1
|
Li G, Wu N, Ghabrial J, Stinnett V, Klausner M, Morsberger L, Long P, Baraban E, Gross JM, Zou YS. Chromoanagenesis in Osteosarcoma. Biomolecules 2025; 15:833. [PMID: 40563473 DOI: 10.3390/biom15060833] [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: 04/25/2025] [Revised: 05/25/2025] [Accepted: 06/04/2025] [Indexed: 06/28/2025] Open
Abstract
Chromoanagenesis is a catastrophic genomic phenomenon involving sudden, extensive rearrangements within one or a few cell cycles. In osteosarcoma, the most prevalent malignant bone tumor in children and adolescents, these events dramatically alter the genomic landscape, frequently disrupting key tumor suppressor genes like TP53 and RB1, amplifying oncogene expression, and propelling tumor progression and evolution. This review elucidates how key chromoanagenic mechanisms, such as chromothripsis and chromoanasynthesis, arise from replication stress and impaired DNA repair pathways, ultimately contributing to genomic instability in osteosarcoma. Chromothripsis features prominently in osteosarcoma, occurring in up to 62% of tumor regions and driving intratumoral heterogeneity through persistent genomic crises. Next-generation sequencing, optical genome mapping, and emerging technologies like single-cell sequencing empower researchers to detect and characterize these complex structural variants, demonstrating how a single catastrophic event can profoundly influence osteosarcoma progression over time. While targeted therapies for osteosarcoma have proven elusive, innovative strategies harnessing comprehensive genomic profiling and patient-derived preclinical models hold promise for uncovering tumor-specific vulnerabilities tied to chromoanagenesis. Ultimately, unraveling how these rapid, large-scale rearrangements fuel osteosarcoma's aggressive nature will not only refine disease classification and prognosis but also pave the way for novel therapeutic approaches to enhance patient outcomes.
Collapse
Affiliation(s)
- Guozhuang Li
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing 100730, China
- Key Laboratory of Big Data for Spinal Deformities, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Nan Wu
- State Key Laboratory of Complex Severe and Rare Diseases, Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing 100730, China
- Key Laboratory of Big Data for Spinal Deformities, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jen Ghabrial
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Victoria Stinnett
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Melanie Klausner
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Laura Morsberger
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Patty Long
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ezra Baraban
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - John M Gross
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ying S Zou
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
2
|
Corriveau ML, Korb JC, Michener SL, Owen NM, Wilson EL, Kubala J, Turner A, Takacs DS, Potocki L, Swann JW, Xue M, Dai H, Chao HT. De Novo Chromosomes 3q and 5q Chromothripsis Leads to a 5q14.3 Microdeletion Syndrome Presentation: Case Report and Review of the Literature. Am J Med Genet A 2025; 197:e63975. [PMID: 39887826 DOI: 10.1002/ajmg.a.63975] [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: 10/10/2024] [Revised: 11/25/2024] [Accepted: 12/13/2024] [Indexed: 02/01/2025]
Abstract
5q14.3 microdeletion syndrome (MIM#613443) is a neurodevelopmental disorder (NDD) involving copy number loss of multiple genes including Myocyte enhancer factor 2C (MEF2C) gene in the q14.3 region of chromosome 5. Chromosomes 5 and 15 chromothripsis involving 5q14.3 was previously reported in one individual with developmental epileptic encephalopathy (DEE). A complex chromothripsis between chromosomes 3, 5, 7, 9, and 18 that involved 5q14.3 was also reported in a pregnancy complicated by brain and kidney anomalies on fetal ultrasound. Here, we report chromothripsis of chromosomes 3q and 5q involving 5q14.3 in a three-year-old female with Lennox-Gastaut syndrome. The chromosomes 3q and 5q chromothripsis was found by trio genome sequencing (GS) and confirmed by fluorescent in situ hybridization (FISH). Notable clinical findings include medically refractory seizures, global developmental delay, increased fluid-attenuated inversion recovery (FLAIR) signal in the left inferior temporal gyrus, and dysmorphic features. Chromothripsis of chromosomes 3 and 5 was previously recognized in renal cell carcinomas. To the best of our knowledge, this is the first reported case of chromosomes 3q and 5q chromothripsis leading to a developmental epileptic encephalopathy (DEE) due to disruption of 5q14.3. These findings expand chromosomes 3 and 5 chromothripsis as a genomic mechanism underlying 5q14.3 microdeletion syndrome.
Collapse
Affiliation(s)
- Melina L Corriveau
- The Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- School of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Joshua C Korb
- The Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- School of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Sydney L Michener
- The Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | - Nichole M Owen
- Baylor Genetics, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | | | | | - Alicia Turner
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Hospital, Houston, Texas, USA
| | - Danielle S Takacs
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Hospital, Houston, Texas, USA
- Department of Pediatrics and Neurology Neurophysiology and Epilepsy, Baylor College of Medicine, Houston, Texas, USA
| | - Lorraine Potocki
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Hospital, Houston, Texas, USA
| | - John W Swann
- The Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| | - Mingshan Xue
- The Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
| | - Hongzheng Dai
- Baylor Genetics, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
| | - Hsiao-Tuan Chao
- The Cain Pediatric Neurology Research Foundation Laboratories, Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
- Division of Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- Texas Children's Hospital, Houston, Texas, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA
- McNair Medical Institute, the Robert and Janice McNair Foundation, Houston, Texas, USA
| |
Collapse
|
3
|
Roessner A, Franke S, Schreier J, Ullmann SR, Karras FS. Non-Coding RNAs in Diagnostic Pathology of High-Grade Central Osteosarcoma. Diagnostics (Basel) 2025; 15:1355. [PMID: 40506925 PMCID: PMC12154195 DOI: 10.3390/diagnostics15111355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2025] [Revised: 05/18/2025] [Accepted: 05/20/2025] [Indexed: 06/16/2025] Open
Abstract
A histological evaluation remains the cornerstone of diagnosing highly malignant osteosarcoma, having demonstrated its efficacy and reliability over several decades. However, despite these advancements, misdiagnoses with severe consequences, including inadequate surgical procedures, continue to occur. Consequently, there is a pressing need to further enhance diagnostic security. Adjunct immunohistochemical approaches have demonstrated significant effectiveness in regard to cancer diagnostics, generally. However, their utility for identifying highly malignant osteosarcoma is limited. Molecular genetic findings have significantly improved the diagnosis of Ewing's sarcoma by identifying specific translocations and have been used to detect specific IDH gene mutations in chondrosarcoma. Nevertheless, molecular genetic alterations in highly malignant osteosarcoma exhibit a high degree of complexity, thereby limiting their diagnostic utility. Given that only 1-2% of the human genome comprises protein-coding sequences, the growing number of non-coding regulatory RNAs, which are increasingly being elucidated, has garnered substantial attention in the field of clinical cancer diagnostics. Over the past several years, patterns of altered non-coding RNA expression have been identified that facilitate the distinction between benign and malignant tumors in various organs. In the field of bone tumors, the experience of this approach has been limited thus far. The divergent expression of microRNAs has demonstrated utility for differentiating osteosarcoma from osteoblastoma and discriminating between osteosarcoma and giant-cell tumors of bone and fibrous dysplasia. However, the application of non-coding RNA expression patterns for the differential diagnosis of osteosarcoma is still in its preliminary stages. This review provides an overview of the current status of non-coding RNAs in osteosarcoma diagnostics, in conjunction with a histological evaluation. The potential of this approach is discussed comprehensively.
Collapse
Affiliation(s)
- Albert Roessner
- Institute of Pathology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; (S.F.); (J.S.); (S.R.U.); (F.S.K.)
| | | | | | | | | |
Collapse
|
4
|
Valtorta S, Granata S, de Pretis S, Bertoli G, Redaelli S, Berno V, Spinelli AE, Diprima S, Rainone P, Coliva A, Todde S, Marfia G, Navone S, Caroli M, Bentivegna A, Di Muzio N, Moresco RM. Radio-chemotherapy and metformin selectively modulate the heterogeneous landscape of glioma with ribosome biogenesis, long non coding RNA and immune-escape markers as major player. Int J Biol Sci 2025; 21:3527-3554. [PMID: 40520013 PMCID: PMC12160513 DOI: 10.7150/ijbs.103194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 04/16/2025] [Indexed: 06/18/2025] Open
Abstract
Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults with a short survival time after standard therapy administration including radiotherapy (RT) associated with temozolomide (TMZ). Here, we investigated the effects of radiochemotherapy in association with metformin (MET), a drug targeting cell metabolism on a syngeneic GBM mouse model using Positron Emission Tomography imaging with [18F]FLT and [18F]VC701 and single-cell RNA-sequencing analysis. The addition of drugs to RT significantly increased survival and [18F]FLT showed an early predictive response of combined therapy. We identified the presence of heterogeneous tumor populations with different treatment sensitivity and a complex immune evasive microenvironment. Tumor cells surviving to treatments showed immune response, among the main differentially modulated biological functions and a potential role of long non-coding RNAs (lncRNAs) in treatment resistance. Association with TMZ or TMZ plus MET reduced the pro-tumor phenotype of immune reaction acting more on myeloid cells the first and on lymphocytes the latter. Off note, MET add-on counteracted the immune-evasive phenotype particularly of T cells suggesting a potential role of MET also in adopted immunity.
Collapse
Affiliation(s)
- Silvia Valtorta
- Institute of Bioimaging and Complex Biological Systems (IBSBC), National Research Council (CNR), 20054 Segrate (MI), Italy
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20312 Milan, Italy
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
| | - Silvia Granata
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20312 Milan, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Stefano de Pretis
- Center for Omics Sciences, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy
| | - Gloria Bertoli
- Institute of Bioimaging and Complex Biological Systems (IBSBC), National Research Council (CNR), 20054 Segrate (MI), Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
| | - Serena Redaelli
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Valeria Berno
- Advanced Light and Electron Microscopy Bioimaging Center ALEMBIC, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy
| | - Antonello E. Spinelli
- Experimental Imaging Center, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy
| | - Santo Diprima
- Center for Omics Sciences, San Raffaele Scientific Institute (IRCCS), 20132 Milan, Italy
| | - Paolo Rainone
- Institute of Bioimaging and Complex Biological Systems (IBSBC), National Research Council (CNR), 20054 Segrate (MI), Italy
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20312 Milan, Italy
| | - Angela Coliva
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20312 Milan, Italy
| | - Sergio Todde
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Unit of Neurosurgery, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, 20122, Milan, Italy
- Aerospace Medicine Institute “A. Mosso”, Italian Air Force, 20138, Milan, Italy
| | - Stefania Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Unit of Neurosurgery, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Manuela Caroli
- Laboratory of Experimental Neurosurgery and Cell Therapy, Unit of Neurosurgery, Fondazione IRCCS Ca' Granda - Ospedale Maggiore Policlinico, 20122, Milan, Italy
| | - Angela Bentivegna
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
- Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Nadia Di Muzio
- Department of Radiation Oncology, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Rosa Maria Moresco
- Institute of Bioimaging and Complex Biological Systems (IBSBC), National Research Council (CNR), 20054 Segrate (MI), Italy
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), 20312 Milan, Italy
- GBM-BI-TRACE (GlioBlastoMa-BIcocca-TRAnslational-CEnter), University of Milano-Bicocca, 20900 Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| |
Collapse
|
5
|
Mcneil TR, Sikder S, Dalal Y. Cancer cells' chamber of secrets: the link between micronuclei, chromothripsis and malignancy. Open Biol 2025; 15:240388. [PMID: 40359993 DOI: 10.1098/rsob.240388] [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: 12/03/2024] [Revised: 02/19/2025] [Accepted: 04/09/2025] [Indexed: 05/15/2025] Open
Abstract
Micronuclei exhibit defective proteomes rendering their chromatin vulnerable to fragmentation. This fragmentation process, known as chromothripsis, promotes tumorigenesis by catalysing the activation of oncogenes and the silencing of tumor suppressors. With this role in mind, micronuclei serve as promising targets for therapeutic intervention. This review will explore recent discoveries regarding how micronuclei form, their function in catalysing chromothripsis and how chromothripsis provides a selective advantage for cancer cells.
Collapse
Affiliation(s)
| | - Sweta Sikder
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Yamini Dalal
- Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| |
Collapse
|
6
|
Li X, Gong Q, Yu J, Liang J, Yao R, Zhou J, Chen Y, Lei Z, Yu Z, Zhang X, Qiu X. Identification of effective anti-osteosarcoma agents via screening of an in-house NQO1-targeted compound library. Bioorg Med Chem 2025; 122:118162. [PMID: 40117702 DOI: 10.1016/j.bmc.2025.118162] [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: 02/07/2025] [Revised: 03/10/2025] [Accepted: 03/16/2025] [Indexed: 03/23/2025]
Abstract
Osteosarcoma is one of the most prevalent malignant bone tumors, and despite advances in treatment, significant improvements in survival rates for osteosarcoma patients remain elusive. There is an urgent need for developing novel molecules for the targeted treatment of osteosarcoma. NAD(P)H:quinone oxidoreductase 1 (NQO1) is highly overexpressed in osteosarcoma. Here, we evaluated a series of in-house NQO1-targeting compounds, including NQO1 substrates and β-lap prodrugs, through phenotypic screening using NQO1-positive methylnitronitrosoguanidine-induced human osteosarcoma cells (MNNG) and NQO1-negative normal human umbilical vein endothelial cells (HUVEC), aiming to identify novel candidate compounds for osteosarcoma therapy. As a result, compound 21, an NQO1 substrate, was identified as a potent anti-osteosarcoma agent that promotes apoptosis and induces cell cycle arrest in osteosarcoma cells in vitro, while significantly inhibiting tumor growth in vivo. These findings suggest that compound 21 holds promise as a candidate for osteosarcoma treatment. Moreover, NQO1-targeting substrates present a promising pathway for the discovery of novel anti-osteosarcoma agents.
Collapse
Affiliation(s)
- Xiang Li
- Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Qijie Gong
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Jianglin Yu
- Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing 210008, China
| | - Jiaqi Liang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Rui Yao
- Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Jian Zhou
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Yaxin Chen
- Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China
| | - Zhijie Lei
- Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing 210008, China
| | - Zhan Yu
- Departemt of Emergency, The Affiliated Jiangning Hospital of NJMU, Nanjing Medical University (NJMU), Nanjing 211100, China
| | - Xiaojin Zhang
- Jiangsu Key Laboratory of Drug Design and Optimization, and Department of Chemistry, China Pharmaceutical University, Nanjing 211198, China.
| | - Xusheng Qiu
- Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing 210008, China; Department of Orthopedics, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing 210008, China.
| |
Collapse
|
7
|
Houben A, Fuchs J, Banaei-Moghaddam AM, Chen J, Kim G, Liu T. Does chromoanagenesis play a role in the origin of B chromosomes? Heredity (Edinb) 2025:10.1038/s41437-025-00758-w. [PMID: 40253498 DOI: 10.1038/s41437-025-00758-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/31/2025] [Indexed: 04/21/2025] Open
Abstract
B chromosomes (Bs) exist in addition to the standard (A) chromosomes in a wide range of species. The process underlying their origin is still unclear. We propose pathways of intra- and interspecific origin of B chromosomes based on known mechanisms of chromosome evolution and available knowledge of their sequence composition in different species. We speculate that a mitotic or meiotic segregation error of one or more A chromosomes initiates, via chromoanagenesis, the formation of a proto-B chromosome. In the second step, proto-B chromosomes accumulate A chromosome- and organelle-derived sequences over time, most likely via DNA double-strand break (DSB) mis-repair. Consequently, the original structure of the early stage proto-B chromosomes becomes masked by continuous sequence incorporation. The similarity between A chromosome sequences integrated into B chromosomes and the original sequences on the donor chromosomes decreases over time if there is no selection pressure on these sequences on B chromosomes. However, besides chromoanagenesis, also other mechanisms leading to the formation of B chromosomes might exist.
Collapse
Affiliation(s)
- Andreas Houben
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK) Gatersleben, 06466, Seeland, Germany.
| | - Jörg Fuchs
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK) Gatersleben, 06466, Seeland, Germany
| | - Ali Mohammad Banaei-Moghaddam
- Laboratory of Genomics and Epigenomics (LGE), Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Jianyong Chen
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK) Gatersleben, 06466, Seeland, Germany
| | - Gihwan Kim
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK) Gatersleben, 06466, Seeland, Germany
| | - Taoran Liu
- Leibniz Institute of Plant Genetics and Crop Plant Research (IPK) Gatersleben, 06466, Seeland, Germany
| |
Collapse
|
8
|
Pellestor F, Ganne B, Gaillard JB, Gatinois V. Chromoplexy: A Pathway to Genomic Complexity and Cancer Development. Int J Mol Sci 2025; 26:3826. [PMID: 40332527 PMCID: PMC12027847 DOI: 10.3390/ijms26083826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Chromoplexy is a phenomenon of complex genome rearrangement, occurring during a single cell event and characterized by the formation of chain rearrangements affecting multiple chromosomes. Unlike other genomic rearrangements such as chromothripsis, which involves a single chromosome, chromoplexy affects several chromosomes at once, creating patterns of complex, balanced translocations, and leading to the formation of fusion genes and the simultaneous disruption of several genes. Chromoplexy was first identified in prostate cancers, but it is now observed in various cancers where gene fusions take place. The precise mechanisms behind chromoplexy remain under investigation. The occurrence of these rearrangements follows multiple double-stranded breaks that appear to occur in certain regions or during particular genome configurations (open chromatin, active transcription area), and which lead to an intricate series of inter- and intra-chromosomal translocations and deletions without significant alterations in the number of copies. Although chromoplexy is considered a very early event in oncogenesis, the phenomenon can be repeated and can constitute a mechanism of clonal tumor progression. The occurrence of chromoplexy supports the equilibrium model punctuated by tumor evolution, characterized by periods of relative stability punctuated by sudden and rapid periods of radical genomic changes.
Collapse
Affiliation(s)
- Franck Pellestor
- Chromosomal Genetics Unit and Chromostem Research Platform, Department of Molecular Genetics and Cytogenomics, Unique Site of Biology (SUB), University Hospital of Montpellier, 371 Avenue du Doyen Gaston Giraud, 34295 Montpellier Cedex 5, France; (B.G.); (J.B.G.); (V.G.)
| | | | | | | |
Collapse
|
9
|
Li Q, Keskus AG, Wagner J, Izydorczyk MB, Timp W, Sedlazeck FJ, Klein AP, Zook JM, Kolmogorov M, Schatz MC. Unraveling the hidden complexity of cancer through long-read sequencing. Genome Res 2025; 35:599-620. [PMID: 40113261 PMCID: PMC12047254 DOI: 10.1101/gr.280041.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Cancer is fundamentally a disease of the genome, characterized by extensive genomic, transcriptomic, and epigenomic alterations. Most current studies predominantly use short-read sequencing, gene panels, or microarrays to explore these alterations; however, these technologies can systematically miss or misrepresent certain types of alterations, especially structural variants, complex rearrangements, and alterations within repetitive regions. Long-read sequencing is rapidly emerging as a transformative technology for cancer research by providing a comprehensive view across the genome, transcriptome, and epigenome, including the ability to detect alterations that previous technologies have overlooked. In this Perspective, we explore the current applications of long-read sequencing for both germline and somatic cancer analysis. We provide an overview of the computational methodologies tailored to long-read data and highlight key discoveries and resources within cancer genomics that were previously inaccessible with prior technologies. We also address future opportunities and persistent challenges, including the experimental and computational requirements needed to scale to larger sample sizes, the hurdles in sequencing and analyzing complex cancer genomes, and opportunities for leveraging machine learning and artificial intelligence technologies for cancer informatics. We further discuss how the telomere-to-telomere genome and the emerging human pangenome could enhance the resolution of cancer genome analysis, potentially revolutionizing early detection and disease monitoring in patients. Finally, we outline strategies for transitioning long-read sequencing from research applications to routine clinical practice.
Collapse
Affiliation(s)
- Qiuhui Li
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Ayse G Keskus
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA
| | - Justin Wagner
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - Michal B Izydorczyk
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Winston Timp
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland 21218, USA
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Texas 77030, USA
- Department of Computer Science, Rice University, Houston, Texas 77251, USA
| | - Alison P Klein
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins Medicine, Baltimore, Maryland 21031, USA
| | - Justin M Zook
- Material Measurement Laboratory, National Institute of Standards and Technology, Gaithersburg, Maryland 20899, USA
| | - Mikhail Kolmogorov
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892, USA;
| | - Michael C Schatz
- Department of Computer Science, Johns Hopkins University, Baltimore, Maryland 21218, USA;
- Sidney Kimmel Comprehensive Cancer Center, Department of Oncology, Johns Hopkins Medicine, Baltimore, Maryland 21031, USA
| |
Collapse
|
10
|
Li Y, Liang J, Ren X, Guo J, Wang X, Wang X, Yu S, Li T, Yang X. FGFR3-TACC3 fusion gene promotes glioblastoma malignant progression through the activation of STAT3 signaling pathway. Front Oncol 2025; 15:1560008. [PMID: 40265014 PMCID: PMC12011601 DOI: 10.3389/fonc.2025.1560008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/20/2025] [Indexed: 04/24/2025] Open
Abstract
Objective The Fibroblast growth factor receptors 3-transforming acidic coiled-coil-containing protein 3 (FGFR3-TACC3, F3-T3) oncogenic fusion gene, identified in malignant tumors such as gliomas and bladder cancer, has been particularly noted in recurrent gliomas where it is considered to drive malignant progression, thus presenting itself as a viable therapeutic target. However, the precise mechanism by which F3-T3 facilitates the malignant progression of glioma is not fully understood. Methods Correction analysis of STAT3 and FGFR3 with major glioma mutation types and pan-cancer analysis was conducted using The Cancer Genome Atlas (TCGA) database. A series of phenotypic experiments, including CCK-8, EdU, colony-formation assay, wound healing assay, and transwell assay were conducted to detect the effects of F3-T3 on proliferation, invasion, and migration of glioma cells. The association between F3-T3 and epithelial-mesenchymal transition (EMT) was investigated through enrichment analysis of the E-MTAB-6037 gene chip database and confirmed by western blot. The underling mechanism were further inferred and validated through RNA sequencing, E-MTAB-6037 gene chip data, and western blot. The relationship between p-STAT3 expression and the WHO grade of glioma was evaluated using immunohistochemistry (IHC) and tissue microarray analysis. Furthermore, the results of vivo experiments and IHC has confirmed the impact of F3-T3 on glioma malignant progression and activation of the STAT3 signaling pathway. Results The experimental results from this study indicate that F3-T3 accelerates the epithelial-mesenchymal transition (EMT) process in glioma cells, thereby promoting their proliferation, invasion, and migration capabilities. Mechanistically, it was determined through RNA sequencing that the signal transducer and activator of transcription 3 (STAT3) signaling pathway is crucial for the malignant progression of F3-T3. This finding was further supported through follow-up experiments conducted after STAT3 knockdown. The role of the STAT3 pathway in gliomas was also reinforced through bioinformatic analysis and immunohistochemistry (IHC) on tissue microarrays (TMA). Further in vivo experiments corroborated the role of F3-T3 in enhancing glioma growth and progression. Conclusion F3-T3 facilitates the proliferation, invasion, migration and EMT of glioma cells, thereby promoting their malignant progression through STAT3 signaling activation. These findings highlight its potential as a therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Yiming Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jianshen Liang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Xiude Ren
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiahe Guo
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Xisen Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Xuya Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Shengping Yu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Tao Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuejun Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Neuro-Oncology, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
11
|
Golas MM, Gunawan B, Gutenberg A, Danner BC, Gerdes JS, Stadelmann C, Füzesi L, Liersch T, Sander B. Cytogenetic signatures favoring metastatic organotropism in colorectal cancer. Nat Commun 2025; 16:3261. [PMID: 40188208 PMCID: PMC11972295 DOI: 10.1038/s41467-025-58413-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Colorectal carcinoma (CRC) exhibits metastatic organotropism, primarily targeting liver, lung, and rarely the brain. Here, we study chromosomal imbalances (CIs) in cohorts of primary CRCs and metastases. Brain metastases show the highest burden of CIs, including aneuploidies and focal CIs, with enrichment of +12p encoding KRAS. Compared to liver and lung metastases, brain metastases present with increased co-occurrence of KRAS mutation and amplification. CRCs with concurrent KRAS mutation and amplification display significant metabolic reprogramming with upregulation of glycolysis, alongside upregulation of cell cycle pathways, including copy number gains of MDM2 and CDK4. Evolutionary modeling suggests early acquisition of many organotropic CIs enriched in both liver and brain metastases, while brain-enriched CIs preferentially emerge later. Collectively, this study supports a model where cytogenetic events in CRCs favor site-specific metastatic colonization. These site-enriched CI patterns may serve as biomarkers for metastatic potential in precision oncology.
Collapse
Affiliation(s)
- Mariola Monika Golas
- Human Genetics, Faculty of Medicine, University of Augsburg, Augsburg, Germany.
- Comprehensive Cancer Center Augsburg, University Medical Center Augsburg, Augsburg, Germany.
| | - Bastian Gunawan
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
- Institute of Pathology Northern Hesse, Kassel, Germany
| | - Angelika Gutenberg
- Department of Neurosurgery, Asklepios Hospital Harburg, Hamburg, Germany
| | - Bernhard C Danner
- Department of Cardiac, Thoracic and Vascular Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Jan S Gerdes
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
- Epilepsy Center Hamburg, Evangelical Hospital Alsterdorf, Neurology and Epileptology, Hamburg, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Laszlo Füzesi
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Torsten Liersch
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Bjoern Sander
- Institute of Pathology, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
12
|
Farooq AR, Zhang AX, Chan-Seng-Yue M, Topham JT, O'Kane GM, Jang GH, Fischer S, Dodd A, Holter S, Wilson J, Grant RC, Aung KL, Zogopoulos G, Elimova E, Prince R, Jang R, Moore M, Biagi J, Tang P, Goodwin R, Bathe OF, Marra M, Laskin J, Renouf DJ, Schaeffer DF, Karasinska JM, Notta F, Gallinger S, Knox JJ, Tsang ES. The tandem duplicator phenotype may be a novel targetable subgroup in pancreatic cancer. NPJ Precis Oncol 2025; 9:100. [PMID: 40185871 PMCID: PMC11971333 DOI: 10.1038/s41698-025-00888-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 03/20/2025] [Indexed: 04/07/2025] Open
Abstract
Tandem duplicator phenotype (TDP) consists of distinct genomic rearrangements where tandem duplications are randomly distributed. In this study, we characterized the prevalence and outcomes of TDP in a large series of prospectively sequenced tumors from patients with pancreatic ductal adenocarcinomas (PDAC). Whole-genome sequencing (WGS) was performed in 530 PDAC cases from the PanCuRx Initiative, COMPASS and PanGen/POG trials in Canada. Of 530 cases, 52 were identified as TDP (9.8%; 13 resected, 39 advanced). Etiological subgroups of TDP included BRCA1 (n = 9), CCNE1 (n = 4), and unknown (n = 39). Presence of TDP was not prognostic in resected specimens (p = 0.77) compared with non-HRD and non-TDP cases, described as typicals. In advanced cases, when stratified for only classical subtype cases, platinum therapy was correlated with longer response in non-BRCA1 TDP vs. typicals (p = 0.0036). There was no difference in overall survival between TDP and typicals (p = 0.5).TDP represents a potential novel targetable subgroup for chemotherapy selection in PDAC.
Collapse
Affiliation(s)
| | - Amy X Zhang
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | - James T Topham
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | | | - Gun Ho Jang
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | - Anna Dodd
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Spring Holter
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Julie Wilson
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | | | | | - Elena Elimova
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | | | - Raymond Jang
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Malcolm Moore
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - James Biagi
- Cancer Centre of Southeastern Ontario/Queen's University, Kingston, ON, Canada
| | - Patricia Tang
- Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Rachel Goodwin
- Division of Medical Oncology, Department of Medicine, The Ottawa Hospital, The University of Ottawa, Ottawa, ON, K1N 6N5, Canada
| | - Oliver F Bathe
- Department of Oncology, Tom Baker Cancer Center, University of Calgary, 1331 29th St NW, Calgary, AB, T2N 4N2, Canada
| | - Marco Marra
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - Janessa Laskin
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - Daniel J Renouf
- BC Cancer, University of British Columbia, Vancouver, BC, Canada
| | - David F Schaeffer
- Department of Pathology, Vancouver General Hospital, Vancouver, BC, Canada
| | | | - Faiyaz Notta
- Ontario Institute for Cancer Research, Toronto, ON, Canada
| | | | | | - Erica S Tsang
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
13
|
Keskus AG, Bryant A, Ahmad T, Yoo B, Aganezov S, Goretsky A, Donmez A, Lansdon LA, Rodriguez I, Park J, Liu Y, Cui X, Gardner J, McNulty B, Sacco S, Shetty J, Zhao Y, Tran B, Narzisi G, Helland A, Cook DE, Chang PC, Kolesnikov A, Carroll A, Molloy EK, Bi C, Walter A, Gibson M, Pushel I, Guest E, Pastinen T, Shafin K, Miga KH, Malikic S, Day CP, Robine N, Sahinalp C, Dean M, Farooqi MS, Paten B, Kolmogorov M. Severus detects somatic structural variation and complex rearrangements in cancer genomes using long-read sequencing. Nat Biotechnol 2025:10.1038/s41587-025-02618-8. [PMID: 40185952 DOI: 10.1038/s41587-025-02618-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/26/2025] [Indexed: 04/07/2025]
Abstract
For the detection of somatic structural variation (SV) in cancer genomes, long-read sequencing is advantageous over short-read sequencing with respect to mappability and variant phasing. However, most current long-read SV detection methods are not developed for the analysis of tumor genomes characterized by complex rearrangements and heterogeneity. Here, we present Severus, a breakpoint graph-based algorithm for somatic SV calling from long-read cancer sequencing. Severus works with matching normal samples, supports unbalanced cancer karyotypes, can characterize complex multibreak SV patterns and produces haplotype-specific calls. On a comprehensive multitechnology cell line panel, Severus consistently outperforms other long-read and short-read methods in terms of SV detection F1 score (harmonic mean of the precision and recall). We also illustrate that compared to long-read methods, short-read sequencing systematically misses certain classes of somatic SVs, such as insertions or clustered rearrangements. We apply Severus to several clinical cases of pediatric leukemia/lymphoma, revealing clinically relevant cryptic rearrangements missed by standard genomic panels.
Collapse
Affiliation(s)
- Ayse G Keskus
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Asher Bryant
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Tanveer Ahmad
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Byunggil Yoo
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | | | - Anton Goretsky
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
- Department of Computer Science, University of Maryland, College Park, MD, USA
| | - Ataberk Donmez
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
- Department of Computer Science, University of Maryland, College Park, MD, USA
| | - Lisa A Lansdon
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Isabel Rodriguez
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| | - Jimin Park
- University of California, Santa Cruz, Genomics Institute, Santa Cruz, CA, USA
| | - Yuelin Liu
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
- Department of Computer Science, University of Maryland, College Park, MD, USA
| | - Xiwen Cui
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Joshua Gardner
- University of California, Santa Cruz, Genomics Institute, Santa Cruz, CA, USA
| | - Brandy McNulty
- University of California, Santa Cruz, Genomics Institute, Santa Cruz, CA, USA
| | - Samuel Sacco
- University of California, Santa Cruz, Genomics Institute, Santa Cruz, CA, USA
| | - Jyoti Shetty
- Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Yongmei Zhao
- Sequencing Facility Bioinformatics Group, Biomedical Informatics and Data Science Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Bao Tran
- Sequencing Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | | | | | | | | | | - Erin K Molloy
- Department of Computer Science, University of Maryland, College Park, MD, USA
| | - Chengpeng Bi
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Adam Walter
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Margaret Gibson
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Irina Pushel
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Erin Guest
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Tomi Pastinen
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Kishwar Shafin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| | - Karen H Miga
- University of California, Santa Cruz, Genomics Institute, Santa Cruz, CA, USA
| | - Salem Malikic
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Chi-Ping Day
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | - Cenk Sahinalp
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Michael Dean
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD, USA
| | - Midhat S Farooqi
- Children's Mercy Hospital, University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Benedict Paten
- University of California, Santa Cruz, Genomics Institute, Santa Cruz, CA, USA
| | - Mikhail Kolmogorov
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
14
|
de Groen PC. The strength of organ, tissue, and body field effects determines the frequency of all neoplasia. Ann N Y Acad Sci 2025; 1546:11-22. [PMID: 40096640 PMCID: PMC11998479 DOI: 10.1111/nyas.15306] [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] [Indexed: 03/19/2025]
Abstract
In 1953, Danely Slaughter proposed the concept of field cancerization, or field effect, to explain the development of additional neoplasia of similar type. A recent theory (de Groen, 2022) states that all DNA is exposed to a constant source of damage, resulting in a constant rate of germline and somatic DNA mutations. If the field effect and constant mutation theories are correct and a single somatic mutation causes the transition from non-neoplastic to neoplastic phenotype, then all rates of neoplasia formation can be modeled by exponential equations containing a single variable that determines the chance of phenotype transition. In this perspective, studies from 1953 till 2021 originating from America, Europe, and Asia about head, chest, abdomen, pelvic, and skin neoplasia were reviewed and showed consistent field effects that are modeled by tapering exponential equations containing a single variable defining field effect strength; Pearson and linear correlation coefficients for observed and modeled data range from 0.994 to 1. Thus, existing data are compatible with a constant rate of DNA damage. Organ-specific, tissue-specific, or body-wide mutagenesis conditions determine the rate of neoplasia development and explain the co-occurrence of seemingly unrelated neoplasia at predictable frequencies. Shared risk factors explain increased risk for additional neoplasia in persons with one neoplastic lesion.
Collapse
Affiliation(s)
- Piet C. de Groen
- Division of Gastroenterology, Hepatology & NutritionUniversity of MinnesotaMinneapolisMinnesotaUSA
| |
Collapse
|
15
|
Goad J, Rajkovic A. Uterine fibroids at single-cell resolution: unveiling cellular heterogeneity to improve understanding of pathogenesis and guide future therapies. Am J Obstet Gynecol 2025; 232:S124-S134. [PMID: 40253076 DOI: 10.1016/j.ajog.2024.08.037] [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: 10/18/2023] [Revised: 07/16/2024] [Accepted: 08/16/2024] [Indexed: 04/21/2025]
Abstract
Uterine leiomyomas or fibroids are benign tumors of the myometrium that affect approximately 70% of reproductive-age women. Fibroids continue to be the leading cause of hysterectomy, resulting in substantial healthcare costs. Genetic complexity and lack of cellular and molecular understanding of fibroids have posed considerable challenges to developing noninvasive treatment options. Over the years, research efforts have intensified to unravel the genetic and cellular diversities within fibroids to deepen our understanding of their origins and progression. Studies using immunostaining and flow cytometry have revealed cellular heterogeneity within these tumors. A correlation has been observed between genetic mutations in fibroids and their size, which is influenced by cellular composition, proliferation rates, and extracellular matrix accumulation. Fibroids with mediator complex subunit 12 (MED12) mutation are composed of smooth muscle cells and fibroblasts equally. In contrast, the fibroids with high-mobility group AT-hook 2 (HMGA2) translocation are 90% composed of smooth muscle cells. More recently, single-cell RNA sequencing in the myometrium and MED12 mutation carrying fibroids has identified further heterogeneity in smooth muscle cells and fibroblast cells, identifying 3 different smooth muscle cell populations and fibroblast cell populations. Although both myometrium and fibroids have similar cellular composition, these cells differs in their transcriptomic profile and have specialized roles, underscoring the complex cellular landscape contributing to fibroid pathogenesis. Furthermore, not all smooth muscle cells in MED12-mutant fibroid carry the MED12 mutation, suggesting that MED12-mutant fibroids might not be monoclonal in nature. This review describes the intricacies of fibroid biology revealed by single-cell RNA sequencing. These advances have identified new cellular targets for potential therapies, provided insights into treatment resistance, and laid the groundwork for more personalized approaches to fibroid management. As we continue to unravel the cellular and molecular complexity of fibroids, we anticipate that this knowledge will translate into more effective and less invasive treatments, ultimately improving outcomes for the millions of women affected by this condition.
Collapse
Affiliation(s)
- Jyoti Goad
- Department of Pathology, University of California San Francisco, San Francisco, CA
| | - Aleksandar Rajkovic
- Department of Pathology, University of California San Francisco, San Francisco, CA; Division of Reproductive Endocrinology and Infertility, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA; Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Francisco, San Francisco, CA.
| |
Collapse
|
16
|
Lu B, Winnall S, Cross W, Barnes CP. Cell-cycle dependent DNA repair and replication unifies patterns of chromosome instability. Nat Commun 2025; 16:3033. [PMID: 40155604 PMCID: PMC11953314 DOI: 10.1038/s41467-025-58245-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
Chromosomal instability (CIN) is pervasive in human tumours and often leads to structural or numerical chromosomal aberrations. Somatic structural variants (SVs) are intimately related to copy number alterations but the two types of variant are often studied independently. Additionally, despite numerous studies on detecting various SV patterns, there are still no general quantitative models of SV generation. To address this issue, we develop a computational cell-cycle model for the generation of SVs from end-joining repair and replication after double-strand break formation. Our model provides quantitative information on the relationship between breakage fusion bridge cycle, chromothripsis, seismic amplification, and extra-chromosomal circular DNA. Given whole-genome sequencing data, the model also allows us to infer important parameters in SV generation with Bayesian inference. Our quantitative framework unifies disparate genomic patterns resulted from CIN, provides a null mutational model for SV, and reveals deeper insights into the impact of genome rearrangement on tumour evolution.
Collapse
Affiliation(s)
- Bingxin Lu
- Department of Cell and Developmental Biology, University College London, Gower Street, London, UK.
- UCL Genetics Institute, University College London, Gower Street, London, UK.
- School of Biosciences, University of Surrey, Stag Hill, Guildford, UK.
- Surrey Institute for People-Centred Artificial Intelligence, University of Surrey, Stag Hill, Guildford, UK.
| | - Samuel Winnall
- Department of Cell and Developmental Biology, University College London, Gower Street, London, UK
| | - William Cross
- Department of Cell and Developmental Biology, University College London, Gower Street, London, UK
- Rare Malignancies and Cancer Evolution Laboratory, School of Biological Sciences, University of Reading, Whiteknights, Reading, UK
| | - Chris P Barnes
- Department of Cell and Developmental Biology, University College London, Gower Street, London, UK.
- UCL Genetics Institute, University College London, Gower Street, London, UK.
| |
Collapse
|
17
|
Berenguer-Rubio A, Such E, Hernández NT, González-Rojo P, Díaz-González Á, Avetisyan G, Gil-Aparicio C, González-López J, Pantoja-Borja N, Rubio-Martínez LA, Hernández-Girón S, Valera-Cuesta MS, Ramírez-Fuentes C, Simonet-Redondo M, Díaz-Beveridge R, de la Calva C, Amaya-Valero JV, Ballester-Ibáñez C, Liquori A, Giner F, Mayordomo-Aranda E. Exploring the Potential of Optical Genome Mapping in the Diagnosis and Prognosis of Soft Tissue and Bone Tumors. Int J Mol Sci 2025; 26:2820. [PMID: 40141463 PMCID: PMC11942867 DOI: 10.3390/ijms26062820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
Sarcomas are rare malignant tumors of mesenchymal origin with a high misdiagnosis rate due to their heterogeneity and low incidence. Conventional diagnostic techniques, such as Fluorescence In Situ Hybridization (FISH) and Next-Generation Sequencing (NGS), have limitations in detecting structural variations (SVs), copy number variations (CNVs), and predicting clinical behavior. Optical genome mapping (OGM) provides high-resolution genome-wide analysis, improving sarcoma diagnosis and prognosis assessment. This study analyzed 53 sarcoma samples using OGM. Ultra-high molecular weight (UHMW) DNA was extracted from core and resection biopsies, and data acquisition was performed with the Bionano Saphyr platform. Bioinformatic pipelines identified structural variations, comparing them with known alterations for each sarcoma subtype. OGM successfully analyzed 62.3% of samples. Diagnostic-defining alterations were found in 95.2% of cases, refining diagnoses and revealing novel oncogenic and tumor suppressor gene alterations. The challenges included DNA extraction and quality issues from some tissue samples. Despite these limitations, OGM proved to be a powerful diagnostic and predictive tool for bone and soft tissue sarcomas, surpassing conventional methods in resolution and scope, enhancing the understanding of sarcoma genetics, and enabling better patient stratification and personalized therapies.
Collapse
Affiliation(s)
- Alejandro Berenguer-Rubio
- Cytogenetic Laboratory, Instituto de Investigación Sanitaria La Fe, 46026 València, Spain; (A.B.-R.); (N.T.H.); (Á.D.-G.); (G.A.); (C.G.-A.)
- Department of Hematology, Hospital Universitari i Politècnic La Fe, 46026 València, Spain;
| | - Esperanza Such
- Cytogenetic Laboratory, Instituto de Investigación Sanitaria La Fe, 46026 València, Spain; (A.B.-R.); (N.T.H.); (Á.D.-G.); (G.A.); (C.G.-A.)
- Department of Hematology, Hospital Universitari i Politècnic La Fe, 46026 València, Spain;
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Neus Torres Hernández
- Cytogenetic Laboratory, Instituto de Investigación Sanitaria La Fe, 46026 València, Spain; (A.B.-R.); (N.T.H.); (Á.D.-G.); (G.A.); (C.G.-A.)
- Department of Hematology, Hospital Universitari i Politècnic La Fe, 46026 València, Spain;
| | - Paula González-Rojo
- Pathology Department, Hospital Universitari i Politècnic La Fe, 46026 València, Spain; (P.G.-R.); (J.G.-L.); (N.P.-B.); (L.A.R.-M.); (S.H.-G.); (M.S.V.-C.); (F.G.); (E.M.-A.)
| | - Álvaro Díaz-González
- Cytogenetic Laboratory, Instituto de Investigación Sanitaria La Fe, 46026 València, Spain; (A.B.-R.); (N.T.H.); (Á.D.-G.); (G.A.); (C.G.-A.)
- Department of Hematology, Hospital Universitari i Politècnic La Fe, 46026 València, Spain;
| | - Gayane Avetisyan
- Cytogenetic Laboratory, Instituto de Investigación Sanitaria La Fe, 46026 València, Spain; (A.B.-R.); (N.T.H.); (Á.D.-G.); (G.A.); (C.G.-A.)
- Department of Hematology, Hospital Universitari i Politècnic La Fe, 46026 València, Spain;
| | - Carolina Gil-Aparicio
- Cytogenetic Laboratory, Instituto de Investigación Sanitaria La Fe, 46026 València, Spain; (A.B.-R.); (N.T.H.); (Á.D.-G.); (G.A.); (C.G.-A.)
- Department of Hematology, Hospital Universitari i Politècnic La Fe, 46026 València, Spain;
| | - Judith González-López
- Pathology Department, Hospital Universitari i Politècnic La Fe, 46026 València, Spain; (P.G.-R.); (J.G.-L.); (N.P.-B.); (L.A.R.-M.); (S.H.-G.); (M.S.V.-C.); (F.G.); (E.M.-A.)
| | - Nicolay Pantoja-Borja
- Pathology Department, Hospital Universitari i Politècnic La Fe, 46026 València, Spain; (P.G.-R.); (J.G.-L.); (N.P.-B.); (L.A.R.-M.); (S.H.-G.); (M.S.V.-C.); (F.G.); (E.M.-A.)
| | - Luis Alberto Rubio-Martínez
- Pathology Department, Hospital Universitari i Politècnic La Fe, 46026 València, Spain; (P.G.-R.); (J.G.-L.); (N.P.-B.); (L.A.R.-M.); (S.H.-G.); (M.S.V.-C.); (F.G.); (E.M.-A.)
| | - Soraya Hernández-Girón
- Pathology Department, Hospital Universitari i Politècnic La Fe, 46026 València, Spain; (P.G.-R.); (J.G.-L.); (N.P.-B.); (L.A.R.-M.); (S.H.-G.); (M.S.V.-C.); (F.G.); (E.M.-A.)
| | - María Soledad Valera-Cuesta
- Pathology Department, Hospital Universitari i Politècnic La Fe, 46026 València, Spain; (P.G.-R.); (J.G.-L.); (N.P.-B.); (L.A.R.-M.); (S.H.-G.); (M.S.V.-C.); (F.G.); (E.M.-A.)
| | - Cristina Ramírez-Fuentes
- Radiology Department, Hospital Universitari i Politècnic La Fe, 46026 València, Spain; (C.R.-F.); (M.S.-R.)
| | - María Simonet-Redondo
- Radiology Department, Hospital Universitari i Politècnic La Fe, 46026 València, Spain; (C.R.-F.); (M.S.-R.)
| | - Roberto Díaz-Beveridge
- Medical Oncology Service, Hospital Universitari i Politècnic La Fe, 46026 València, Spain;
| | - Carolina de la Calva
- Orthopaedics and Traumatology Department, Hospital Universitari i Politècnic La Fe, 46026 València, Spain; (C.d.l.C.); (J.V.A.-V.)
| | - José Vicente Amaya-Valero
- Orthopaedics and Traumatology Department, Hospital Universitari i Politècnic La Fe, 46026 València, Spain; (C.d.l.C.); (J.V.A.-V.)
| | | | - Alessandro Liquori
- Department of Hematology, Hospital Universitari i Politècnic La Fe, 46026 València, Spain;
| | - Francisco Giner
- Pathology Department, Hospital Universitari i Politècnic La Fe, 46026 València, Spain; (P.G.-R.); (J.G.-L.); (N.P.-B.); (L.A.R.-M.); (S.H.-G.); (M.S.V.-C.); (F.G.); (E.M.-A.)
| | - Empar Mayordomo-Aranda
- Pathology Department, Hospital Universitari i Politècnic La Fe, 46026 València, Spain; (P.G.-R.); (J.G.-L.); (N.P.-B.); (L.A.R.-M.); (S.H.-G.); (M.S.V.-C.); (F.G.); (E.M.-A.)
| |
Collapse
|
18
|
Ma Y, Shih CH, Cheng J, Chen HC, Wang LJ, Tan Y, Zhang Y, Brown DD, Oesterreich S, Lee AV, Chiu YC, Chen YC. High-Throughput Empirical and Virtual Screening To Discover Novel Inhibitors of Polyploid Giant Cancer Cells in Breast Cancer. Anal Chem 2025; 97:5498-5506. [PMID: 40040372 PMCID: PMC11923954 DOI: 10.1021/acs.analchem.4c05138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Therapy resistance in breast cancer is increasingly attributed to polyploid giant cancer cells (PGCCs), which arise through whole genome doubling and exhibit heightened resilience to standard treatments. Characterized by enlarged nuclei and increased DNA content, these cells tend to be dormant under therapeutic stress, driving disease relapse. Despite their critical role in resistance, strategies to effectively target PGCCs are limited, largely due to the lack of high-throughput methods for assessing their viability. Traditional assays lack the sensitivity needed to detect PGCC-specific elimination, prompting the development of novel approaches. To address this challenge, we developed a high-throughput single-cell morphological analysis workflow designed to differentiate compounds that selectively inhibit non-PGCCs, PGCCs, or both. Using this method, we screened a library of 2726 FDA Phase 1-approved drugs, identifying promising anti-PGCC candidates, including proteasome inhibitors, FOXM1, CHK, and macrocyclic lactones. Notably, RNA-Seq analysis of cells treated with the macrocyclic lactone Pyronaridine revealed AXL inhibition as a potential strategy for targeting PGCCs. Although our single-cell morphological analysis pipeline is powerful, empirical testing of all existing compounds is impractical and inefficient. To overcome this limitation, we trained a machine learning model to predict anti-PGCC efficacy in silico, integrating chemical fingerprints and compound descriptions from prior publications and databases. The model demonstrated a high correlation with experimental outcomes and predicted efficacious compounds in an expanded library of over 6,000 drugs. Among the top-ranked predictions, we experimentally validated five compounds as potent PGCC inhibitors using cell lines and patient-derived models. These findings underscore the synergistic potential of integrating high-throughput empirical screening with machine learning-based virtual screening to accelerate the discovery of novel therapies, particularly for targeting therapy-resistant PGCCs in breast cancer.
Collapse
Affiliation(s)
- Yushu Ma
- UPMC
Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, Pennsylvania 15232, United States
- Department
of Computational and Systems Biology, University
of Pittsburgh, 3420 Forbes
Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Chien-Hung Shih
- UPMC
Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, Pennsylvania 15232, United States
| | - Jinxiong Cheng
- UPMC
Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, Pennsylvania 15232, United States
- Department
of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O’Hara Street, Pittsburgh, Pennsylvania 15260, United States
| | - Hsiao-Chun Chen
- UPMC
Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, Pennsylvania 15232, United States
- Department
of Computational and Systems Biology, University
of Pittsburgh, 3420 Forbes
Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Li-Ju Wang
- UPMC
Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, Pennsylvania 15232, United States
| | - Yanhao Tan
- UPMC
Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, Pennsylvania 15232, United States
- Division
of Malignant Hematology and Medical Oncology, Department of Medicine, University of Pittsburgh, 5150 Centre Avenue, Pittsburgh, Pennsylvania 15232, United States
| | - Yuan Zhang
- UPMC
Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, Pennsylvania 15232, United States
- Department
of Immunology, University of Pittsburgh, 5051 Centre Ave, Pittsburgh, Pennsylvania 15213, United States
| | - Daniel D. Brown
- Institute
for Precision Medicine, University of Pittsburgh, 5051 Centre Ave, Pittsburgh, Pennsylvania 15213, United States
| | - Steffi Oesterreich
- UPMC
Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, Pennsylvania 15232, United States
- Department
of Pharmacology and Chemical Biology, University
of Pittsburgh, 4200 Fifth
Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Adrian V. Lee
- UPMC
Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, Pennsylvania 15232, United States
- Institute
for Precision Medicine, University of Pittsburgh, 5051 Centre Ave, Pittsburgh, Pennsylvania 15213, United States
- Department
of Pharmacology and Chemical Biology, University
of Pittsburgh, 4200 Fifth
Avenue, Pittsburgh, Pennsylvania 15213, United States
| | - Yu-Chiao Chiu
- UPMC
Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, Pennsylvania 15232, United States
- Department
of Computational and Systems Biology, University
of Pittsburgh, 3420 Forbes
Avenue, Pittsburgh, Pennsylvania 15260, United States
- Division
of Malignant Hematology and Medical Oncology, Department of Medicine, University of Pittsburgh, 5150 Centre Avenue, Pittsburgh, Pennsylvania 15232, United States
- CMU-Pitt
Ph.D. Program in Computational Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Yu-Chih Chen
- UPMC
Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, Pennsylvania 15232, United States
- Department
of Computational and Systems Biology, University
of Pittsburgh, 3420 Forbes
Avenue, Pittsburgh, Pennsylvania 15260, United States
- Department
of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O’Hara Street, Pittsburgh, Pennsylvania 15260, United States
- CMU-Pitt
Ph.D. Program in Computational Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
19
|
Xian W, Wang S, Xie J, Yamamoto Y, Khorrami M, Zhang Y, Montes RC, Desales C, Khorrami M, Mory Z, Hoffman A, Su A, Nguyen C, Davies PJA, Stephan C, Pan S, Wu W, Liu Y, Siegelman J, Waters RE, Ross WA, Song S, Metersky M, Beer DG, Crum CP, Stewart AJ, Vincent M, Russell R, Izard RA, Ho KY, Hung-Sen Lai J, Bachovchin WW, Ajani JA, McKeon FD. Evolution of Esophageal Adenocarcinoma From Precursor Lesion Stem Cells. Gastroenterology 2025:S0016-5085(25)00521-9. [PMID: 40090599 DOI: 10.1053/j.gastro.2025.02.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND & AIMS Metastatic cancers arise from a decades-long succession of increasingly virulent precursor lesions, each of which represents prospective targets for therapeutic intervention. This evolutionary process has been particularly vivid in esophageal adenocarcinoma (EAC), as this cancer and associated precursor lesions, including Barrett's esophagus (BE), low-grade dysplasia (LGD), and high-grade dysplasia (HGD), coexist in an accessible, 2-dimensional pattern in esophageal mucosa. Given the durability of these precursor lesions, it is likely that they, like EAC, rely on stem cells for their regenerative growth. To assess the role of stem cells in the evolution of EAC, we apply technology that selectively clones stem cells from the gastrointestinal tract to patient-matched endoscopic biopsies from each of the precursor lesions implicated in EAC. METHODS Histologically validated, endoscopic biopsy series including EAC, HGD, LGD, BE, and normal esophageal mucosa were obtained from patients presenting with EAC. Rare (1:1000) cells from each of these lesions proved clonogenic and were assessed by in vitro differentiation, tumorigenicity in mice, and by molecular genetics. RESULTS Each of the lesions in the evolution of EAC possesses a discrete set of clonogenic cells marked by immaturity, enormous proliferative potential, and lesion-specific differentiation fate. DNA sequencing of these clones reveals intralesional heterogeneity and clonal resolution of the mutation progression within a given patient from BE, LGD, HGD, and EAC. High-throughput chemical screens against BE stem cells reveal drug combinations that are similarly effective against stem cells of LGD, HGD, and EAC. CONCLUSIONS All lesions in the evolution of EAC possess discrete populations of stem cells that are potential therapeutic targets.
Collapse
Affiliation(s)
- Wa Xian
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Shan Wang
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Jingzhong Xie
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Yusuke Yamamoto
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | - Melina Khorrami
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Yanting Zhang
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | | | - Caycel Desales
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Melika Khorrami
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Zaal Mory
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Ashley Hoffman
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Amber Su
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Crystal Nguyen
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | | | | | - Shuang Pan
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts
| | - Wengen Wu
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts
| | - Yuxin Liu
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts
| | - Jeremy Siegelman
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
| | - Rebecca E Waters
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - William A Ross
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shumei Song
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark Metersky
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine, University of Connecticut Health Center, Farmington, Connecticut
| | - David G Beer
- Departments of Thoracic Surgery and Radiation Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Christopher P Crum
- Department of Pathology, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts
| | - Alexander J Stewart
- School of Mathematics and Statistics, University of St. Andrews, North Haugh, United Kingdom
| | | | | | | | - Khek Yu Ho
- Department of Medicine, National University of Singapore, Singapore
| | - Jack Hung-Sen Lai
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts; Department of Developmental, Molecular and Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, Massachusetts
| | - William W Bachovchin
- Sackler School of Graduate Biomedical Science, Tufts University, Boston, Massachusetts; Department of Developmental, Molecular and Chemical Biology, Tufts University Graduate School of Biomedical Sciences, Boston, Massachusetts
| | - Jaffer A Ajani
- Department of Gastrointestinal Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Frank D McKeon
- Department of Biology and Biochemistry, University of Houston, Houston, Texas.
| |
Collapse
|
20
|
Di Patria L, Habel N, Olaso R, Fernandes R, Brenner C, Stefanovska B, Fromigue O. C-terminal binding protein-2 triggers CYR61-induced metastatic dissemination of osteosarcoma in a non-hypoxic microenvironment. J Exp Clin Cancer Res 2025; 44:83. [PMID: 40038783 PMCID: PMC11881356 DOI: 10.1186/s13046-025-03350-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/23/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Osteosarcoma is the most prevalent cancer-related bone disease diagnosed in the pediatric age group. The rapid development of metastatic lesions and resistance to chemotherapy remain major mechanisms responsible for the failure of treatments and poor outcome. We established that the expression level of Cysteine-rich protein 61 (CYR61/CCN1) correlates to tumor neo-vascularization and dissemination in preclinical and clinical osteosarcoma samples. The aim of this study was to investigate the CYR61-related mechanisms leading to the acquisition of metastatic capacity by osteosarcoma cells. METHODS Transcriptomic data issued from RNA-seq were subjected to pathways and gene set enrichment analyses. Murine and human cell lines with overexpressed or downregulated C-terminal Binding protein 2 (CtBP2) were established by lentiviral transduction. Cell metabolic activity was assessed by Seahorse XF Analyzer; cell replication rate by BrdU incorporation assay; stemness by clonogenicity assay and RT-qPCR detection of markers; cell migration by wound healing assay and Boyden chambers system; cell invasion using Matrigel coated Boyden chambers or fluorescence microscopy of Matrigel embedded 3D spheroids. FFPE samples derived from syngeneic tumor cells grafts into BALB/c mice were analyzed by IHC. The protein interactome was predicted in silico using the STRING database. RESULTS GSEA revealed that CYR61 modulate the transcription process. The in vitro expression level of CtBP2 and Cyr61 correlated positively in a panel of osteosarcoma cell lines. In silico analysis of protein-protein interaction network revealed a link with stemness markers. Variations in CtBP2 expression levels influenced stemness markers expression levels, cell clonogenicity, cell migration, Matrix Metalloproteinase activity and cell invasion. Surprisingly, while induction of CtBP2 expression under CYR61 correlated with the metastatic dissemination process in vivo, it occurred only at the invasive front of tumors. Hypoxic conditions in central tumor region interfered with CtBP2 induction of expression. CONCLUSIONS Our findings identify for the first time that CtBP2 acts as a required critical inducing factor in the CYR61-related metastatic progression of osteosarcoma, by favoring cell migration and invasiveness. Moreover, we demonstrate that while CtBP2 is a downstream transcriptional target of CYR61 signaling cascade, it occurs only under non-hypoxic conditions. The present study suggests that CtBP2 may represent a potential pivotal target for therapeutic management of metastases spreading in osteosarcoma.
Collapse
Affiliation(s)
- Laura Di Patria
- Inserm UMR981, Gustave Roussy Cancer Campus, Molecular Predictors and New Targets in Oncology, Université Paris Saclay, 39 Rue Camille Desmoulins, Villejuif, F-94805, France
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Nadia Habel
- Inserm UMR981, Gustave Roussy Cancer Campus, Molecular Predictors and New Targets in Oncology, Université Paris Saclay, 39 Rue Camille Desmoulins, Villejuif, F-94805, France
- Present Address : Centre de Traitement de L'Information Génétique (CTIG), INRAE, Jouy en Josas, France
| | - Robert Olaso
- Université Paris Saclay, CEA, Centre National de Recherche en Génomique Humaine (CNRGH), Evry, France
| | - Romain Fernandes
- CNRS UMR9018, Gustave Roussy, Metabolic and Systemic Aspects of Oncogenesis for New Therapeutic Approaches, Université Paris Saclay, Villejuif, France
| | - Catherine Brenner
- CNRS UMR9018, Gustave Roussy, Metabolic and Systemic Aspects of Oncogenesis for New Therapeutic Approaches, Université Paris Saclay, Villejuif, France
| | - Bojana Stefanovska
- Inserm UMR981, Gustave Roussy Cancer Campus, Molecular Predictors and New Targets in Oncology, Université Paris Saclay, 39 Rue Camille Desmoulins, Villejuif, F-94805, France
- Present Address: Department of Biochemistry and Structural Biology, Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Olivia Fromigue
- Inserm UMR981, Gustave Roussy Cancer Campus, Molecular Predictors and New Targets in Oncology, Université Paris Saclay, 39 Rue Camille Desmoulins, Villejuif, F-94805, France.
| |
Collapse
|
21
|
Mao X, Rao G, Li G, Chen S. Insights into Extrachromosomal DNA in Cancer: Biogenesis, Methodologies, Functions, and Therapeutic Potential. Adv Biol (Weinh) 2025; 9:e2400433. [PMID: 39945006 DOI: 10.1002/adbi.202400433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 02/01/2025] [Indexed: 03/17/2025]
Abstract
Originating from, but independent of, linear chromosomes, extrachromosomal DNA (ecDNA) exists in a more active state of transcription and autonomous replication. It plays a crucial role in the development of malignancies and therapy resistance. Since its discovery in eukaryotic cells more than half a century ago, the biological characteristics and functions of ecDNA have remained unclear due to limitations in detection methods. However, recent advancements in research tools have transformed ecDNA research. It is believed that ecDNA exhibits greater activity in the abnormal amplification of oncogenes, thereby driving cancer progression through their overexpression. Notably, compared to linear DNA, ecDNA can also function as a genomic element with regulatory roles, including both trans- and cis-acting functions. Its critical roles in tumorigenesis, evolution, progression, and drug resistance in malignant tumors are increasingly recognized. This review provides a comprehensive summary of the evolutionary context of ecDNA and highlights significant progress in understanding its biological functions and potential applications as a therapeutic target in malignant tumors.
Collapse
Affiliation(s)
- Xudong Mao
- Department of Urology, The Affiliated Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, P. R. China
| | - Guocheng Rao
- Department of Endocrinology & Metabolism, Daepartment of Biotherapy, Center for Diabetes and Metabolism Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center of Biotherapy, Chengdu, Sichuan, 610000, P. R. China
| | - Gonghui Li
- Department of Urology, The Affiliated Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, P. R. China
| | - Shihan Chen
- Department of Endocrinology, The Affiliated Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, P. R. China
| |
Collapse
|
22
|
Wei Q, Hu S, Loghavi S, Toruner GA, Ravandi-Kashani F, Tang Z, Li S, Xu J, Daver N, Medeiros LJ, Tang G. Chromoanagenesis Is Frequently Associated With Highly Complex Karyotypes, Extensive Clonal Heterogeneity, and Treatment Refractoriness in Acute Myeloid Leukemia. Am J Hematol 2025; 100:417-426. [PMID: 39871121 DOI: 10.1002/ajh.27575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/03/2024] [Accepted: 12/23/2024] [Indexed: 01/29/2025]
Abstract
Chromoanagenesis (CAG) encompasses a spectrum of catastrophic genomic events, including chromothripsis, chromoanasynthesis, and chromoplexy. We studied CAG in 410 patients with a diagnosis of acute myeloid leukemia (AML), 292 newly diagnosed (ND), and 118 refractory/relapsed, using optical genome mapping. CAG was identified by the presence of clusters (with 10 or more breakpoints) of structural abnormalities and/or segmental copy number alterations within one or more chromosomal regions. CAG was detected in 65 (16%) patients. Compared with patients without CAG, those with CAG showed significantly (p < 0.0001) higher frequencies of highly complex karyotype (92% vs. 11%), monosomal karyotype (88% vs. 12%), extensive clonal heterogeneity (75% vs. 7%), gene amplification (49% vs. 1%), and TP53 deletion/mutation (92% vs. 9%). Overall, CAG was detected in about two-thirds of AML patients who exhibited the abovementioned high-risk cytogenetic abnormalities/karyotype. Among the 42 patients with ND AML and CAG, 36 received treatments and follow-ups, and 28 (78%) had no or only partial response to therapy. Among patients with ND AML, those with CAG had a shorter overall survival than those without CAG (median survival: 5 vs. 14 months, p < 0.0001). However, in multivariate analysis, CAG did not appear to be an independent risk factor for survival. These results indicate that CAG is frequently associated with high-risk chromosomal alterations and genomic instability in AML and may contribute to treatment refractoriness and inferior survival in this subset of AML patients.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/therapy
- Female
- Male
- Middle Aged
- Aged
- Adult
- Aged, 80 and over
- Chromothripsis
- Chromosome Aberrations
- Adolescent
- Young Adult
- Drug Resistance, Neoplasm/genetics
- Karyotype
Collapse
Affiliation(s)
- Qing Wei
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Shimin Hu
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Gokce A Toruner
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Farhad Ravandi-Kashani
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhenya Tang
- Department of Pathology, Microbiology and Immunology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Shaoying Li
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jie Xu
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Naval Daver
- Department of Leukemia, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Guilin Tang
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
23
|
Mohamed AM, Eid M, Eid O, Hussein SH, Mahmoud W, Mahrous R, Refaat K, Farid M. Generation of Dual-Color FISH probes targeting 9p21, Xp21, and 17p13.1 loci as diagnostic markers for some genetic disorders and cancer in Egypt. J Genet Eng Biotechnol 2025; 23:100449. [PMID: 40074450 PMCID: PMC11720894 DOI: 10.1016/j.jgeb.2024.100449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 11/18/2024] [Accepted: 12/05/2024] [Indexed: 03/14/2025]
Abstract
INTRODUCTION The fluorescence in situ hybridization (FISH) is a very important technique, as it can diagnose many genetic disorders and cancers. Molecular cytogenetic analysis (FISH) can diagnose numerical chromosome aberrations, sex chromosomes anomalies, and many genetic disorders. AIM With the limited number of commercially available probes that do not cover all research needs and the high prices of the commercial probes, our goal is to apply recent technologies to produce FISH probes that can accurately and sensitively diagnose genetic diseases and cancer in Egypt and establishing the inhouse production of different FISH probes. We intend to adhere to the published guidelines and validation procedures to ensure the production of accurate FISH probes for clinical diagnosis. METHODS We used specific DNA segments extracted from BAC clones, and we performed nick translation to label the segment with fluorescence labeled dye. The second method involved the use of specific primers for the centromere of certain chromosomes and using PCR technique for amplification and labeling. The probes were tested on metaphase and interphase cells derived from cultured human peripheral blood samples. We followed standard guidelines to test the adequacy of probe slide hybridization, proper probe localization, probe sensitivity and specificity, probe reproducibility, cut-off values, and overall probe validation. RESULTS In this research, we presented the generation of three dual-color probes, each probe has a control locus. We offered three dual-color probes targeted 9p21, Xp21 and 17p13.1 loci. chromosome 9p21probe for diagnosis of structural abnormalities in chromosome 9, the Xp21 to test for structural abnormalities of chromosome X, and the 17p13.1 for TP53 gene to detect the loss of p53. We also produced probes for Down syndrome specific region, Rb gene and centromeres for chromosomes X, 17, and 18. CONCLUSION The produced probes are specific and sensitive and can be produced at the commercial level in the laboratory. The production of FISH probes in Egypt can be used as a powerful diagnostic marker for genetic disorders and cancers and our work can be consider as a base to start national project to produce our needs of FISH probes.
Collapse
Affiliation(s)
- Amal M Mohamed
- Human Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Egypt.
| | - Maha Eid
- Human Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Egypt
| | - Ola Eid
- Human Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Egypt
| | - Shymaa H Hussein
- Human Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Egypt
| | - Wael Mahmoud
- Human Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Egypt
| | - Rana Mahrous
- Human Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Egypt
| | - Khaled Refaat
- Human Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Egypt
| | - Marwa Farid
- Human Genetics Department, Human Genetics and Genome Research Institute, National Research Centre, Egypt
| |
Collapse
|
24
|
Takimoto N, Ishii Y, Mitsumoto T, Takasu S, Namiki M, Toyoda T, Shibutani M, Ogawa K. Involvement of nuclear atrophy of binucleated hepatocytes in the large micronucleus formation induced by rat hepatocarcinogen acetamide. Toxicol Appl Pharmacol 2025; 496:117243. [PMID: 39870197 DOI: 10.1016/j.taap.2025.117243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/09/2025] [Accepted: 01/22/2025] [Indexed: 01/29/2025]
Abstract
Acetamide is a hepatocarcinogen in rats. We previously revealed that acetamide induces characteristic large micronuclei in rat liver, suggesting the possible involvement of chromosome aberrations in acetamide-induced hepatocarcinogenesis. To elucidate the mechanism of large micronuclei formation, in this study we examined time-dependent changes in rat hepatocytes after administration of acetamide. Male 6-week-old F344 rats were gavaged with a single-dose administration of acetamide. A liver micronucleus test showed large micronuclei formation 48 and 72 h after acetamide administration. Histopathological analysis showed binucleated hepatocytes with a unilateral atrophic nucleus beginning 6 h after acetamide administration, and the number reached a maximum at 24 h. At 48 h, the number of binucleated hepatocytes with an atrophic nucleus decreased, and apoptotic hepatocytes and large micronucleated hepatocytes appeared. The changes in the frequency of these abnormal binucleated hepatocytes demonstrated a transition from atrophic nuclei to large micronuclei. Immunohistopathological examinations of binucleated hepatocytes showed loss of nuclear lamina, accumulation of barrier-to-autointegration factor (BAF) and chromatin condensation with heterochromatinization at the atrophic site of nuclei. Results of a BrdU-labeling assay were negative. The abnormal expression of BAF in morphologically normal nuclei suggested that nuclear envelope aberration in hepatocytes was an initial event of the nuclear atrophy. In addition, lack of involvement of cell division in the nuclear atrophy and large micronucleus formation was also demonstrated by BrdU-labeling assay. Overall, our data suggest that large micronuclei induced by acetamide are formed in binucleated hepatocytes through nuclear atrophy.
Collapse
Affiliation(s)
- Norifumi Takimoto
- Division of Pathology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan; Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Yuji Ishii
- Division of Pathology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan.
| | - Tatsuya Mitsumoto
- Division of Pathology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan; Faculty of Animal Health Technology, Yamazaki University of Animal Health Technology, 4-7-2, Minami-osawa, Hachioji, Tokyo 192-0364, Japan
| | - Shinji Takasu
- Division of Pathology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Moeka Namiki
- Division of Pathology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Takeshi Toyoda
- Division of Pathology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan
| | - Makoto Shibutani
- Laboratory of Veterinary Pathology, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan
| | - Kumiko Ogawa
- Division of Pathology, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki-shi, Kanagawa 210-9501, Japan
| |
Collapse
|
25
|
Eugen-Olsen RB, Hariprakash J, Oestergaard V, Regenberg B. Molecular mechanisms of extrachromosomal circular DNA formation. Nucleic Acids Res 2025; 53:gkaf122. [PMID: 40037708 PMCID: PMC11879418 DOI: 10.1093/nar/gkaf122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 01/26/2025] [Accepted: 02/06/2025] [Indexed: 03/06/2025] Open
Abstract
Recent research reveals that eukaryotic genomes form circular DNA from all parts of their genome, some large enough to carry whole genes. In organisms like yeast and in human cancers, it is often observed that extrachromosomal circular DNA (eccDNA) benefits the individual cell by providing resources for rapid cellular growth. However, our comprehension of eccDNA remains incomplete, primarily due to their transient nature. Early studies suggest they arise when DNA breaks and is subsequently repaired incorrectly. In this review, we provide an overview of the evidence for molecular mechanisms that lead to eccDNA formation in human cancers and yeast, focusing on nonhomologous end joining, alternative end joining, and homologous recombination repair pathways. Furthermore, we present hypotheses in the form of molecular eccDNA formation models and consider cellular conditions which may affect eccDNA generation. Finally, we discuss the framework for future experimental evidence.
Collapse
Affiliation(s)
- Rasmus A B Eugen-Olsen
- Department of Biology, University of Copenhagen, Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Judith M Hariprakash
- Department of Biology, University of Copenhagen, Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Vibe H Oestergaard
- Department of Biology, University of Copenhagen, Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Birgitte Regenberg
- Department of Biology, University of Copenhagen, Copenhagen, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
26
|
Nichols A, Norman R, Chen Y, Choi Y, Striepen J, Salataj E, Toufektchan E, Koche R, Maciejowski J. Mitotic transcription ensures ecDNA inheritance through chromosomal tethering. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637945. [PMID: 39990406 PMCID: PMC11844496 DOI: 10.1101/2025.02.12.637945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Extrachromosomal DNA (ecDNA) are circular DNA bodies that play critical roles in tumor progression and treatment resistance by amplifying oncogenes across a wide range of cancer types. ecDNA lack centromeres and are thus not constrained by typical Mendelian segregation, enabling their unequal accumulation within daughter cells and associated increases in copy number. Despite intrinsic links to their oncogenic potential, the fidelity and mechanisms of ecDNA inheritance are poorly understood. Here, we show that ecDNA are protected against cytosolic mis-segregation through mitotic clustering and by tethering to the telomeric and subtelomeric regions of mitotic chromosomes. ecDNA-chromosome tethering depends on BRD4 transcriptional co-activation and mitotic transcription of the long non-coding RNA PVT1 , which is co-amplified with MYC in colorectal and prostate cancer cell lines. Disruption of ecDNA-chromosome tethering through BRD4 inhibition, PVT1 depletion, or inhibiting mitotic transcription results in cytosolic mis-segregation, ecDNA reintegration, and the formation of homogeneously staining regions (HSRs). We propose that nuclear inheritance of ecDNA is facilitated by an RNA-mediated physical tether that links ecDNA to mitotic chromosomes and thus protects against cytosolic mis-segregation and chromosomal integration.
Collapse
|
27
|
Simovic-Lorenz M, Ernst A. Chromothripsis in cancer. Nat Rev Cancer 2025; 25:79-92. [PMID: 39548283 DOI: 10.1038/s41568-024-00769-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 11/17/2024]
Abstract
Chromothripsis is a mutational phenomenon in which a single catastrophic event generates extensive rearrangements of one or a few chromosomes. This extreme form of genome instability has been detected in 30-50% of cancers. Studies conducted in the past few years have uncovered insights into how chromothripsis arises and deciphered some of the cellular and molecular consequences of chromosome shattering. This Review discusses the defining features of chromothripsis and describes its prevalence across different cancer types as indicated by the manifestations of chromothripsis detected in human cancer samples. The different mechanistic models of chromothripsis, derived from in vitro systems that enable causal inference through experimental manipulation, are discussed in detail. The contribution of chromothripsis to cancer development, the selective advantages that cancer cells might gain from chromothripsis, the evolutionary trajectories of chromothriptic tumours, and the potential vulnerabilities and therapeutic opportunities presented by chromothriptic cells are also highlighted.
Collapse
Affiliation(s)
- Milena Simovic-Lorenz
- Group Genome Instability in Tumors, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Aurélie Ernst
- Group Genome Instability in Tumors, German Cancer Research Center, Heidelberg, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
28
|
Lu YY, Lu W, Zheng J, Luo JS. High APEX1 Expression Facilitates Osteosarcoma Cell Proliferation. Asian Pac J Cancer Prev 2025; 26:453-463. [PMID: 40022689 PMCID: PMC12118012 DOI: 10.31557/apjcp.2025.26.2.453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 02/06/2025] [Indexed: 03/03/2025] Open
Abstract
Osteosarcoma (OS) is a serious malignancy affecting children and young adults; however, there is limited improvement in the survival of patients with OS over the past four decades. Molecular targeted therapy is a promising treatment strategy for OS. Apurinic/apyrimidinic exonuclease 1 (APEX1)-a key factor for DNA damage repair-is associated with OS proliferation, but the underlying molecular mechanism remains unclear. APEX1 expression in OS tissues and paired paracancerous tissues and in human osteoblast cell line hFOB1.19 and OS cell lines was determined using real-time quantitative PCR (RT-qPCR). APEX1-shRNA and NC-shRNA lentiviral vectors were constructed and transfected into MG-63 cells. The effects of APEX1 knockdown on MG-63 cell proliferation and apoptosis were assessed using MTT, xenograft tumor growth, and terminal deoxynucleotidyl transferase dUTP nick end labeling assays. Expression changes of apoptosis- and angiogenesis-related genes due to APEX1 knockdown were detected using RT-qPCR and immunohistochemistry. To preliminarily determine the mechanism by which APEX1 affects OS cell proliferation, transcription factors were predicted using three databases, and construction of protein-protein interaction network, gene ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses were performed. APEX1 expression was higher in OS tissues than in paracancerous tissues. APEX1 expression was also higher in OS cell lines than in hFOB1.19 cells, with the highest APEX1 expression observed in MG-63 cells. APEX1 knockdown mediated by APEX1-shRNA lentivirus markedly suppressed MG-63 cell proliferation both in vitro and in vivo and induced their apoptosis. APEX1 knockdown downregulated CD31 expression but had no effect on the expression of P53 and Caspase3. Bioinformatics analyses suggested that USF1 or SP1 regulates APEX1 transcription and its recruitment in DNA damage response pathways, affecting OS cell proliferation. Thus, high APEX1 expression in OS facilitates cell proliferation likely via CD31, and USF1 or SP1 may regulate APEX1 transcription and its recruitment in DNA damage response pathways.
Collapse
Affiliation(s)
| | | | | | - Ju Shu Luo
- Division of Spinal Surgery, The First People’s Hospital of Yulin (The Sixth Affiliated Hospital of Guangxi Medical University), No. 495 Mid-way of education, Yulin, 537000, Guangxi, China.
| |
Collapse
|
29
|
Koeppel J, Ferreira R, Vanderstichele T, Riedmayr LM, Peets EM, Girling G, Weller J, Murat P, Liberante FG, Ellis T, Church GM, Parts L. Randomizing the human genome by engineering recombination between repeat elements. Science 2025; 387:eado3979. [PMID: 39883775 DOI: 10.1126/science.ado3979] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 08/09/2024] [Indexed: 02/01/2025]
Abstract
We lack tools to edit DNA sequences at scales necessary to study 99% of the human genome that is noncoding. To address this gap, we applied CRISPR prime editing to insert recombination handles into repetitive sequences, up to 1697 per cell line, which enables generating large-scale deletions, inversions, translocations, and circular DNA. Recombinase induction produced more than 100 stochastic megabase-sized rearrangements in each cell. We tracked these rearrangements over time to measure selection pressures, finding a preference for shorter variants that avoided essential genes. We characterized 29 clones with multiple rearrangements, finding an impact of deletions on expression of genes in the variant but not on nearby genes. This genome-scrambling strategy enables large deletions, sequence relocations, and the insertion of regulatory elements to explore genome dispensability and organization.
Collapse
Affiliation(s)
| | - Raphael Ferreira
- Harvard Medical School, Department of Genetics, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA, USA
| | | | - Lisa Maria Riedmayr
- Harvard Medical School, Department of Genetics, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
| | | | | | | | | | | | - Tom Ellis
- Wellcome Sanger Institute, Hinxton, UK
- Department of Bioengineering, Imperial College London, London, UK
| | - George McDonald Church
- Harvard Medical School, Department of Genetics, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Harvard-MIT Program in Health Sciences and Technology, Cambridge, Massachusetts, USA
- Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
30
|
Gardner AL, Zheng L, Howland K, Saunders A, Ramirez A, Parker P, Iloegbunam C, Morgan D, Jost TA, Brock A. Mapping cell-cell fusion at single-cell resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.11.627873. [PMID: 39896473 PMCID: PMC11785005 DOI: 10.1101/2024.12.11.627873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Cell-cell fusion is a tightly controlled process in the human body known to be involved in fertilization, placental development, muscle growth, bone remodeling, and viral response. Fusion between cancer cells results first in a whole-genome doubled state, which may be followed by the generation of aneuploidies; these genomic alterations are known drivers of tumor evolution. The role of cell-cell fusion in cancer progression and treatment response has been understudied due to limited experimental systems for tracking and analyzing individual fusion events. To meet this need, we developed a molecular toolkit to map the origins and outcomes of individual cell fusion events within a tumor cell population. This platform, ClonMapper Duo ('CMDuo'), identifies cells that have undergone cell-cell fusion through a combination of reporter expression and engineered fluorescence-associated index sequences paired to randomly generated nucleotide barcodes. scRNA-seq of the indexed barcodes enables the mapping of each set of parental cells and fusion progeny throughout the cell population. In triple-negative breast cancer cells CMDuo uncovered subclonal transcriptomic hybridization and unveiled distinct cell-states which arise in direct consequence of homotypic cell-cell fusion. CMDuo is a platform that enables mapping of cell-cell fusion events in high-throughput single cell data and enables the study of cell fusion in disease progression and therapeutic response.
Collapse
Affiliation(s)
- Andrea L Gardner
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Lan Zheng
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Kennedy Howland
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Andrew Saunders
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Andrea Ramirez
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Patrik Parker
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Chisom Iloegbunam
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Daylin Morgan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Tyler A Jost
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Amy Brock
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
31
|
Espejo Valle-Inclan J, De Noon S, Trevers K, Elrick H, van Belzen IAEM, Zumalave S, Sauer CM, Tanguy M, Butters T, Muyas F, Rust AG, Amary F, Tirabosco R, Giess A, Sosinsky A, Elgar G, Flanagan AM, Cortés-Ciriano I. Ongoing chromothripsis underpins osteosarcoma genome complexity and clonal evolution. Cell 2025; 188:352-370.e22. [PMID: 39814020 DOI: 10.1016/j.cell.2024.12.005] [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: 03/29/2024] [Revised: 08/05/2024] [Accepted: 12/07/2024] [Indexed: 01/18/2025]
Abstract
Osteosarcoma is the most common primary cancer of the bone, with a peak incidence in children and young adults. Using multi-region whole-genome sequencing, we find that chromothripsis is an ongoing mutational process, occurring subclonally in 74% of osteosarcomas. Chromothripsis generates highly unstable derivative chromosomes, the ongoing evolution of which drives the acquisition of oncogenic mutations, clonal diversification, and intra-tumor heterogeneity across diverse sarcomas and carcinomas. In addition, we characterize a new mechanism, termed loss-translocation-amplification (LTA) chromothripsis, which mediates punctuated evolution in about half of pediatric and adult high-grade osteosarcomas. LTA chromothripsis occurs when a single double-strand break triggers concomitant TP53 inactivation and oncogene amplification through breakage-fusion-bridge cycles. It is particularly prevalent in osteosarcoma and is not detected in other cancers driven by TP53 mutation. Finally, we identify the level of genome-wide loss of heterozygosity as a strong prognostic indicator for high-grade osteosarcoma.
Collapse
Affiliation(s)
| | - Solange De Noon
- Research Department of Pathology, University College London Cancer Institute, London WC1E 6DD, UK; Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK
| | - Katherine Trevers
- Research Department of Pathology, University College London Cancer Institute, London WC1E 6DD, UK; Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK
| | - Hillary Elrick
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK
| | - Ianthe A E M van Belzen
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK
| | - Sonia Zumalave
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK
| | - Carolin M Sauer
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK
| | - Mélanie Tanguy
- Scientific Research and Development, Genomics England, One Canada Square, London E14 5AB, UK
| | - Thomas Butters
- Research Department of Pathology, University College London Cancer Institute, London WC1E 6DD, UK; Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK
| | - Francesc Muyas
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK
| | - Alistair G Rust
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK
| | - Fernanda Amary
- Research Department of Pathology, University College London Cancer Institute, London WC1E 6DD, UK; Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK
| | - Roberto Tirabosco
- Research Department of Pathology, University College London Cancer Institute, London WC1E 6DD, UK; Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK
| | - Adam Giess
- Scientific Research and Development, Genomics England, One Canada Square, London E14 5AB, UK
| | | | - Greg Elgar
- Scientific Research and Development, Genomics England, One Canada Square, London E14 5AB, UK
| | - Adrienne M Flanagan
- Research Department of Pathology, University College London Cancer Institute, London WC1E 6DD, UK; Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore HA7 4LP, UK.
| | - Isidro Cortés-Ciriano
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton CB10 1SA, UK.
| |
Collapse
|
32
|
Liu Z, Wang Y, Peng Z, Li H, Wang H, Wu Y, Jiang X, Fu P. Fusion of tumor cells and mesenchymal stem/stroma cells: a source of tumor heterogeneity, evolution and recurrence. Med Oncol 2025; 42:52. [PMID: 39838167 DOI: 10.1007/s12032-024-02595-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 12/28/2024] [Indexed: 01/23/2025]
Abstract
The heterogeneity and evolution of tumors remain significant obstacles in cancer treatment, contributing to both therapy resistance and relapse. Mesenchymal stem/stromal cells (MSCs) are multipotent stromal cells within the tumor microenvironment that interact with tumor cells through various mechanisms, including cell fusion. While previous research has largely focused on the effects of MSC-tumor cell fusion on tumor proliferation, migration, and tumorigenicity, emerging evidence indicates that its role in tumor maintenance, evolution, and recurrence, particularly under stress conditions, may be even more pivotal. This review examines the connection between MSC-tumor cell fusion and several critical factors like tumor heterogeneity, cancer stem cells, and therapy resistance, highlighting the crucial role of cell fusion in tumor survival, evolution, and recurrence. Additionally, we explore potential therapeutic strategies aimed at targeting this process.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yihao Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zesheng Peng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Li
- Department of Cataract, Nanyang Eye Hospital, Nanyang, 473000, China
| | - Haofei Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuyi Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Peng Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
33
|
Collins RL, Talkowski ME. Diversity and consequences of structural variation in the human genome. Nat Rev Genet 2025:10.1038/s41576-024-00808-9. [PMID: 39838028 DOI: 10.1038/s41576-024-00808-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2024] [Indexed: 01/23/2025]
Abstract
The biomedical community is increasingly invested in capturing all genetic variants across human genomes, interpreting their functional consequences and translating these findings to the clinic. A crucial component of this endeavour is the discovery and characterization of structural variants (SVs), which are ubiquitous in the human population, heterogeneous in their mutational processes, key substrates for evolution and adaptation, and profound drivers of human disease. The recent emergence of new technologies and the remarkable scale of sequence-based population studies have begun to crystalize our understanding of SVs as a mutational class and their widespread influence across phenotypes. In this Review, we summarize recent discoveries and new insights into SVs in the human genome in terms of their mutational patterns, population genetics, functional consequences, and impact on human traits and disease. We conclude by outlining three frontiers to be explored by the field over the next decade.
Collapse
Affiliation(s)
- Ryan L Collins
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael E Talkowski
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
34
|
Zeng Y, Lv W, Tao H, Li C, Jiang S, Liang Y, Chen C, Yu T, Li Y, Wu S, Cui X, Liang N, Wang P, Xu H, Dong J, Teng H, Chen K, Mu K, Fan T, Cen X, Xu Z, Zhu M, Wang W, Mi J, Xiang X, Dong W, Yang H, Bolund L, Lin L, Song J, Song X, Luo Y, Lin C, Han P. Mapping the chromothripsis landscape in urothelial carcinoma unravels great intratumoral and intertumoral heterogeneity. iScience 2025; 28:111510. [PMID: 39790556 PMCID: PMC11714673 DOI: 10.1016/j.isci.2024.111510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 08/24/2024] [Accepted: 11/28/2024] [Indexed: 01/12/2025] Open
Abstract
Chromothripsis, a hallmark of cancer, is characterized by extensive and localized DNA rearrangements involving one or a few chromosomes. However, its genome-wide frequency and characteristics in urothelial carcinoma (UC) remain largely unknown. Here, by analyzing single-regional and multi-regional whole-genome sequencing (WGS), we present the chromothripsis blueprint in 488 UC patients. Chromothripsis events exhibit significant intertumoral heterogeneity, being detected in 41% of UC patients, with an increase from 30% in non-muscle-invasive disease (Ta/1) to 53% in muscle-invasive disease (T2-4). The presence of chromothripsis correlates with an unstable cancer genome and poor clinical outcomes. Analysis of multi-regional WGS data from 52 patients revealed pronounced intratumoral heterogeneity with chromothripsis events detectable only in specific tumor regions rather than uniformly across all areas. Chromothripsis events evolve under positive selection and contribute to tumor dissemination. This study presents a comprehensive genome-wide chromothripsis landscape in UC, highlighting the significance of chromothripsis in UC development.
Collapse
Affiliation(s)
- Yuchen Zeng
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Wei Lv
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- Department of Urology & Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China
- College of Life Sciences, University of Chinese Academy of Science, Beijing 100049, China
- Department of Biomedicine, Aarhus University, 8200 Aarhus, Denmark
| | - Huiying Tao
- The 2nd Medical College of Binzhou Medical University, Yantai, Shandong 264003, China
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Conghui Li
- Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Shiqi Jiang
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Yuan Liang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Chen Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Tianxi Yu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Yue Li
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Shuang Wu
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Xin Cui
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Ning Liang
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Ping Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing 100101, China
| | - Huixin Xu
- Department of Biomedicine, Aarhus University, 8200 Aarhus, Denmark
| | - Jingjing Dong
- Department of General Medicine, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Huajing Teng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ke Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430000, China
| | - Kai Mu
- The Second Hospital, Cheeloo College of Medicine, Shandong University, Shandong 250033, China
| | - Tianda Fan
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xiaoping Cen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
- College of Life Sciences, University of Chinese Academy of Science, Beijing 100049, China
| | - Zhe Xu
- College of Life Sciences, University of Chinese Academy of Science, Beijing 100049, China
| | - Ming Zhu
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing 100084, China
| | - Wenting Wang
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Jia Mi
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong 264003, China
| | - Xi Xiang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| | - Wei Dong
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Huanming Yang
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Lars Bolund
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Lin Lin
- Department of Biomedicine, Aarhus University, 8200 Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Jinzhao Song
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Yonglun Luo
- Department of Biomedicine, Aarhus University, 8200 Aarhus, Denmark
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Chunhua Lin
- Department of Urology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, China
| | - Peng Han
- Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518107, Guangdong, China
| |
Collapse
|
35
|
Rekhtman N, Tischfield SE, Febres-Aldana CA, Lee JJK, Chang JC, Herzberg BO, Selenica P, Woo HJ, Vanderbilt CM, Yang SR, Xu F, Bowman AS, da Silva EM, Noronha AM, Mandelker DL, Mehine M, Mukherjee S, Blanco-Heredia J, Orgera JJ, Nanjangud GJ, Baine MK, Aly RG, Sauter JL, Travis WD, Savari O, Moreira AL, Falcon CJ, Bodd FM, Wilson CE, Sienty JV, Manoj P, Sridhar H, Wang L, Choudhury NJ, Offin M, Yu HA, Quintanal-Villalonga A, Berger MF, Ladanyi M, Donoghue MT, Reis-Filho JS, Rudin CM. Chromothripsis-Mediated Small Cell Lung Carcinoma. Cancer Discov 2025; 15:83-104. [PMID: 39185963 PMCID: PMC11726019 DOI: 10.1158/2159-8290.cd-24-0286] [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: 03/01/2024] [Revised: 06/13/2024] [Accepted: 08/23/2024] [Indexed: 08/27/2024]
Abstract
Small cell lung carcinoma (SCLC) is a highly aggressive malignancy that is typically associated with tobacco exposure and inactivation of RB1 and TP53 genes. Here, we performed detailed clinicopathologic, genomic, and transcriptomic profiling of an atypical subset of SCLC that lacked RB1 and TP53 co-inactivation and arose in never/light smokers. We found that most cases were associated with chromothripsis-massive, localized chromosome shattering-recurrently involving chromosome 11 or 12 and resulting in extrachromosomal amplification of CCND1 or co-amplification of CCND2/CDK4/MDM2, respectively. Uniquely, these clinically aggressive tumors exhibited genomic and pathologic links to pulmonary carcinoids, suggesting a previously uncharacterized mode of SCLC pathogenesis via transformation from lower-grade neuroendocrine tumors or their progenitors. Conversely, SCLC in never-smokers harboring inactivated RB1 and TP53 exhibited hallmarks of adenocarcinoma-to-SCLC derivation, supporting two distinct pathways of plasticity-mediated pathogenesis of SCLC in never-smokers. Significance: Here, we provide the first detailed description of a unique SCLC subset lacking RB1/TP53 alterations and identify extensive chromothripsis and pathogenetic links to pulmonary carcinoids as its hallmark features. This work defines atypical SCLC as a novel entity among lung cancers, highlighting its exceptional histogenesis, clinicopathologic characteristics, and therapeutic vulnerabilities. See related commentary by Nadeem and Drapkin, p. 8.
Collapse
Affiliation(s)
- Natasha Rekhtman
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sam E. Tischfield
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christopher A. Febres-Aldana
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jake June-Koo Lee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jason C. Chang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Benjamin O. Herzberg
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center and the Herbert Irving Comprehensive Cancer Center, New York, New York
| | - Pier Selenica
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Hyung Jun Woo
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Chad M. Vanderbilt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Soo-Ryum Yang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fei Xu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anita S. Bowman
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Edaise M. da Silva
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anne Marie Noronha
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Diana L. Mandelker
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Miika Mehine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Semanti Mukherjee
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Juan Blanco-Heredia
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John J. Orgera
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gouri J. Nanjangud
- Department of Molecular Cytogenetics Core Facility, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marina K. Baine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rania G. Aly
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jennifer L. Sauter
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William D. Travis
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Omid Savari
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, University Hospitals Cleveland Medical Center- Case Western Reserve University, Cleveland, Ohio
| | - Andre L. Moreira
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, New York University Grossman School of Medicine, New York, New York
| | - Christina J. Falcon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Francis M. Bodd
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christina E. Wilson
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Druckenmiller Center for Lung Cancer Research, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jacklynn V. Sienty
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Division of Biostatistics Research Scientists, New York University, New York, New York
| | - Parvathy Manoj
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Harsha Sridhar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lu Wang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Noura J. Choudhury
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Michael Offin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Helena A. Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | | | - Michael F. Berger
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mark T.A. Donoghue
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jorge S. Reis-Filho
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charles M. Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
36
|
Schwalbe EC, Lindsey JC, Danilenko M, Hill RM, Crosier S, Ryan SL, Williamson D, Castle J, Hicks D, Kool M, Milde T, Korshunov A, Pfister SM, Bailey S, Clifford SC. Molecular and clinical heterogeneity within MYC-family amplified medulloblastoma is associated with survival outcomes: A multicenter cohort study. Neuro Oncol 2025; 27:222-236. [PMID: 39377358 PMCID: PMC11726341 DOI: 10.1093/neuonc/noae178] [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] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND MYC/MYCN are the most frequent oncogene amplifications in medulloblastoma (MB) and its primary biomarkers of high-risk (HR) disease. However, while many patients' MYC(N)-amplified tumors are treatment-refractory, some achieve long-term survival. We therefore investigated clinicobiological heterogeneity within MYC(N)-amplified MB and determined its relevance for improved disease management. METHODS We characterized the clinical and molecular correlates of MYC- (MYC-MB; n = 64) and MYCN-amplified MBs (MYCN-MB; n = 95), drawn from >1600 diagnostic cases. RESULTS Most MYC-MBs were molecular group 3 (46/58; 79% assessable) and aged ≥3 years at diagnosis (44/64 [69%]). We identified a "canonical" very high-risk (VHR) MYC-amplified group (n = 51/62; 82%) with dismal survival irrespective of treatment (11% 5-year progression-free survival [PFS]), defined by co-occurrence with ≥1 additional established risk factor(s) (subtotal surgical-resection [STR], metastatic disease, LCA pathology), and commonly group 3/4 subgroup 2 with a high proportion of amplified cells. The majority of remaining noncanonical MYC-MBs survived (i.e. non-group 3/group 3 without other risk features; 11/62 (18%); 61% 5-year PFS). MYCN survival was primarily related to molecular group; MYCN-amplified SHH MB, and group 3/4 MB with additional risk factors, respectively defined VHR and HR groups (VHR, 39% [35/89]; 20% 5-year PFS/HR, 33% [29/89]; 46% 5-year PFS). Twenty-two out of 35 assessable MYCN-amplified SHH tumors harbored TP53 mutations; 9/12 (75%) with data were germline. MYCN-amplified group 3/4 MB with no other risk factors (28%; 25/89) had 70% 5-year PFS. CONCLUSIONS MYC(N)-amplified MB displays significant clinicobiological heterogeneity. Diagnostics incorporating molecular groups, subgroups, and clinical factors enable their risk assessment. VHR "canonical" MYC tumors are essentially incurable and SHH-MYCN-amplified MBs fare extremely poorly (20% survival at 5 years); both require urgent development of alternative treatment strategies. Conventional risk-adapted therapies are appropriate for more responsive groups, such as noncanonical MYC and non-SHH-MYCN MB.
Collapse
Affiliation(s)
- Edward C Schwalbe
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne, UK
| | - Janet C Lindsey
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Marina Danilenko
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Rebecca M Hill
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Stephen Crosier
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Sarra L Ryan
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Daniel Williamson
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Jemma Castle
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Debbie Hicks
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Marcel Kool
- Hopp Children´s Cancer Center (KiTZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Till Milde
- Hopp Children´s Cancer Center (KiTZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Andrey Korshunov
- Hopp Children´s Cancer Center (KiTZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Stefan M Pfister
- Hopp Children´s Cancer Center (KiTZ), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, Heidelberg University Hospital, Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Simon Bailey
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| | - Steven C Clifford
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Newcastle upon Tyne, UK
| |
Collapse
|
37
|
Lee C, Oh JS. Novel BRCA1-PLK1-CIP2A axis orchestrates homologous recombination-mediated DNA repair to maintain chromosome integrity during oocyte meiosis. Nucleic Acids Res 2025; 53:gkae1207. [PMID: 39657788 PMCID: PMC11754672 DOI: 10.1093/nar/gkae1207] [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: 06/12/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024] Open
Abstract
Double-strand breaks (DSBs) are a formidable threat to genome integrity, potentially leading to cancer and various genetic diseases. The prolonged lifespan of mammalian oocytes increases their susceptibility to DNA damage over time. While somatic cells suppress DSB repair during mitosis, oocytes exhibit a remarkable capacity to repair DSBs during meiotic maturation. However, the precise mechanisms underlying DSB repair in oocytes remain poorly understood. Here, we describe the pivotal role of the BRCA1-PLK1-CIP2A axis in safeguarding genomic integrity during meiotic maturation in oocytes. We found that inhibition of homologous recombination (HR) severely impaired chromosome integrity by generating chromosome fragments during meiotic maturation. Notably, HR inhibition impaired the recruitment of CIP2A to damaged chromosomes, and the depletion of CIP2A led to chromosome fragmentation following DSB induction. Moreover, BRCA1 depletion impaired chromosomal recruitment of CIP2A, but not vice versa. Importantly, the impaired chromosomal recruitment of CIP2A could be rescued by PLK1 inhibition. Consequently, our findings not only underscore the importance of the chromosomal recruitment of CIP2A in preventing chromosome fragmentation, but also demonstrate the regulatory role of the BRCA1-PLK1-CIP2A axis in this process during oocyte meiotic maturation.
Collapse
Affiliation(s)
- Crystal Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, South Korea
| | - Jeong Su Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, South Korea
| |
Collapse
|
38
|
Liu Y, Zhu F, Li X, Guan X, Hou Y, Feng Y, Dong X, Li Y. SpatialSNV: A novel method for identifying and analyzing spatially resolved SNVs in tumor microenvironments. Gigascience 2025; 14:giaf065. [PMID: 40515555 PMCID: PMC12166308 DOI: 10.1093/gigascience/giaf065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 04/14/2025] [Accepted: 05/18/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND The dynamics of single-nucleotide variants (SNVs) play a critical role in understanding tumor development, yet their influence on shaping tumor microenvironments remains largely unexplored. Spatial transcriptomics offers an opportunity to map SNVs within the tumor context, potentially uncovering new insights into tumor microenvironment dynamics. RESULTS This study developed SpatialSNV for identifying effective SNVs across tumor sections using multiple spatial transcriptomics platforms. The analysis revealed that SNVs reflect regional tumor evolutionary traces and extend beyond RNA expression changes. The tumor margins exhibited a distinct mutational profile, with novel SNVs diminishing in a distance-dependent manner from the tumor boundary. These mutations were significantly linked to inflammatory and hypoxic microenvironments. Furthermore, spatially correlated SNV groups were identified, exhibiting distinct spatial patterns and implicating specific roles in tumor-immune system crosstalk. Among these, critical SNVs such as S100A11L40P in colorectal cancer were identified as tumor region-specific mutations. This mutation, located within exonic nonsynonymous regions, may produce neoantigens presented by HLAs, marking it as a potential therapeutic target. CONCLUSIONS SpatialSNV represents a promising framework for unraveling the mechanisms underlying tumor-immune crosstalk within the tumor microenvironment by leveraging spatial transcriptomics and SNV-based tissue domain characterization. This approach is designed to be scalable, integrative, and adaptable, making it accessible to researchers aiming to explore tumor heterogeneity and identify therapeutic targets.
Collapse
Affiliation(s)
- Yi Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou, Zhejiang 310030, China
| | - Fan Zhu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou, Zhejiang 310030, China
| | - Xinxing Li
- BGI Research, Hangzhou, Zhejiang 310030, China
| | - Xiangyu Guan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Hangzhou, Zhejiang 310030, China
| | - Yong Hou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
- BGI Research, Shenzhen, Guangdong 518083, China
| | - Yu Feng
- BGI Research, Hangzhou, Zhejiang 310030, China
| | - Xuan Dong
- BGI Research, Hangzhou, Zhejiang 310030, China
| | - Young Li
- BGI Research, Hangzhou, Zhejiang 310030, China
- Key Laboratory of Spatial Omics of Zhejiang Province, Hangzhou 310030, China
| |
Collapse
|
39
|
Chen X, Agustinus AS, Li J, DiBona M, Bakhoum SF. Chromosomal instability as a driver of cancer progression. Nat Rev Genet 2025; 26:31-46. [PMID: 39075192 DOI: 10.1038/s41576-024-00761-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/31/2024]
Abstract
Chromosomal instability (CIN) refers to an increased propensity of cells to acquire structural and numerical chromosomal abnormalities during cell division, which contributes to tumour genetic heterogeneity. CIN has long been recognized as a hallmark of cancer, and evidence over the past decade has strongly linked CIN to tumour evolution, metastasis, immune evasion and treatment resistance. Until recently, the mechanisms by which CIN propels cancer progression have remained elusive. Beyond the generation of genomic copy number heterogeneity, recent work has unveiled additional tumour-promoting consequences of abnormal chromosome segregation. These mechanisms include complex chromosomal rearrangements, epigenetic reprogramming and the induction of cancer cell-intrinsic inflammation, emphasizing the multifaceted role of CIN in cancer.
Collapse
Affiliation(s)
- Xuelan Chen
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Albert S Agustinus
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Pharmacology Graduate Program, Weill Cornell Medicine, New York, NY, USA
| | - Jun Li
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Melody DiBona
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samuel F Bakhoum
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
40
|
Jakobsen MZ, Brøndum RF, Gregersen H, Due H, Dybkær K. A systematic literature review on clonal evolution events preceding relapse in multiple myeloma. Crit Rev Oncol Hematol 2025; 205:104560. [PMID: 39549892 DOI: 10.1016/j.critrevonc.2024.104560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024] Open
Abstract
Despite considerable treatment advances, multiple myeloma (MM) remains an incurable hematological cancer due to treatment resistance. A systematic literature search was conducted to identify determinants for clonal evolution driving relapse and drug resistance in MM. A total of 631 non-duplicate publications were screened of which 28 articles were included for data extraction. Genetic alterations, mutational signatures, evolutionary trajectories, and non-genetic determinants were identified as key topics to characterize clonal evolution in relapsed MM. A variety of factors led to clonal diversification and increased tumor mutation burden, such as MAPK-Ras mutations and incremental changes related to chromosomal bands 1 and 17, while mutational signature analyses revealed that APOBEC activity and melphalan treatment leave a distinct impact on the clonal composition in MM genomes. To capture and dissect tumor heterogeneity, our review suggests combining methods or using technical approaches with high resolution to assess the impact of clonal evolution.
Collapse
Affiliation(s)
- Maja Zimmer Jakobsen
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Rasmus Froberg Brøndum
- Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark; Center for Clinical Data Science, Aalborg University, and Aalborg University Hospital, Aalborg, Denmark
| | - Henrik Gregersen
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Hanne Due
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark
| | - Karen Dybkær
- Department of Hematology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark; Clinical Cancer Research Center, Aalborg University Hospital, Aalborg, Denmark.
| |
Collapse
|
41
|
Kadali VN, Shoshani O. Aberrant nuclei with amplified DNA in cancer. Trends Cancer 2025; 11:9-11. [PMID: 39370335 DOI: 10.1016/j.trecan.2024.09.005] [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: 07/31/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 10/08/2024]
Abstract
Gene amplification in the form of extrachromosomal DNA (ecDNA) or intrachromosomal homogenous staining regions (HSRs) is an emerging hallmark in cancer. Recent studies implicate abnormal nuclear structures in the biogenesis and evolution of amplified DNA. Here, we discuss how the interplay between aberrant nuclei and gene amplification drives cancer therapy resistance and metastasis.
Collapse
Affiliation(s)
| | - Ofer Shoshani
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
42
|
Yang QL, Xie Y, Qiao K, Lim JYS, Wu S. Modern biology of extrachromosomal DNA: A decade-long voyage of discovery. Cell Res 2025; 35:11-22. [PMID: 39748050 PMCID: PMC11701097 DOI: 10.1038/s41422-024-01054-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/07/2024] [Indexed: 01/04/2025] Open
Abstract
Genomic instability is a hallmark of cancer and is a major driving force of tumorigenesis. A key manifestation of genomic instability is the formation of extrachromosomal DNAs (ecDNAs) - acentric, circular DNA molecules ranging from 50 kb to 5 Mb in size, distinct from chromosomes. Ontological studies have revealed that ecDNA serves as a carrier of oncogenes, immunoregulatory genes, and enhancers, capable of driving elevated transcription of its cargo genes and cancer heterogeneity, leading to rapid tumor evolution and therapy resistance. Although ecDNA was documented over half a century ago, the past decade has witnessed a surge in breakthrough discoveries about its biological functions. Here, we systematically review the modern biology of ecDNA uncovered over the last ten years, focusing on how discoveries during this pioneering stage have illuminated our understanding of ecDNA-driven transcription, heterogeneity, and cancer progression. Furthermore, we discuss ongoing efforts to target ecDNA as a novel approach to cancer therapy. This burgeoning field is entering a new phase, poised to reshape our knowledge of cancer biology and therapeutic strategies.
Collapse
Affiliation(s)
- Qing-Lin Yang
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yipeng Xie
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kailiang Qiao
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jun Yi Stanley Lim
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sihan Wu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
43
|
Koltsova AS, Pendina AA, Malysheva OV, Trusova ED, Staroverov DA, Yarmolinskaya MI, Polenov NI, Glotov AS, Kogan IY, Efimova OA. In Vitro Effect of Estrogen and Progesterone on Cytogenetic Profile of Uterine Leiomyomas. Int J Mol Sci 2024; 26:96. [PMID: 39795954 PMCID: PMC11720186 DOI: 10.3390/ijms26010096] [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: 10/30/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/13/2025] Open
Abstract
In the present study, we aimed to investigate intratumoral karyotype diversity as well as the estrogen/progesterone effect on the cytogenetic profile of uterine leiomyomas (ULs). A total of 15 UL samples obtained from 15 patients were cultured in the media supplemented with estrogen and/or progesterone and without adding hormones. Conventional cytogenetic analysis of culture samples revealed clonal chromosomal abnormalities in 11 out of 15 ULs. Cytogenetic findings were presented by simple and complex chromosomal rearrangements (64% and 36% of cases, respectively) verified through FISH and aCGH. In most ULs with complex chromosomal rearrangements, the breakpoints did not feature clusterization on a single chromosome but were evenly distributed across rearranged chromosomes. The number of breakpoints showed a strong positive correlation with the number of rearranged chromosomes. Moreover, both abovementioned parameters were in a linear dependency from the number of karyotypically different clones per UL. This suggests that complex chromosomal rearrangements in ULs predominantly originate through sequential events rather than one hit. The results of UL cytogenetic analysis depended on the presence of estrogen and/or progesterone in the culture medium. The greatest variety of cytogenetically different cell clones was detected in the samples cultured without hormone supplementation. Their counterparts cultured with progesterone supplementation showed a sharp decrease in clone number, whereas such a decrease induced by estrogen or estrogen-progesterone supplementation was insignificant. These findings suggest that estrogen-progesterone balance is crucial for forming a UL cytogenetic profile, which, in turn, may underlie the unique response of the every karyotypically abnormal UL to medications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Olga A. Efimova
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductology, 199034 St. Petersburg, Russia; (A.S.K.)
| |
Collapse
|
44
|
Wang Z, Yu J, Zhu W, Hong X, Xu Z, Mao S, Huang L, Han P, He C, Song C, Xiang X. Unveiling the mysteries of extrachromosomal circular DNA: from generation to clinical relevance in human cancers and health. Mol Cancer 2024; 23:276. [PMID: 39707444 DOI: 10.1186/s12943-024-02187-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/26/2024] [Indexed: 12/23/2024] Open
Abstract
Extrachromosomal circular DNAs (eccDNAs) are a type of circular DNAs originating from but independent of chromosomal DNAs. Nowadays, with the rapid development of sequencing and bioinformatics, the accuracy of eccDNAs detection has significantly improved. This advancement has consequently enhanced the feasibility of exploring the biological characteristics and functions of eccDNAs. This review elucidates the potential mechanisms of eccDNA generation, the existing methods for their detection and analysis, and their basic features. Furthermore, it focuses on the biological functions of eccDNAs in regulating gene expression under both physiological and pathological conditions. Additionally, the review summarizes the clinical implications of eccDNAs in human cancers and health.
Collapse
Affiliation(s)
- Zilong Wang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- Department of Andrology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Jiaying Yu
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Wenli Zhu
- School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Xiaoning Hong
- Clinical Big Data Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Zhen Xu
- Department of Andrology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Shuang Mao
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Lei Huang
- School of Medicine, Sun Yat-Sen University, Shenzhen, 518107, China
| | - Peng Han
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China
- Department of Biology, University of Copenhagen, Copenhagen, 2200, Denmark
| | - Chunxiao He
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Changze Song
- Department of Andrology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| | - Xi Xiang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|
45
|
Hashimoto A, Hashimoto S. Plasticity and Tumor Microenvironment in Pancreatic Cancer: Genetic, Metabolic, and Immune Perspectives. Cancers (Basel) 2024; 16:4094. [PMID: 39682280 DOI: 10.3390/cancers16234094] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer has long been believed to be a genetic disease caused by the accumulation of mutations in key genes involved in cellular processes. However, recent advances in sequencing technology have demonstrated that cells with cancer driver mutations are also present in normal tissues in response to aging, environmental damage, and chronic inflammation, suggesting that not only intrinsic factors within cancer cells, but also environmental alterations are important key factors in cancer development and progression. Pancreatic cancer tissue is mostly comprised of stromal cells and immune cells. The desmoplasmic microenvironment characteristic of pancreatic cancer is hypoxic and hypotrophic. Pancreatic cancer cells may adapt to this environment by rewiring their metabolism through epigenomic changes, enhancing intrinsic plasticity, creating an acidic and immunosuppressive tumor microenvironment, and inducing noncancerous cells to become tumor-promoting. In addition, pancreatic cancer has often metastasized to local and distant sites by the time of diagnosis, suggesting that a similar mechanism is operating from the precancerous stage. Here, we review key recent findings on how pancreatic cancers acquire plasticity, undergo metabolic reprogramming, and promote immunosuppressive microenvironment formation during their evolution. Furthermore, we present the following two signaling pathways that we have identified: one based on the small G-protein ARF6 driven by KRAS/TP53 mutations, and the other based on the RNA-binding protein Arid5a mediated by inflammatory cytokines, which promote both metabolic reprogramming and immune evasion in pancreatic cancer. Finally, the striking diversity among pancreatic cancers in the relative importance of mutational burden and the tumor microenvironment, their clinical relevance, and the potential for novel therapeutic strategies will be discussed.
Collapse
Affiliation(s)
- Ari Hashimoto
- Department of Molecular Biology, Graduate School of Medicine, Hokkaido University, Sapporo 060-8638, Japan
| | - Shigeru Hashimoto
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0818, Japan
| |
Collapse
|
46
|
Minowa T, Murata K, Mizue Y, Murai A, Nakatsugawa M, Sasaki K, Tokita S, Kubo T, Kanaseki T, Tsukahara T, Handa T, Sato S, Horimoto K, Kato J, Hida T, Hirohashi Y, Uhara H, Torigoe T. Single-cell profiling of acral melanoma infiltrating lymphocytes reveals a suppressive tumor microenvironment. Sci Transl Med 2024; 16:eadk8832. [PMID: 39630887 DOI: 10.1126/scitranslmed.adk8832] [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: 09/16/2023] [Revised: 06/12/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Acral lentiginous melanoma (ALM) is the most common melanoma subtype in non-Caucasians. Despite advances in cancer immunotherapy, current immune checkpoint inhibitors remain unsatisfactory for ALM. Hence, we conducted comprehensive immune profiling using single-cell phenotyping with reactivity screening of the T cell receptors of tumor-infiltrating T lymphocytes (TILs) in ALM. Compared with cutaneous melanoma, ALM showed a lower frequency of tumor-reactive CD8 clusters and an enrichment of regulatory T cells with direct tumor recognition ability, suggesting a suppressive immune microenvironment in ALM. Tumor-reactive CD8 TILs showed heterogeneous expression of coinhibitory molecules, including KLRC1 (NKG2A), in subpopulations with therapeutic implications. Overall, our study provides a foundation for enhancing the efficacy of immunotherapy in ALM.
Collapse
Affiliation(s)
- Tomoyuki Minowa
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Kenji Murata
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Yuka Mizue
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Aiko Murai
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Munehide Nakatsugawa
- Department of Pathology, Tokyo Medical University Hachioji Medical Center, 193-0998 Hachioji, Tokyo, Japan
| | - Kenta Sasaki
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Serina Tokita
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Terufumi Kubo
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Takayuki Kanaseki
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
- Joint Research Center for Immunoproteogenomics, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Tomohide Tsukahara
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Toshiya Handa
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Sayuri Sato
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Kohei Horimoto
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Junji Kato
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Tokimasa Hida
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Yoshihiko Hirohashi
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| | - Hisashi Uhara
- Department of Dermatology, Sapporo Medical University School of Medicine, 060-8543 Sapporo, Hokkaido, Japan
| | - Toshihiko Torigoe
- Department of Pathology, Sapporo Medical University School of Medicine, 060-8556 Sapporo, Hokkaido, Japan
| |
Collapse
|
47
|
Koeppel J, Weller J, Vanderstichele T, Parts L. Engineering structural variants to interrogate genome function. Nat Genet 2024; 56:2623-2635. [PMID: 39533047 DOI: 10.1038/s41588-024-01981-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Structural variation, such as deletions, duplications, inversions and complex rearrangements, can have profound effects on gene expression, genome stability, phenotypic diversity and disease susceptibility. Structural variants can encompass up to millions of bases and have the potential to rearrange substantial segments of the genome. They contribute considerably more to genetic diversity in human populations and have larger effects on phenotypic traits than point mutations. Until recently, our understanding of the effects of structural variants was driven mainly by studying naturally occurring variation. New genome-engineering tools capable of generating deletions, insertions, inversions and translocations, together with the discovery of new recombinases and advances in creating synthetic DNA constructs, now enable the design and generation of an extended range of structural variation. Here, we discuss these tools and examples of their application and highlight existing challenges that will need to be overcome to fully harness their potential.
Collapse
|
48
|
Leppä AM, Grimes K, Jeong H, Huang FY, Andrades A, Waclawiczek A, Boch T, Jauch A, Renders S, Stelmach P, Müller-Tidow C, Karpova D, Sohn M, Grünschläger F, Hasenfeld P, Benito Garagorri E, Thiel V, Dolnik A, Rodriguez-Martin B, Bullinger L, Mrózek K, Eisfeld AK, Krämer A, Sanders AD, Korbel JO, Trumpp A. Single-cell multiomics analysis reveals dynamic clonal evolution and targetable phenotypes in acute myeloid leukemia with complex karyotype. Nat Genet 2024; 56:2790-2803. [PMID: 39587361 PMCID: PMC11631769 DOI: 10.1038/s41588-024-01999-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/15/2024] [Indexed: 11/27/2024]
Abstract
Chromosomal instability is a major driver of intratumoral heterogeneity (ITH), promoting tumor progression. In the present study, we combined structural variant discovery and nucleosome occupancy profiling with transcriptomic and immunophenotypic changes in single cells to study ITH in complex karyotype acute myeloid leukemia (CK-AML). We observed complex structural variant landscapes within individual cells of patients with CK-AML characterized by linear and circular breakage-fusion-bridge cycles and chromothripsis. We identified three clonal evolution patterns in diagnosis or salvage CK-AML (monoclonal, linear and branched polyclonal), with 75% harboring multiple subclones that frequently displayed ongoing karyotype remodeling. Using patient-derived xenografts, we demonstrated varied clonal evolution of leukemic stem cells (LSCs) and further dissected subclone-specific drug-response profiles to identify LSC-targeting therapies, including BCL-xL inhibition. In paired longitudinal patient samples, we further revealed genetic evolution and cell-type plasticity as mechanisms of disease progression. By dissecting dynamic genomic, phenotypic and functional complexity of CK-AML, our findings offer clinically relevant avenues for characterizing and targeting disease-driving LSCs.
Collapse
Affiliation(s)
- Aino-Maija Leppä
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Karen Grimes
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Hyobin Jeong
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Republic of Korea
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Frank Y Huang
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Alvaro Andrades
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | - Alexander Waclawiczek
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Tobias Boch
- University Hospital Mannheim, Heidelberg University, Mannheim, Germany
| | - Anna Jauch
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Simon Renders
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Patrick Stelmach
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Darja Karpova
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Markus Sohn
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
| | - Florian Grünschläger
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Patrick Hasenfeld
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| | | | - Vera Thiel
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Anna Dolnik
- Charité Medical Department, Division of Hematology, Oncology and Tumor Immunology, Berlin, Germany
| | | | - Lars Bullinger
- Charité Medical Department, Division of Hematology, Oncology and Tumor Immunology, Berlin, Germany
| | - Krzysztof Mrózek
- Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Clara D. Bloomfield Center for Leukemia Outcomes Research, Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Ann-Kathrin Eisfeld
- Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
- Clara D. Bloomfield Center for Leukemia Outcomes Research, Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Alwin Krämer
- Clinical Cooperation Unit Molecular Hematology/Oncology, German Cancer Research Center (DKFZ) and Department of Internal Medicine V, University of Heidelberg, Heidelberg, Germany
| | - Ashley D Sanders
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
- Charité-Universitätsmedizin, Berlin, Germany
| | - Jan O Korbel
- European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany.
- Bridging Research Division on Mechanisms of Genomic Variation and Data Science, German Cancer Research Center, Heidelberg, Germany.
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
49
|
Frankenbach-Désor T, Niesner I, Ahmed P, Dürr HR, Klein A, Knösel T, Gospos J, McGovern JA, Hutmacher DW, Holzapfel BM, Mayer-Wagner S. Tissue-engineered patient-derived osteosarcoma models dissecting tumour-bone interactions. Cancer Metastasis Rev 2024; 44:8. [PMID: 39592467 PMCID: PMC11599440 DOI: 10.1007/s10555-024-10218-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024]
Abstract
Osteosarcoma is the most common malignant bone tumor, primarily affecting children and young adults. For these young patients, the current treatment options for osteosarcoma impose considerable constraints on daily life with significant morbidity and a low survival rate. Despite ongoing research efforts, the 5-year survival rate of first-diagnosed patients without metastases has not changed in the past four decades. The demand for novel treatments is currently still unmet, in particular for effective second-line therapy. Therefore, there is an urgent need for advanced preclinical models and drug-testing platforms that take into account the complex disease characteristics, the high heterogeneity of the tumour and the interactions with the bone microenvironment. In this review, we provide a comprehensive overview about state-of-the-art tissue-engineered and patient-specific models for osteosarcoma. These sophisticated platforms for advanced therapy trials aim to improve treatment outcomes for future patients by modelling the patient's disease state in a more accurate and complex way, thus improving the quality of preclinical research studies.
Collapse
Affiliation(s)
- Tina Frankenbach-Désor
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
| | - Isabella Niesner
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Parveen Ahmed
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Hans Roland Dürr
- Department of Orthopaedics and Trauma Surgery, Orthopaedic Oncology, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Alexander Klein
- Department of Orthopaedics and Trauma Surgery, Orthopaedic Oncology, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Thomas Knösel
- Institute of Pathology, Ludwig-Maximilians-Universität (LMU) Munich, Thalkirchner Str. 36, 80337, Munich, Germany
| | - Jonathan Gospos
- Centre for Biomedical Technologies, School of Medical, Mechanical and Process Engineering, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD, 4059, Australia
- Max Planck Queensland Center for the Materials Science of Extracellular Matrices, Queensland University of Technology (QUT), 2 George Street, Brisbane, QLD, 4000, Australia
| | - Jacqui A McGovern
- Centre for Biomedical Technologies, School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, 37 Kent Street, Woolloongabba, QLD, 4102, Australia
- Max Planck Queensland Center for the Materials Science of Extracellular Matrices, Queensland University of Technology (QUT), 2 George Street, Brisbane, QLD, 4000, Australia
| | - Dietmar W Hutmacher
- Centre for Biomedical Technologies, School of Medical, Mechanical and Process Engineering, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, QLD, 4059, Australia
- Max Planck Queensland Center for the Materials Science of Extracellular Matrices, Queensland University of Technology (QUT), 2 George Street, Brisbane, QLD, 4000, Australia
| | - Boris M Holzapfel
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Susanne Mayer-Wagner
- Department of Orthopaedics and Trauma Surgery, Musculoskeletal University Center Munich (MUM), LMU University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| |
Collapse
|
50
|
Pflughaupt P, Abdullah A, Masuda K, Sahakyan A. Towards the genomic sequence code of DNA fragility for machine learning. Nucleic Acids Res 2024; 52:12798-12816. [PMID: 39441076 PMCID: PMC11602142 DOI: 10.1093/nar/gkae914] [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: 03/18/2024] [Revised: 09/20/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024] Open
Abstract
Genomic DNA breakages and the subsequent insertion and deletion mutations are important contributors to genome instability and linked diseases. Unlike the research in point mutations, the relationship between DNA sequence context and the propensity for strand breaks remains elusive. Here, by analyzing the differences and commonalities across myriads of genomic breakage datasets, we extract the sequence-linked rules and patterns behind DNA fragility. We show the overall deconvolution of the sequence influence into short-, mid- and long-range effects, and the stressor-dependent differences in defining the range and compositional effects on DNA fragility. We summarize and release our feature compendium as a library that can be seamlessly incorporated into genomic machine learning procedures, where DNA fragility is of concern, and train a generalized DNA fragility model on cancer-associated breakages. Structural variants (SVs) tend to stabilize regions in which they emerge, with the effect most pronounced for pathogenic SVs. In contrast, the effects of chromothripsis are seen across regions less prone to breakages. We find that viral integration may bring genome fragility, particularly for cancer-associated viruses. Overall, this work offers novel insights into the genomic sequence basis of DNA fragility and presents a powerful machine learning resource to further enhance our understanding of genome (in)stability and evolution.
Collapse
Affiliation(s)
- Patrick Pflughaupt
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Adib A Abdullah
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Kairi Masuda
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Aleksandr B Sahakyan
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| |
Collapse
|