databases and our own RNA-seq data, and in vitro experiments. We found that ARHGAP11A was significantly upregulated in pancreatic adenocarcinoma and correlated with poor clinical outcomes of pancreatic adenocarcinoma patients. Functional enrichment analyses revealed that ARHGAP11A-related genes were enriched in pathways of cell cycle and cell apoptosis. Further molecular functional experiments on ARHGAP11A knockdown pancreatic adenocarcinoma cell lines demonstrated that ARHGAP11A knockdown significantly inhibited cell proliferation, promoted cell cycle arrest in the G1/S stage, and induced cell apoptosis. In comparison, overexpression of ARHGAP11A could remarkably promote the proliferation, cell cycle progression, and apoptosis resistance of pancreatic adenocarcinoma cells. In conclusion, our study demonstrates that ARHGAP11A plays an oncogenic role in pancreatic adenocarcinoma, thus providing a novel therapeutic target and prognostic biomarker for patients with pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Qiu-Ai Shu
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xin-di Huang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Jian Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xi-Ru Liang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Ya-Lan Wang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China
| | - Jin-Min Yu
- Division of Gastroenterology, Xi'an Central Hospital, Xi'an, Shaanxi 710003, China
| | - Zi-Wei Wang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Na Liu
- Department of Gastroenterology, Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, Hainan 570100, China.
| | - Jin-Hai Wang
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China.
| | - Ning Xie
- Department of Gastroenterology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710004, China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China; The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China.
| |
Collapse
|
3
|
Kratochvilová L, Dinová A, Valková N, Dobrovolná M, Sánchez-Murcia PA, Brázda V. Chromatin Immunoprecipitation Reveals p53 Binding to G-Quadruplex DNA Sequences in Myeloid Leukemia Cell Lines. ACS BIO & MED CHEM AU 2025; 5:283-298. [PMID: 40255281 PMCID: PMC12006861 DOI: 10.1021/acsbiomedchemau.4c00124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 04/22/2025]
Abstract
Clarifying functions of the p53 protein is a crucial aspect of cancer research. We analyzed the binding sites of p53 wild-type (WT) protein and its oncologically significant mutants and evaluated their transactivation properties using a functional yeast assay. Unlike the binding sites as determined in myeloid leukemia cell lines by chromatin immunoprecipitation of p53-R175H, p53-Y220C, p53-M237I, p53-R248Q, and p53-R273H mutants, the target sites of p53-WT and p53-R282W were significantly associated with putative G-quadruplex sequences (PQSs). Guanine-quadruplex (G-quadruplex or G4) formation in these sequences was evaluated by using a set of biophysical methods. G4s can modulate gene expression induced by p53. At low p53 expression level, PQS upstream of the p53-response element (RE) leads to greater gene expression induced by p53-R282W compared to that for the RE without PQS. Meanwhile, p53-WT protein expression is decreased by the PQS presence. At a high p53 expression level, the presence of PQS leads to a decreased expression of the reporter regardless of the distance and localization of the G4 from the RE.
Collapse
Affiliation(s)
- Libuše Kratochvilová
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic
- Department
of Food Chemistry and Biotechnology, Faculty of Chemistry, Brno University of Technology, Purkyňova 118, Brno 612 00, Czech Republic
| | - Alessandra Dinová
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic
| | - Natália Valková
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic
| | - Michaela Dobrovolná
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic
- Department
of Food Chemistry and Biotechnology, Faculty of Chemistry, Brno University of Technology, Purkyňova 118, Brno 612 00, Czech Republic
| | - Pedro A. Sánchez-Murcia
- Laboratory
of Computer-Aided Molecular Design, Division of Medicinal Chemistry, Otto-Loewi Research Center, Neue Stiftingtalstr. 6/III, Graz A-8010, Austria
- BioTechMed-Graz, Mozartgasse 12/II, Graz A-8010, Austria
| | - Václav Brázda
- Institute
of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 65, Czech Republic
- Department
of Food Chemistry and Biotechnology, Faculty of Chemistry, Brno University of Technology, Purkyňova 118, Brno 612 00, Czech Republic
| |
Collapse
|
4
|
Parra RG, Komives EA, Wolynes PG, Ferreiro DU. Frustration In Physiology And Molecular Medicine. ARXIV 2025:arXiv:2502.03851v1. [PMID: 39975445 PMCID: PMC11838788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Molecules provide the ultimate language in terms of which physiology and pathology must be understood. Myriads of proteins participate in elaborate networks of interactions and perform chemical activities coordinating the life of cells. To perform these often amazing tasks, proteins must move and we must think of them as dynamic ensembles of three dimensional structures formed first by folding the polypeptide chains so as to minimize the conflicts between the interactions of their constituent amino acids. It is apparent however that, even when completely folded, not all conflicting interactions have been resolved so the structure remains 'locally frustrated'. Over the last decades it has become clearer that this local frustration is not just a random accident but plays an essential part of the inner workings of protein molecules. We will review here the physical origins of the frustration concept and review evidence that local frustration is important for protein physiology, protein-protein recognition, catalysis and allostery. Also, we highlight examples showing how alterations in the local frustration patterns can be linked to distinct pathologies. Finally we explore the extensions of the impact of frustration in higher order levels of organization of systems including gene regulatory networks and the neural networks of the brain.
Collapse
Affiliation(s)
- R. Gonzalo Parra
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Spain
| | | | - Peter G. Wolynes
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005
| | - Diego U. Ferreiro
- Protein Physiology Lab, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales, Consejo Nacional de Investigaciones Científicas y Técnicas - Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| |
Collapse
|
5
|
Ghosh P, Kundu A, Ganguly D. From experimental studies to computational approaches: recent trends in designing novel therapeutics for amyloidogenesis. J Mater Chem B 2025; 13:858-881. [PMID: 39664012 DOI: 10.1039/d4tb01890g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Amyloidosis is a condition marked by misfolded proteins that build up in tissues and eventually destroy organs. It has been connected to a number of fatal illnesses, including non-neuropathic and neurodegenerative conditions, which in turn have a significant influence on the worldwide health sector. The inability to identify the underlying etiology of amyloidosis has hampered efforts to find a treatment for the condition. Despite the identification of a multitude of putative pathogenic variables that may operate independently or in combination, the molecular mechanisms responsible for the development and progression of the disease remain unclear. A thorough investigation into protein aggregation and the impacts of toxic aggregated species will help to clarify the cytotoxicity of aggregation-mediated cellular apoptosis and lay the groundwork for future studies aimed at creating effective treatments and medications. This review article provides a thorough summary of the combination of various experimental and computational approaches to modulate amyloid aggregation. Further, an overview of the latest developments of novel therapeutic agents is given, along with a discussion of the possible obstacles and viewpoints on this developing field. We believe that the information provided by this review will help scientists create innovative treatment strategies that affect the way proteins aggregate.
Collapse
Affiliation(s)
- Pooja Ghosh
- Centre for Interdisciplinary Sciences, JIS Institute of Advanced Studies & Research (JISIASR) Kolkata, JIS University, GP Block, Sector-5, Salt Lake, Kolkata 700091, West Bengal, India.
| | - Agnibin Kundu
- Department of Medicine, District Hospital Howrah, 10, Biplabi Haren Ghosh Sarani Lane, Howrah 711101, West Bengal, India
| | - Debabani Ganguly
- Centre for Health Science & Technology, JIS Institute of Advanced Studies & Research (JISIASR) Kolkata, JIS University, GP Block, Sector-5, Salt Lake, Kolkata 700091, West Bengal, India.
| |
Collapse
|
6
|
Chai D, Wang J, Lim JM, Xie X, Yu X, Zhao D, Maza PAM, Wang Y, Cyril-Remirez D, Young KH, Li Y. Lipid nanoparticles deliver DNA-encoded biologics and induce potent protective immunity. Mol Cancer 2025; 24:12. [PMID: 39806486 PMCID: PMC11727718 DOI: 10.1186/s12943-024-02211-8] [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: 09/27/2024] [Accepted: 12/25/2024] [Indexed: 01/16/2025] Open
Abstract
Lipid nanoparticles (LNPs) for mRNA delivery have advanced significantly, but LNP-mediated DNA delivery still faces clinical challenges. This study compared various LNP formulations for delivering DNA-encoded biologics, assessing their expression efficacy and the protective immunity generated by LNP-encapsulated DNA in different models. The LNP formulation used in Moderna's Spikevax mRNA vaccine (LNP-M) demonstrated a stable nanoparticle structure, high expression efficiency, and low toxicity. Notably, a DNA vaccine encoding the spike protein, delivered via LNP-M, induced stronger antigen-specific antibody and T cell immune responses compared to electroporation. Single-cell RNA sequencing (scRNA-seq) analysis revealed that the LNP-M/pSpike vaccine enhanced CD80 activation signaling in CD8+ T cells, NK cells, macrophages, and DCs, while reducing the immunosuppressive signals. The enrichment of TCR and BCR by LNP-M/pSpike suggested an increase in immune response specificity and diversity. Additionally, LNP-M effectively delivered DNA-encoded antigens, such as mouse PD-L1 and p53R172H, or monoclonal antibodies targeting mouse PD1 and human p53R282W. This approach inhibited tumor growth or metastasis in several mouse models. The long-term anti-tumor effects of LNP-M-delivered anti-p53R282W antibody relied on memory CD8+ T cell responses and enhanced MHC-I signaling from APCs to CD8+ T cells. These results highlight LNP-M as a promising and effective platform for delivering DNA-based vaccines and cancer immunotherapies.
Collapse
Affiliation(s)
- Dafei Chai
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Junhao Wang
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jing Ming Lim
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xiaohui Xie
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xinfang Yu
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dan Zhao
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Perry Ayn Mayson Maza
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yifei Wang
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dana Cyril-Remirez
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Ken H Young
- Department of Pathology, Division of Hematopathology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Yong Li
- Department of Medicine, Section of Epidemiology and Population Sciences, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
7
|
Russo A, Moy J, Khin M, Dorsey TR, Lopez Carrero A, Burdette JE. Loss of phosphatase and tensin homolog ( PTEN) increases Lysyl oxidase-like 2 ( LOXL2) expression enhancing the growth of fallopian tube epithelial cells as three-dimensional spheroids. CANCER PATHOGENESIS AND THERAPY 2025; 3:68-75. [PMID: 39872364 PMCID: PMC11763906 DOI: 10.1016/j.cpt.2024.03.003] [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: 01/03/2024] [Revised: 03/15/2024] [Accepted: 03/21/2024] [Indexed: 01/30/2025]
Abstract
Background High-grade serous ovarian cancer (HGSOC) accounts for 70-80% of all ovarian cancer-related deaths. Multiple studies have suggested that the fallopian tube epithelium (FTE) serves as the cell of origin of HGSOC. Phosphatase and tensin homolog (PTEN) is a tumor suppressor and its loss is sufficient to induce numerous tumorigenic changes in FTE, including increased migration, formation of multicellular tumor spheroids (MTSs), and ovarian colonization. In murine oviductal epithelial (MOE) cells (the equivalent of human FTE) loss of PTEN results in the upregulation of transcripts associated with the extracellular matrix, with a specific focus on the elevation of lysyl oxidase-like 2 (LOXL2). Although LOXL2 is known to drive transformation and invasion in solid tumors and is associated with a poor prognosis in ovarian cancer, its specific role in the tumorigenesis of ovarian cancer originating from FTE remains unclear. Therefore, we aim to investigate whether LOXL2 mediates tumorigenesis from the fallopian tube epithelium. Methods In this study, we utilized clustered regularly interspaced short palindromic repeats (CRISPR)-CRISPR-associated protein 9 (CAS9) technology to delete LOXL2 in PTEN-deficient MOE cells to understand its role in mediating the oncogenic effects of PTEN loss. In addition, CRISPR-CAS9 was used to delete LOXL2 in OVCAR8 ovarian cancer cells. We monitored the changes in tumorigenic properties, such as migration, invasion, and growth of three-dimensional (3D) spheroids, to assess whether the loss of LOXL2 resulted in any changes. Results We found that a reduction in LOXL2 expression did not significantly change the migration or invasive capabilities of PTEN-depleted MOE or human ovarian cancer cells. However, we found that a reduction in LOXL2 expression resulted in a significant reduction in 3D MTS formation and survival in both lines. Conclusions These results reveal for the first time that PTEN loss in FTE cells increases LOXL2 expression through downregulation of Pax2, and LOXL2 deletion blocks 3D spheroid formation.
Collapse
Affiliation(s)
- Angela Russo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, IL 60607, USA
| | - Junlone Moy
- College of Pharmacy, University of Illinois, Chicago, IL 60612, USA
| | - Manead Khin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, IL 60607, USA
| | | | - Alfredo Lopez Carrero
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, IL 60607, USA
| | - Joanna E. Burdette
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois, Chicago, IL 60607, USA
| |
Collapse
|
8
|
Qiu L, Ma Z, Wu X. Mutant p53-Mediated Tumor Secretome: Bridging Tumor Cells and Stromal Cells. Genes (Basel) 2024; 15:1615. [PMID: 39766882 PMCID: PMC11675497 DOI: 10.3390/genes15121615] [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: 11/01/2024] [Revised: 12/06/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
The tumor secretome comprises the totality of protein factors secreted by various cell components within the tumor microenvironment, serving as the primary medium for signal transduction between tumor cells and between tumor cells and stromal cells. The deletion or mutation of the p53 gene leads to alterations in cellular secretion characteristics, contributing to the construction of the tumor microenvironment in a cell non-autonomous manner. This review discusses the critical roles of mutant p53 in regulating the tumor secretome to remodel the tumor microenvironment, drive tumor progression, and influence the plasticity of cancer-associated fibroblasts (CAFs) as well as the dynamics of tumor immunity by focusing on both secreted protein expression and secretion pathways. The aim is to provide new insights for targeted cancer therapies.
Collapse
Affiliation(s)
| | | | - Xiaoming Wu
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming 650500, China; (L.Q.); (Z.M.)
| |
Collapse
|
9
|
Xu W, Ma W, Wang D, Zhou X, Wang K, Mu K. Integrated multi-omics profiling reveals a clinically relevant molecular feature and potential therapeutic target on phyllodes tumors of breast. Transl Oncol 2024; 46:101998. [PMID: 38761630 PMCID: PMC11112002 DOI: 10.1016/j.tranon.2024.101998] [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: 12/29/2023] [Revised: 04/19/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024] Open
Abstract
Phyllodes tumors (PTs) has an increased risk of local relapse and distant metastases. Molecular features correlating to histologic grade and aggressive behavior of PTs are poorly characterized. Here, whole exome sequencing (WES) was performed to explore genetic mutations in 61 samples of fibroepithelial breast tumors, including 16 fibroadenomas (FAs), 18 benign PTs, 19 borderline PTs, and 8 malignant PTs. Our work clearly shows that FA, benign PT, borderline PT, and malignant PT are independent entities at the genomic level. They may exist as hidden sub-clones carrying specific genetic alterations. Malignant PT-specific mutations present a multi-gene co-mutational pattern suggesting a synergistic effect of co-mutated genes in processes associated with malignant behavior. Moreover, we made a combined genomic and transcriptomic analysis, which presented a mutated gene-based interaction with expression profiles. We found that EGFR mutations (c.710C > T, c.758A > G, c.1295A > G, and c.2156G > C) serve as a hub of interaction network in borderline PTs, which suggests EGFR tyrosine kinase inhibitors (EGFRi) might be effective for borderline PTs. We found TP53 mutations (c.730G > T, c.844C > T, and c.1019delA) serves as a hub event of molecular changes of malignant PTs. Thus, our study based on the omics platforms of genome and transcriptome provides a better understanding of relapse process and the potential targeted therapy in PTs, which is pivotal in improving molecular-guided patient selection and designing clinically relevant combination strategies.
Collapse
Affiliation(s)
- Wei Xu
- Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China; Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Wei Ma
- Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China
| | - Depeng Wang
- Department of Pathology, First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, 261000, China
| | - Xingchen Zhou
- Department of Pathology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250021, China
| | - Kangyu Wang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Kun Mu
- Department of Pathology, School of Basic Medical Sciences, Shandong University, Jinan, 250012, China; Department of Pathology, Qilu Hospital of Shandong University, Jinan, 250012, China.
| |
Collapse
|
10
|
Vieira IA, Pezzi EH, Bandeira IC, Reis LB, de Araújo Rocha YM, Fernandes BV, Siebert M, Miyamoto KN, Siqueira MB, Achatz MI, Galvão HDCR, Garcia FADO, Campacci N, Carraro DM, Formiga MN, Vianna FSL, Palmero EI, Macedo GS, Ashton-Prolla P. Functional pri-miR-34b/c rs4938723 and KRAS 3'UTR rs61764370 SNPs: Novel phenotype modifiers in Li-Fraumeni Syndrome? Gene 2024; 898:148069. [PMID: 38070788 DOI: 10.1016/j.gene.2023.148069] [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: 07/25/2023] [Revised: 11/14/2023] [Accepted: 12/06/2023] [Indexed: 12/25/2023]
Abstract
PURPOSE Li-Fraumeni Syndrome (LFS) is a rare cancer predisposing condition caused by germline pathogenic TP53 variants, in which core tumors comprise sarcomas, breast, brain and adrenocortical neoplasms. Clinical manifestations are highly variable in carriers of the Brazilian germline founder variant TP53 p.R337H, possibly due to the influence of modifier genes such as miRNA genes involved in the regulation of the p53 pathway. Herein, we investigated the potential phenotypic effects of two miRNA-related functional SNPs, pri-miR-34b/c rs4938723 and 3'UTR KRAS rs61764370, in a cohort of 273 LFS patients from Southern and Southeastern Brazil. METHODS The genotyping of selected SNPs was performed by TaqMan® allelic discrimination and subsequently custom TaqMan® genotyping results were confirmed by Sanger sequencing in all SNP-positive LFS patients. RESULTS Although the KRAS SNP showed no effect as a phenotype modulator, the rs4938723 CC genotype was significantly associated with development of LFS non-core tumors (first tumor diagnosis) in p.R337H carriers (p = 0.039). Non-core tumors were also more frequently diagnosed in carriers of germline TP53 DNA binding domain variants harboring the rs4938723 C variant allele. Previous studies described pri-miR-34b/c rs4938723 C as a risk allele for sporadic occurrence of thyroid and prostate cancers (non-core tumors of the LFS spectrum). CONCLUSION With this study, we presented additional evidence about the importance of analyzing miRNA genes that could indirectly regulate p53 expression, and, therefore, may modulate the LFS phenotype, such as those of the miR-34 family.
Collapse
Affiliation(s)
- Igor Araujo Vieira
- Post-Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Genomic Medicine Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Rio Grande do Sul, Brazil; Health School, Universidade do Vale do Rio dos Sinos (UNISINOS), São Leopoldo 93022-750, Brazil.
| | - Eduarda Heidrich Pezzi
- Genomic Medicine Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Larissa Brussa Reis
- Post-Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Genomic Medicine Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Yasminne Marinho de Araújo Rocha
- Genomic Medicine Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Bruna Vieira Fernandes
- Genomic Medicine Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Marina Siebert
- Experimental Research Center, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Monique Banik Siqueira
- Health School, Universidade do Vale do Rio dos Sinos (UNISINOS), São Leopoldo 93022-750, Brazil
| | - Maria I Achatz
- Centro de Oncologia, Hospital Sírio-Libanês, São Paulo, Brazil
| | | | | | - Natalia Campacci
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil; Genomic Medicine Service from Hospital Beneficência Portuguesa de São Paulo, São Paulo, Brazil
| | | | | | - Fernanda Sales Luiz Vianna
- Post-Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Genomic Medicine Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Rio Grande do Sul, Brazil; Department of Genetics, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil
| | - Edenir Inez Palmero
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, São Paulo, Brazil; Department of Genetics, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Gabriel S Macedo
- Genomic Medicine Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Rio Grande do Sul, Brazil; Hospital Moinhos de Vento (HMV), Porto Alegre, Rio Grande do Sul, Brazil; Experimental Research Center, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Patricia Ashton-Prolla
- Post-Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Genomic Medicine Laboratory, Experimental Research Center, Hospital de Clínicas de Porto Alegre (HCPA), Porto Alegre, Rio Grande do Sul, Brazil; Department of Genetics, UFRGS, Porto Alegre, Rio Grande do Sul, Brazil; Medical Genetics Service, HCPA, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
11
|
King C, Fowler JC, Abnizova I, Sood RK, Hall MWJ, Szeverényi I, Tham M, Huang J, Young SM, Hall BA, Birgitte Lane E, Jones PH. Somatic mutations in facial skin from countries of contrasting skin cancer risk. Nat Genet 2023; 55:1440-1447. [PMID: 37537257 PMCID: PMC10484783 DOI: 10.1038/s41588-023-01468-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 07/06/2023] [Indexed: 08/05/2023]
Abstract
The incidence of keratinocyte cancer (basal cell and squamous cell carcinomas of the skin) is 17-fold lower in Singapore than the UK1-3, despite Singapore receiving 2-3 times more ultraviolet (UV) radiation4,5. Aging skin contains somatic mutant clones from which such cancers develop6,7. We hypothesized that differences in keratinocyte cancer incidence may be reflected in the normal skin mutational landscape. Here we show that, compared to Singapore, aging facial skin from populations in the UK has a fourfold greater mutational burden, a predominant UV mutational signature, increased copy number aberrations and increased mutant TP53 selection. These features are shared by keratinocyte cancers from high-incidence and low-incidence populations8-13. In Singaporean skin, most mutations result from cell-intrinsic processes; mutant NOTCH1 and NOTCH2 are more strongly selected than in the UK. Aging skin in a high-incidence country has multiple features convergent with cancer that are not found in a low-risk country. These differences may reflect germline variation in UV-protective genes.
Collapse
Affiliation(s)
| | | | | | | | - Michael W J Hall
- Wellcome Sanger Institute, Hinxton, UK
- Department of Oncology, University of Cambridge, Hutchinson Research Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Ildikó Szeverényi
- Skin Research Institute of Singapore and Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Institute of Aquaculture and Environmental Safety, Georgikon Campus, Hungarian University of Agricultural and Life Sciences, Keszthely, Hungary
| | - Muly Tham
- Skin Research Institute of Singapore and Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jingxiang Huang
- Skin Research Institute of Singapore and Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | | | - Benjamin A Hall
- Department of Medical Physics and Biomedical Engineering, University College London, London, UK
| | - E Birgitte Lane
- Skin Research Institute of Singapore and Institute of Medical Biology, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Philip H Jones
- Wellcome Sanger Institute, Hinxton, UK.
- Department of Oncology, University of Cambridge, Hutchinson Research Centre, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
12
|
Romanovsky E, Kluck K, Ourailidis I, Menzel M, Beck S, Ball M, Kazdal D, Christopoulos P, Schirmacher P, Stiewe T, Stenzinger A, Budczies J. Homogenous TP53mut-associated tumor biology across mutation and cancer types revealed by transcriptome analysis. Cell Death Discov 2023; 9:126. [PMID: 37059713 PMCID: PMC10104808 DOI: 10.1038/s41420-023-01413-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 04/16/2023] Open
Abstract
TP53 is the most frequently mutated gene in human cancer. While no TP53-targeting drugs have been approved in the USA or Europe so far, preclinical and clinical studies are underway to investigate targeting of specific or all TP53 mutations, for example, by restoration of the functionality of mutated TP53 (TP53mut) or protecting wildtype TP53 (TP53wt) from negative regulation. We performed a comprehensive mRNA expression analysis in 24 cancer types of TCGA to extract (i) a consensus expression signature shared across TP53 mutation types and cancer types, (ii) differential gene expression patterns between tumors harboring different TP53 mutation types such as loss of function, gain of function or dominant-negative mutations, and (iii) cancer-type-specific patterns of gene expression and immune infiltration. Analysis of mutational hotspots revealed both similarities across cancer types and cancer type-specific hotspots. Underlying ubiquitous and cancer type-specific mutational processes with the associated mutational signatures contributed to explaining this observation. Virtually no genes were differentially expressed between tumors harboring different TP53 mutation types, while hundreds of genes were over- and underexpressed in TP53mut compared to TP53wt tumors. A consensus list included 178 genes that were overexpressed and 32 genes that were underexpressed in the TP53mut tumors of at least 16 of the investigated 24 cancer types. In an association analysis of immune infiltration with TP53 mutations in 32 cancer subtypes, decreased immune infiltration was observed in six subtypes, increased infiltration in two subtypes, a mixed pattern of decreased and increased immune cell populations in four subtypes, while immune infiltration was not associated with TP53 status in 20 subtypes. The analysis of a large cohort of human tumors complements results from experimental studies and supports the view that TP53 mutations should be further evaluated as predictive markers for immunotherapy and targeted therapies.
Collapse
Affiliation(s)
- Eva Romanovsky
- Institute of Pathology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Klaus Kluck
- Institute of Pathology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Iordanis Ourailidis
- Institute of Pathology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Michael Menzel
- Institute of Pathology, Heidelberg University Hospital, 69120, Heidelberg, Germany
- Center for Personalized Medicine (ZPM) Heidelberg, 69120, Heidelberg, Germany
| | - Susanne Beck
- Institute of Pathology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Markus Ball
- Institute of Pathology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Daniel Kazdal
- Institute of Pathology, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) Heidelberg, member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Peter Schirmacher
- Institute of Pathology, Heidelberg University Hospital, 69120, Heidelberg, Germany
- Center for Personalized Medicine (ZPM) Heidelberg, 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg partner site, Heidelberg, Germany
| | - Thorsten Stiewe
- Institute of Molecular Oncology, member of the German Center for Lung Research (DZL), Philipps-University, 35037, Marburg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, Heidelberg University Hospital, 69120, Heidelberg, Germany
- Center for Personalized Medicine (ZPM) Heidelberg, 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg partner site, Heidelberg, Germany
| | - Jan Budczies
- Institute of Pathology, Heidelberg University Hospital, 69120, Heidelberg, Germany.
- Center for Personalized Medicine (ZPM) Heidelberg, 69120, Heidelberg, Germany.
- German Cancer Consortium (DKTK), Heidelberg partner site, Heidelberg, Germany.
| |
Collapse
|
13
|
Liang F, Luo Q, Han H, Zhang J, Yang Y, Chen J. Long noncoding RNA LINC01088 inhibits esophageal squamous cell carcinoma progression by targeting the NPM1-HDM2-p53 axis. Acta Biochim Biophys Sin (Shanghai) 2023; 55:367-381. [PMID: 36942988 PMCID: PMC10160232 DOI: 10.3724/abbs.2023021] [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: 02/18/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is characterized by extensive metastasis and poor prognosis. Long noncoding RNAs (lncRNAs) have been shown to play important roles in ESCC. However, the specific roles of lncRNAs in ESCC tumorigenesis and metastasis remain largely unknown. Here, we investigate LINC01088 in ESCC. Differentially expressed LINC01088 levels are screened from the GEO database. We find that LINC01088 is expressed at low level in collected clinical samples and is correlated with vascular tumor emboli and poor overall survival time of patients after surgery. LINC01088 inhibits not only ESCC cell migration and invasion in vitro, but also tumorigenesis and metastasis in vivo. Mechanistically, LINC01088 directly interacts with nucleophosmin (NPM1) and increases the expression of NPM1 in the nucleoplasm compared to that in the nucleolar region. LINC01088 decreases mutant p53 (mut-p53) expression and rescues the transcriptional activity of p53 by targeting the NPM1-HDM2-p53 axis. LINC01088 may also interfere with the DNA repair function of NPM1 by affecting its translocation. Our results highlight the potential of LINC01088 as a prognostic biomarker and therapeutic target of ESCC.
Collapse
Affiliation(s)
- Fan Liang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Qiuli Luo
- College of Life Science and Bioengineering, Beijing University of Technology, Beijing 100020, China
| | - Haibo Han
- Department of Clinical Laboratory, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jianzhi Zhang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yue Yang
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jinfeng Chen
- Department of Thoracic Surgery II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
14
|
Cabral ERM, Pacanhella MF, Lengert AVH, dos Reis MB, Leal LF, de Lima MA, da Silva ALV, Pinto IA, Reis RM, Pinto MT, Cárcano FM. Somatic mutation detection and KRAS amplification in testicular germ cell tumors. Front Oncol 2023; 13:1133363. [PMID: 37007070 PMCID: PMC10060882 DOI: 10.3389/fonc.2023.1133363] [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: 12/28/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Testicular Germ Cell Tumors (TGCT) are the most common cancer among young adult men. The TGCT histopathology is diverse, and the frequency of genomic alterations, along with their prognostic role, remains largely unexplored. Herein, we evaluate the mutation profile of a 15-driver gene panel and copy number variation of KRAS in a large series of TGCT from a single reference cancer center. MATERIALS AND METHODS A cohort of 97 patients with TGCT, diagnosed at the Barretos Cancer Hospital, was evaluated. Real-time PCR was used to assess copy number variation (CNV) of the KRAS gene in 51 cases, and the mutation analysis was performed using the TruSight Tumor 15 (Illumina) panel (TST15) in 65 patients. Univariate analysis was used to compare sample categories in relation to mutational frequencies. Survival analysis was conducted by the Kaplan-Meier method and log-rank test. RESULTS KRAS copy number gain was a very frequent event (80.4%) in TGCT and presented a worse prognosis compared with the group with no KRAS copy gain (10y-OS, 90% vs. 81.5%, p = 0.048). Among the 65 TGCT cases, different variants were identified in 11 of 15 genes of the panel, and the TP53 gene was the most recurrently mutated driver gene (27.7%). Variants were also detected in genes such as KIT, KRAS, PDGFRA, EGFR, BRAF, RET, NRAS, PIK3CA, MET, and ERBB2, with some of them potentially targetable. CONCLUSION Although larger studies incorporating collaborative networks may shed the light on the molecular landscape of TGCT, our findings unveal the potential of actionable variants in clinical management for applying targeted therapies.
Collapse
Affiliation(s)
| | | | - Andre V. H. Lengert
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - Mariana B. dos Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - Leticia F. Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
- Barretos School of Health Sciences Dr. Paulo Prata – FACISB, Barretos, Brazil
| | - Marcos A. de Lima
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | | | - Icaro A. Pinto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - Rui M. Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, Braga, Portugal
- 3ICVS/3B’s-PT Government Associate Laboratory, Braga, Portugal
| | - Mariana T. Pinto
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
| | - Flavio M. Cárcano
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, Brazil
- Division of Genitourinary Medical Oncology, Oncoclínicas, Belo Horizonte, Brazil
| |
Collapse
|
15
|
Asl ER, Rostamzadeh D, Duijf PHG, Mafi S, Mansoori B, Barati S, Cho WC, Mansoori B. Mutant P53 in the formation and progression of the tumor microenvironment: Friend or foe. Life Sci 2023; 315:121361. [PMID: 36608871 DOI: 10.1016/j.lfs.2022.121361] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 12/20/2022] [Accepted: 12/29/2022] [Indexed: 01/07/2023]
Abstract
TP53 is the most frequently mutated gene in human cancer. It encodes the tumor suppressor protein p53, which suppresses tumorigenesis by acting as a critical transcription factor that can induce the expression of many genes controlling a plethora of fundamental cellular processes, including cell cycle progression, survival, apoptosis, and DNA repair. Missense mutations are the most frequent type of mutations in the TP53 gene. While these can have variable effects, they typically impair p53 function in a dominant-negative manner, thereby altering intra-cellular signaling pathways and promoting cancer development. Additionally, it is becoming increasingly apparent that p53 mutations also have non-cell autonomous effects that influence the tumor microenvironment (TME). The TME is a complex and heterogeneous milieu composed of both malignant and non-malignant cells, including cancer-associated fibroblasts (CAFs), adipocytes, pericytes, different immune cell types, such as tumor-associated macrophages (TAMs) and T and B lymphocytes, as well as lymphatic and blood vessels and extracellular matrix (ECM). Recently, a large body of evidence has demonstrated that various types of p53 mutations directly affect TME. They fine-tune the inflammatory TME and cell fate reprogramming, which affect cancer progression. Notably, re-educating the p53 signaling pathway in the TME may be an effective therapeutic strategy in combating cancer. Therefore, it is timely to here review the recent advances in our understanding of how TP53 mutations impact the fate of cancer cells by reshaping the TME.
Collapse
Affiliation(s)
- Elmira Roshani Asl
- Department of Biochemistry, Saveh University of Medical Sciences, Saveh, Iran
| | - Davoud Rostamzadeh
- Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj, Iran; Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Pascal H G Duijf
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia; Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD, Australia; Centre for Data Science, Queensland University of Technology, Brisbane, QLD, Australia; Cancer and Aging Research Program, Queensland University of Technology, Brisbane, QLD, Australia; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Sahar Mafi
- Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj, Iran; Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Behnaz Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, Hong Kong
| | - Behzad Mansoori
- The Wistar Institute, Molecular & Cellular Oncogenesis Program, Philadelphia, PA, United States.
| |
Collapse
|
16
|
Bizzarri AR. Conformational Heterogeneity and Frustration of the Tumor Suppressor p53 as Tuned by Punctual Mutations. Int J Mol Sci 2022; 23:12636. [PMID: 36293489 PMCID: PMC9604312 DOI: 10.3390/ijms232012636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 12/02/2022] Open
Abstract
The conformational heterogeneity of the p53 tumor suppressor, the wild-type (p53wt) and mutated forms, was investigated by a computational approach, including the modeling and all atoms of the molecular dynamics (MD) simulations. Four different punctual mutations (p53R175H, p53R248Q, p53R273H, and p53R282W) which are known to affect the DNA binding and belong to the most frequent hot-spot mutations in human cancers, were taken into consideration. The MD trajectories of the wild-type and mutated p53 forms were analyzed by essential dynamics to extract the relevant collective motions and by the frustration method to evaluate the degeneracy of the energy landscape. We found that p53 is characterized by wide collective motions and its energy landscape exhibits a rather high frustration level, especially in the regions involved in the binding to physiological ligands. Punctual mutations give rise to a modulation of both the collective motions and the frustration of p53, with different effects depending on the mutation. The regions of p53wt and of the mutated forms characterized by a high frustration level are also largely involved in the collective motions. Such a correlation is discussed also in connection with the intrinsic disordered character of p53 and with its central functional role.
Collapse
Affiliation(s)
- Anna Rita Bizzarri
- Biophysics and Nanoscience Centre, DEB, Università della Tuscia, Largo dell'Università, 01100 Viterbo, Italy
| |
Collapse
|
17
|
Chantre-Justino M, Alves G, Delmonico L. Clinical applications of liquid biopsy in HPV‐negative and HPV‐positive head and neck squamous cell carcinoma: advances and challenges. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2022; 3:533-552. [PMID: 36071985 PMCID: PMC9446158 DOI: 10.37349/etat.2022.00099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/22/2022] [Indexed: 12/02/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) represent the most common epithelial tumors that arise from mucosa of the oral cavity, pharynx, and larynx. The development of HNSCCs is usually associated with tobacco use, alcohol consumption, and human papillomavirus (HPV) infection. Most HNSCCs are diagnosed in advanced states, leading to a worse clinical outcome. Screening tests based on potential biomarkers associated with HNSCCs could improve this scenario. Liquid biopsy has emerged as a promising area of cancer investigation, offering a minimally invasive approach to track circulating biomarkers in body fluids that could potentially contribute to the diagnosis, predict prognosis, and monitor response to treatment. This review will discuss translational studies describing the clinical applications of liquid biopsy in HPV-negative and HPV-positive HNSCCs focused on circulating nucleic acids [cell-free DNA (cfDNA) and cell-free RNA (cfRNA)], circulating tumor cells (CTCs), and extracellular vesicles (EVs), which can be found in plasma, serum, and saliva.
Collapse
Affiliation(s)
- Mariana Chantre-Justino
- 1Research Division, National Institute of Traumatology and Orthopaedics (INTO), Rio de Janeiro 20940-070, Brazil 2Circulating Biomarkers Laboratory, Pathology Department, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | - Gilda Alves
- 2Circulating Biomarkers Laboratory, Pathology Department, Faculty of Medical Sciences, Rio de Janeiro State University, Rio de Janeiro 20550-170, Brazil
| | - Lucas Delmonico
- 3Oncoclínicas Precision Medicine, Vila Nova Conceição, São Paulo 04513-020, Brazil
| |
Collapse
|
18
|
Long Q, Yuan Y, Li M. RNA-SSNV: A Reliable Somatic Single Nucleotide Variant Identification Framework for Bulk RNA-Seq Data. Front Genet 2022; 13:865313. [PMID: 35846154 PMCID: PMC9279659 DOI: 10.3389/fgene.2022.865313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 05/17/2022] [Indexed: 11/13/2022] Open
Abstract
The usage of expressed somatic mutations may have a unique advantage in identifying active cancer driver mutations. However, accurately calling mutations from RNA-seq data is difficult due to confounding factors such as RNA-editing, reverse transcription, and gap alignment. In the present study, we proposed a framework (named RNA-SSNV, https://github.com/pmglab/RNA-SSNV) to call somatic single nucleotide variants (SSNV) from tumor bulk RNA-seq data. Based on a comprehensive multi-filtering strategy and a machine-learning classification model trained with comprehensively curated features, RNA-SSNV achieved the best precision–recall rate (0.880–0.884) in a testing dataset and robustly retained 0.94 AUC for the precision–recall curve in three validation adult-based TCGA (The Cancer Genome Atlas) datasets. We further showed that the somatic mutations called by RNA-SSNV tended to have a higher functional impact and therapeutic power in known driver genes. Furthermore, VAF (variant allele fraction) analysis revealed that subclonal harboring expressed mutations had evolutional selection advantage and RNA had higher detection power to rescue DNA-omitted mutations. In sum, RNA-SSNV will be a useful approach to accurately call expressed somatic mutations for a more insightful analysis of cancer drive genes and carcinogenic mechanisms.
Collapse
Affiliation(s)
- Qihan Long
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Center for Precision Medicine, Sun Yat-Sen University, Guangzhou, China
- Center for Disease Genome Research, Sun Yat-Sen University, Guangzhou, China
| | - Yangyang Yuan
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Center for Precision Medicine, Sun Yat-Sen University, Guangzhou, China
- Center for Disease Genome Research, Sun Yat-Sen University, Guangzhou, China
| | - Miaoxin Li
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Center for Precision Medicine, Sun Yat-Sen University, Guangzhou, China
- Center for Disease Genome Research, Sun Yat-Sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Key Laboratory of Tropical Disease Control (SYSU), Ministry of Education, Guangzhou, China
- *Correspondence: Miaoxin Li,
| |
Collapse
|
19
|
Slabáková E, Kahounová Z, Procházková J, Souček K. Regulation of Neuroendocrine-like Differentiation in Prostate Cancer by Non-Coding RNAs. Noncoding RNA 2021; 7:ncrna7040075. [PMID: 34940756 PMCID: PMC8704250 DOI: 10.3390/ncrna7040075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 11/18/2021] [Accepted: 11/29/2021] [Indexed: 12/21/2022] Open
Abstract
Neuroendocrine prostate cancer (NEPC) represents a variant of prostate cancer that occurs in response to treatment resistance or, to a much lesser extent, de novo. Unravelling the molecular mechanisms behind transdifferentiation of cancer cells to neuroendocrine-like cancer cells is essential for development of new treatment opportunities. This review focuses on summarizing the role of small molecules, predominantly microRNAs, in this phenomenon. A published literature search was performed to identify microRNAs, which are reported and experimentally validated to modulate neuroendocrine markers and/or regulators and to affect the complex neuroendocrine phenotype. Next, available patients’ expression datasets were surveyed to identify deregulated microRNAs, and their effect on NEPC and prostate cancer progression is summarized. Finally, possibilities of miRNA detection and quantification in body fluids of prostate cancer patients and their possible use as liquid biopsy in prostate cancer monitoring are discussed. All the addressed clinical and experimental contexts point to an association of NEPC with upregulation of miR-375 and downregulation of miR-34a and miR-19b-3p. Together, this review provides an overview of different roles of non-coding RNAs in the emergence of neuroendocrine prostate cancer.
Collapse
|
20
|
Gomes AS, Ramos H, Inga A, Sousa E, Saraiva L. Structural and Drug Targeting Insights on Mutant p53. Cancers (Basel) 2021; 13:3344. [PMID: 34283062 PMCID: PMC8268744 DOI: 10.3390/cancers13133344] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/29/2021] [Accepted: 06/29/2021] [Indexed: 12/20/2022] Open
Abstract
p53 is a transcription factor with a pivotal role in cell homeostasis and fate. Its impairment is a major event in tumor onset and development. In fact, about half of human cancers bear TP53 mutations that not only halt the normal function of p53, but also may acquire oncogenic gain of functions that favor tumorigenesis. Although considered undruggable for a long time, evidence has proven the capability of many compounds to restore a wild-type (wt)-like function to mutant p53 (mutp53). However, they have not reached the clinic to date. Structural studies have strongly contributed to the knowledge about p53 structure, stability, dynamics, function, and regulation. Importantly, they have afforded relevant insights into wt and mutp53 pharmacology at molecular levels, fostering the design and development of p53-targeted anticancer therapies. Herein, we provide an integrated view of mutp53 regulation, particularly focusing on mutp53 structural traits and on targeting agents capable of its reactivation, including their biological, biochemical and biophysical features. With this, we expect to pave the way for the development of improved small molecules that may advance precision cancer therapy by targeting p53.
Collapse
Affiliation(s)
- Ana Sara Gomes
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal; (A.S.G.); (H.R.)
| | - Helena Ramos
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal; (A.S.G.); (H.R.)
| | - Alberto Inga
- Laboratory of Transcriptional Networks, Department CIBIO, University of Trento, Via Sommarive 9, 38123 Trento, Italy;
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal;
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, University of Porto, Novo Edifício do Terminal de Cruzeiros do Porto de Leixões, Avenida General Norton de Matos, S/N, 4450-208 Matosinhos, Portugal
| | - Lucília Saraiva
- LAQV/REQUIMTE, Laboratório de Microbiologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, 4050-313 Porto, Portugal; (A.S.G.); (H.R.)
| |
Collapse
|
21
|
Meng D, Guo K, Zhang D, Zhao C, Sun C, Zhang F. Ring finger 20/ring finger 40/WW domain-containing adaptor with coiled-coil complex interacts with p53 to regulate gene transcription in DNA damage response. Oncol Lett 2021; 21:436. [PMID: 33868474 PMCID: PMC8045150 DOI: 10.3892/ol.2021.12697] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 12/17/2020] [Indexed: 01/21/2023] Open
Abstract
p53 is one of the most important tumor suppressor genes, and its primary function is to act as a transcriptional activator to control cell cycle arrest, DNA repair and cellular metabolism by recognizing and binding to specific DNA sequences. Defects in the ring finger (RNF)20/RNF40/WW domain-containing adaptor with coiled-coil (WAC) complex, one of the histone H2B ubiquitination E3 ligases, have been reported to be a key factor in oncogenesis, cancer cell migration and invasion. Histone H2B mono-ubiquitination has been demonstrated to be essential for maintaining the functionality of the p53 tumor suppressor protein. The aim of the present study was to identify any sites in the p53 DNA-binding domain (DBD) specific to the RNF20/RNF40/WAC complex that may be involved in the gene regulation in DNA damage response. The results demonstrated that p53 and the RNF20/RNF40/WAC complex interacted with each other, and the coiled-coil regions in RNF20, RNF40 and WAC were identified to directly interact with p53. The R282 site in the p53 DBD, one of the frequent missense mutations associated with p53 mutation-dependent cancer, was demonstrated to be the key binding site for the RNF20/RNF40/WAC complex. Furthermore, knockout of RNF20/RNF40 suppressed the expression levels of p53 and its target genes in HCT116 cells compared with those in wild-type HCT116 cells. Consistent with these results, the R282W mutation in p53 inhibited the expression levels of p53 and its downstream genes by inactivating the interaction between p53 and RNF20/RNF40 compared with those in wild-type HCT116 cells. In conclusion, the results of the present study revealed the molecular mechanism of the interaction between the RNF20/RNF40/WAC complex and p53, and demonstrated that these proteins regulated gene transcription in the DNA damage response.
Collapse
Affiliation(s)
- Danni Meng
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Kun Guo
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Die Zhang
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Cheng Zhao
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Chuanwen Sun
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| | - Feng Zhang
- College of Life Sciences, Shanghai Normal University, Shanghai 200234, P.R. China
| |
Collapse
|
22
|
Shen E, Han Y, Cai C, Liu P, Chen Y, Gao L, Huang Q, Shen H, Zeng S, He M. Low expression of NLRP1 is associated with a poor prognosis and immune infiltration in lung adenocarcinoma patients. Aging (Albany NY) 2021; 13:7570-7588. [PMID: 33658393 PMCID: PMC7993699 DOI: 10.18632/aging.202620] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 12/19/2020] [Indexed: 02/06/2023]
Abstract
NLRP1 (NLR family, pyrin domain containing 1), the first NLR protein, described to form an inflammasome, plays critical roles in innate immunity and inflammation. However, NLRP1 has not been reported to be linked to LUAD (lung adenocarcinoma) risk, prognosis, immunotherapy or any other treatments. This research aimed to explore the prognostic value and mechanism of NLRP1 in LUAD. We performed bioinformatics analysis on LUAD data downloaded from TCGA (The Cancer Genome Atlas) and GEO (Gene Expression Omnibus), and jointly analyzed with online databases such as TCGAportal, LinkedOmics, TIMER, ESTIMATE and TISIDB. NLRP1 expression of LUAD tissue was considerably lower than that in normal lung tissue. Decreased NLRP1 expression of LUAD was associated with relatively high pathological, T and N stages. Kaplan-Meier survival analysis indicated that patients with low NLRP1 expression had a worse prognosis than those with high expression. Multivariate Cox analysis further showed that NLRP1 expression level was an independent prognostic factor of LUAD. Moreover, the level of NLRP1 expression was positively linked to the degree of infiltration of various TIICs (tumor-infiltrating immune cells). Our findings confirmed that reduced expression of NLRP1 was significantly related to poor prognosis and low degree of immune cell infiltration in LUAD patients.
Collapse
Affiliation(s)
- Edward Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,Department of Life Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Ying Han
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Changjing Cai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Ping Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Yihong Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Le Gao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Qiaoqiao Huang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Hong Shen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Shan Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Min He
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,Key Laboratory for Molecular Radiation Oncology of Hunan Province, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
23
|
Evaluating the Influence of a G-Quadruplex Prone Sequence on the Transactivation Potential by Wild-Type and/or Mutant P53 Family Proteins through a Yeast-Based Functional Assay. Genes (Basel) 2021; 12:genes12020277. [PMID: 33672023 PMCID: PMC7919268 DOI: 10.3390/genes12020277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 02/06/2023] Open
Abstract
P53, P63, and P73 proteins belong to the P53 family of transcription factors, sharing a common gene organization that, from the P1 and P2 promoters, produces two groups of mRNAs encoding proteins with different N-terminal regions; moreover, alternative splicing events at C-terminus further contribute to the generation of multiple isoforms. P53 family proteins can influence a plethora of cellular pathways mainly through the direct binding to specific DNA sequences known as response elements (REs), and the transactivation of the corresponding target genes. However, the transcriptional activation by P53 family members can be regulated at multiple levels, including the DNA topology at responsive promoters. Here, by using a yeast-based functional assay, we evaluated the influence that a G-quadruplex (G4) prone sequence adjacent to the p53 RE derived from the apoptotic PUMA target gene can exert on the transactivation potential of full-length and N-terminal truncated P53 family α isoforms (wild-type and mutant). Our results show that the presence of a G4 prone sequence upstream or downstream of the P53 RE leads to significant changes in the relative activity of P53 family proteins, emphasizing the potential role of structural DNA features as modifiers of P53 family functions at target promoter sites.
Collapse
|
24
|
Cruz O, Caloretti V, Salvador H, Celis V, Santa-Maria V, Morales La Madrid A, Suñol M, Puerta P, Muchart J, Krauel L, Lavarino C. Synchronous choroid plexus papilloma and Wilms tumor in a girl, disclosing a Li-Fraumeni syndrome. Hered Cancer Clin Pract 2021; 19:1. [PMID: 33407742 PMCID: PMC7789227 DOI: 10.1186/s13053-020-00158-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 12/11/2020] [Indexed: 11/26/2022] Open
Abstract
Background Li-Fraumeni Syndrome (LFS) is a cancer predisposition syndrome characterized by the early-onset of multiple primary cancers which can occur at different moments (metachronous onset) or, more rarely, coincidentally (synchronous onset). Here we describe a previously unreported patient with presentation of synchronous Wilms tumor and Choroid plexus papilloma, leading to the diagnosis of a Li-Fraumeni Syndrome (LFS). Case presentation A 6-year-old girl without previous complains presented with abdominal pain. Abdominal US and MRI showed a left renal tumor with subcapsular hematoma. Due to mild headaches, the diagnostic workup included a brain MRI that unexpectedly identified a large left parietal lobe tumor. Histopathological analysis determined the diagnosis of classic Wilms tumor and choroid-plexus papilloma (CPP), respectively. Both neoplasms showed intense nuclear p53 immunostaining associated with the pathogenic TP53 mutation c.844C > T (p.Arg282Trp). Our patient and her father shared the same heterozygous germline TP53 mutation, confirming the diagnosis of familiar Li-Fraumeni syndrome in the girl. The treatment was tailored to simultaneous tumor presentations. Conclusions LFS has been associated with Choroid plexus carcinoma (CPC), but rarely with CPP as in our patient. That suggests that it may be advisable to consider the possibility of analyzing TP53 mutation, not only in all patients with CPC, but also in some patients with CPP, especially when histological or clinical evidences point out to perform this study. The dissimilar presentation of LFS among our patient’s father, not having so far any neoplasia diagnosed, while her daughter presented precociously with two simultaneous different tumors, could be related to possible effects of modifier genes on the underlying mutant p53 genotype. Supplementary Information The online version contains supplementary material available at 10.1186/s13053-020-00158-7.
Collapse
Affiliation(s)
- Ofelia Cruz
- Department of Pediatric Oncology, Hospital Sant Joan de Déu, Esplugues de Llobregat, Passeig Sant Joan de Deu 2, 08950, Barcelona, Spain.
| | - Victoria Caloretti
- Department of Pediatric Oncology, Hospital Sant Joan de Déu, Esplugues de Llobregat, Passeig Sant Joan de Deu 2, 08950, Barcelona, Spain
| | - Hector Salvador
- Department of Pediatric Oncology, Hospital Sant Joan de Déu, Esplugues de Llobregat, Passeig Sant Joan de Deu 2, 08950, Barcelona, Spain
| | - Veronica Celis
- Department of Pediatric Oncology, Hospital Sant Joan de Déu, Esplugues de Llobregat, Passeig Sant Joan de Deu 2, 08950, Barcelona, Spain
| | - Vicente Santa-Maria
- Department of Pediatric Oncology, Hospital Sant Joan de Déu, Esplugues de Llobregat, Passeig Sant Joan de Deu 2, 08950, Barcelona, Spain
| | - Andrés Morales La Madrid
- Department of Pediatric Oncology, Hospital Sant Joan de Déu, Esplugues de Llobregat, Passeig Sant Joan de Deu 2, 08950, Barcelona, Spain
| | - Mariona Suñol
- Department of Pathology, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Patricia Puerta
- Department of Neurosurgery, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Jordi Muchart
- Department of Diagnostic Imaging, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Lucas Krauel
- Department of Surgery, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| | - Cinzia Lavarino
- Laboratory of Molecular Oncology, Hospital Sant Joan de Déu, Esplugues de Llobregat, Barcelona, Spain
| |
Collapse
|
25
|
Rogoża-Janiszewska E, Malińska K, Górski B, Scott RJ, Cybulski C, Kluźniak W, Lener M, Jakubowska A, Gronwald J, Huzarski T, Lubiński J, Dębniak T. Prevalence of germline TP53 variants among early-onset breast cancer patients from Polish population. Breast Cancer 2020; 28:226-235. [PMID: 32888145 PMCID: PMC7796867 DOI: 10.1007/s12282-020-01151-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 08/20/2020] [Indexed: 11/28/2022]
Abstract
Background The objective of this study was to determine spectrum and prevalence of germline mutations in TP53 gene among Polish women with early-onset breast cancer (BC), which has not been determined until now. Methods A cohort of 100 females with BC diagnosed ≤ 30 years of age and with a positive family history of cancer was used as a discovery cohort. 1880 women with BC ≤ 45 years old and a control group of 2000 healthy women were genotyped as a replication phase of this study. Results Four heterozygous pathogenic missense mutations were detected in a group of 100 patients with early-onset breast cancer. On the basis of software prediction and available literature data, all these variants were defined as pathogenic. None of these TP53 variants were detected among 1880 breast cancer patients and 2000 healthy controls. No large mutations were found among early-onset cases using MLPA reaction. Conclusion Germline pathogenic TP53 variants were found in 4% early-onset Polish BC patients. No founder mutations were identified in Polish population. To improve the treatment and surveillance screening, the search for germline TP53 pathogenic variants is recommended for all female BC cases diagnosed ≤ 30 years old. Electronic supplementary material The online version of this article (10.1007/s12282-020-01151-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Emilia Rogoża-Janiszewska
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland.
| | - Karolina Malińska
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Bohdan Górski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Rodney J Scott
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, NSW, Australia.,Department of Molecular Medicine, NSW Health Pathology-Hunter, Newcastle, NSW, Australia
| | - Cezary Cybulski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Wojciech Kluźniak
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Marcin Lener
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Anna Jakubowska
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Jacek Gronwald
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Tomasz Huzarski
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Jan Lubiński
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Tadeusz Dębniak
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
26
|
Nagam SL, Jinka R. Impact of TP53 polymorphisms (IVS3 + 40-41ins16, EX4 215G > C and IVS6 + 62G > A) on the risk of endometrial cancer, survival and hazard ratio of the patients. Meta Gene 2020. [DOI: 10.1016/j.mgene.2020.100684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
|
27
|
Madan E, Parker TM, Pelham CJ, Palma AM, Peixoto ML, Nagane M, Chandaria A, Tomás AR, Canas-Marques R, Henriques V, Galzerano A, Cabral-Teixeira J, Selvendiran K, Kuppusamy P, Carvalho C, Beltran A, Moreno E, Pati UK, Gogna R. HIF-transcribed p53 chaperones HIF-1α. Nucleic Acids Res 2019; 47:10212-10234. [PMID: 31538203 PMCID: PMC6821315 DOI: 10.1093/nar/gkz766] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/14/2019] [Accepted: 09/02/2019] [Indexed: 02/06/2023] Open
Abstract
Chronic hypoxia is associated with a variety of physiological conditions such as rheumatoid arthritis, ischemia/reperfusion injury, stroke, diabetic vasculopathy, epilepsy and cancer. At the molecular level, hypoxia manifests its effects via activation of HIF-dependent transcription. On the other hand, an important transcription factor p53, which controls a myriad of biological functions, is rendered transcriptionally inactive under hypoxic conditions. p53 and HIF-1α are known to share a mysterious relationship and play an ambiguous role in the regulation of hypoxia-induced cellular changes. Here we demonstrate a novel pathway where HIF-1α transcriptionally upregulates both WT and MT p53 by binding to five response elements in p53 promoter. In hypoxic cells, this HIF-1α-induced p53 is transcriptionally inefficient but is abundantly available for protein-protein interactions. Further, both WT and MT p53 proteins bind and chaperone HIF-1α to stabilize its binding at its downstream DNA response elements. This p53-induced chaperoning of HIF-1α increases synthesis of HIF-regulated genes and thus the efficiency of hypoxia-induced molecular changes. This basic biology finding has important implications not only in the design of anti-cancer strategies but also for other physiological conditions where hypoxia results in disease manifestation.
Collapse
Affiliation(s)
- Esha Madan
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Taylor M Parker
- Department of Surgery, Simon Cancer Research Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Christopher J Pelham
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, MO 63110, USA
| | - Antonio M Palma
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Maria L Peixoto
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Masaki Nagane
- Department of Biochemistry, School of Veterinary Medicine, Azabu University, 1-17-71 Fuchinobe, Chuo-ku, Sagamihara, Kanagawa, 252-5201, Japan
| | - Aliya Chandaria
- Biosciences unit, College of Life and Environmental Sciences, University of Exeter, Stocker Road Exeter EX4 4QD, UK
| | - Ana R Tomás
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | | | | | | | | | - Karuppaiyah Selvendiran
- Division of Gynecologic Oncology, Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Periannan Kuppusamy
- Department of Radiology and Medicine, 601 Rubin Building, Norris Cotton Cancer Center, Geisel School of Medicine, Dartmouth College, 1 Medical Center Drive, Lebanon, NH 03756, USA
| | - Carlos Carvalho
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Antonio Beltran
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Eduardo Moreno
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| | - Uttam K Pati
- Transcription and Human Biology Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rajan Gogna
- Champalimaud Centre for the Unknown, 1400-038 Lisbon, Portugal
| |
Collapse
|
28
|
Huang Y, Liu N, Liu J, Liu Y, Zhang C, Long S, Luo G, Zhang L, Zhang Y. Mutant p53 drives cancer chemotherapy resistance due to loss of function on activating transcription of PUMA. Cell Cycle 2019; 18:3442-3455. [PMID: 31726940 DOI: 10.1080/15384101.2019.1688951] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
P53 is a critical tumor suppressor gene, activating p53 and its downstream targets to induce apoptosis is a promising way for cancer therapy. However, more than 50% of cancer patients have p53 mutations, which may cause cancer therapy resistance, and the underline mechanism is poorly understood. Here, we found that cell viability decrease and apoptosis induced by p53-dependent traditional drugs in colon cancer cells were eliminated in p53 mutant cells. Mutant p53 did not up-regulate the expression of its direct downstream targets PUMA and p21, due to the inhibition of PUMA transcription. Furthermore, mutant p53 could not bind to the promoter of PUMA to activate its transcription like WT p53 did, while overexpressed WT p53 rescued PUMA-induced subsequent apoptosis. In conclusion, our findings demonstrate mutant p53 may cause chemo-resistance of tumor because of inactivating PUMA transcription, which prompts some new insights for clinical therapy of cancer patients with mutant p53.Abbreviations: CRC: Colorectal cancer; CDKs: Cyclin-dependent kinases; PUMA: p53 up-regulated modulator of apoptosis; PDGF: the platelet-derived growth factor; WT p53: wild-type p53 protein; mutp53: mutant p53 proteins; BAX: Bcl-2-associated X protein; NOXA: Phorbol-12-myristate-13-acetate-induced protein 1.
Collapse
Affiliation(s)
- Yuan Huang
- College of Biology, Hunan University, Changsha, China
| | - Nannan Liu
- College of Biology, Hunan University, Changsha, China
| | - Jing Liu
- College of Biology, Hunan University, Changsha, China
| | - Yeying Liu
- College of Biology, Hunan University, Changsha, China
| | - Chuchu Zhang
- College of Biology, Hunan University, Changsha, China
| | - Shuaiyu Long
- College of Biology, Hunan University, Changsha, China
| | - Guang Luo
- College of Biology, Hunan University, Changsha, China
| | - Lingling Zhang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yingjie Zhang
- College of Biology, Hunan University, Changsha, China.,Shenzhen Institute, Hunan University, Shenzhen, China
| |
Collapse
|
29
|
Krassowski M, Paczkowska M, Cullion K, Huang T, Dzneladze I, Ouellette BFF, Yamada JT, Fradet-Turcotte A, Reimand J. ActiveDriverDB: human disease mutations and genome variation in post-translational modification sites of proteins. Nucleic Acids Res 2019; 46:D901-D910. [PMID: 29126202 PMCID: PMC5753267 DOI: 10.1093/nar/gkx973] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 10/18/2017] [Indexed: 12/18/2022] Open
Abstract
Interpretation of genetic variation is needed for deciphering genotype-phenotype associations, mechanisms of inherited disease, and cancer driver mutations. Millions of single nucleotide variants (SNVs) in human genomes are known and thousands are associated with disease. An estimated 21% of disease-associated amino acid substitutions corresponding to missense SNVs are located in protein sites of post-translational modifications (PTMs), chemical modifications of amino acids that extend protein function. ActiveDriverDB is a comprehensive human proteo-genomics database that annotates disease mutations and population variants through the lens of PTMs. We integrated >385,000 published PTM sites with ∼3.6 million substitutions from The Cancer Genome Atlas (TCGA), the ClinVar database of disease genes, and human genome sequencing projects. The database includes site-specific interaction networks of proteins, upstream enzymes such as kinases, and drugs targeting these enzymes. We also predicted network-rewiring impact of mutations by analyzing gains and losses of kinase-bound sequence motifs. ActiveDriverDB provides detailed visualization, filtering, browsing and searching options for studying PTM-associated mutations. Users can upload mutation datasets interactively and use our application programming interface in pipelines. Integrative analysis of mutations and PTMs may help decipher molecular mechanisms of phenotypes and disease, as exemplified by case studies of TP53, BRCA2 and VHL. The open-source database is available at https://www.ActiveDriverDB.org.
Collapse
Affiliation(s)
- Michal Krassowski
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warsaw, Poland
| | - Marta Paczkowska
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Kim Cullion
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Tina Huang
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Irakli Dzneladze
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - B F Francis Ouellette
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Department of Cell and Systems Biology, University of Toronto, Toronto, Ontario, Canada
| | - Joseph T Yamada
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Amelie Fradet-Turcotte
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Québec, Canada
| | - Jüri Reimand
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Ekalaksananan T, Wongjampa W, Phusingha P, Chuerduangphui J, Vatanasapt P, Promthet S, Patarapadungkit N, Pientong C. Comprehensive Data of P53 R282 Gene Mutation with Human Papillomaviruses (HPV)-Associated Oral Squamous Cell Carcinoma (OSCC). Pathol Oncol Res 2019; 26:1191-1199. [DOI: 10.1007/s12253-019-00673-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/22/2019] [Indexed: 12/01/2022]
|
31
|
Luo X, Deng C, Liu F, Liu X, Lin T, He D, Wei G. HnRNPL promotes Wilms tumor progression by regulating the p53 and Bcl2 pathways. Onco Targets Ther 2019; 12:4269-4279. [PMID: 31213844 PMCID: PMC6549776 DOI: 10.2147/ott.s203046] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 04/16/2019] [Indexed: 12/17/2022] Open
Abstract
Background: Wilms tumor (WT) is the most common renal tumor in children with diffusely anaplastic or unfavorable histology, indicative of a poor prognosis. Heterogeneous nuclear ribonucleoprotein L (hnRNPL) is an RNA-binding protein (RBP) and a regulator of alternative RNA splicing that plays an important role in the occurrence and development of several cancers. Methods: Next generation sequencing technologies was used to discovery differentially expressed genes between WT and adjacent nontumors. The gene ontology (GO) analysis was performed to uncover the biological functions of differentially expressed genes, and the kyoto encyclopedia of genes and genomes (KEGG) pathway enrichment analysis was applied to find out the related signal pathways. Expression levelsof hnRNPL with WT tissues and cells were determined by RT-qPCR.After silencing hnRNPL, the expression of hnRNPL, p53 and Bcl-2 were detected by RT-qPCR and Western blot in WT cell line. The regulatory effects of hnRNPLon proliferative and apoptotic potentials of WT cells were evaluated by MTT and flow cytometry, respectively. RNA-binding protein immuno-precipitation was used to confirm the direct interaction of hnRNPL with p53 mRNA. Mouse xenograft models ofhnRNPL knockdown were established to test the functions in the growth of WT in vivo. Results: High levels of hnRNPL were expressed in WT tissues and cells. Functional analysis revealed that hnRNPL silencing suppressed cell proliferation and promoted cell apoptosis in WT. Molecular mechanism exploration indicated that hnRNPL directly targeted p53. Moreover, knockdown of hnRNPL inhibited the expression of p53 and Bcl2 in WT. Additionally, hnRNPL silencing inhibited the growth of xenograft tumors in vivo. Conclusion: HnRNPL act as p53 mRNA-binding protein, which plays an important role in the proliferation and apoptosis of WT through p53 and Bcl2 pathways and these findings provide new insights into the mechanism of WT pathogenesis.
Collapse
Affiliation(s)
- Xin Luo
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, People's Republic of China
| | - Changkai Deng
- Department of Pediatric Surgery, Chengdu Women and Children's Central Hospital, Chengdu 610073, People's Republic of China
| | - Feng Liu
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, People's Republic of China.,Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Chongqing 400014, People's Republic of China
| | - Xing Liu
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, People's Republic of China.,Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Chongqing 400014, People's Republic of China
| | - Tao Lin
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, People's Republic of China.,Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Chongqing 400014, People's Republic of China
| | - Dawei He
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, People's Republic of China.,Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Chongqing 400014, People's Republic of China
| | - Guanghui Wei
- Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing Key Laboratory of Children Urogenital Development and Tissue Engineering, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Chongqing 400014, People's Republic of China.,Department of Pediatric Urology Surgery, Children's Hospital of Chongqing Medical University, Chongqing 400014, People's Republic of China
| |
Collapse
|
32
|
Candido S, Abrams SL, Steelman LS, Lertpiriyapong K, Martelli AM, Cocco L, Ratti S, Follo MY, Murata RM, Rosalen PL, Bueno-Silva B, de Alencar SM, Lombardi P, Mao W, Montalto G, Cervello M, Rakus D, Gizak A, Lin HL, Libra M, Akula SM, McCubrey JA. Effects of the MDM-2 inhibitor Nutlin-3a on PDAC cells containing and lacking WT-TP53 on sensitivity to chemotherapy, signal transduction inhibitors and nutraceuticals. Adv Biol Regul 2019; 72:22-40. [PMID: 30898612 DOI: 10.1016/j.jbior.2019.03.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 03/09/2019] [Accepted: 03/11/2019] [Indexed: 06/09/2023]
Abstract
Mutations at the TP53 gene are readily detected (approximately 50-75%) in pancreatic ductal adenocarcinoma (PDAC) patients. TP53 was previously thought to be a difficult target as it is often mutated, deleted or inactivated on both chromosomes in certain cancers. In the following study, the effects of restoration of wild-type (WT) TP53 activity on the sensitivities of MIA-PaCa-2 pancreatic cancer cells to the MDM2 inhibitor nutlin-3a in combination with chemotherapy, targeted therapy, as well as, nutraceuticals were examined. Upon introduction of the WT-TP53 gene into MIA-PaCa-2 cells, which contain a TP53 gain of function (GOF) mutation, the sensitivity to the MDM2 inhibitor increased. However, effects of nutlin-3a were also observed in MIA-PaCa-2 cells lacking WT-TP53, as upon co-treatment with nutlin-3a, the sensitivity to certain inhibitors, chemotherapeutic drugs and nutraceuticals increased. Interestingly, co-treatment with nutlin-3a and certain chemotherapeutic drug such as irinotecan and oxaliplatin resulted in antagonistic effects in cells both lacking and containing WT-TP53 activity. These studies indicate the sensitizing abilities that WT-TP53 activity can have in PDAC cells which normally lack WT-TP53, as well as, the effects that the MDM2 inhibitor nutlin-3a can have in both cells containing and lacking WT-TP53 to various therapeutic agents.
Collapse
Affiliation(s)
- Saverio Candido
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy; Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy
| | - Stephen L Abrams
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA, 27834
| | - Linda S Steelman
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA, 27834
| | - Kvin Lertpiriyapong
- Weill Cornell Medicine and the Hospital for Special Surgery, New York City, New York, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Matilde Y Follo
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Ramiro M Murata
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy; Department of Foundational Sciences, School of Dental Medicine, East Carolina University, USA
| | - Pedro L Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Bruno Bueno-Silva
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil; Dental Research Division, Guarulhos University, Guarulhos, Brazil
| | | | - Paolo Lombardi
- Naxospharma, Via Giuseppe Di Vittorio 70, Novate Milanese, 20026, Italy
| | - Weifeng Mao
- College of Basic Medical Sciences, Dalian Medical University, Dalian, Liaoning, China
| | - Giuseppe Montalto
- Dipartimento di Promozione Della Salute, Materno-Infantile, Medicina Interna e Specialistica di Eccellenza (PROMISE), University of Palermo, Palermo, Italy; Consiglio Nazionale Delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale Delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw, Poland
| | - Agnieska Gizak
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, Wroclaw, Poland
| | - Heng-Liang Lin
- Catholic Fu Jen University Hospital, New Taipei City, Taiwan
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy; Research Center for Prevention, Diagnosis and Treatment of Cancer (PreDiCT), University of Catania, Catania, Italy
| | - Shaw M Akula
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA, 27834.
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA, 27834.
| |
Collapse
|
33
|
Cohen J, Vo NTK, Seymour CB, Mothersill CE. Parallel comparison of pre-conditioning and post-conditioning effects in human cancers and keratinocytes upon acute gamma irradiation. Int J Radiat Biol 2019; 95:170-178. [PMID: 30496014 DOI: 10.1080/09553002.2019.1547850] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE To determine and compare the effects of pre-conditioning and post-conditioning towards gamma radiation responses in human cancer cells and keratinocytes. MATERIAL AND METHODS The clonogenic survival of glioblastoma cells (T98G), keratinocytes (HaCaT), and colorectal carcinoma cells (HCT116 p53+/+ and p53-/-) was assessed following gamma ray exposure from a Cs-137 source. The priming dose preceded the challenge dose in pre-conditioning whereas the priming dose followed the challenge dose in post-conditioning. The priming dose was either 5 mGy or 0.1 Gy. The challenge dose was 0.5-5 Gy. RESULTS In both pre- and post-conditioning where the priming dose was 0.1 Gy and the challenge dose was 4 Gy, RAR developed in T98G but not in HaCaT cells. In HCT116 p53+/+, pre-conditioning had either no effect or a radiosensitizing effect and whereas post-conditioning induced either radiosensitizing or radioadaptive effect. The different observed outcomes were dependent on dose, the time interval between the priming and challenge dose, and the time before the first irradiation. Post-conditioning effects could occur with a priming dose as low as 5 mGy in HCT116 p53+/+ cells. When HCT116 cells had no p53 protein expression, the radiosensitizing or radioadaptive response by the conditioning effect was abolished. CONCLUSIONS The results suggest that radiation conditioning responses are complex and depend on at least the following factors: the magnitude of priming/challenge dose, the time interval between priming and challenge dose, p53 status, cell seeding time prior to the first radiation treatment. This work is the first parallel comparison demonstrating the potential outcomes of pre- and post-conditioning in different human cell types using environmentally and medically relevant radiation doses.
Collapse
Affiliation(s)
- Jason Cohen
- a Radiation Sciences , McMaster University , Hamilton , Canada
| | - Nguyen T K Vo
- b Department of Biology , McMaster University , Hamilton , Canada
| | - Colin B Seymour
- b Department of Biology , McMaster University , Hamilton , Canada
| | | |
Collapse
|
34
|
Fischer NW, Prodeus A, Gariépy J. Survival in males with glioma and gastric adenocarcinoma correlates with mutant p53 residual transcriptional activity. JCI Insight 2018; 3:121364. [PMID: 30089713 DOI: 10.1172/jci.insight.121364] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 06/21/2018] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND There is currently no clinical distinction between different TP53 mutations, despite increasing evidence that not all mutations have equally deleterious effects on the activity of the encoded tumor suppressor protein p53. The objective of this study was to determine whether these biological differences have clinical significance. METHODS This retrospective cohort analysis included 2,074 patients with sporadic TP53 mutations (403 unique mutations) and 1,049 germline TP53 mutation carriers (188 unique mutations). Survival was projected by stratifying patients according to their p53 mutant-specific residual transcriptional activity scores. RESULTS Pan-cancer survival analyses revealed a strong association between increased mutant p53 residual activity and improved survival in males with glioma and gastric adenocarcinoma (P = 0.002 and P = 0.02) that was not present in the female cohorts (P = 0.16 and P = 0.50). Male glioma and gastric cancer patients with TP53 mutations resulting in >5% transcriptional activity had 3.1-fold (95% CI, 2.4-3.8; P = 0.002; multivariate analysis hazard ratio [HR]) and 4.6-fold (95% CI, 3.7-5.6; P = 0.001; multivariate analysis HR) lower risk of death as compared with patients harboring inactive (0% activity) p53 mutants. The correlation between mutant p53 residual activity with survival was recapitulated in the dataset of germline TP53 mutation carriers (HR = 3.0, 95% CI, 2.7-3.4, P < 0.001 [females]; HR = 2.2, 95% CI, 1.8-2.6, P < 0.001 [males]), where brain and gastric tumors were more common among males (P < 0.001 and P = 0.001, respectively). CONCLUSION The retention of mutant p53 transcriptional activity prognosticates superior survival for men with glioma and gastric adenocarcinoma harboring sporadic TP53 mutations. Among germline TP53 mutation carriers, increased residual transcriptional activity is correlated with prolonged lifetime cancer survival and delayed tumor onset, and males are more prone to develop brain and gastric tumors. FUNDING Canadian Institutes of Health Research (no. 148556).
Collapse
Affiliation(s)
- Nicholas W Fischer
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Aaron Prodeus
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Jean Gariépy
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
35
|
Abrams SL, Lertpiriyapong K, Yang LV, Martelli AM, Cocco L, Ratti S, Falasca M, Murata RM, Rosalen PL, Lombardi P, Libra M, Candido S, Montalto G, Cervello M, Steelman LS, McCubrey JA. Introduction of WT-TP53 into pancreatic cancer cells alters sensitivity to chemotherapeutic drugs, targeted therapeutics and nutraceuticals. Adv Biol Regul 2018; 69:16-34. [PMID: 29980405 DOI: 10.1016/j.jbior.2018.06.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 06/20/2018] [Accepted: 06/26/2018] [Indexed: 06/08/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive, highly metastatic malignancy and accounts for 85% of pancreatic cancers. PDAC patients have poor prognosis with a five-year survival of only 5-10%. Mutations at the TP53 gene are readily detected in pancreatic tumors isolated from PDAC patients. We have investigated the effects of restoration of wild-type (WT) TP53 activity on the sensitivity of pancreatic cancer cells to: chemotherapy, targeted therapy, as well as, nutraceuticals. Upon introduction of the WT-TP53 gene into the MIA-PaCa-2 pancreatic cancer cell line, the sensitivity to drugs used to treat pancreatic cancer cells such as: gemcitabine, fluorouracil (5FU), cisplatin, irinotecan, oxaliplatin, and paclitaxel increased significantly. Likewise, the sensitivity to drugs used to treat other cancers such as: doxorubicin, mitoxantrone, and 4 hydroxy tamoxifen (4HT) also increased upon introduction of WT-TP53 into MIA-PaCa-2 cells. Furthermore, the sensitivity to certain inhibitors which target: PI3K/mTORC1, PDK1, SRC, GSK-3, and biochemical processes such as proteasomal degradation and the nutraceutical berberine as increased upon introduction of WT-TP53. Furthermore, in some cases, cells with WT-TP53 were more sensitive to the combination of drugs and suboptimal doses of the MDM2 inhibitor nutlin-3a. However, TP53-independent effects of nutlin-3a were observed upon treatment with either a proteasomal or a PI3K/mTOR inhibitor. These studies indicate the sensitizing effects that WT-TP53 can have in PDAC cells which normally lack WT-TP53 to various therapeutic agents and suggest approaches to improve PDAC therapy.
Collapse
Affiliation(s)
- Stephen L Abrams
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Kvin Lertpiriyapong
- Department of Comparative Medicine, Brody School of Medicine at East Carolina University, USA
| | - Li V Yang
- Department of Internal Medicine, Hematology/Oncology Section, Brody School of Medicine at East Carolina University, USA
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Lucio Cocco
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Stefano Ratti
- Department of Biomedical and Neuromotor Sciences, Università di Bologna, Bologna, Italy
| | - Marco Falasca
- Metabolic Signalling Group, School of Pharmacy & Biomedical Sciences, Curtin Health Innovation Research Institute, Faculty of Health Sciences, Curtin University, Perth, Western Australia 6102, Australia
| | - Ramiro M Murata
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA; Department of Foundational Sciences, School of Dental Medicine, East Carolina University, USA
| | - Pedro L Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, State University of Campinas, Piracicaba, Brazil
| | - Paolo Lombardi
- Naxospharma, Via Giuseppe Di Vittorio 70, Novate Milanese 20026, Italy; Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences - Pathology & Oncology Section, University of Catania, Catania, Italy
| | - Saverio Candido
- Department of Biomedical and Biotechnological Sciences - Pathology & Oncology Section, University of Catania, Catania, Italy
| | - Giuseppe Montalto
- Biomedical Department of Internal Medicine and Specialties, University of Palermo, Palermo, Italy; Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Melchiorre Cervello
- Consiglio Nazionale delle Ricerche, Istituto di Biomedicina e Immunologia Molecolare "Alberto Monroy", Palermo, Italy
| | - Linda S Steelman
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - James A McCubrey
- Department of Microbiology & Immunology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|
36
|
Tornesello ML, Annunziata C, Tornesello AL, Buonaguro L, Buonaguro FM. Human Oncoviruses and p53 Tumor Suppressor Pathway Deregulation at the Origin of Human Cancers. Cancers (Basel) 2018; 10:213. [PMID: 29932446 PMCID: PMC6071257 DOI: 10.3390/cancers10070213] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 06/19/2018] [Accepted: 06/22/2018] [Indexed: 12/18/2022] Open
Abstract
Viral oncogenesis is a multistep process largely depending on the complex interplay between viruses and host factors. The oncoviruses are capable of subverting the cell signaling machinery and metabolic pathways and exploit them for infection, replication, and persistence. Several viral oncoproteins are able to functionally inactivate the tumor suppressor p53, causing deregulated expression of many genes orchestrated by p53, such as those involved in apoptosis, DNA stability, and cell proliferation. The Epstein⁻Barr virus (EBV) BZLF1, the high-risk human papillomavirus (HPV) E6, and the hepatitis C virus (HCV) NS5 proteins have shown to directly bind to and degrade p53. The hepatitis B virus (HBV) HBx and the human T cell lymphotropic virus-1 (HTLV-1) Tax proteins inhibit p53 activity through the modulation of p300/CBP nuclear factors, while the Kaposi's sarcoma herpesvirus (HHV8) LANA, vIRF-1 and vIRF-3 proteins have been shown to destabilize the oncosuppressor, causing a decrease in its levels in the infected cells. The large T antigen of the Merkel cell polyomavirus (MCPyV) does not bind to p53 but significantly reduces p53-dependent transcription. This review describes the main molecular mechanisms involved in the interaction between viral oncoproteins and p53-related pathways as well as in the development of therapeutic strategies targeting such interactions.
Collapse
Affiliation(s)
- Maria Lina Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", via Mariano Semmola, 80131 Napoli, Italy.
| | - Clorinda Annunziata
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", via Mariano Semmola, 80131 Napoli, Italy.
| | - Anna Lucia Tornesello
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", via Mariano Semmola, 80131 Napoli, Italy.
| | - Luigi Buonaguro
- Cancer Immunomodulation Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", via Mariano Semmola, 80131 Napoli, Italy.
| | - Franco Maria Buonaguro
- Molecular Biology and Viral Oncology Unit, Istituto Nazionale Tumori IRCCS "Fondazione G. Pascale", via Mariano Semmola, 80131 Napoli, Italy.
| |
Collapse
|
37
|
LSD1 Inhibition Prolongs Survival in Mouse Models of MPN by Selectively Targeting the Disease Clone. Hemasphere 2018; 2:e54. [PMID: 31723778 PMCID: PMC6745991 DOI: 10.1097/hs9.0000000000000054] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 04/27/2018] [Indexed: 12/27/2022] Open
Abstract
Supplemental Digital Content is available in the text Despite recent advances, the myeloproliferative neoplasms (MPNs) are attended by considerable morbidity and mortality. Janus kinase (Jak) inhibitors such as ruxolitinib manage symptoms but do not substantially change the natural history of the disease. In this report, we show the effects of IMG-7289, an irreversible inhibitor of the epigenetically active lysine-specific demethylase 1 (LSD1) in mouse models of MPN. Once-daily treatment with IMG-7289 normalized or improved blood cell counts, reduced spleen volumes, restored normal splenic architecture, and reduced bone marrow fibrosis. Most importantly, LSD1 inhibition lowered mutant allele burden and improved survival. IMG-7289 selectively inhibited proliferation and induced apoptosis of JAK2V617F cells by concomitantly increasing expression and methylation of p53, and, independently, the pro-apoptotic factor PUMA and by decreasing the levels of its antiapoptotic antagonist BCLXL. These data provide a molecular understanding of the disease-modifying activity of the LSD1 inhibitor IMG-7289 that is currently undergoing clinical evaluation in patients with high-risk myelofibrosis. Moreover, low doses of IMG-7289 and ruxolitinib synergize in normalizing the MPN phenotype in mice, offering a rationale for investigating combination therapy.
Collapse
|
38
|
Stiewe T, Haran TE. How mutations shape p53 interactions with the genome to promote tumorigenesis and drug resistance. Drug Resist Updat 2018; 38:27-43. [PMID: 29857816 DOI: 10.1016/j.drup.2018.05.001] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/27/2018] [Accepted: 05/03/2018] [Indexed: 12/31/2022]
Abstract
The tumor suppressive transcription factor p53 regulates a wide array of cellular processes that confer upon cells an essential protection against cancer development. Wild-type p53 regulates gene expression by directly binding to DNA in a sequence-specific manner. p53 missense mutations are the most common mutations in malignant cells and can be regarded as synonymous with anticancer drug resistance and poor prognosis. The current review provides an overview of how the extraordinary variety of more than 2000 different mutant p53 proteins, known as the p53 mutome, affect the interaction of p53 with DNA. We discuss how the classification of p53 mutations to loss of function (LOF), gain of function (GOF), and dominant-negative (DN) inhibition of a remaining wild-type allele, hides a complex p53 mutation spectrum that depends on the distinctive nature of each mutant protein, requiring different therapeutic strategies for each mutant p53 protein. We propose to regard the different mutant p53 categories as continuous variables, that may not be independent of each other. In particular, we suggest here to consider GOF mutations as a special subset of LOF mutations, especially when mutant p53 binds to DNA through cooperation with other transcription factors, and we present a model for GOF mechanism that consolidates many observations on the GOF phenomenon. We review how novel mutant p53 targeting approaches aim to restore a wild-type-like DNA interaction and to overcome resistance to cancer therapy.
Collapse
Affiliation(s)
- Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, 35037 Marburg, Germany.
| | - Tali E Haran
- Department of Biology, Technion-Israel Institute of Technology, Technion City, Haifa 32000, Israel.
| |
Collapse
|
39
|
Romano FJ, Rossetti S, Conteduca V, Schepisi G, Cavaliere C, Di Franco R, La Mantia E, Castaldo L, Nocerino F, Ametrano G, Cappuccio F, Malzone G, Montanari M, Vanacore D, Quagliariello V, Piscitelli R, Pepe MF, Berretta M, D'Aniello C, Perdonà S, Muto P, Botti G, Ciliberto G, Veneziani BM, De Falco F, Maiolino P, Caraglia M, Montella M, De Giorgi U, Facchini G. Role of DNA repair machinery and p53 in the testicular germ cell cancer: a review. Oncotarget 2018; 7:85641-85649. [PMID: 27821802 PMCID: PMC5356765 DOI: 10.18632/oncotarget.13063] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/19/2016] [Indexed: 12/21/2022] Open
Abstract
Notwithstanding the peculiar sensitivity to cisplatin-based treatment, resulting in a very high percentage of cures even in advanced stages of the disease, still we do not know the biological mechanisms that make Testicular Germ Cell Tumor (TGCT) “unique” in the oncology scene. p53 and MDM2 seem to play a pivotal role, according to several in vitro observations, but no correlation has been found between their mutational or expression status in tissue samples and patients clinical outcome. Furthermore, other players seem to be on stage: DNA Damage Repair Machinery (DDR) , especially Homologous Recombination (HR) proteins, above all Ataxia Telangiectasia Mutated (ATM), cooperates with p53 in response to DNA damage, activating apoptotic cascade and contributing to cell “fate”. Homologous Recombination deficiency has been assumed to be a Germ Cell Tumor characteristic underlying platinum-sensitivity, whereby Poly(ADP-ribose) polymerase (PARP), an enzyme involved in HR DNA repair, is an intriguing target: PARP inhibitors have already entered in clinical practice of other malignancies and trials are recruiting TGCT patients in order to validate their role in this disease. This paper aims to summarize evidence, trying to outline an overview of DDR implications not only in TGCT curability, but also in resistance to chemotherapy.
Collapse
Affiliation(s)
- Francesco Jacopo Romano
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy
| | - Sabrina Rossetti
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Naples, Italy
| | - Vincenza Conteduca
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy
| | - Giuseppe Schepisi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy
| | - Carla Cavaliere
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Department of Onco-Ematology Medical Oncology, S.G. Moscati Hospital of Taranto, Taranto, Italy
| | - Rossella Di Franco
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Elvira La Mantia
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - Luigi Castaldo
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Department of Uro-Gynaecological Oncology, Division of Urology, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Naples, Italy
| | - Flavia Nocerino
- Epidemiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Gianluca Ametrano
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Francesca Cappuccio
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Psicology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Gabriella Malzone
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - Micaela Montanari
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Daniela Vanacore
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy
| | - Vincenzo Quagliariello
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy
| | - Raffaele Piscitelli
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Pharmacy Unit, Istituto Nazionale Tumori, Istituto Nazionale Tumori-Fondazione G. Pascale Naples, Italy
| | - Maria Filomena Pepe
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - Massimiliano Berretta
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, Aviano, Italy
| | - Carmine D'Aniello
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Division of Medical Oncology, A.O.R.N. dei COLLI "Ospedali Monaldi-Cotugno-CTO", Napoli, Italy
| | - Sisto Perdonà
- Department of Uro-Gynaecological Oncology, Division of Urology, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Naples, Italy
| | - Paolo Muto
- Radiation Oncology, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Gerardo Botti
- Pathology Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - Gennaro Ciliberto
- Scientific Directorate, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Bianca Maria Veneziani
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples "Federico II", Naples, Italy
| | - Francesco De Falco
- Psicology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Piera Maiolino
- Pharmacy Unit, Istituto Nazionale Tumori, Istituto Nazionale Tumori-Fondazione G. Pascale Naples, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy
| | - Maurizio Montella
- Epidemiology Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori 'Fondazione Giovanni Pascale', IRCCS, Napoli, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori IRCCS, Meldola, Italy
| | - Gaetano Facchini
- Progetto ONCONET2.0, Linea Progettuale 14 per L'implementazione della Prevenzione e Diagnosi Precoce del Tumore alla Prostata e Testicolo, Regione Campania, Italy.,Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori 'Fondazione G. Pascale', IRCCS, Naples, Italy
| |
Collapse
|