1
|
Stevers NO, Kim S, Yuan JB, Barger CJ, Hong C, Lenzo O, McKinney AM, Wu SH, Lee YJ, Kwok DW, Suwala AK, Appin CL, Gordan JD, Molinaro AM, Song JS, Costello JF. The immortality mechanism of TERT promoter mutant cancers is self-reinforcing and reversible. Mol Cell 2025:S1097-2765(25)00465-4. [PMID: 40513567 DOI: 10.1016/j.molcel.2025.05.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 03/26/2025] [Accepted: 05/21/2025] [Indexed: 06/16/2025]
Abstract
Activating mutations in the telomerase reverse transcriptase (TERT) promoter are prevalent in cancer and enable limitless cell division characteristic of immortal cells. Solving the immortality mechanism represents a major step toward selective reversal in cancer cells. TERT promoter (TERTp) mutations create a de novo E26 transformation-specific (ETS) transcription factor binding motif. Here, we analyzed 53 cell lines representing 16 cancer types and 6 recurrent TERTp mutations and found that the GA-binding protein (GABP) tetramer is responsible for promoter activation in all cases. Surprisingly, TERT expression is maintained after tetramer depletion. Further investigation revealed an underlying network of auto-suppression among the GABP subunits. Release from it drives TERT maintenance via upregulated GABP dimers or a paralogous tetramer. The GABPB1L tetramer is therefore a pan-cancer, pan-mutation activator of the mutant TERT promoter, but it is replaceable. Domains shared by the three GABP complexes, rather than solely the B1L tetramer, are mutation-specific vulnerabilities.
Collapse
Affiliation(s)
- Nicholas O Stevers
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Somang Kim
- Department of Physics, University of Illinois, Urbana-Champaign, Champaign, IL, USA; Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Champaign, IL, USA; Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Champaign, IL, USA
| | - Jimmy Bo Yuan
- Department of Physics, University of Illinois, Urbana-Champaign, Champaign, IL, USA; Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Champaign, IL, USA; Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Champaign, IL, USA
| | - Carter J Barger
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Chibo Hong
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Olivia Lenzo
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Andrew M McKinney
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Samuel H Wu
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Yu Jin Lee
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Darwin W Kwok
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Abigail K Suwala
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Christina L Appin
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - John D Gordan
- Quantitative Biosciences Institute, University of California, San Francisco, San Francisco, CA, USA; Department of Medicine, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA; Division of Hematology/Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Annette M Molinaro
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA; Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Jun S Song
- Department of Physics, University of Illinois, Urbana-Champaign, Champaign, IL, USA; Institute for Genomic Biology, University of Illinois, Urbana-Champaign, Champaign, IL, USA; Cancer Center at Illinois, University of Illinois, Urbana-Champaign, Champaign, IL, USA.
| | - Joseph F Costello
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Rittavee Y, Ratanaprasert N, Ahmed S, Anekpuritanang T, Muanprasat C, Pongsapich W, Jinawath N. Biomarkers for predicting second primary malignancy risk in head and neck squamous cell carcinoma: An integrated molecular perspective. Crit Rev Oncol Hematol 2025; 210:104711. [PMID: 40157583 DOI: 10.1016/j.critrevonc.2025.104711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Second primary malignancies (SPMs) threaten long-term survival in head and neck squamous cell carcinoma (HNSCC). Advancing our understanding of the molecular events driving these secondary tumors is essential. This review has explicated molecular drivers of SPM development, including epigenetic alterations such as DNA hypermethylation, genetic polymorphisms affecting detoxification pathways, and shifts in gene and protein expression profiles. Disruptions in the p53 signaling pathway, immune-related pathways, and impairments in glutathione S-transferase-mediated detoxification, emerge as central contributors to SPM risk. Additionally, direct comparisons of tumor specimens with adjacent or distant normal mucosa highlight field cancerization biomarkers, underscoring widespread carcinogen-induced damage. Loss of heterozygosity at chromosome arm 13q, p53 overexpression in tumor-distant epithelia, and proteomic abnormalities in ostensibly healthy mucosa collectively promote a tumor-prone field that encourages the formation of independent secondary tumors. This interplay underscores a multifactorial landscape of SPM pathogenesis, involving genetic susceptibility, environmental exposures, and intricate epigenetic and transcriptomic networks. By recognizing and validating reliable biomarkers, clinicians may pinpoint high-risk patients with greater precision, intervene earlier, and customize follow-up protocols and treatment regimens. Ultimately, translating these insights into routine practice promises a more proactive, individualized approach to preventing SPMs in HNSCC.
Collapse
Affiliation(s)
- Yutthana Rittavee
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | - Narin Ratanaprasert
- Department of Otorhinolaryngology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Thailand
| | - Saad Ahmed
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | | | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand
| | - Warut Pongsapich
- Department of Otorhinolaryngology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Thailand
| | - Natini Jinawath
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand; Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Thailand; Integrative Computational Bioscience (ICBS) Center, Mahidol University, Nakhon Pathom, Thailand.
| |
Collapse
|
3
|
Pepe D, Janssens X, Timcheva K, Marrón-Liñares GM, Verbelen B, Konstantakos V, De Groote D, De Bie J, Verhasselt A, Dewaele B, Godderis A, Cools C, Franco-Tolsau M, Royaert J, Verbeeck J, Kampen KR, Subramanian K, Cabrerizo Granados D, Menschaert G, De Keersmaecker K. Reannotation of cancer mutations based on expressed RNA transcripts reveals functional non-coding mutations in melanoma. Am J Hum Genet 2025:S0002-9297(25)00146-6. [PMID: 40359938 DOI: 10.1016/j.ajhg.2025.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
The role of synonymous mutations in cancer pathogenesis is currently underexplored. We developed a method to detect significant clusters of synonymous and missense mutations in public cancer genomics data. In melanoma, we show that 22% (11/50) of these mutation clusters are misannotated as coding mutations because the reference transcripts used for their annotation are not expressed. Instead, these mutations are actually non-coding. This, for instance, applies to the mutation clusters targeting known cancer genes kinetochore localized astrin (SPAG5) binding protein (KNSTRN) and BCL2-like 12 (BCL2L12), each affecting 4%-5% of melanoma tumors. For the latter, we show that these mutations are functional non-coding mutations that target the shared promoter region of interferon regulatory factor 3 (IRF3) and BCL2L12. This results in downregulation of IRF3, BCL2L12, and tumor protein p53 (TP53) expression in a CRISPR-Cas9 primary melanocyte model and in melanoma tumors. In individuals with melanoma, these mutations were also associated with a worse response to immunotherapy. Finally, we propose a simple automated method to more accurately annotate cancer mutations based on expressed transcripts. This work shows the importance of integrating DNA- and RNA-sequencing data to properly annotate mutations and identifies a number of previously overlooked and wrongly annotated functional non-coding mutations in melanoma.
Collapse
Affiliation(s)
- Daniele Pepe
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Xander Janssens
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Kalina Timcheva
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Grecia M Marrón-Liñares
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Benno Verbelen
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Vasileios Konstantakos
- Department of Human Genetics, KU Leuven, Leuven, Belgium; VIB Center for AI & Computational Biology (VIB.AI), Leuven, Belgium; VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Dylan De Groote
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Jolien De Bie
- Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | | | - Barbara Dewaele
- Department of Human Genetics, KU Leuven, Leuven, Belgium; Center for Human Genetics, University Hospitals Leuven, Leuven, Belgium
| | | | - Charlotte Cools
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Mireia Franco-Tolsau
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Jonathan Royaert
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Jelle Verbeeck
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Kim R Kampen
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium; Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Karthik Subramanian
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - David Cabrerizo Granados
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium
| | - Gerben Menschaert
- OHMX.bio NV, Evergem, Belgium; Department of Data Analysis and Mathematical Modelling, Ghent University, Ghent, Belgium
| | - Kim De Keersmaecker
- Department of Oncology, KU Leuven, Leuven, Belgium; Leuven Cancer Institute (LKI), Leuven, Belgium.
| |
Collapse
|
4
|
Bollu VS, Chen YC, Zhang F, Gowda K, Amin S, Sharma AK, Schell TD, Zhu J, Robertson GP. Managing telomerase and telomere dysfunction in acral melanoma. Pharmacol Res 2025; 215:107700. [PMID: 40097124 DOI: 10.1016/j.phrs.2025.107700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Acral Lentiginous Melanoma is a rare and aggressive subtype of melanoma that commonly affects the palms, soles, and nail beds. It is more prevalent in individuals with darker skin tones, including Asian, African, and Hispanic populations. Unlike cutaneous melanomas, acral melanoma is not associated with UV exposure and has a distinct genetic and molecular profile, underscoring the need for tailored research and treatment strategies. Standard treatments, such as surgery, chemotherapy, immunotherapy, and targeted therapies, have shown limited success for this melanoma subtype, highlighting the urgency of developing more effective interventions. Telomerase is an enzyme that extends telomeres and is a key target in acral melanoma which exhibits' high telomerase activity, driven by mutations in the telomerase reverse transcriptase TERT promoter, which contributes to uncontrolled tumor cell proliferation, cancer cell immortality, and resistance to conventional therapies. Therefore, targeting telomerase presents a promising therapeutic avenue for acral melanoma patients who do not respond well to current treatments. Several approaches for targeting telomerase deregulation have been developed, and their potential for the management of acral melanoma is discussed in this review. Specifically, the promise of telomerase-targeted therapies for acral melanoma is emphasized and explores how these strategies could improve outcomes for patients with this challenging skin cancer. By focusing on the role of telomerase in tumorigenesis and treatment resistance, telomerase-targeted strategies hold potential as a foundational component of therapies for acral melanoma, complementing existing approaches.
Collapse
Affiliation(s)
- Vishnu Sravan Bollu
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Yu-Chi Chen
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Fan Zhang
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pharmaceutical Sciences, Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, WA 99202, United States
| | - Krishne Gowda
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Shantu Amin
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Arun K Sharma
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Todd D Schell
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Jiyue Zhu
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pharmaceutical Sciences, Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, WA 99202, United States
| | - Gavin P Robertson
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Dermatology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Surgery, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Foreman Foundation for Melanoma Research, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Melanoma and Skin Cancer Center, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Melanoma Therapeutics Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States.
| |
Collapse
|
5
|
Udutha S, Taglang C, Batsios G, Gillespie AM, Tran M, Hoeve JT, Graeber TG, Viswanath P. Combined inhibition of de novo glutathione and nucleotide biosynthesis is synthetically lethal in glioblastoma. Cell Rep 2025; 44:115596. [PMID: 40253695 DOI: 10.1016/j.celrep.2025.115596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 11/15/2024] [Accepted: 03/30/2025] [Indexed: 04/22/2025] Open
Abstract
Understanding the mechanisms by which oncogenic events alter metabolism will help identify metabolic weaknesses that can be targeted for therapy. Telomerase reverse transcriptase (TERT) is essential for telomere maintenance in most cancers. Here, we show that TERT acts via the transcription factor forkhead box O1 (FOXO1) to upregulate glutamate-cysteine ligase (GCLC), the rate-limiting enzyme for de novo biosynthesis of glutathione (GSH, reduced) in multiple cancer models, including glioblastoma (GBM). Genetic ablation of GCLC or pharmacological inhibition using buthionine sulfoximine (BSO) reduces GSH synthesis from [U-13C]-glutamine in GBMs. However, GCLC inhibition drives de novo pyrimidine nucleotide biosynthesis by upregulating the glutamine-utilizing enzymes glutaminase (GLS) and carbamoyl-phosphate synthetase 2, aspartate transcarbamoylase, and dihydroorotatase (CAD) in an MYC-driven manner. Combining BSO with the glutamine antagonist JHU-083 is synthetically lethal in vitro and in vivo and significantly extends the survival of mice bearing intracranial GBM xenografts. Collectively, our studies advance our understanding of oncogene-induced metabolic vulnerabilities in GBMs.
Collapse
Affiliation(s)
- Suresh Udutha
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Céline Taglang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Georgios Batsios
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Anne Marie Gillespie
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Meryssa Tran
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Johanna Ten Hoeve
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA; Crump Institute for Molecular Imaging, Los Angeles, CA, USA; UCLA Metabolomics Center, Los Angeles, CA, USA
| | - Thomas G Graeber
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA; Crump Institute for Molecular Imaging, Los Angeles, CA, USA; UCLA Metabolomics Center, Los Angeles, CA, USA
| | - Pavithra Viswanath
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
6
|
Cook DJ, Trejo-Lopez JA, Pitel BA, Saha N, Koganti T, Hardcastle J, Smoley S, Isaacson M, Gandham M, Sivasankaran G, Dasari S, Fung KM, Powell SZ, Trivedi D, Abdullaev Z, Aldape K, Quezado M, Pratt D, Cimino PJ, Edgar MA, Vaubel RA, Raghunathan A, Giannini C, Jenkins RB, Ida CM. Expanding the spectrum of TERT promoter mutations in CNS tumors: A case series of non-canonical mutations. J Neuropathol Exp Neurol 2025:nlaf022. [PMID: 40105650 DOI: 10.1093/jnen/nlaf022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Affiliation(s)
- David J Cook
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Jorge A Trejo-Lopez
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Beth A Pitel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Neiladri Saha
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Tejaswi Koganti
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Jayson Hardcastle
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Stephanie Smoley
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Matthew Isaacson
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Mallika Gandham
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Gopinath Sivasankaran
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Surendra Dasari
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Suzanne Z Powell
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, United States
| | - Darshan Trivedi
- Department of Anatomic Pathology, Ochsner Medical Center, New Orleans, LA, United States
| | - Zied Abdullaev
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Martha Quezado
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Drew Pratt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Patrick Joseph Cimino
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| | - Mark A Edgar
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, United States
| | - Rachael A Vaubel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Aditya Raghunathan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Caterina Giannini
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Robert B Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Cristiane M Ida
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
7
|
Flynn A, Pattison AD, Balachander S, Boehm E, Bowen B, Dwight T, Rossello FJ, Hofmann O, Martelotto L, Zethoven M, Kirschner LS, Else T, Fishbein L, Gill AJ, Tischler AS, Giordano T, Prodanov T, Noble JR, Reddel RR, Trainer AH, Ghayee HK, Bourdeau I, Elston M, Ishak D, Ngeow Yuen Yie J, Hicks RJ, Crona J, Åkerström T, Stålberg P, Dahia P, Grimmond S, Clifton-Bligh R, Pacak K, Tothill RW. Multi-omic analysis of SDHB-deficient pheochromocytomas and paragangliomas identifies metastasis and treatment-related molecular profiles. Nat Commun 2025; 16:2632. [PMID: 40097403 PMCID: PMC11914184 DOI: 10.1038/s41467-025-57595-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
Hereditary SDHB-mutant pheochromocytomas (PC) and paragangliomas (PG) are rare tumours with a high propensity to metastasize although their clinical behaviour is unpredictable. To characterize the genomic landscape of these tumours and identify metastasis biomarkers, we perform multi-omic analysis on 94 tumours from 79 patients using seven molecular methods. Sympathetic (chromaffin cell) and parasympathetic (non-chromaffin cell) PCPG have distinct molecular profiles reflecting their cell-of-origin and biochemical profile. TERT and ATRX-alterations are associated with metastatic PCPG and these tumours have an increased mutation load, and distinct transcriptional and telomeric features. Most PCPG have quiet genomes with some rare co-operative driver events, including EPAS1/HIF-2α mutations. Two mechanisms of acquired resistance to DNA alkylating chemotherapies are identifiable; MGMT overexpression and mismatch repair-deficiency causing hypermutation. Our comprehensive multi-omic analysis of SDHB-mutant PCPG therefore identifies features of metastatic disease and treatment response, expanding our understanding of these rare neuroendocrine tumours.
Collapse
Affiliation(s)
- Aidan Flynn
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Andrew D Pattison
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Shiva Balachander
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Emma Boehm
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Blake Bowen
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Trisha Dwight
- Kolling Institute of Medical Research, Royal North Shore Hospital St Leonards NSW, Melbourne, Australia
| | - Fernando J Rossello
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
- Murdoch Children's Research Institute, The Royal Children's Hospital, Melbourne, VIC, 3052, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, Murdoch Children's Research Institute, Melbourne, VIC, 3052, Australia
- Australian Regenerative Medicine Institute, Monash University, Victoria, Australia
| | - Oliver Hofmann
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Luciano Martelotto
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | | | - Lawrence S Kirschner
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | | | - Lauren Fishbein
- Department of Medicine, Division of Endocrinology, Metabolism, Diabetes, University of Colorado, Aurora, CO, USA
| | - Anthony J Gill
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards NSW, Sydney, Australia
| | | | | | - Tamara Prodanov
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Jane R Noble
- Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Roger R Reddel
- Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Alison H Trainer
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Hans Kumar Ghayee
- University of Florida and Malcom Randall VA Medical Center, Gainesville, FL, USA
| | - Isabelle Bourdeau
- Division of endocrinology and Research Center, Center hospitalier de l'Université de Montréal, Montreal, Canada
| | - Marianne Elston
- Waikato Clinical Campus, University of Auckland, Hamilton, New Zealand
| | - Diana Ishak
- Cancer Genetics Service, National Cancer Center Singapore, Singapore, Singapore
| | - Joanne Ngeow Yuen Yie
- Cancer Genetics Service, National Cancer Center Singapore, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Rodney J Hicks
- St Vincent's Dept of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Joakim Crona
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Tobias Åkerström
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Peter Stålberg
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Patricia Dahia
- Div. Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio (UTHSCSA), San Antonio, TX, USA
| | - Sean Grimmond
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Roderick Clifton-Bligh
- Kolling Institute of Medical Research, Royal North Shore Hospital St Leonards NSW, Melbourne, Australia.
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia.
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA.
| | - Richard W Tothill
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
8
|
Jin K, Xu J, Zhang L, Liu Z, Su X, Xu Z, Ding Y, Liu H, Chang Y, Xu L, Wang Z, Zhu Y, Xu J. TERT promoter mutations or protein overexpression define an aggressive subset with favourable immunotherapeutic response in advanced urothelial carcinoma. BMJ ONCOLOGY 2025; 4:e000586. [PMID: 40099003 PMCID: PMC11911668 DOI: 10.1136/bmjonc-2024-000586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
Objective Telomerase reverse transcriptase (TERT) gene promoter mutation (TPM) is a key non-coding somatic alteration in urothelial carcinoma (UC) that plays a critical role in telomerase activation. Despite its importance, the prognostic value of TPM has shown mixed results in previous studies. Methods and analysis This study included 155 UC patients from two local clinical centres and 1652 patients from four public datasets, along with matched clinical annotation. Immunohistochemistry of TERT and immune-related markers was performed on tissue microarrays, and transcriptomic and genomic data were analysed to evaluate immune microenvironment characteristics and mutational profiles associated with TPM. We assessed the association of TPM or TERT overexpression (OE) with clinical outcomes, genomics and immunological profiles across tumour stages. Results In early-stage UC, TPM or TERT OE was not significantly associated with patient outcomes. However, in advanced urothelial carcinoma (aUC), TPM or TERT OE was linked to markedly worse overall survival (OS) and a poor response to platinum-based chemotherapy. Notably, despite this unfavourable prognosis, these patients exhibited a more favourable response to anti-PD-1/PD-L1 immunotherapy. aUC with TPM or TERT OE was characterised by an immune-evasive microenvironment, including infiltration of exhausted CD8+ T cells and elevated PD-1 and PD-L1 expression. Furthermore, genomic analysis further revealed a higher APOBEC mutational signature and a lower clock-like mutational signature in aUC with TPM or TERT OE. Conclusion In this retrospective study, TPM or TERT OE identifies a more aggressive subset of patients with poor OS and an immune-evasive microenvironment but a better response to immunotherapy in aUC.
Collapse
Affiliation(s)
- Kaifeng Jin
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jingtong Xu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lingkai Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zhaopei Liu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaohe Su
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ziyue Xu
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yawei Ding
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hailong Liu
- Department of Urology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Chang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Le Xu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zewei Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jiejie Xu
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
9
|
Ishahak M, Han RH, Annamalai D, Woodiwiss T, McCornack C, Cleary RT, DeSouza PA, Qu X, Dahiya S, Kim AH, Millman JR. Genetically Engineered Brain Organoids Recapitulate Spatial and Developmental States of Glioblastoma Progression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410110. [PMID: 39836549 PMCID: PMC11905097 DOI: 10.1002/advs.202410110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/18/2024] [Indexed: 01/23/2025]
Abstract
Glioblastoma (GBM) is an aggressive form of brain cancer that is highly resistant to therapy due to significant intra-tumoral heterogeneity. The lack of robust in vitro models to study early tumor progression has hindered the development of effective therapies. Here, this study develops engineered GBM organoids (eGBOs) harboring GBM subtype-specific oncogenic mutations to investigate the underlying transcriptional regulation of tumor progression. Single-cell and spatial transcriptomic analyses revealed that these mutations disrupt normal neurodevelopment gene regulatory networks resulting in changes in cellular composition and spatial organization. Upon xenotransplantation into immunodeficient mice, eGBOs form tumors that recapitulate the transcriptional and spatial landscape of human GBM samples. Integrative single-cell trajectory analysis of both eGBO-derived tumor cells and patient GBM samples reveal the dynamic gene expression changes in developmental cell states underlying tumor progression. This analysis of eGBOs provides an important validation of engineered cancer organoid models and demonstrates their utility as a model of GBM tumorigenesis for future preclinical development of therapeutics.
Collapse
Affiliation(s)
- Matthew Ishahak
- Division of EndocrinologyMetabolism and Lipid ResearchWashington University School of Medicine660 South Euclid Avenue, Campus Box 8127St. LouisMO63110USA
| | - Rowland H. Han
- Department of GeneticsWashington University School of Medicine4515 McKinley Ave.St. LouisMO63110USA
| | - Devi Annamalai
- Department of GeneticsWashington University School of Medicine4515 McKinley Ave.St. LouisMO63110USA
| | - Timothy Woodiwiss
- Department of Neurological SurgeryUniversity of Iowa Healthcare1800 John Pappajohn PavilionIowa CityIA52242USA
| | - Colin McCornack
- Department of GeneticsWashington University School of Medicine4515 McKinley Ave.St. LouisMO63110USA
| | - Ryan T. Cleary
- Department of GeneticsWashington University School of Medicine4515 McKinley Ave.St. LouisMO63110USA
| | - Patrick A. DeSouza
- Department of GeneticsWashington University School of Medicine4515 McKinley Ave.St. LouisMO63110USA
| | - Xuan Qu
- Department of GeneticsWashington University School of Medicine4515 McKinley Ave.St. LouisMO63110USA
| | - Sonika Dahiya
- Division of NeuropathologyWashington University School of Medicine660 South Euclid Avenue, Campus Box 8118St. LouisMO63110USA
| | - Albert H. Kim
- Department of GeneticsWashington University School of Medicine4515 McKinley Ave.St. LouisMO63110USA
- Taylor Family Department of NeurosurgeryWashington University School of Medicine660 South Euclid Avenue, Campus Box 8057St. LouisMO63110USA
- The Brain Tumor Center at Siteman Cancer Center4921 Parkview PlaceSt. LouisMO63110USA
| | - Jeffrey R. Millman
- Division of EndocrinologyMetabolism and Lipid ResearchWashington University School of Medicine660 South Euclid Avenue, Campus Box 8127St. LouisMO63110USA
- Department of Biomedical EngineeringWashington University1 Brookings Drive, Campus Box 1097St. LouisMO63130USA
| |
Collapse
|
10
|
Xu Y, Gao J, Wang N, Zedenius J, Nilsson IL, Lui WO, Xu D, Juhlin CC, Larsson C, Mu N. BRAF-induced EHF Expression Affects TERT in Aggressive Papillary Thyroid Cancer. J Clin Endocrinol Metab 2025; 110:693-705. [PMID: 39183149 PMCID: PMC11834717 DOI: 10.1210/clinem/dgae589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 07/10/2024] [Accepted: 08/22/2024] [Indexed: 08/27/2024]
Abstract
CONTEXT BRAFV600E and TERT promoter mutations in papillary thyroid carcinoma (PTC) have a synergistic effect on prognosis. This effect is believed to arise from MAPK activation triggered by BRAFV600E, leading to the upregulation of ETS transcription factors that bind to the mutant TERT promoter. OBJECTIVES To explore the role of ETS factors in relation to clinical features, BRAFV600E, and TERT promoter mutations in PTC. DESIGN Transcriptomic data for 28 ETS factors were analyzed in the PTC cohort of The Cancer Genome Atlas (n = 399) and subsequently validated in a local cohort (n = 93). In vitro experiments were performed to investigate the regulatory role in relation to BRAFV600E and TERT expression. RESULTS The Cancer Genome Atlas identified ETS1, ERG, FLI1, GABPA, EHF, ETV6, and SPDEF as differentially expressed genes between stages I + II and III + IV. In both cohorts, EHF was consistently associated with adverse clinical features, BRAFV600E and TERT promoter mutation/expression. Notably, in BRAFV600E mutated PTC, high EHF expression was associated with shorter disease-free survival. Cases harboring concurrent BRAFV600E, TERT promoter mutations, and high EHF expression exhibited the shortest disease-free survival. In cells harboring concurrent BRAFV600E and TERT promoter mutation, overexpression of EHF significantly increased TERT expression, whereas knockdown or pharmacological inhibition of BRAF significantly decreased both EHF and TERT expression. In addition, chromatin immunoprecipitation and quantitative PCR analysis suggested a potential binding of EHF in TERT promoter mutant cells but not in TERT promoter wild-type cells. CONCLUSION The ETS transcription factor EHF is associated with poor prognosis in PTC. This is potentially mediated by BRAF-induced upregulation of EHF, which in turn increases TERT expression in TERT promoter mutated cells.
Collapse
Affiliation(s)
- Yiyi Xu
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm SE-171 64, Sweden
| | - Jiwei Gao
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm SE-171 64, Sweden
- The Cancer Hospital of the University of Chinese Academy of Sciences, (Zhejiang Cancer Hospital), Hangzhou 310022, China
| | - Na Wang
- Department of Medicine-Huddinge, Karolinska Institutet, Stockholm SE-141 83, Sweden
| | - Jan Zedenius
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm SE-171 76, Sweden
- Department of Breast, Endocrine Tumors and Sarcoma, Karolinska University Hospital, Stockholm SE-171 64, Sweden
| | - Inga-Lena Nilsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm SE-171 76, Sweden
- Department of Breast, Endocrine Tumors and Sarcoma, Karolinska University Hospital, Stockholm SE-171 64, Sweden
| | - Weng-Onn Lui
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm SE-171 64, Sweden
| | - Dawei Xu
- Department of Medicine-Solna, Karolinska Institutet, Stockholm SE-171 76, Sweden
| | - C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm SE-171 64, Sweden
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm SE-171 64, Sweden
| | - Catharina Larsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm SE-171 64, Sweden
| | - Ninni Mu
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm SE-171 64, Sweden
- Clinical Genetics, Karolinska University Hospital, Stockholm SE-171 76, Sweden
| |
Collapse
|
11
|
Pazhakh V, Fox LC, Elzen ND, Emerson MR, Cohen SB, Bryan TM, Norris K, Baird DM, Cochrane T, Mackintosh J, Scott A, Blombery P. A novel TERT variant associated with a telomere biology disorder and challenges in variant classification. EJHAEM 2025; 6:e1066. [PMID: 39866942 PMCID: PMC11760216 DOI: 10.1002/jha2.1066] [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: 11/19/2024] [Accepted: 12/21/2024] [Indexed: 01/28/2025]
Abstract
Telomere biology disorders (TBDs) are inherited conditions associated with multisystem manifestations. We describe clinical and functional characterisation of a novel TERT variant. Whole-genome sequencing was performed along with single telomere length analysis (STELA). Telomerase activity and processivity were assessed. A novel TERT variant (K710R) was detected in a patient with classic TBD features showing reduced telomerase activity and processivity. Despite clinical and functional evidence, the variant was classified as a variant of uncertain significance. We have described a novel TERT variant and highlighted the need for further refinement of variant classification specific for TBDs.
Collapse
Affiliation(s)
- Vahid Pazhakh
- Department of PathologyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - Lucy C. Fox
- Department of PathologyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Clinical Haematology DepartmentPeter MacCallum Cancer Centre and Royal Melbourne HospitalMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Nicole Den Elzen
- Department of PathologyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - Matthew R. Emerson
- Children's Medical Research InstituteFaculty of Medicine and HealthUniversity of SydneyWestmeadNew South WalesAustralia
| | - Scott B. Cohen
- Children's Medical Research InstituteFaculty of Medicine and HealthUniversity of SydneyWestmeadNew South WalesAustralia
| | - Tracy M. Bryan
- Children's Medical Research InstituteFaculty of Medicine and HealthUniversity of SydneyWestmeadNew South WalesAustralia
| | - Kevin Norris
- TeloNostiX Ltd, Central Biotechnology ServicesCardiffUK
| | - Duncan M. Baird
- Division of Cancer and GeneticsCardiff University School of MedicineUniversity Hospital of WalesCardiffUK
| | - Tara Cochrane
- Department of HaematologyGold Coast University HospitalGriffith UniversityGold CoastQueenslandAustralia
| | - John Mackintosh
- Department of Thoracic MedicineThe Prince Charles HospitalBrisbaneQueenslandAustralia
| | - Ashleigh Scott
- Department of Haematology and Bone Marrow TransplantRoyal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
| | - Piers Blombery
- Department of PathologyPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Clinical Haematology DepartmentPeter MacCallum Cancer Centre and Royal Melbourne HospitalMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoriaAustralia
| |
Collapse
|
12
|
Murugan AK, Kannan S, Alzahrani AS. TERT promoter mutations in gliomas: Molecular roles in tumorigenesis, metastasis, diagnosis, prognosis, therapeutic targeting, and drug resistance. Biochim Biophys Acta Rev Cancer 2025; 1880:189243. [PMID: 39674418 DOI: 10.1016/j.bbcan.2024.189243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Telomerase reverse transcriptase (TERT), a critical player in cellular immortalization, has emerged as a focal point of investigation due to its frequent promoter mutations in various human malignancies. TERT promoter mutations exhibit a significant role in tumorigenesis, fostering unbridled cellular proliferation and survival. This comprehensive review delves into the landscape of TERT promoter mutations and their profound implications in cancer, particularly within the context of gliomas. This article meticulously examines the intricate interplay between TERT promoter mutations and the metastatic cascade, shedding light on their capacity to orchestrate invasive behavior in gliomas. Moreover, this review describes the recent trends in therapeutic targeting of the TERT and dissects the evolving landscape of drug resistance associated with TERT mutations, providing insights into potential therapeutic challenges. In addition, the diagnostic and prognostic implications of TERT promoter mutations in gliomas are scrutinized, unraveling their potential as robust biomarkers. It also discusses the recent advancements in molecular diagnostics, illustrating the promise of TERT mutations as diagnostic tools and prognostic indicators. This review collectively aims to contribute to a deeper understanding of TERT promoter mutations in gliomas, offering a foundation for future research endeavors and paving the way for innovative strategies in glioma management.
Collapse
Affiliation(s)
- Avaniyapuram Kannan Murugan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia.
| | - Siddarth Kannan
- School of Medicine, University of Central Lancashire, Preston PR1 2HE, UK
| | - Ali S Alzahrani
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
13
|
Zhang G, Yan S, Liu Y, Du Z, Min Q, Qin S. PROTACs coupled with oligonucleotides to tackle the undruggable. Bioanalysis 2025; 17:261-276. [PMID: 39895280 PMCID: PMC11864318 DOI: 10.1080/17576180.2025.2459528] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 01/24/2025] [Indexed: 02/04/2025] Open
Abstract
Undruggable targets account for roughly 85% of human disease-related targets and represent a category of therapeutic targets that are difficult to tackle with traditional methods, but their considerable clinical importance. These targets are generally defined by planar functional interfaces and the absence of efficient ligand-binding pockets, making them unattainable for conventional pharmaceutical strategies. The advent of oligonucleotide-based proteolysis-targeting chimeras (PROTACs) has instilled renewed optimism in addressing these challenges. These PROTACs facilitate the targeted degradation of undruggable entities, including transcription factors (TFs) and RNA-binding proteins (RBPs), via proteasome-dependent mechanisms, thereby presenting novel therapeutic approaches for diseases linked to these targets. This review offers an in-depth examination of recent progress in the integration of PROTAC technology with oligonucleotides to target traditionally undruggable proteins, emphasizing the design principles and mechanisms of action of these innovative PROTACs.
Collapse
Affiliation(s)
- Guangshuai Zhang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Si Yan
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Yan Liu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Ziwei Du
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Qin Min
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
| | - Shuanglin Qin
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, P.R.China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Research Center for Precision Medication of Chinese Medicine, FuRong Laboratory, Hunan University of Chinese Medicine, Changsha, P.R. China
| |
Collapse
|
14
|
Smith C, Asnafi V, Touzart A. Neo-enhancers in T-cell acute lymphoblastic Leukaemia (T-ALL) and beyond. Int J Cancer 2025. [PMID: 39749749 DOI: 10.1002/ijc.35315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
T-cell acute lymphoblastic leukaemia (T-ALL) is a rare aggressive haematological malignancy characterised by the clonal expansion of immature T-cell precursors. It accounts for 15% of paediatric and 25% of adult ALL. T-ALL is associated with the overexpression of major transcription factors (TLX1/3, TAL1, HOXA) that drive specific transcriptional programmes and constitute the molecular classifying subgroups of T-ALL. Although the dysregulation of transcription factor oncogenes is frequently associated with chromosomal translocations in T-ALL, epigenetic dysregulation resulting in changes to post-translational modifications of histones has also been reported. This includes non-coding intergenic mutations that form oncogenic neo-enhancers. This review will focus on the known epigenetically activating intergenic mutations reported in T-ALL, and will discuss the wider implications of neo-enhancer mutations in cancer.
Collapse
Affiliation(s)
- Charlotte Smith
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Université Paris Cité, CNRS, INSERM U1151, Institut Necker Enfants-Malades (INEM), Paris, France
| | - Vahid Asnafi
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Université Paris Cité, CNRS, INSERM U1151, Institut Necker Enfants-Malades (INEM), Paris, France
| | - Aurore Touzart
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
- Université Paris Cité, CNRS, INSERM U1151, Institut Necker Enfants-Malades (INEM), Paris, France
| |
Collapse
|
15
|
Xing H, Liu D, Li J, Ge Y, Guo X, Chen W, Zhao D, Shi Y, Li Y, Wang Y, Wang Y, Xia Y, Wu J, Liang T, Wang H, Liu Q, Jin S, Qu T, Guo S, Li H, Yang T, Zhang K, Wang Y, Ma W. TERTp Mutation and its Prognostic Value in Glioma Patients Under the 2021 WHO Classification: A Real-World Study. Cancer Med 2025; 14:e70533. [PMID: 39804195 PMCID: PMC11727134 DOI: 10.1002/cam4.70533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/16/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The 2021 WHO Classification of Central Nervous System Tumors introduces more molecular markers for glioma reclassification, including TERT promoter (TERTp) mutation as a key feature in glioblastoma diagnosis. AIMS Given the changes in the entities included in each subtype under the new classification, this research investigated the distribution, prognostic value, and correlations with other molecular alterations of TERTp mutation in different subgroups under this latest classification. METHODS All glioma patients admitted to Peking Union Medical College Hospital for surgical resection or biopsy from 2011 to 2022 were included. Samples were analyzed for TERTp mutation and 59 other gene alterations and chromosome copy number variations. RESULTS A total of 207 patients were included. The occurrence of TERTp mutations varied with percentages of 4.55%, 100%, and 77.92% in astrocytoma, oligodendroglioma, and glioblastoma, respectively. 65% of all adult-type glioma patients and 42.6% of IDH-wildtype histology grade 2 or 3 patients were TERTp-mutant. Survival analysis showed that TERTp mutation was a predictor of better prognosis in IDH-mutant grade 2 gliomas (median OS (mOS): not reached (NA) (95% CI: NA-NA) vs. 75.9 (95% CI: 55.4-NA) months, HR = 0.077 (95% CI: 0.01-0.64), p = 0.003), while poor OS was associated with all Grade 4 gliomas (mOS: 17.5 (95% CI: 12.6-24.2) vs. 40.5 (95% CI: 24.4-83.8) months, HR = 2.014 (95% CI: 1.17-3.47), p = 0.01) and all IDH-wildtype histology grade 2 or 3 gliomas (median OS: 12.6 (95% CI: 11-24.2) vs. 83.8 (95% CI: 35.2-NA) months, HR = 3.768 (95% CI: 1.83-7.78), p < 0.001). Moreover, TERTp mutation tended to co-occur with EGFR, KRAS, and MET in glioblastoma. In the IDH-mutant subgroup, it tended to co-occur with CIC and FUBP1 alterations, while being mutually exclusive with ATRX and TP53 alterations. These correlations may further refine prognostic predictions.
Collapse
Affiliation(s)
- Hao Xing
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Delin Liu
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Junlin Li
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yulu Ge
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xiaopeng Guo
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- China Anti‐Cancer Association Specialty Committee of GliomaBeijingChina
| | - Wenlin Chen
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Dachun Zhao
- Department of PathologyPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yixin Shi
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yilin Li
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yaning Wang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yuekun Wang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yu Xia
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jiaming Wu
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Tingyu Liang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Hai Wang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Qianshu Liu
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Shanmu Jin
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Tian Qu
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Siying Guo
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Huanzhang Li
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Tianrui Yang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Kun Zhang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yu Wang
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- China Anti‐Cancer Association Specialty Committee of GliomaBeijingChina
| | - Wenbin Ma
- Department of Neurosurgery, Center for Malignant Brain Tumors, National Glioma MDT AlliancePeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- China Anti‐Cancer Association Specialty Committee of GliomaBeijingChina
| |
Collapse
|
16
|
Casalini R, Romei C, Ciampi R, Ramone T, Prete A, Gambale C, Matrone A, Torregrossa L, Ugolini C, Elisei R. Minor role of TP53 and TERT promoter mutations in medullary thyroid carcinoma: report of new cases and revision of the literature. Endocrine 2025; 87:243-251. [PMID: 39179735 DOI: 10.1007/s12020-024-03990-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/02/2024] [Indexed: 08/26/2024]
Abstract
PURPOSE Aims of this study were to investigate the prevalence of TP53 and TERT mutations in Medullary Thyroid carcinoma (MTC) and their role in inducing aggressiveness in positive cases. METHODS We performed a literature search in PubMed to identify studies investigating the prevalence of TERT and TP53 mutations in MTC. We also included data on MTC cases (n = 193) obtained at our center and unpublished. The in-silico pathogenicity of the TP53 mutations has been evaluated by predictor tools. RESULTS We identified a total of 25 and 11 published papers: all together 1280 cases have been investigated for the presence of TP53 mutations and 974 for TERT promoter mutation. Twenty-five out of 1280 (2%) cases had a TP53 mutation while only 3/974 MTC cases (0.3%) have been found to be positive for TERT promoter mutations. Among all, we identified 19 different TP53 mutations that in 12 cases were demonstrated to have an in silico predicted high pathogenic role and a high impact on protein function. Three non-sense and 4 probably not damaging mutations were also reported. The pathogenic role of the TERT promoter mutations has been previously in vitro determined. No correlation between TP53 and/or TERT mutations and aggressiveness of MTC has been demonstrated. CONCLUSION The prevalence of TP53 and TERT promoter mutations is very low in MTC. The reported mutations are pathogenic in the majority of cases. Because of their rarity it is not possible to clarify if they play or not a role in the pathogenesis and/or aggressiveness of this specific thyroid tumor.
Collapse
Affiliation(s)
- Roberta Casalini
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Cristina Romei
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Raffaele Ciampi
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Teresa Ramone
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Alessandro Prete
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Carla Gambale
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Antonio Matrone
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy
| | - Liborio Torregrossa
- Department of Surgical, Medical, Molecular Pathology and Critical Area, Unit of Pathology, University Hospital of Pisa, Pisa, Italy
| | - Clara Ugolini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, Unit of Pathology, University Hospital of Pisa, Pisa, Italy
| | - Rossella Elisei
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University Hospital of Pisa, Pisa, Italy.
| |
Collapse
|
17
|
Huang Y, Wang Y, Li R, Liu Y, Yang Y. miR-450b-5p promotes development of endometriosis by inhibiting the GABPA/HOXD10 axis. iScience 2024; 27:111487. [PMID: 39759007 PMCID: PMC11696641 DOI: 10.1016/j.isci.2024.111487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 10/18/2024] [Accepted: 11/25/2024] [Indexed: 01/07/2025] Open
Abstract
Despite decades of research, the pathogenesis of endometriosis remains unclear. Recent studies have shown that microRNAs play an important role in this condition. In this study, we found that the expression level of miR-450b-5p was increased in ectopic endometrial tissues and that GA-binding protein A (GABPA) and HOXD10 expression levels were decreased. Overexpression of miR-450b-5p or knockdown of GABPA significantly promoted the proliferation and invasion of hEM15A cells and inhibited apoptosis in vivo and in vitro. Furthermore, GABPA was shown to be a direct target of miR-450b-5p and to bind directly to the promoter of the HOXD10 gene, regulating its transcription. Finally, intraperitoneal injection of HOXD10-overexpressing lentivirus in mice significantly attenuated ectopic endometrial lesions. miR-450b-5p directly targets GABPA, regulates expression of HOXD10, and promotes the growth of ectopic endometriotic lesions. Therefore, the miR-450b-5p/GABPA/HOXD10 signaling pathway may be a potential target for treatment of heterotopic endothelial cell disease.
Collapse
Affiliation(s)
- Yi Huang
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Yidan Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China
- Gansu International Scientific and Technological Cooperation, Base of Reproductive Medicine Transformation Application, Key Laboratory for Reproductive Medicine and Embryo, Lanzhou 730000, China
| | - Ruiyun Li
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China
| | - Yongmei Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China
| | - Yuan Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou 730000, China
- Reproductive Medicine Center, The First Hospital of Lanzhou University, Lanzhou 730000, China
| |
Collapse
|
18
|
Sadr Z, Ghasemi M, Jafarpour S, Seyfi R, Ghasemi A, Boustanipour E, Khorshid HRK, Ehtesham N. Beginning at the ends: telomere and telomere-based cancer therapeutics. Mol Genet Genomics 2024; 300:1. [PMID: 39638969 DOI: 10.1007/s00438-024-02206-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
Telomeres, which are situated at the terminal ends of chromosomes, undergo a reduction in length with each cellular division, ultimately reaching a critical threshold that triggers cellular senescence. Cancer cells circumvent this senescence by utilizing telomere maintenance mechanisms (TMMs) that grant them a form of immortality. These mechanisms can be categorized into two primary processes: the reactivation of telomerase reverse transcriptase and the alternative lengthening of telomeres (ALT) pathway, which is dependent on homologous recombination (HR). Various strategies have been developed to inhibit telomerase activation in 85-95% of cancers, including the use of antisense oligonucleotides such as small interfering RNAs and endogenous microRNAs, agents that simulate telomere uncapping, expression modulators, immunotherapeutic vaccines targeting telomerase, reverse transcriptase inhibitors, stabilization of G-quadruplex structures, and gene therapy approaches. Conversely, in the remaining 5-15% of human cancers that rely on ALT, mechanisms involve modifications in the chromatin environment surrounding telomeres, upregulation of TERRA long non-coding RNA, enhanced activation of the ataxia telangiectasia and Rad-3-related protein kinase signaling pathway, increased interactions with nuclear receptors, telomere repositioning driven by HR, and recombination events between non-sister chromatids, all of which present potential targets for therapeutic intervention. Additionally, combinatorial therapy has emerged as a strategy that employs selective agents to simultaneously target both telomerase and ALT, aiming for optimal clinical outcomes. Given the critical role of anti-TMM strategies in cancer treatment, this review provides an overview of the latest insights into the structure and function of telomeres, their involvement in tumorigenesis, and the advancements in TMM-based cancer therapies.
Collapse
Affiliation(s)
- Zahra Sadr
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoumeh Ghasemi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Soheyla Jafarpour
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reyhaneh Seyfi
- Department of Stem Cells Technology and Tissue Regeneration, Faculty of Interdisciplinary Science and Technologies, Tarbiat Modares University, Tehran, Iran
| | - Aida Ghasemi
- Neuromuscular Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Boustanipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Naeim Ehtesham
- Department of Medical Genetics, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran.
| |
Collapse
|
19
|
Elliott K, Singh VK, Bäckerholm A, Ögren L, Lindberg M, Soczek KM, Hoberg E, Luijts T, Van den Eynden J, Falkenberg M, Doudna J, Ståhlberg A, Larsson E. Mechanistic basis of atypical TERT promoter mutations. Nat Commun 2024; 15:9965. [PMID: 39557834 PMCID: PMC11574208 DOI: 10.1038/s41467-024-54158-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 10/31/2024] [Indexed: 11/20/2024] Open
Abstract
Non-coding mutations in the TERT promoter (TERTp), typically at one of two bases -124 and -146 bp upstream of the start codon, are among the most prevalent driver mutations in human cancer. Several additional recurrent TERTp mutations have been reported but their functions and origins remain largely unexplained. Here, we show that atypical TERTp mutations arise secondary to canonical TERTp mutations in a two-step process. Canonical TERTp mutations create de novo binding sites for ETS family transcription factors that induce favourable conditions for DNA damage formation by UV light, thus creating a hotspot effect but only after a first mutational hit. In agreement, atypical TERTp mutations co-occur with canonical driver mutations in large cancer cohorts and arise subclonally specifically on the TERTp driver mutant chromosome homolog of melanoma cells treated with UV light in vitro. Our study gives an in-depth view of TERTp mutations in cancer and provides a mechanistic explanation for atypical TERTp mutations.
Collapse
Affiliation(s)
- Kerryn Elliott
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Vinod Kumar Singh
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Alan Bäckerholm
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Linnea Ögren
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Markus Lindberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Katarzyna M Soczek
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Emily Hoberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tom Luijts
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Jimmy Van den Eynden
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent, Ghent, Belgium
| | - Maria Falkenberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jennifer Doudna
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Anders Ståhlberg
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, Sweden
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Erik Larsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
20
|
Zhang W, Lin S, Wang Z, Zhang W, Xing M. Coexisting RET/PTC and TERT Promoter Mutation Predict Poor Prognosis but Effective RET and MEK Targeting in Thyroid Cancer. J Clin Endocrinol Metab 2024; 109:3166-3175. [PMID: 38735658 PMCID: PMC11570377 DOI: 10.1210/clinem/dgae327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/09/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
CONTEXT The role of RET/PTC rearrangement in the clinical outcomes of papillary thyroid cancer (PTC) is controversial and remains to be clearly undefined. OBJECTIVE This work aimed to investigate the role of coexisting RET/PTC rearrangement and TERT promoter mutation in the prognosis and therapeutic targeting in PTC. METHODS A total of 669 PTC patients with complete clinical follow-up and genetic data were pooled from thyroid cancer data sets TCGA-THCA, MSK-MetTropism, and MSK-IMPACT, from whom 163 patients (112 women and 47 men, 4 unknown) with wild-type (WT) BRAF/RAS were identified, with a median age (interquartile range [IQR]) of 46.00 (33.00-61.00) years and a median follow-up time (IQR) of 16.13 (8.09-27.91) months for comparative genotype cohort analysis of mortality. RESULTS There was a significant concurrence index between RET/PTC and TERT promoter mutations, being 2.040 (95% CI, 1.110-3.747; P = .023). Mortality occurred in 5 of 100 (5%) patients harboring neither mutation, 2 of 18 (11.1%) patients harboring a TERT promoter mutation alone, 0 of 31 (0%) patients harboring a RET/PTC alone, and 7 of 14 (50%) patients harboring both genetic alterations, corresponding to hazard ratios (95% CI) of 1 (reference), 2.469 (0.405-14.022), 3.296e-09 (0-inf), and 9.019 (2.635-30.870), respectively, which remained essentially unchanged after adjustment for patient race, sex, and age. Similar results were observed with BRAF/RAS and TERT promoter mutations. Mechanistically, RET/PTC used the MAP kinase pathway to upregulate the mutated TERT, but not the WT TERT, and, correspondingly, targeting RET and MEK could suppress mutated TERT but not the WT TERT. CONCLUSION Coexisting RET/PTC and TERT promoter mutation identify PTC as a unique clinical entity with high mortality, providing new implications for genetic-based prognostication and potential therapeutic targeting of RET and MEK guided by RET/PTC and TERT status.
Collapse
Affiliation(s)
- Wei Zhang
- Thyroid Research Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, PR China
| | - Shuhuang Lin
- Thyroid Research Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, PR China
| | - Zhuo Wang
- Thyroid Research Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, PR China
| | - Wenyong Zhang
- Thyroid Research Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, PR China
| | - Mingzhao Xing
- Thyroid Research Institute, School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong 518055, PR China
| |
Collapse
|
21
|
Wu Y, Wu Z, Yao M, Liu L, Song Y, Ma L, Liu C. GABPA inhibits tumorigenesis in clear cell renal cell carcinoma by regulating ferroptosis through ACSL4. Sci Rep 2024; 14:26521. [PMID: 39489850 PMCID: PMC11532409 DOI: 10.1038/s41598-024-78441-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common genitourinary malignancy characterized by dysregulated cellular metabolism leading to aberrant glucose metabolism, fatty acid accumulation, and excessive reactive oxygen species production. ccRCC cells exhibit an augmented oxidative stress response. Complex interactions between iron metabolism and lipid homeostasis in ccRCC cells require a counteracting response that enables ferroptosis evasion and survival maintenance. Additionally, abnormal GA-binding protein transcription factor subunit alpha (GABPA) expression is associated with ccRCC occurrence and development, but its impact on ferroptosis-related molecular mechanisms remains unclear. Herein, we examined the impact of the GABPA-ACSL4 pathway on ferroptosis in ccRCC through bioinformatics analysis, as well as in vitro and in vivo experiments. In contrast to that in adjacent normal tissues, GABPA expression was significantly downregulated in ccRCC tissues, and this downregulation was linked to poor overall survival. Increased GABPA expression suppressed ccRCC cell proliferation, migration, and invasion. Moreover, GABPA overexpression increased the susceptibility of ccRCC cells to ferroptosis. Additionally, GABPA directly bound to the promoter region of ACSL4, promoting ferroptosis. Thus, inducing the GABPA-ACSL4 pathway activates ferroptosis, inhibits proliferation or metastasis, and exerts anticancer activity in ccRCC. These findings have important implications for regulating ccRCC occurrence and development.
Collapse
Affiliation(s)
- Yaqian Wu
- Department of Urology, Peking University Third Hospital, Beijing, 100191, People's Republic of China
- Department of Pathology, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, People's Republic of China
| | - Zonglong Wu
- Department of Urology, Peking University Third Hospital, Beijing, 100191, People's Republic of China
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Mengfei Yao
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Li Liu
- School of Nursing, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Beijing, 100191, People's Republic of China
| | - Yimeng Song
- Department of Urology, Peking University Third Hospital, Beijing, 100191, People's Republic of China
| | - Lulin Ma
- Department of Urology, Peking University Third Hospital, Beijing, 100191, People's Republic of China.
| | - Cheng Liu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, People's Republic of China.
| |
Collapse
|
22
|
Shanmugam R, Majee P, Shi W, Ozturk MB, Vaiyapuri TS, Idzham K, Raju A, Shin SH, Fidan K, Low JL, Chua JY, Kong YC, Qi OY, Tan E, Chok AY, Seow-En I, Wee I, Macalinao DC, Chong DQ, Chang HY, Lee F, Leow WQ, Murata-Hori M, Xiaoqian Z, Shumei C, Tan CS, Dasgupta R, Tan IB, Tergaonkar V. Iron-(Fe3+)-Dependent Reactivation of Telomerase Drives Colorectal Cancers. Cancer Discov 2024; 14:1940-1963. [PMID: 38885349 PMCID: PMC11450372 DOI: 10.1158/2159-8290.cd-23-1379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 04/15/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024]
Abstract
Over-consumption of iron-rich red meat and hereditary or genetic iron overload are associated with an increased risk of colorectal carcinogenesis, yet the mechanistic basis of how metal-mediated signaling leads to oncogenesis remains enigmatic. Using fresh colorectal cancer samples we identify Pirin, an iron sensor, that overcomes a rate-limiting step in oncogenesis, by reactivating the dormant human telomerase reverse transcriptase (hTERT) subunit of the telomerase holoenzyme in an iron-(Fe3+)-dependent manner and thereby drives colorectal cancers. Chemical genetic screens combined with isothermal dose-response fingerprinting and mass spectrometry identified a small molecule SP2509 that specifically inhibits Pirin-mediated hTERT reactivation in colorectal cancers by competing with iron-(Fe3+) binding. Our findings, first to document how metal ions reactivate telomerase, provide a molecular mechanism for the well-known association between red meat and increased incidence of colorectal cancers. Small molecules like SP2509 represent a novel modality to target telomerase that acts as a driver of 90% of human cancers and is yet to be targeted in clinic. Significance: We show how iron-(Fe3+) in collusion with genetic factors reactivates telomerase, providing a molecular mechanism for the association between iron overload and increased incidence of colorectal cancers. Although no enzymatic inhibitors of telomerase have entered the clinic, we identify SP2509, a small molecule that targets telomerase reactivation and function in colorectal cancers.
Collapse
Affiliation(s)
- Raghuvaran Shanmugam
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
| | - Prativa Majee
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
| | - Wei Shi
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
| | - Mert B. Ozturk
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
| | - Thamil S. Vaiyapuri
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
| | - Khaireen Idzham
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
| | - Anandhkumar Raju
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
| | - Seung H. Shin
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
| | - Kerem Fidan
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
| | - Joo-Leng Low
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore, Republic of Singapore.
| | - Joelle Y.H. Chua
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
| | - Yap C. Kong
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore, Republic of Singapore.
| | - Ong Y. Qi
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
| | - Emile Tan
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Republic of Singapore.
| | - Aik Y. Chok
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Republic of Singapore.
| | - Isaac Seow-En
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Republic of Singapore.
| | - Ian Wee
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Republic of Singapore.
| | - Dominique C. Macalinao
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Republic of Singapore.
| | - Dawn Q. Chong
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Republic of Singapore.
| | - Hong Y. Chang
- Experimental Drug Development Center, Agency for Science, Technology, and Research (A*STAR), Singapore, Republic of Singapore.
| | - Fiona Lee
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore, Republic of Singapore.
| | - Wei Q. Leow
- Department of Colorectal Surgery, Singapore General Hospital, Singapore, Republic of Singapore.
| | - Maki Murata-Hori
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore, Republic of Singapore.
| | - Zhang Xiaoqian
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore, Republic of Singapore.
| | - Chia Shumei
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore, Republic of Singapore.
| | - Chris S.H. Tan
- Department of Chemistry, College of Science, Southern University of Science and Technology, Shenzhen, China.
| | - Ramanuj Dasgupta
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore, Republic of Singapore.
| | - Iain B. Tan
- Genome Institute of Singapore, Agency for Science, Technology, and Research (A*STAR), Singapore, Republic of Singapore.
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Republic of Singapore.
- Cancer and Stem Cell Biology, Duke-National University of Singapore, Singapore, Republic of Singapore.
| | - Vinay Tergaonkar
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Republic of Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Republic of Singapore.
| |
Collapse
|
23
|
Kageler L, Aquilanti E. Discovery of telomerase inhibitors: existing strategies and emerging innovations. Biochem Soc Trans 2024; 52:1957-1968. [PMID: 39194999 DOI: 10.1042/bst20230264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024]
Abstract
Telomerase, crucial for maintaining telomere length, is an attractive target for cancer therapy due to its role in cellular immortality. Despite three decades of research efforts, no small-molecule telomerase inhibitors have been clinically approved, highlighting the extensive challenges in developing effective telomerase-based therapeutics. This review examines conventional and emerging methods to measure telomerase activity and discusses existing inhibitors, including oligonucleotides and small molecules. Furthermore, this review highlights recent breakthroughs in structural studies of telomerase using cryo-electron microscopy, which can facilitate improved structure-based drug design. Altogether, advancements in structural methodologies and high-throughput screening offer promising prospects for telomerase-based cancer therapeutic development.
Collapse
Affiliation(s)
- Lauren Kageler
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, U.S.A
| | - Elisa Aquilanti
- Division of Neuro Oncology, Dana Farber Cancer Institute, Boston, MA, U.S.A
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, U.S.A
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, U.S.A
| |
Collapse
|
24
|
Sun X, Xiao C, Wang X, Wu S, Yang Z, Sui B, Song Y. Role of post-translational modifications of Sp1 in cancer: state of the art. Front Cell Dev Biol 2024; 12:1412461. [PMID: 39228402 PMCID: PMC11368732 DOI: 10.3389/fcell.2024.1412461] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 07/16/2024] [Indexed: 09/05/2024] Open
Abstract
Specific protein 1 (Sp1) is central to regulating transcription factor activity and cell signaling pathways. Sp1 is highly associated with the poor prognosis of various cancers; it is considered a non-oncogene addiction gene. The function of Sp1 is complex and contributes to regulating extensive transcriptional activity, apart from maintaining basal transcription. Sp1 activity and stability are affected by post-translational modifications (PTMs), including phosphorylation, ubiquitination, acetylation, glycosylation, and SUMOylation. These modifications help to determine genetic programs that alter the Sp1 structure in different cells and increase or decrease its transcriptional activity and DNA binding stability in response to pathophysiological stimuli. Investigating the PTMs of Sp1 will contribute to a deeper understanding of the mechanism underlying the cell signaling pathway regulating Sp1 stability and the regulatory mechanism by which Sp1 affects cancer progression. Furthermore, it will facilitate the development of new drug targets and biomarkers, thereby elucidating considerable implications in the prevention and treatment of cancer.
Collapse
Affiliation(s)
- Xutao Sun
- Department of Typhoid, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengpu Xiao
- Department of Chinese Internal Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xinyang Wang
- Department of Pneumology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Siyu Wu
- Department of Pneumology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhendong Yang
- Department of Pneumology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Bowen Sui
- Department of Pneumology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yunjia Song
- Department of Pharmacology, School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
25
|
Gnanapragasam A, Kirbizakis E, Li A, White KH, Mortenson KL, Cavalcante de Moura J, Jawhar W, Yan Y, Falter R, Russett C, Giannias B, Camilleri-Broët S, Bertos N, Cools-Lartigue J, Garzia L, Sangwan V, Ferri L, Zhang X, Bailey SD. HiChIP-Based Epigenomic Footprinting Identifies a Promoter Variant of UXS1 That Confers Genetic Susceptibility to Gastroesophageal Cancer. Cancer Res 2024; 84:2377-2389. [PMID: 38748784 PMCID: PMC11247317 DOI: 10.1158/0008-5472.can-23-2397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 02/01/2024] [Accepted: 05/08/2024] [Indexed: 07/16/2024]
Abstract
Genome-wide association studies (GWAS) have identified more than a hundred single nucleotide variants (SNV) associated with the risk of gastroesophageal cancer (GEC). The majority of the identified SNVs map to noncoding regions of the genome. Uncovering the causal SNVs and genes they modulate could help improve GEC prevention and treatment. Herein, we used HiChIP against histone 3 lysine 27 acetylation (H3K27ac) to simultaneously annotate active promoters and enhancers, identify the interactions between them, and detect nucleosome-free regions (NFR) harboring potential causal SNVs in a single assay. The application of H3K27ac HiChIP in GEC relevant models identified 61 potential functional SNVs that reside in NFRs and interact with 49 genes at 17 loci. The approach led to a 67% reduction in the number of SNVs in linkage disequilibrium at these 17 loci, and at 7 loci, a single putative causal SNV was identified. One SNV, rs147518036, located within the promoter of the UDP-glucuronate decarboxylase 1 (UXS1) gene, seemed to underlie the GEC risk association captured by the rs75460256 index SNV. The rs147518036 SNV creates a GABPA DNA recognition motif, resulting in increased promoter activity, and CRISPR-mediated inhibition of the UXS1 promoter reduced the viability of the GEC cells. These findings provide a framework that simplifies the identification of potentially functional regulatory SNVs and target genes underlying risk-associated loci. In addition, the study implicates increased expression of the enzyme UXS1 and activation of its metabolic pathway as a predisposition to gastric cancer, which highlights potential therapeutic avenues to treat this disease. Significance: Epigenomic footprinting using a histone posttranslational modification targeted 3D genomics methodology elucidates functional noncoding sequence variants and their target genes at cancer risk loci.
Collapse
Affiliation(s)
- Ansley Gnanapragasam
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
| | - Eftyhios Kirbizakis
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
| | - Anna Li
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
| | - Kyle H White
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
- Department of Pathology, McGill University, Montreal, Canada
| | | | - Juliana Cavalcante de Moura
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
| | - Wajih Jawhar
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
- Department of Surgery, McGill University, Montreal, Canada
| | - Yifei Yan
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
| | - Reilly Falter
- Department of Experimental Medicine, McGill University, Montreal, Canada
| | - Colleen Russett
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
| | - Betty Giannias
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
| | - Sophie Camilleri-Broët
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | - Nicholas Bertos
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
| | - Jonathan Cools-Lartigue
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
- Department of Pathology, McGill University, Montreal, Canada
| | - Livia Garzia
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
- Department of Surgery, McGill University, Montreal, Canada
- Department of Pathology, McGill University, Montreal, Canada
| | - Veena Sangwan
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
- Department of Pathology, McGill University, Montreal, Canada
| | - Lorenzo Ferri
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
- Department of Pathology, McGill University, Montreal, Canada
| | - Xiaoyang Zhang
- Department of Experimental Medicine, McGill University, Montreal, Canada
| | - Swneke D Bailey
- The Cancer Research Program, Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
- Department of Surgery, McGill University, Montreal, Canada
- Department of Pathology, McGill University, Montreal, Canada
| |
Collapse
|
26
|
Nevarez AJ, Mudla A, Diaz SA, Hao N. Using deep learning to decipher the impact of telomerase promoter mutations on the dynamic metastatic morpholome. PLoS Comput Biol 2024; 20:e1012271. [PMID: 39078811 PMCID: PMC11288469 DOI: 10.1371/journal.pcbi.1012271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 06/22/2024] [Indexed: 08/02/2024] Open
Abstract
Melanoma showcases a complex interplay of genetic alterations and intra- and inter-cellular morphological changes during metastatic transformation. While pivotal, the role of specific mutations in dictating these changes still needs to be fully elucidated. Telomerase promoter mutations (TERTp mutations) significantly influence melanoma's progression, invasiveness, and resistance to various emerging treatments, including chemical inhibitors, telomerase inhibitors, targeted therapy, and immunotherapies. We aim to understand the morphological and phenotypic implications of the two dominant monoallelic TERTp mutations, C228T and C250T, enriched in melanoma metastasis. We developed isogenic clonal cell lines containing the TERTp mutations and utilized dual-color expression reporters steered by the endogenous Telomerase promoter, giving us allelic resolution. This approach allowed us to monitor morpholomic variations induced by these mutations. TERTp mutation-bearing cells exhibited significant morpholome differences from their wild-type counterparts, with increased allele expression patterns, augmented wound-healing rates, and unique spatiotemporal dynamics. Notably, the C250T mutation exerted more pronounced changes in the morpholome than C228T, suggesting a differential role in metastatic potential. Our findings underscore the distinct influence of TERTp mutations on melanoma's cellular architecture and behavior. The C250T mutation may offer a unique morpholomic and systems-driven advantage for metastasis. These insights provide a foundational understanding of how a non-coding mutation in melanoma metastasis affects the system, manifesting in cellular morpholome.
Collapse
Affiliation(s)
- Andres J. Nevarez
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Anusorn Mudla
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Sabrina A. Diaz
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Nan Hao
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, California, United States of America
| |
Collapse
|
27
|
Sang Y, Hu G, Xue J, Chen M, Hong S, Liu R. Risk stratification by combining common genetic mutations and TERT promoter methylation in papillary thyroid cancer. Endocrine 2024; 85:304-312. [PMID: 38356100 DOI: 10.1007/s12020-024-03722-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/30/2024] [Indexed: 02/16/2024]
Abstract
PURPOSE Risk stratification based on somatic mutations in TERT promoter and BRAF/RAS has been well established for papillary thyroid cancer (PTC), and there is emerging evidence showed that TERT promoter methylation was frequently observed in thyroid cancer patients with adverse features. This study was aimed to comprehensive explore the prognostic value of BRAF/RAS mutations, TERT promoter mutations, and TERT promoter methylation in PTC. METHODS The relationships of BRAF/RAS mutations, TERT promoter mutations, and TERT promoter methylation with clinical characteristics and outcomes of PTC were analyzed in 382 patients with PTC. RESULTS TERT promoter mutation and hypermethylation were collectively observed in 52 (13.6%) samples and associated with BRAF/RAS mutation, aggressive clinical characteristics, and poor clinical outcomes of PTC. Coexistence of BRAF/RAS and TERT alterations was found in 45 of 382 (11.8%) PTC patients and strongly associated with old patient age, extrathyroidal extension, advanced pathologic T stage and metastasis. Importantly, patients with both BRAF/RAS and TERT alterations had higher rates of tumor recurrence (13.6% vs 1.5%, P = 0.042) and disease progression (24.4% vs 3.3%, P < 0.001) than patients without any alterations, and cox regression analysis revealed that the coexistence of BRAF/RAS and TERT alterations, but not BRAF/RAS or TERT alterations alone, increased the risk of progression-free interval with an adjusted HR of 10.35 (95% CI: 1.79-59.81, P = 0.009). CONCLUSIONS This study suggested that comprehensively analysis of BRAF/RAS mutations, TERT promoter mutation and methylation is an effective strategy to identify high-risk patients with PTC.
Collapse
Affiliation(s)
- Ye Sang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Second Road, Guangzhou, China
| | - Guanghui Hu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Second Road, Guangzhou, China
| | - Junyu Xue
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Second Road, Guangzhou, China
| | - Mengke Chen
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Second Road, Guangzhou, China
| | - Shubin Hong
- Department of Endocrinology, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Second Road, Guangzhou, China
| | - Rengyun Liu
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan Second Road, Guangzhou, China.
| |
Collapse
|
28
|
Flynn A, Pattison AD, Balachander S, Boehm E, Bowen B, Dwight T, Rosello F, Hofmann O, Martelotto L, Zethoven M, Kirschner LS, Else T, Fishbein L, Gill AJ, Tischler AS, Giordano T, Prodanov T, Noble JR, Reddel RR, Trainer AH, Ghayee HK, Bourdeau I, Elston M, Ishak D, Ngeow Yuen Yie J, Hicks RJ, Crona J, Åkerström T, Stålberg P, Dahia P, Grimmond S, Clifton-Bligh R, Pacak K, Tothill RW. Multi-omic analysis of SDHB-deficient pheochromocytomas and paragangliomas identifies metastasis and treatment-related molecular profiles. RESEARCH SQUARE 2024:rs.3.rs-4410500. [PMID: 38978571 PMCID: PMC11230496 DOI: 10.21203/rs.3.rs-4410500/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Hereditary SDHB-mutant pheochromocytomas (PC) and paragangliomas (PG) are rare tumours with a high propensity to metastasize although their clinical behaviour is unpredictable. To characterize the genomic landscape of these tumours and identify metastasis biomarkers, we performed multi-omic analysis on 94 tumours from 79 patients using seven molecular methods. Sympathetic (chromaffin cell) and parasympathetic (non-chromaffin cell) PCPG had distinct molecular profiles reflecting their cell-of-origin and biochemical profile. TERT and ATRX-alterations were associated with metastatic PCPG and these tumours had an increased mutation load, and distinct transcriptional and telomeric features. Most PCPG had quiet genomes with some rare co-operative driver events observed, including EPAS1/HIF-2α mutations. Two mechanisms of acquired resistance to DNA alkylating chemotherapies were also detected - MGMT overexpression and mismatch repair-deficiency causing hypermutation. Our comprehensive multi-omic analysis of SDHB-mutant PCPG therefore identified features of metastatic disease and treatment response, expanding our understanding of these rare neuroendocrine tumours.
Collapse
Affiliation(s)
- Aidan Flynn
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Andrew D. Pattison
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Shiva Balachander
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Emma Boehm
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Blake Bowen
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Trisha Dwight
- Kolling Institute of Medical Research, Royal North Shore Hospital St Leonards NSW, Australia
| | | | - Oliver Hofmann
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Luciano Martelotto
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | | | - Lawrence S. Kirschner
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | | | - Lauren Fishbein
- Department of Medicine, Division of Endocrinology, Metabolism, Diabetes, University of Colorado, Aurora, CO, USA
| | - Anthony J Gill
- Sydney Medical School, University of Sydney, Sydney NSW, Australia
- NSW Health Pathology, Department of Anatomical Pathology, Royal North Shore Hospital, St Leonards NSW, Australia
| | | | | | - Tamara Prodanov
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Jane R Noble
- Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Roger R Reddel
- Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Alison H. Trainer
- Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, VIC, Australia
| | - Hans Kumar Ghayee
- University of Florida and Malcom Randall VA Medical Center, Gainesville, FL, USA
| | | | - Marianne Elston
- Waikato Clinical Campus, University of Auckland, Hamilton, New Zealand
| | | | | | - Rodney J Hicks
- St Vincent’s Dept of Medicine, University of Melbourne, VIC, Australia
| | - Joakim Crona
- 18a Department of Medical Sciences, 18b Department of Surgical Sciences, Uppsala University, Sweden
| | - Tobias Åkerström
- 18a Department of Medical Sciences, 18b Department of Surgical Sciences, Uppsala University, Sweden
| | - Peter Stålberg
- 18a Department of Medical Sciences, 18b Department of Surgical Sciences, Uppsala University, Sweden
| | - Patricia Dahia
- Div. Hematology and Medical Oncology, Department of Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio (UTHSCSA), TX, USA
| | - Sean Grimmond
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
| | - Roderick Clifton-Bligh
- Kolling Institute of Medical Research, Royal North Shore Hospital St Leonards NSW, Australia
- Sydney Medical School, University of Sydney, Sydney NSW, Australia
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Richard W Tothill
- Centre for Cancer Research and Department of Clinical Pathology, University of Melbourne, VIC, Australia
- Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| |
Collapse
|
29
|
Graham MK, Xu B, Davis C, Meeker AK, Heaphy CM, Yegnasubramanian S, Dyer MA, Zeineldin M. The TERT Promoter is Polycomb-Repressed in Neuroblastoma Cells with Long Telomeres. CANCER RESEARCH COMMUNICATIONS 2024; 4:1533-1547. [PMID: 38837897 PMCID: PMC11188873 DOI: 10.1158/2767-9764.crc-22-0287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 05/04/2023] [Accepted: 05/22/2024] [Indexed: 06/07/2024]
Abstract
Acquiring a telomere maintenance mechanism is a hallmark of high-risk neuroblastoma and commonly occurs by expressing telomerase (TERT). Telomerase-negative neuroblastoma has long telomeres and utilizes the telomerase-independent alternative lengthening of telomeres (ALT) mechanism. Conversely, no discernable telomere maintenance mechanism is detected in a fraction of neuroblastoma with long telomeres. Here, we show, unlike most cancers, DNA of the TERT promoter is broadly hypomethylated in neuroblastoma. In telomerase-positive neuroblastoma cells, the hypomethylated DNA promoter is approximately 1.5 kb. The TERT locus shows active chromatin marks with low enrichment for the repressive mark, H3K27me3. MYCN, a commonly amplified oncogene in neuroblstoma, binds to the promoter and induces TERT expression. Strikingly, in neuroblastoma with long telomeres, the hypomethylated region spans the entire TERT locus, including multiple nearby genes with enrichment for the repressive H3K27me3 chromatin mark. Furthermore, subtelomeric regions showed enrichment of repressive chromatin marks in neuroblastomas with long telomeres relative to those with short telomeres. These repressive marks were even more evident at the genic loci, suggesting a telomere position effect (TPE). Inhibiting H3K27 methylation by three different EZH2 inhibitors induced the expression of TERT in cell lines with long telomeres and H3K27me3 marks in the promoter region. EZH2 inhibition facilitated MYCN binding to the TERT promoter in neuroblastoma cells with long telomeres. Taken together, these data suggest that epigenetic regulation of TERT expression differs in neuroblastoma depending on the telomere maintenance status, and H3K27 methylation is important in repressing TERT expression in neuroblastoma with long telomeres. SIGNIFICANCE The epigenetic landscape of the TERT locus is unique in neuroblastoma. The DNA at the TERT locus, unlike other cancer cells and similar to normal cells, are hypomethylated in telomerase-positive neuroblastoma cells. The TERT locus is repressed by polycomb repressive complex-2 complex in neuroblastoma cells that have long telomeres and do not express TERT. Long telomeres in neuroblastoma cells are also associated with repressive chromatin states at the chromosomal termini, suggesting TPE.
Collapse
Affiliation(s)
- Mindy K. Graham
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Urology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Christine Davis
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alan K. Meeker
- Department of Urology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher M. Heaphy
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Srinivasan Yegnasubramanian
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael A. Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee
- Howard Hughes Medical Institute, Chevy Chase, Maryland
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Maged Zeineldin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
30
|
Iñiguez-Muñoz S, Llinàs-Arias P, Ensenyat-Mendez M, Bedoya-López AF, Orozco JIJ, Cortés J, Roy A, Forsberg-Nilsson K, DiNome ML, Marzese DM. Hidden secrets of the cancer genome: unlocking the impact of non-coding mutations in gene regulatory elements. Cell Mol Life Sci 2024; 81:274. [PMID: 38902506 PMCID: PMC11335195 DOI: 10.1007/s00018-024-05314-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 12/07/2023] [Accepted: 06/06/2024] [Indexed: 06/22/2024]
Abstract
Discoveries in the field of genomics have revealed that non-coding genomic regions are not merely "junk DNA", but rather comprise critical elements involved in gene expression. These gene regulatory elements (GREs) include enhancers, insulators, silencers, and gene promoters. Notably, new evidence shows how mutations within these regions substantially influence gene expression programs, especially in the context of cancer. Advances in high-throughput sequencing technologies have accelerated the identification of somatic and germline single nucleotide mutations in non-coding genomic regions. This review provides an overview of somatic and germline non-coding single nucleotide alterations affecting transcription factor binding sites in GREs, specifically involved in cancer biology. It also summarizes the technologies available for exploring GREs and the challenges associated with studying and characterizing non-coding single nucleotide mutations. Understanding the role of GRE alterations in cancer is essential for improving diagnostic and prognostic capabilities in the precision medicine era, leading to enhanced patient-centered clinical outcomes.
Collapse
Affiliation(s)
- Sandra Iñiguez-Muñoz
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Pere Llinàs-Arias
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Miquel Ensenyat-Mendez
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Andrés F Bedoya-López
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain
| | - Javier I J Orozco
- Saint John's Cancer Institute, Providence Saint John's Health Center, Santa Monica, CA, USA
| | - Javier Cortés
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, 08017, Barcelona, Spain
- Medica Scientia Innovation Research SL (MEDSIR), 08018, Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, 28670, Madrid, Spain
| | - Ananya Roy
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Karin Forsberg-Nilsson
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- University of Nottingham Biodiscovery Institute, Nottingham, UK
| | - Maggie L DiNome
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Diego M Marzese
- Cancer Epigenetics Laboratory at the Cancer Cell Biology Group, Institut d'Investigació Sanitària Illes Balears (IdISBa), Palma, Spain.
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
31
|
Liu Z, Hu Y, Xie H, Chen K, Wen L, Fu W, Zhou X, Tang F. Single-Cell Chromatin Accessibility Analysis Reveals the Epigenetic Basis and Signature Transcription Factors for the Molecular Subtypes of Colorectal Cancers. Cancer Discov 2024; 14:1082-1105. [PMID: 38445965 DOI: 10.1158/2159-8290.cd-23-1445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/06/2024] [Accepted: 03/04/2024] [Indexed: 03/07/2024]
Abstract
Colorectal cancer is a highly heterogeneous disease, with well-characterized subtypes based on genome, DNA methylome, and transcriptome signatures. To chart the epigenetic landscape of colorectal cancers, we generated a high-quality single-cell chromatin accessibility atlas of epithelial cells for 29 patients. Abnormal chromatin states acquired in adenomas were largely retained in colorectal cancers, which were tightly accompanied by opposite changes of DNA methylation. Unsupervised analysis on malignant cells revealed two epigenetic subtypes, exactly matching the iCMS classification, and key iCMS-specific transcription factors (TFs) were identified, including HNF4A and PPARA for iCMS2 tumors and FOXA3 and MAFK for iCMS3 tumors. Notably, subtype-specific TFs bind to distinct target gene sets and contribute to both interpatient similarities and diversities for both chromatin accessibilities and RNA expressions. Moreover, we identified CpG-island methylator phenotypes and pinpointed chromatin state signatures and TF regulators for the CIMP-high subtype. Our work systematically revealed the epigenetic basis of the well-known iCMS and CIMP classifications of colorectal cancers. SIGNIFICANCE Our work revealed the epigenetic basis of the well-known iCMS and CIMP classifications of colorectal cancers. Moreover, interpatient minor similarities and major diversities of chromatin accessibility signatures of TF target genes can faithfully explain the corresponding interpatient minor similarities and major diversities of RNA expression signatures of colorectal cancers, respectively. This article is featured in Selected Articles from This Issue, p. 897.
Collapse
Affiliation(s)
- Zhenyu Liu
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery, Third Hospital, Peking University, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
| | - Yuqiong Hu
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery, Third Hospital, Peking University, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
- Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Haoling Xie
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery, Third Hospital, Peking University, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Kexuan Chen
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery, Third Hospital, Peking University, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
| | - Lu Wen
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery, Third Hospital, Peking University, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
| | - Wei Fu
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery, Third Hospital, Peking University, Beijing, China
- Peking University Third Hospital Cancer Center, Beijing, China
| | - Xin Zhou
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery, Third Hospital, Peking University, Beijing, China
- Peking University Third Hospital Cancer Center, Beijing, China
| | - Fuchou Tang
- School of Life Sciences, Biomedical Pioneering Innovation Center, Department of General Surgery, Third Hospital, Peking University, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| |
Collapse
|
32
|
Waitkus MS, Erman EN, Reitman ZJ, Ashley DM. Mechanisms of telomere maintenance and associated therapeutic vulnerabilities in malignant gliomas. Neuro Oncol 2024; 26:1012-1024. [PMID: 38285162 PMCID: PMC11145458 DOI: 10.1093/neuonc/noae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Indexed: 01/30/2024] Open
Abstract
A majority of cancers (~85%) activate the enzyme telomerase to maintain telomere length over multiple rounds of cellular division. Telomerase-negative cancers activate a distinct, telomerase-independent mechanism of telomere maintenance termed alternative lengthening of telomeres (ALT). ALT uses homologous recombination to maintain telomere length and exhibits features of break-induced DNA replication. In malignant gliomas, the activation of either telomerase or ALT is nearly ubiquitous in pediatric and adult tumors, and the frequency with which these distinct telomere maintenance mechanisms (TMMs) is activated varies according to genetically defined glioma subtypes. In this review, we summarize the current state of the field of TMMs and their relevance to glioma biology and therapy. We review the genetic alterations and molecular mechanisms leading to telomerase activation or ALT induction in pediatric and adult gliomas. With this background, we review emerging evidence on strategies for targeting TMMs for glioma therapy. Finally, we comment on critical gaps and issues for moving the field forward to translate our improved understanding of glioma telomere maintenance into better therapeutic strategies for patients.
Collapse
Affiliation(s)
- Matthew S Waitkus
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Elise N Erman
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Zachary J Reitman
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - David M Ashley
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
33
|
Ng YB, Akincilar SC. Shaping DNA damage responses: Therapeutic potential of targeting telomeric proteins and DNA repair factors in cancer. Curr Opin Pharmacol 2024; 76:102460. [PMID: 38776747 DOI: 10.1016/j.coph.2024.102460] [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: 12/10/2022] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 05/25/2024]
Abstract
Shelterin proteins regulate genomic stability by preventing inappropriate DNA damage responses (DDRs) at telomeres. Unprotected telomeres lead to persistent DDR causing cell cycle inhibition, growth arrest, and apoptosis. Cancer cells rely on DDR to protect themselves from DNA lesions and exogenous DNA-damaging agents such as chemotherapy and radiotherapy. Therefore, targeting DDR machinery is a promising strategy to increase the sensitivity of cancer cells to existing cancer therapies. However, the success of these DDR inhibitors depends on other mutations, and over time, patients develop resistance to these therapies. This suggests the need for alternative approaches. One promising strategy is co-inhibiting shelterin proteins with DDR molecules, which would offset cellular fitness in DNA repair in a mutation-independent manner. This review highlights the associations and dependencies of the shelterin complex with the DDR proteins and discusses potential co-inhibition strategies that might improve the therapeutic potential of current inhibitors.
Collapse
Affiliation(s)
- Yu Bin Ng
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore
| | - Semih Can Akincilar
- Laboratory of NFκB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Republic of Singapore.
| |
Collapse
|
34
|
Liu X, Wang J, Wu LJ, Trinh B, Tsai RYL. IMPDH Inhibition Decreases TERT Expression and Synergizes the Cytotoxic Effect of Chemotherapeutic Agents in Glioblastoma Cells. Int J Mol Sci 2024; 25:5992. [PMID: 38892179 PMCID: PMC11172490 DOI: 10.3390/ijms25115992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/21/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
IMP dehydrogenase (IMPDH) inhibition has emerged as a new target therapy for glioblastoma multiforme (GBM), which remains one of the most refractory tumors to date. TCGA analyses revealed distinct expression profiles of IMPDH isoenzymes in various subtypes of GBM and low-grade glioma (LGG). To dissect the mechanism(s) underlying the anti-tumor effect of IMPDH inhibition in adult GBM, we investigated how mycophenolic acid (MPA, an IMPDH inhibitor) treatment affected key oncogenic drivers in glioblastoma cells. Our results showed that MPA decreased the expression of telomerase reverse transcriptase (TERT) in both U87 and U251 cells, and the expression of O6-methylguanine-DNA methyltransferase (MGMT) in U251 cells. In support, MPA treatment reduced the amount of telomere repeats in U87 and U251 cells. TERT downregulation by MPA was associated with a significant decrease in c-Myc (a TERT transcription activator) in U87 but not U251 cells, and a dose-dependent increase in p53 and CCCTC-binding factor (CTCF) (TERT repressors) in both U87 and U251 cells. In U251 cells, MPA displayed strong cytotoxic synergy with BCNU and moderate synergy with irinotecan, oxaliplatin, paclitaxel, or temozolomide (TMZ). In U87 cells, MPA displayed strong cytotoxic synergy with all except TMZ, acting primarily through the apoptotic pathway. Our work expands the mechanistic potential of IMPDH inhibition to TERT/telomere regulation and reveals a synthetic lethality between MPA and anti-GBM drugs.
Collapse
Affiliation(s)
- Xiaoqin Liu
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (X.L.); (J.W.); (L.J.W.); (B.T.)
| | - Junying Wang
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (X.L.); (J.W.); (L.J.W.); (B.T.)
| | - Laura J. Wu
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (X.L.); (J.W.); (L.J.W.); (B.T.)
| | - Britni Trinh
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (X.L.); (J.W.); (L.J.W.); (B.T.)
| | - Robert Y. L. Tsai
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX 77030, USA; (X.L.); (J.W.); (L.J.W.); (B.T.)
- Department of Translational Medical Sciences, College of Medicine, Texas A&M University Health Science Center, Houston, TX 77030, USA
| |
Collapse
|
35
|
Chen H, Han Z, Su J, Song X, Ma Q, Lin Y, Ran Z, Li X, Mou R, Wang Y, Li D. Ferroptosis and hepatocellular carcinoma: the emerging role of lncRNAs. Front Immunol 2024; 15:1424954. [PMID: 38846953 PMCID: PMC11153672 DOI: 10.3389/fimmu.2024.1424954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 06/09/2024] Open
Abstract
Hepatocellular carcinoma is the most common form of primary liver cancer and poses a significant challenge to the medical community because of its high mortality rate. In recent years, ferroptosis, a unique form of cell death, has garnered widespread attention. Ferroptosis, which is characterized by iron-dependent lipid peroxidation and mitochondrial alterations, is closely associated with the pathological processes of various diseases, including hepatocellular carcinoma. Long non-coding RNAs (lncRNAs), are a type of functional RNA, and play crucial regulatory roles in a variety of biological processes. In this manuscript, we review the regulatory roles of lncRNAs in the key aspects of ferroptosis, and summarize the research progress on ferroptosis-related lncRNAs in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Haoran Chen
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Zhongyu Han
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Junyan Su
- The First People’s Hospital of Longquanyi District, Chengdu, China
| | - Xuanliang Song
- The First People’s Hospital of Longquanyi District, Chengdu, China
| | - Qingquan Ma
- The First People’s Hospital of Longquanyi District, Chengdu, China
| | - Yumeng Lin
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Zijin Ran
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Xueping Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rongkun Mou
- Department of General Surgery, The Third Hospital of Mianyang, Mianyang, China
| | - Yi Wang
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| | - Dongxuan Li
- Department of General Surgery, Chengdu Xinhua Hospital Affiliated to North Sichuan Medical College, Chengdu, China
| |
Collapse
|
36
|
Liu R, Zhu G, Tan J, Shen X, Xing M. Genetic trio of BRAF and TERT alterations and rs2853669TT in papillary thyroid cancer aggressiveness. J Natl Cancer Inst 2024; 116:694-701. [PMID: 38113409 PMCID: PMC11077312 DOI: 10.1093/jnci/djad265] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/20/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND BRAF V600E and TERT promoter alterations are core components in current genetics-based risk assessment for precision management of papillary thyroid cancer. It remains unknown whether this approach could achieve even better precision through a widely recognized prognostic single-nucleotide variation (SNV, formerly SNP), rs2853669T>C, in the TERT promoter. METHODS The genetic status of alterations and SNV were examined by sequencing genomic DNA from papillary thyroid cancer in 608 patients (427 women and 181 men) aged 47 years (interquartile range = 37-57), with a median follow-up time of 75 months (interquartile range = 36-123), and their relationship with clinical outcomes was analyzed. A luciferase reporter assay was performed to examine TERT promoter activities. RESULTS TERT promoter alterations showed a strong association with papillary thyroid cancer recurrence in the presence of genotype TT of rs2853669 (adjusted hazard ratio [HR] = 2.12, 95% confidence interval [CI] = 1.10 to 4.12) but not TC/CC (adjusted HR = 1.17, 95% CI = 0.56 to 2.41). TERT and BRAF alterations commonly coexisted and synergistically promoted papillary thyroid cancer recurrence. With this genetic duet, TT of rs2853669 showed a robustly higher disease recurrence than TC/CC (adjusted HR = 14.26, 95% CI = 2.86 to 71.25). Patients with the genetic trio of BRAF V600E, TERT alteration, and TT of rs2853669 had a recurrence of 76.5% vs recurrence of 8.4% with neither variation and with TC/CC (HR = 13.48, 95% CI = 6.44 to 28.21). T allele of rs2853669 strongly increased TERT promoter activities, particularly the variant promoters. CONCLUSIONS The SNV rs2853669T>C dramatically refines the prognostic power of BRAF V600E and TERT promoter alterations to a higher precision, suggesting the need for including this SNV in the current genetics-based risk prognostication of papillary thyroid cancer.
Collapse
Affiliation(s)
- Rengyun Liu
- Laboratory for Cellular and Molecular Thyroid Research, Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Guangwu Zhu
- Laboratory for Cellular and Molecular Thyroid Research, Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jie Tan
- Laboratory for Cellular and Molecular Thyroid Research, Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaopei Shen
- Laboratory for Cellular and Molecular Thyroid Research, Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mingzhao Xing
- Laboratory for Cellular and Molecular Thyroid Research, Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
37
|
Tang Q, Hu G, Sang Y, Chen Y, Wei G, Zhu M, Chen M, Li S, Liu R, Peng Z. Therapeutic targeting of PLK1 in TERT promoter-mutant hepatocellular carcinoma. Clin Transl Med 2024; 14:e1703. [PMID: 38769666 PMCID: PMC11106514 DOI: 10.1002/ctm2.1703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Hotspot mutations in the promoter of telomerase reverse transcriptase (TERT) gene are the most common genetic variants in hepatocellular carcinoma (HCC) and associated with poor prognosis of the disease. However, no drug was currently approved for treating TERT promoter mutation positive HCC patients. Here, we aim to explore the potential therapeutic strategy for targeting TERT promoter mutation in HCC. METHODS The Liver Cancer Model Repository database was used for screening potential drugs to selectively suppress the growth of TERT promoter mutant HCC cells. RNA-seq, CRISPR-Cas9 technology and siRNA transfection were performed for mechanistic studies. Cell counting kit-8 (CCK8) assay and the xenograft tumour models were used for cell growth detection in vitro and in vivo, respectively. Cell apoptosis and cell cycle arrest were analysed by Annexin V-FITC staining and/or propidium iodide staining. RESULTS PLK1 inhibitors were remarkably more sensitive to HCC cells harbouring TERT promoter mutation than wild-type cells in vitro and in vivo, which were diminished after TERT promoter mutation was edited to the wild-type nucleotide. Comparing the HCC cells with wild-type promoter of TERT, PLK1 inhibitors specifically downregulated Smad3 to regulate TERT for inducing apoptosis and G2/M arrest in TERT mutant HCC cells. Moreover, knockout of Smad3 counteracted the effects of PLK1 inhibitors in TERT mutant HCC cells. Finally, a cooperative effect of PLK1 and Smad3 inhibition was observed in TERT mutant cells. CONCLUSIONS PLK1 inhibition selectively suppressed the growth of TERT mutant HCC cells through Smad3, thus contributed to discover a novel therapeutic strategy to treat HCC patients harbouring TERT promoter mutations. KEY POINTS TERT promoter mutation confers sensitivity to PLK1 inhibitors in HCC. The selective growth inhibition of TERT mutant HCC cells induced by PLK1 inhibitor was mediated by Smad3. Combined inhibition of PLK1 and Smad3 showed a cooperative anti-tumor effect in TERT mutant HCC cells.
Collapse
Affiliation(s)
- Qin Tang
- Department of Radiation OncologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Guanghui Hu
- Institute of Precision MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Ye Sang
- Institute of Precision MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Yulu Chen
- Institute of Precision MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Guangyan Wei
- Department of Radiation OncologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Meiyan Zhu
- Department of Radiation OncologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Mengke Chen
- Institute of Precision MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Shiyong Li
- Institute of Precision MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Rengyun Liu
- Institute of Precision MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Zhenwei Peng
- Department of Radiation OncologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
- Cancer CenterThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
38
|
Mu Z, Zhang X, Sun D, Sun Y, Shi C, Ju G, Kai Z, Huang L, Chen L, Liang J, Lin Y. Characterizing Genetic Alterations Related to Radioiodine Avidity in Metastatic Thyroid Cancer. J Clin Endocrinol Metab 2024; 109:1231-1240. [PMID: 38060243 PMCID: PMC11031230 DOI: 10.1210/clinem/dgad697] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 10/31/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
CONTEXT Patients with differentiated thyroid cancer (DTC) with distant metastasis (DM) are usually not recognized as radioactive iodine (RAI)-refractory DTC in a timely manner. The elucidation of genetic features related to RAI uptake patterns may shed light on the early recognition of RAI-refractory DTC. OBJECTIVE This work aimed to elucidate the underlying molecular features behind different RAI uptake patterns. METHODS A total of 214 patients with DM-DTC were retrospectively included in the analysis. RAI uptake patterns were defined as initially RAI refractory (I-RAIR) and initially RAI avid (I-RAIA) according to the first post-treatment scan, then I-RAIA was further divided into continually RAIA (C-RAIA), partly RAIR (P-RAIR), and gradually RAIR (G-RAIR) according to subsequent scans. The molecular subtype groups-BRAFV600E mutated, RAS mutated, fusions, and others-were classified according to main driver genes status. RESULTS BRAF, TERT promoter, and TP53 mutations are more frequently detected in the I-RAIR pattern while RET fusions and RAS mutations are more frequent in the I-RAIA pattern. A late-hit mutation including TERT, TP53, or PIK3CA is more common in I-RAIR than that in I-RAIA (50.0% vs 26.9%, P = .001), particularly for those with RAS mutations in the I-RAIR group, always accompanied by TERT promoter. Isolated RET fusions accounts for 10% of I-RAIR. When compared among driver gene groups, BRAFV600E-mutated tumors have a higher rate of the I-RAIR pattern (64.4%) than RAS-mutated (4.5%, P < .001) and fusion-positive (20.7%, P < .001) tumors. In I-RAIA subgroups, BRAFV600E-mutated tumors have lower prevalence of the C-RAIA pattern than those with RAS mutation or fusions. CONCLUSION Patients with the I-RAIR pattern predominantly featured mutations of the BRAF and/or TERT promoter, of which RAS mutations were usually accompanied by late-hit mutations, while fusions mostly occurred alone.
Collapse
Affiliation(s)
- Zhuanzhuan Mu
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Xin Zhang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Di Sun
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Yuqing Sun
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Cong Shi
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Gaoda Ju
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
- Department of Medical Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
- Department of Oncology, Peking University International Hospital, Peking University, Beijing, 102206, China
| | - Zhentian Kai
- Department of Bioinformatics, Zhejiang Shaoxing Topgen Biomedical Technology Co., Ltd, Shanghai, 201321, China
| | - Lisha Huang
- Department of Medicine, Zhejiang Shaoxing Topgen Biomedical Technology Co., Ltd, Shanghai, 201321, China
| | - Libo Chen
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Jun Liang
- Department of Medical Oncology, Key Laboratory of Carcinogenesis & Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, 100142, China
- Department of Oncology, Peking University International Hospital, Peking University, Beijing, 102206, China
| | - Yansong Lin
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Beijing, 100730, China
| |
Collapse
|
39
|
Appin CL, Hong C, Suwala AK, Hilz S, Mathur R, Solomon DA, Smirnov IV, Stevers NO, Shai A, Wang A, Berger MS, Chang SM, Phillips JJ, Costello JF. Whole tumor analysis reveals early origin of the TERT promoter mutation and intercellular heterogeneity in TERT expression. Neuro Oncol 2024; 26:640-652. [PMID: 38141254 PMCID: PMC10995505 DOI: 10.1093/neuonc/noad231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Indexed: 12/25/2023] Open
Abstract
BACKGROUND The TERT promoter mutation (TPM) is acquired in most IDH-wildtype glioblastomas (GBM) and IDH-mutant oligodendrogliomas (OD) enabling tumor cell immortality. Previous studies on TPM clonality show conflicting results. This study was performed to determine whether TPM is clonal on a tumor-wide scale. METHODS We investigated TPM clonality in relation to presumed early events in 19 IDH-wildtype GBM and 10 IDH-mutant OD using 3-dimensional comprehensive tumor sampling. We performed Sanger sequencing on 264 tumor samples and deep amplicon sequencing on 187 tumor samples. We obtained tumor purity and copy number estimates from whole exome sequencing. TERT expression was assessed by RNA-seq and RNAscope. RESULTS We detected TPM in 100% of tumor samples with quantifiable tumor purity (219 samples). Variant allele frequencies (VAF) of TPM correlate positively with chromosome 10 loss in GBM (R = 0.85), IDH1 mutation in OD (R = 0.87), and with tumor purity (R = 0.91 for GBM; R = 0.90 for OD). In comparison, oncogene amplification was tumor-wide for MDM4- and most EGFR-amplified cases but heterogeneous for MYCN and PDGFRA, and strikingly high in low-purity samples. TPM VAF was moderately correlated with TERT expression (R = 0.52 for GBM; R = 0.65 for OD). TERT expression was detected in a subset of cells, solely in TPM-positive samples, including samples equivocal for tumor. CONCLUSIONS On a tumor-wide scale, TPM is among the earliest events in glioma evolution. Intercellular heterogeneity of TERT expression, however, suggests dynamic regulation during tumor growth. TERT expression may be a tumor cell-specific biomarker.
Collapse
Affiliation(s)
- Christina L Appin
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Chibo Hong
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Abigail K Suwala
- Department of Neurological Surgery, University of California, San Francisco, California, USA
- Department of Neuropathology, University of Heidelberg, Institute of Pathology, Heidelberg, Germany
| | - Stephanie Hilz
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Radhika Mathur
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - David A Solomon
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Ivan V Smirnov
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Nicholas O Stevers
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Anny Shai
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Albert Wang
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Susan M Chang
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Joanna J Phillips
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Joseph F Costello
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| |
Collapse
|
40
|
Mu Z, Zhang X, Liang D, Fang J, Chen G, Guo W, Sun D, Sun Y, Kai Z, Huang L, Liang J, Lin Y. Risk stratification for radioactive iodine refractoriness using molecular alterations in distant metastatic differentiated thyroid cancer. Chin J Cancer Res 2024; 36:25-35. [PMID: 38455372 PMCID: PMC10915639 DOI: 10.21147/j.issn.1000-9604.2024.01.03] [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: 09/12/2023] [Accepted: 02/01/2024] [Indexed: 03/09/2024] Open
Abstract
Objective Patients with radioactive iodine-refractory differentiated thyroid cancer (RAIR-DTC) are often diagnosed with delay and constrained to limited treatment options. The correlation between RAI refractoriness and the underlying genetic characteristics has not been extensively studied. Methods Adult patients with distant metastatic DTC were enrolled and assigned to undergo next-generation sequencing of a customized 26-gene panel (ThyroLead). Patients were classified into RAIR-DTC or non-RAIR groups to determine the differences in clinicopathological and molecular characteristics. Molecular risk stratification (MRS) was constructed based on the association between molecular alterations identified and RAI refractoriness, and the results were classified as high, intermediate or low MRS. Results A total of 220 patients with distant metastases were included, 63.2% of whom were identified as RAIR-DTC. Genetic alterations were identified in 90% of all the patients, with BRAF (59.7% vs. 17.3%), TERT promoter (43.9% vs. 7.4%), and TP53 mutations (11.5% vs. 3.7%) being more prevalent in the RAIR-DTC group than in the non-RAIR group, except for RET fusions (15.8% vs. 39.5%), which had the opposite pattern. BRAF and TERT promoter are independent predictors of RAIR-DTC, accounting for 67.6% of patients with RAIR-DTC. MRS was strongly associated with RAI refractoriness (P<0.001), with an odds ratio (OR) of high to low MRS of 7.52 [95% confidence interval (95% CI), 3.96-14.28; P<0.001] and an OR of intermediate to low MRS of 3.20 (95% CI, 1.01-10.14; P=0.041). Conclusions Molecular alterations were associated with RAI refractoriness, with BRAF and TERT promoter mutations being the predominant contributors, followed by TP53 and DICER1 mutations. MRS might serve as a valuable tool for both prognosticating clinical outcomes and directing precision-based therapeutic interventions.
Collapse
Affiliation(s)
- Zhuanzhuan Mu
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Xin Zhang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Dongquan Liang
- Pepperdine University Graduate School of Education and Psychology, Los Angeles 90045, USA
| | - Jugao Fang
- Department of Head and Neck Surgery, Affiliated Beijing Tongren Hospital, Capital Medical University, Beijing 100730, China
| | - Ge Chen
- Department of General Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing 100730, China
| | - Wenting Guo
- Central Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing 100730, China
| | - Di Sun
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Yuqing Sun
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| | - Zhentian Kai
- Department of Bioinformatics, Zhejiang Shaoxing Topgen Biomedical Technology Co., Ltd, Shanghai 201321, China
| | - Lisha Huang
- Department of Medicine, Zhejiang Shaoxing Topgen Biomedical Technology Co., Ltd, Shanghai 201321, China
| | - Jun Liang
- Department of Oncology, Peking University International Hospital, Beijing 102206, China
| | - Yansong Lin
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Chinese Academy of Medical Sciences & PUMC, Beijing 100730, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing 100730, China
| |
Collapse
|
41
|
Shin E, Kwon Y, Jung E, Kim YJ, Kim C, Hong S, Kim J. TM4SF19 controls GABP-dependent YAP transcription in head and neck cancer under oxidative stress conditions. Proc Natl Acad Sci U S A 2024; 121:e2314346121. [PMID: 38315837 PMCID: PMC10873613 DOI: 10.1073/pnas.2314346121] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/04/2024] [Indexed: 02/07/2024] Open
Abstract
Tobacco and alcohol are risk factors for human papillomavirus-negative head and neck squamous cell carcinoma (HPV- HNSCC), which arises from the mucosal epithelium of the upper aerodigestive tract. Notably, despite the mutagenic potential of smoking, HPV- HNSCC exhibits a low mutational load directly attributed to smoking, which implies an undefined role of smoking in HPV- HNSCC. Elevated YAP (Yes-associated protein) mRNA is prevalent in HPV- HNSCC, irrespective of the YAP gene amplification status, and the mechanism behind this upregulation remains elusive. Here, we report that oxidative stress, induced by major risk factors for HPV- HNSCC such as tobacco and alcohol, promotes YAP transcription via TM4SF19 (transmembrane 4 L six family member 19). TM4SF19 modulates YAP transcription by interacting with the GABP (Guanine and adenine-binding protein) transcription factor complex. Mechanistically, oxidative stress induces TM4SF19 dimerization and topology inversion in the endoplasmic reticulum membrane, which in turn protects the GABPβ1 subunit from proteasomal degradation. Conversely, depletion of TM4SF19 impairs the survival, proliferation, and migration of HPV- HNSCC cells, highlighting the potential therapeutic relevance of targeting TM4SF19. Our findings reveal the roles of the key risk factors of HPV- HNSCC in tumor development via oxidative stress, offering implications for upcoming therapeutic approaches in HPV- HNSCC.
Collapse
Affiliation(s)
- Eunbie Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Korea
| | - Yongsoo Kwon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Korea
| | - Eunji Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Korea
| | - Yong Joon Kim
- Department of Ophthalmology, Severance Hospital, Institute of Vision Research, Yonsei University College of Medicine, Seoul03722, South Korea
| | - Changgon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Korea
| | - Semyeong Hong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Korea
| | - Joon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Korea
| |
Collapse
|
42
|
Koca DS, Kolpakov V, Ihlow J, von Laffert M, Erb-Eigner K, Herbst H, Kriese K, Schweizer L, Bertelmann E. Prevalence of TERT Promoter Mutations in Orbital Solitary Fibrous Tumors. Curr Issues Mol Biol 2024; 46:1467-1484. [PMID: 38392213 PMCID: PMC10887834 DOI: 10.3390/cimb46020095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/24/2024] Open
Abstract
The orbital manifestation of a solitary fibrous tumor (SFT) is exceptionally rare and poses specific challenges in diagnosis and treatment. Its rather exceptional behavior among all SFTs comprises a high tendency towards local recurrence, but it rarely culminates in metastatic disease. This raises the question of prognostic factors in orbital SFTs (oSFTs). Telomerase reverse transcriptase (TERT)-promoter mutations have previously been linked to an unfavorable prognosis in SFTs of other locations. We analyzed the prevalence of TERT promoter mutations of SFTs in the orbital compartment. We performed a retrospective, descriptive clinico-histopathological analysis of nine cases of oSFTs between the years of 2017 and 2021. A TERT promoter mutation was present in one case, which was classified with intermediate metastatic risk. Local recurrence or progress occurred in six cases after primary resection; no distant metastases were reported. Multimodal imaging repeatedly showed particular morphologic patterns, including tubular vascular structures and ADC reduction. The prevalence of the TERT promoter mutation in oSFT was 11%, which is similar to the prevalence of extra-meningeal SFTs of the head and neck and lower than that in other extra-meningeal compartments. In the present study, the TERT promoter mutation in oSFT manifested in a case with an unfavorable prognosis, comprising aggressive local tumor growth, local recurrence, and eye loss.
Collapse
Affiliation(s)
- David Sinan Koca
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Vladimir Kolpakov
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Jana Ihlow
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Berlin Institute of Health Charité Clinician Scientist Program, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin Institute of Health Biomedical Innovation Academy, Anna-Louisa-Karsch-Str., 210178 Berlin, Germany
| | - Maximilian von Laffert
- Institute of Pathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Department of Diagnostics, Institute of Pathology, Universitätsklinikum Leipzig AöR, Liebigstraße 26, 04103 Leipzig, Germany
| | - Katharina Erb-Eigner
- Department of Radiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Hermann Herbst
- Department of Pathology, Vivantes Hospital Neukölln, Vivantes Netzwerk für Gesundheit GmbH Berlin, Rudower Straße 48, 12351 Berlin, Germany
| | - Karen Kriese
- Department of Pathology, Vivantes Hospital Neukölln, Vivantes Netzwerk für Gesundheit GmbH Berlin, Rudower Straße 48, 12351 Berlin, Germany
| | - Leonille Schweizer
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117 Berlin, Germany
- German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Edinger Institute, Institute of Neurology, University of Frankfurt am Main, 60528 Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt-Mainz, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), 60596 Frankfurt am Main, Germany
| | - Eckart Bertelmann
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
43
|
Beaufrère A, Paisley S, Ba I, Laouirem S, Priori V, Cazier H, Favre L, Cauchy F, Lesurtel M, Calderaro J, Kannengiesser C, Paradis V. Differential diagnosis of small hepatocellular nodules in cirrhosis: surrogate histological criteria of TERT promoter mutations. Histopathology 2024; 84:473-481. [PMID: 37903649 DOI: 10.1111/his.15086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 09/25/2023] [Accepted: 10/14/2023] [Indexed: 11/01/2023]
Abstract
AIMS The differential diagnosis of small hepatocellular nodules in cirrhosis between dysplastic nodules and hepatocellular carcinoma (HCC) remains challenging on biopsy. As TERT promoter (pTERT) mutations may indicate the nodules already engaged in the malignant process, the aim of this study was to identify histological criteria associated with pTERT mutations by detecting these mutations by ddPCR in small formalin-fixed paraffin-embedded (FFPE) hepatocellular nodules arising in cirrhosis. METHODS AND RESULTS We built a bicentric cohort data set of 339 hepatocellular nodules < 2 cm from cirrhotic samples, divided into a test cohort of 299 resected samples and a validation cohort of 40 biopsies. Pathological review, based on the evaluation of 14 histological criteria, classified all nodules. pTERT mutations were identified by ddPCR in FFPE samples. Among the 339 nodules, ddPCR revealed pTERT mutations in 105 cases (31%), including 90 and 15 cases in the test and validation cohorts, respectively. On multivariate analysis, three histological criteria were associated with pTERT mutations in the test cohort: increased cell density (P = 0.003), stromal invasion (P = 0.036) and plate-thickening anomalies (P < 0.001). With the combination of at least two of these major criteria, the AUC for predicting pTERT mutations was 0.84 in the test cohort (sensitivity: 86%, specificity: 83%) and 0.81 in the validation cohort (sensitivity: 87%, specificity: 76%). CONCLUSIONS We identified three histological criteria as surrogate markers of pTERT mutations that may be used in routine biopsy to more clearly classify small hepatocellular nodules arising in cirrhosis.
Collapse
Affiliation(s)
- Aurélie Beaufrère
- Université Paris Cité, Paris, France
- AP-HP.Nord, Department of Pathology, FHU MOSAIC, Beaujon Hospital, Clichy, France
- Centre de Recherche sur l'Inflammation, INSERM UMR 1149, Paris, France
| | - Sarah Paisley
- AP-HP.Nord, Department of Pathology, FHU MOSAIC, Beaujon Hospital, Clichy, France
- Centre de Recherche sur l'Inflammation, INSERM UMR 1149, Paris, France
| | - Ibrahima Ba
- AP-HP.Nord, Department of Molecular Genetics, Bichat Hospital, Paris, France
| | - Samira Laouirem
- Centre de Recherche sur l'Inflammation, INSERM UMR 1149, Paris, France
| | - Victoria Priori
- Centre de Recherche sur l'Inflammation, INSERM UMR 1149, Paris, France
| | - Hélène Cazier
- Centre de Recherche sur l'Inflammation, INSERM UMR 1149, Paris, France
| | - Loëtitia Favre
- AP-HP, Department of Pathology, Henri Mondor Hospital, Créteil, France
| | - François Cauchy
- Centre de Recherche sur l'Inflammation, INSERM UMR 1149, Paris, France
| | - Mickael Lesurtel
- Université Paris Cité, Paris, France
- AP-HP.Nord, Department of HPB Surgery an d Liver Transplantation, Beaujon Hospital, Clichy, France
| | - Julien Calderaro
- AP-HP, Department of Pathology, Henri Mondor Hospital, Créteil, France
| | | | - Valérie Paradis
- Université Paris Cité, Paris, France
- AP-HP.Nord, Department of Pathology, FHU MOSAIC, Beaujon Hospital, Clichy, France
- Centre de Recherche sur l'Inflammation, INSERM UMR 1149, Paris, France
| |
Collapse
|
44
|
Duan M, Song S, Wasserman H, Lee PH, Liu KJ, Gordân R, He Y, Mao P. High UV damage and low repair, but not cytosine deamination, stimulate mutation hotspots at ETS binding sites in melanoma. Proc Natl Acad Sci U S A 2024; 121:e2310854121. [PMID: 38241433 PMCID: PMC10823218 DOI: 10.1073/pnas.2310854121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/20/2023] [Indexed: 01/21/2024] Open
Abstract
Noncoding mutation hotspots have been identified in melanoma and many of them occur at the binding sites of E26 transformation-specific (ETS) proteins; however, their formation mechanism and functional impacts are not fully understood. Here, we used UV (Ultraviolet) damage sequencing data and analyzed cyclobutane pyrimidine dimer (CPD) formation, DNA repair, and CPD deamination in human cells at single-nucleotide resolution. Our data show prominent CPD hotspots immediately after UV irradiation at ETS binding sites, particularly at sites with a conserved TTCCGG motif, which correlate with mutation hotspots identified in cutaneous melanoma. Additionally, CPDs are repaired slower at ETS binding sites than in flanking DNA. Cytosine deamination in CPDs to uracil is suggested as an important step for UV mutagenesis. However, we found that CPD deamination is significantly suppressed at ETS binding sites, particularly for the CPD hotspot on the 5' side of the ETS motif, arguing against a role for CPD deamination in promoting ETS-associated UV mutations. Finally, we analyzed a subset of frequently mutated promoters, including the ribosomal protein genes RPL13A and RPS20, and found that mutations in the ETS motif can significantly reduce the promoter activity. Thus, our data identify high UV damage and low repair, but not CPD deamination, as the main mechanism for ETS-associated mutations in melanoma and uncover important roles of often-overlooked mutation hotspots in perturbing gene transcription.
Collapse
Affiliation(s)
- Mingrui Duan
- Department of Internal Medicine, University of New Mexico Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM87131
| | - Shenghan Song
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM87131
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico, Albuquerque, NM87131
| | - Hana Wasserman
- Program in Computational Biology and Bioinformatics, Department of Biostatistics and Bioinformatics, Duke University, Durham, NC27708
| | - Po-Hsuen Lee
- Department of Internal Medicine, University of New Mexico Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM87131
| | - Ke Jian Liu
- Department of Pathology, Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY11794-7263
| | - Raluca Gordân
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC27708
- Department of Computer Science, Duke University, Durham, NC27708
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC27708
| | - Yi He
- Department of Chemistry and Chemical Biology, University of New Mexico, Albuquerque, NM87131
- Translational Informatics Division, Department of Internal Medicine, University of New Mexico, Albuquerque, NM87131
| | - Peng Mao
- Department of Internal Medicine, University of New Mexico Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM87131
| |
Collapse
|
45
|
Chatterjee S, G K, Y PK. Salivary Chloride Intracellular Channel 1 (CLIC1) as a Hub of Gene-Gene Interactome of Periodontitis With Diabetes Mellitus. Cureus 2024; 16:e51877. [PMID: 38327933 PMCID: PMC10849158 DOI: 10.7759/cureus.51877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/08/2024] [Indexed: 02/09/2024] Open
Abstract
Background and introduction Periodontal disease is one of the most prevalent chronic conditions that affects the oral cavity. Identifying and predicting biomarkers is essential for the prevention of high-morbidity oral diseases. The genomic interaction network identifies common hub genes involved in crucial protein formation in periodontal inflammation. Diabetes mellitus is a metabolic disorder that has a double-edged sword relationship with periodontitis. Chloride intracellular channel 1 (CLIC1) was identified as a hub gene linking the pathogenesis of periodontitis and diabetes mellitus using a bioinformatic tool. Therefore, this current study aimed to assess the concentration of the pro-inflammatory biomarker CLIC1 in saliva among individuals with periodontal health and those with periodontal disease linked to diabetes mellitus. Materials and methods Differentially expressed genes (DEGs) in periodontitis were identified using datasets retrieved from the Gene Expression Omnibus (GEO) database. DEGs were combined to build the network, and GeneMANIA was used to find and rank the interconnecting genes. CLIC1 was identified as the hub gene, and clinical validation was done using patient samples. The study involved 30 participants. Based on clinical and radiographic periodontal findings, they were split into three groups: healthy (group 1, n=10), with periodontitis but no diabetes mellitus (group 2, n=10), and with periodontitis and diabetes mellitus (group 3, n=10). The collection of saliva samples, followed by quantifying these samples, was performed using an enzyme-linked immunosorbent assay (ELISA). Results From network graph analysis, it was discovered that CLIC1 functions as a hub gene in the majority of toll-like receptor pathways. The mean concentration of CLIC1 in saliva increased consistently as the disease was observed in periodontitis patients and periodontitis patients with diabetes mellitus. Conclusion CLIC1 concentrations were positively correlated with periodontitis in individuals with diabetes. Therefore, CLIC1 could be a diagnostic biomarker for patients with periodontitis. However, large-scale studies are needed to confirm more positive associations.
Collapse
Affiliation(s)
- Shubhangini Chatterjee
- Department of Periodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Karthikeyan G
- Department of Periodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Pradeep Kumar Y
- Department of Periodontics, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
46
|
Di Nunno V, Aprile M, Bartolini S, Gatto L, Tosoni A, Ranieri L, De Biase D, Asioli S, Franceschi E. The Biological and Clinical Role of the Telomerase Reverse Transcriptase Gene in Glioblastoma: A Potential Therapeutic Target? Cells 2023; 13:44. [PMID: 38201248 PMCID: PMC10778438 DOI: 10.3390/cells13010044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Glioblastoma IDH-wildtype represents the most lethal and frequent primary tumor of the central nervous system. Thanks to important scientific efforts, we can now investigate its deep genomic assessment, elucidating mutated genes and altered biological mechanisms in addition to its clinical aggressiveness. The telomerase reverse transcriptase gene (TERT) is the most frequently altered gene in solid tumors, including brain tumors and GBM IDH-wildtype. In particular, it can be observed in approximately 80-90% of GBM IDH-wildtype cases. Its clonal distribution on almost all cancer cells makes this gene an optimal target. However, the research of effective TERT inhibitors is complicated by several biological and clinical obstacles which can be only partially surmounted. Very recently, novel immunological approaches leading to TERT inhibition have been investigated, offering the potential to develop an effective target for this altered protein. Here, we perform a narrative review investigating the biological role of TERT alterations on glioblastoma and the principal obstacles associated with TERT inhibitions in this population. Moreover, we discuss possible combination treatment strategies to overcome these limitations.
Collapse
Affiliation(s)
- Vincenzo Di Nunno
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy (E.F.)
| | - Marta Aprile
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, 40138 Bologna, Italy
| | - Stefania Bartolini
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy (E.F.)
| | - Lidia Gatto
- Department of Oncology, Azienda Unità Sanitaria Locale (AUSL) Bologna, 40139 Bologna, Italy
| | - Alicia Tosoni
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy (E.F.)
| | - Lucia Ranieri
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy (E.F.)
| | - Dario De Biase
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Pharmacy and Biotechnology (FaBit), University of Bologna, 40126 Bologna, Italy
| | - Sofia Asioli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Surgical Pathology Section, Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy (E.F.)
| |
Collapse
|
47
|
Zhao M, Guan Z, Gong L, Liu F, Gu W, Liu L, Jiang K, Cai J, Feng C, Kuick CH, Chang KTE, Wang J, Tang H, Yin M, Mao J. Rapid detection of telomerase expression of neuroblastoma in paraffin-embedded tissue: combination of in situ hybridisation and quantitative PCR. Pathology 2023; 55:958-965. [PMID: 37741703 DOI: 10.1016/j.pathol.2023.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 05/24/2023] [Accepted: 07/03/2023] [Indexed: 09/25/2023]
Abstract
Neuroblastoma is a heterogeneous paediatric malignant tumour. Telomere maintenance mechanism (TMM) by telomerase activation or alternative lengthening of telomeres (ALT) is a hallmark of high-risk neuroblastoma. However, the prior assays for telomerase, such as TERT expression by RNA sequencing or microarrays, may not be easy to perform in many histopathology laboratories in hospitals. The aims of this study are to assess the utility of ultrasensitive single-cell RNA in situ hybridisation (RNAscope), immunohistochemistry, and RT-qPCR on formalin-fixed, paraffin-embedded tumour samples as diagnostic tools for detecting TERT expression in neuroblastoma. In this study, we detected MYCN amplification in 22 of 222 cases (10%), TERT rearrangements in 18 of 220 cases (8%), and ALT activation in 39 of 222 cases (18%) using fluorescence in situ hybridisation (FISH). By RNA in situ hybridisation, 36 of 210 (17%) pretreatment neuroblastomas were found to have TERT overexpression, which was significantly associated with the high-risk group (33/78, 42%), TERT rearrangements (16/18, 89%), and MYCN amplification (13/22, 59%). None of the tumours with ALT showed TERT staining. In our study, 19 of the 55 MYCN non-amplified high-risk neuroblastomas displayed TERT mRNA expression, including 13 of the 14 TERT rearrangements, none of the 30 ALT-positive cases, and a significant proportion (6/11, 55%) that did not have the aforementioned genomic anomalies. RT-qPCR results correlated well with RNAscope levels (Spearman's rho=0.621, p<0.001, n=94). In conclusion, TERT RNA in situ hybridisation and RT-qPCR are suitable methods to evaluate TERT expression in neuroblastoma. The combination of detection of the genomic alterations and TERT mRNA expression is a powerful strategy for TMM activation detection, which can categorise neuroblastomas into multiple clinical subgroups for risk stratification in routine histopathology practice.
Collapse
Affiliation(s)
- Manli Zhao
- Department of Pathology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Zhonghai Guan
- Department of Surgical Oncology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Liang Gong
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China
| | - Fei Liu
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Weizhong Gu
- Department of Pathology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Lei Liu
- Department of Pathology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Kewen Jiang
- Biobank, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Jiabin Cai
- Department of Surgical Oncology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Chunyue Feng
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Chik Hong Kuick
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore
| | - Kenneth Tou En Chang
- Department of Pathology and Laboratory Medicine, KK Women's and Children's Hospital, Singapore
| | - Jinhu Wang
- Department of Surgical Oncology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Hongfeng Tang
- Department of Pathology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China
| | - Minzhi Yin
- Department of Pathology, Shanghai Children's Medical Centre, Shanghai, China.
| | - Jianhua Mao
- Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
| |
Collapse
|
48
|
Webersinke G, Burghofer J, Malli T, Rammer M, Jahn SW, Niendorf A, Tavassoli FA, Moinfar F. TERT Promoter Mutation c.-124C>T Commonly Occurs in Low-Grade Fibromatosis-like Metaplastic Breast Carcinoma. Arch Pathol Lab Med 2023; 147:1451-1457. [PMID: 36897999 DOI: 10.5858/arpa.2022-0159-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2022] [Indexed: 03/12/2023]
Abstract
CONTEXT.— Low-grade fibromatosis-like metaplastic carcinoma (FLMC) is a very rare subtype of triple-negative metaplastic (spindle cell) breast carcinoma. It is characterized by the proliferation of spindle cells closely resembling fibromatosis, which represents a benign fibroblastic/myofibroblastic breast proliferation. Unlike most triple-negative and basal-like breast cancers, FLMC has a very low potential for metastases, but demonstrates frequent local recurrences. OBJECTIVE.— To genetically characterize FLMC. DESIGN.— To this end, we analyzed 7 cases by targeted next-generation sequencing for 315 cancer-related genes and performed comparative microarray copy number analysis in 5 of these cases. RESULTS.— All cases shared TERT alterations (6 patients with recurrent c.-124C>T TERT promoter mutation and 1 patient with copy number gain encompassing the TERT locus), had oncogenic PIK3CA/PIK3R1 mutations (activation of the PI3K/AKT/mTOR pathway), and lacked mutations in TP53. TERT was overexpressed in all FLMCs. CDKN2A/B loss or mutation was observed in 4 of 7 cases (57%). Furthermore, tumors displayed chromosomal stability, with only few copy number variations and a low tumor mutational burden. CONCLUSIONS— We conclude that FLMCs typically show the recurrent TERT promoter mutation c.-124C>T, activation of the PI3K/AKT/mTOR pathway, low genomic instability, and wild-type TP53. In conjunction with previous data of metaplastic (spindle cell) carcinoma with and without fibromatosis-like morphology, FLMC is most likely distinguished by TERT promoter mutation. Thus, our data support the notion of a distinct subgroup within low-grade metaplastic breast cancer with spindle cell morphology and associated TERT mutations.
Collapse
Affiliation(s)
- Gerald Webersinke
- From the Laboratory for Molecular Genetic Diagnostics, Ordensklinikum Linz/Hospital of the Sisters of Charity, Linz, Austria (Webersinke, Burghofer, Malli, Rammer)
| | - Jonathan Burghofer
- From the Laboratory for Molecular Genetic Diagnostics, Ordensklinikum Linz/Hospital of the Sisters of Charity, Linz, Austria (Webersinke, Burghofer, Malli, Rammer)
| | - Theodora Malli
- From the Laboratory for Molecular Genetic Diagnostics, Ordensklinikum Linz/Hospital of the Sisters of Charity, Linz, Austria (Webersinke, Burghofer, Malli, Rammer)
| | - Melanie Rammer
- From the Laboratory for Molecular Genetic Diagnostics, Ordensklinikum Linz/Hospital of the Sisters of Charity, Linz, Austria (Webersinke, Burghofer, Malli, Rammer)
| | - Stephan Wenzel Jahn
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria (Jahn, Moinfar)
| | - Axel Niendorf
- Department of Pathology and Molecular Diagnostic, Hamburg-West, Hamburg, Germany (Niendorf)
| | - Fattaneh A Tavassoli
- Department of Pathology, Yale University School of Medicine, New Haven Connecticut (Tavassoli)
| | - Farid Moinfar
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria (Jahn, Moinfar)
- Department of Pathology and Molecular Pathology, Vinzenz Pathologieverbund Linz, Austria (Moinfar)
- The Vincent Academy of Pathology, Linz, Austria (Moinfar)
| |
Collapse
|
49
|
Chua BH, Zaal Anuar N, Ferry L, Domrane C, Wittek A, Mukundan VT, Jha S, Butter F, Tenen DG, Defossez PA, Kappei D. E4F1 and ZNF148 are transcriptional activators of the -57A > C and wild-type TERT promoter. Genome Res 2023; 33:1893-1905. [PMID: 37918959 PMCID: PMC10760450 DOI: 10.1101/gr.277724.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 10/23/2023] [Indexed: 11/04/2023]
Abstract
Point mutations within the TERT promoter are the most recurrent somatic noncoding mutations identified across different cancer types, including glioblastoma, melanoma, hepatocellular carcinoma, and bladder cancer. They are most abundant at -146C > T and -124C > T, and rarer at -57A > C, with the latter originally described as a familial case, but subsequently shown also to occur somatically. All three mutations create de novo E26-specific (ETS) binding sites and result in activation of the TERT gene, allowing cancer cells to achieve replicative immortality. Here, we used a systematic proteomics screen to identify transcription factors preferentially binding to the -146C > T, -124C > T, and -57A > C mutations. Although we confirmed binding of multiple ETS factors to the mutant -146C > T and -124C > T sequences, we identified E4F1 as a -57A > C-specific binder and ZNF148 as a TERT wild-type (WT) promoter binder that showed reduced interaction with the -124C > T allele. Both proteins are activating transcription factors that bind specifically to the -57A > C and WT (at position 124) TERT promoter sequence in corresponding cell lines, and up-regulate TERT transcription and telomerase activity. Our work describes new regulators of TERT gene expression with possible roles in cancer.
Collapse
Affiliation(s)
- Boon Haow Chua
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117596 Singapore
| | - Nurkaiyisah Zaal Anuar
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore
| | - Laure Ferry
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, F-75013 Paris, France
| | - Cecilia Domrane
- Université Paris Cité, CNRS, Epigenetics and Cell Fate, F-75013 Paris, France
| | - Anna Wittek
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore
| | - Vineeth T Mukundan
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore
| | - Sudhakar Jha
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117596 Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, Oklahoma 74078, USA
| | - Falk Butter
- Institute of Molecular Biology (IMB), 55128 Mainz, Germany
- Institute of Molecular Virology and Cell Biology (IMVZ), Friedrich Loeffler Institute, 17493 Greifswald, Germany
| | - Daniel G Tenen
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore
- Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | - Dennis Kappei
- Cancer Science Institute of Singapore, National University of Singapore, 117599 Singapore;
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 117596 Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore
| |
Collapse
|
50
|
Godoy PM, Oyedeji A, Mudd JL, Morikis VA, Zarov AP, Longmore GD, Fields RC, Kaufman CK. Functional analysis of recurrent CDC20 promoter variants in human melanoma. Commun Biol 2023; 6:1216. [PMID: 38030698 PMCID: PMC10686982 DOI: 10.1038/s42003-023-05526-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Small nucleotide variants in non-coding regions of the genome can alter transcriptional regulation, leading to changes in gene expression which can activate oncogenic gene regulatory networks. Melanoma is heavily burdened by non-coding variants, representing over 99% of total genetic variation, including the well-characterized TERT promoter mutation. However, the compendium of regulatory non-coding variants is likely still functionally under-characterized. We developed a pipeline to identify hotspots, i.e. recurrently mutated regions, in melanoma containing putatively functional non-coding somatic variants that are located within predicted melanoma-specific regulatory regions. We identified hundreds of statistically significant hotspots, including the hotspot containing the TERT promoter variants, and focused on a hotspot in the promoter of CDC20. We found that variants in the promoter of CDC20, which putatively disrupt an ETS motif, lead to lower transcriptional activity in reporter assays. Using CRISPR/Cas9, we generated an indel in the CDC20 promoter in human A375 melanoma cell lines and observed decreased expression of CDC20, changes in migration capabilities, increased growth of xenografts, and an altered transcriptional state previously associated with a more proliferative and less migratory state. Overall, our analysis prioritized several recurrent functional non-coding variants that, through downregulation of CDC20, led to perturbation of key melanoma phenotypes.
Collapse
Affiliation(s)
- Paula M Godoy
- Division of Medical Oncology, Department of Medicine and Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Abimbola Oyedeji
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in Saint Louis, St. Louis, MO, USA
| | - Jacqueline L Mudd
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in Saint Louis, St. Louis, MO, USA
| | - Vasilios A Morikis
- Departments of Medicine (Oncology) and Cell Biology and Physiology and the ICCE Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Anna P Zarov
- Division of Medical Oncology, Department of Medicine and Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gregory D Longmore
- Siteman Cancer Center, Washington University in Saint Louis, St. Louis, MO, USA
- Departments of Medicine (Oncology) and Cell Biology and Physiology and the ICCE Institute, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Ryan C Fields
- Department of Surgery, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University in Saint Louis, St. Louis, MO, USA
| | - Charles K Kaufman
- Division of Medical Oncology, Department of Medicine and Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Siteman Cancer Center, Washington University in Saint Louis, St. Louis, MO, USA.
| |
Collapse
|