1
|
Thulasinathan B, Suvilesh KN, Maram S, Grossmann E, Ghouri Y, Teixeiro EP, Chan J, Kaif JT, Rachagani S. The impact of gut microbial short-chain fatty acids on colorectal cancer development and prevention. Gut Microbes 2025; 17:2483780. [PMID: 40189834 PMCID: PMC11980463 DOI: 10.1080/19490976.2025.2483780] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/18/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer is a long-term illness that involves an imbalance in cellular and immune functions. It can be caused by a range of factors, including exposure to environmental carcinogens, poor diet, infections, and genetic alterations. Maintaining a healthy gut microbiome is crucial for overall health, and short-chain fatty acids (SCFAs) produced by gut microbiota play a vital role in this process. Recent research has established that alterations in the gut microbiome led to decreased production of SCFA's in lumen of the colon, which associated with changes in the intestinal epithelial barrier function, and immunity, are closely linked to colorectal cancer (CRC) development and its progression. SCFAs influence cancer progression by modifying epigenetic mechanisms such as DNA methylation, histone modifications, and non-coding RNA functions thereby affecting tumor initiation and metastasis. This suggests that restoring SCFA levels in colon through microbiota modulation could serve as an innovative strategy for CRC prevention and treatment. This review highlights the critical relationship between gut microbiota and CRC, emphasizing the potential of targeting SCFAs to enhance gut health and reduce CRC risk.
Collapse
Affiliation(s)
- Boobalan Thulasinathan
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
| | - Kanve N. Suvilesh
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
| | - Sumanas Maram
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
| | - Erik Grossmann
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Department of Medicine, Digestive Centre, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
| | - Yezaz Ghouri
- Department of Medicine, Digestive Centre, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
| | - Emma Pernas Teixeiro
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Joshua Chan
- Chemical and Biological Engineering, Colorado State University, Fort Collins, CO, USA
| | - Jussuf T. Kaif
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
- Siteman Cancer Centre, Washington University, St. Louis, MO, USA
| | - Satyanarayana Rachagani
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
- Siteman Cancer Centre, Washington University, St. Louis, MO, USA
| |
Collapse
|
2
|
Irshad H, Azhar MA, Qvortrup K. Thiazole modified covalent triazine framework as carcinogenic metabolites adsorbent: A DFT insight. J Mol Graph Model 2025; 137:109009. [PMID: 40081004 DOI: 10.1016/j.jmgm.2025.109009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 03/05/2025] [Accepted: 03/08/2025] [Indexed: 03/15/2025]
Abstract
The potential of a novel thiazole-modified covalent triazine framework (S-CTF) as surface for the adsorption and sensing of the carcinogenic metabolites acrylamide (AM), 2-amino-3,8-dimethylimidazo-[4,5-f]quinoxaline (MEIQX), 2-amino-1-methyl-6-phenylimidazole[4,5-f]pyridine (PhlP) and 3-amino-1,4-dimethyl-5H-pyrido[4,3-b]indole (Trp-P-1) is explored. The selectivity, sensitivity, and adsorption properties of the S-CTF surface are investigated through noncovalent interaction (NCI), quantum theory of atoms in molecules (QTAIM) and symmetry adapted perturbation theory (SAPT0) analyses. All the analytes were found to be physiosorbed on the surface of the sensor with the following strength of interaction: MEIQX@S-CTF = PhlP@S-CTF > Trp-P-1@S-CTF > AM@S-CTF. Evaluation of the electronic properties was done by natural bond orbital (NBO), electron density difference (EDD), frontier molecular orbital (FMO) and density of states (DOS) analyses. Through SAPT0 analysis, MEIQX@S-CTF has shown to have the highest ESAPT0 energy data (-24.58 kcal/mol) whereas FMO analysis reveals that the S-CTF surface shows the highest sensing power for Trp-P-1 among all analytes.
Collapse
Affiliation(s)
- Hasher Irshad
- Department of Chemistry, Technical University of Denmark, 2800, Kongens Lyngby, Denmark
| | | | - Katrine Qvortrup
- Department of Chemistry, Technical University of Denmark, 2800, Kongens Lyngby, Denmark.
| |
Collapse
|
3
|
Rao Z, Wu C, Liao Y, Ye C, Huang S, Zhao D. POCALI: Prediction and Insight on CAncer LncRNAs by Integrating Multi-Omics Data with Machine Learning. SMALL METHODS 2025:e2401987. [PMID: 40405764 DOI: 10.1002/smtd.202401987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 04/27/2025] [Indexed: 05/24/2025]
Abstract
Long non-coding RNAs (lncRNAs) are receiving increasing attention as biomarkers for cancer diagnosis and therapy. Although there are many computational methods to identify cancer lncRNAs, they do not comprehensively integrate multi-omics features for predictions or systematically evaluate the contribution of each omics to the multifaceted landscape of cancer lncRNAs. In this study, an algorithm, POCALI, is developed to identify cancer lncRNAs by integrating 44 omics features across six categories. The contributions of different omics are explored to identifying cancer lncRNAs and, more specifically, how each feature contributes to a single prediction. The model is evaluated and benchmarked POCALI with existing methods. Finally, the cancer phenotype and genomics characteristics of the predicted novel cancer lncRNAs are validated. POCALI identifies secondary structure and gene expression-related features as strong predictors of cancer lncRNAs, and epigenomic features as moderate predictors. POCALI performed better than other methods, especially in terms of sensitivity, and predicted more candidates. Novel POCALI-predicted cancer lncRNAs have strong relationships with cancer phenotypes, similar to known cancer lncRNAs. Overall, this study facilitates the identification of previously undetected cancer lncRNAs and the comprehensive exploration of the multifaceted feature contributions to cancer lncRNA prediction.
Collapse
Affiliation(s)
- Ziyan Rao
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Chenyang Wu
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Yunxi Liao
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Chuan Ye
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Shaodong Huang
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Dongyu Zhao
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| |
Collapse
|
4
|
Steffensen LB, Kavan S, Jensen PS, Pedersen MK, Bøttger SM, Larsen MJ, Dembic M, Bergman O, Matic L, Hedin U, Andersen LVB, Lindholt JS, Houlind KC, Riber LP, Thomassen M, Rasmussen LM. Mutational landscape of atherosclerotic plaques reveals large clonal cell populations. JCI Insight 2025; 10:e188281. [PMID: 40198128 DOI: 10.1172/jci.insight.188281] [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: 10/21/2024] [Accepted: 04/04/2025] [Indexed: 04/10/2025] Open
Abstract
The notion of clonal cell populations in human atherosclerosis has been suggested but not demonstrated. Somatic mutations are used to define cellular clones in tumors. Here, we characterized the mutational landscape of human carotid plaques through whole-exome sequencing to explore the presence of clonal cell populations. Somatic mutations were identified in 12 of 13 investigated plaques, while no mutations were detected in 11 non-atherosclerotic arteries. Mutated clones often constituted over 10% of the sample cell population, with genes related to the contractile apparatus enriched for mutations. In carriers of clonal hematopoiesis of indeterminate potential (CHIP), hematopoietic clones had infiltrated the plaque tissue and constituted substantial fractions of the plaque cell population alongside locally expanded clones. Our findings establish somatic mutations as a common feature of human atherosclerosis and demonstrate the existence of mutated clones expanding locally, as well as CHIP clones invading from the circulation. While our data do not support plaque monoclonality, we observed a pattern suggesting the coexistence of multiple mutated clones of considerable size spanning different regions of plaques. Mutated clones are likely to be relevant to disease development, and somatic mutations will serve as a convenient tool to uncover novel pathological processes of atherosclerosis in future studies.
Collapse
Affiliation(s)
- Lasse Bach Steffensen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
| | - Stephanie Kavan
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
- Department of Clinical Biochemistry and Pharmacology, and
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
| | - Pia Søndergaard Jensen
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
- Department of Clinical Biochemistry and Pharmacology, and
| | - Matilde Kvist Pedersen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
| | - Steffen Møller Bøttger
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
| | - Martin Jakob Larsen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
- Department of Clinical Research, and
| | - Maja Dembic
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
- Department of Clinical Research, and
- Department of Mathematics and Computer Science (IMADA), University of Southern Denmark, Odense, Denmark
| | - Otto Bergman
- Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Ljubica Matic
- Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institute and Karolinska University Hospital, Stockholm, Sweden
| | - Lars van Brakel Andersen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
| | - Jes Sanddal Lindholt
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
- Department of Cardiothoracic Surgery, Odense University Hospital, Odense, Denmark
| | | | - Lars Peter Riber
- Department of Cardiothoracic Surgery, Odense University Hospital, Odense, Denmark
| | - Mads Thomassen
- Department of Clinical Genetics, Odense University Hospital, Odense, Denmark
- Clinical Genome Center, Department of Clinical Research
| | - Lars Melholt Rasmussen
- Centre for Individualized Medicine in Arterial Diseases (CIMA)
- Department of Clinical Biochemistry and Pharmacology, and
| |
Collapse
|
5
|
Zhang T, Sang J, Hoang PH, Zhao W, Rosenbaum J, Johnson KE, Klimczak LJ, McElderry J, Klein A, Wirth C, Bergstrom EN, Díaz-Gay M, Vangara R, Colon-Matos F, Hutchinson A, Lawrence SM, Cole N, Zhu B, Przytycka TM, Shi J, Caporaso NE, Homer R, Pesatori AC, Consonni D, Imielinski M, Chanock SJ, Wedge DC, Gordenin DA, Alexandrov LB, Harris RS, Landi MT. APOBEC affects tumor evolution and age at onset of lung cancer in smokers. Nat Commun 2025; 16:4711. [PMID: 40394004 DOI: 10.1038/s41467-025-59923-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 05/02/2025] [Indexed: 05/22/2025] Open
Abstract
Most solid tumors harbor somatic mutations attributed to off-target activities of APOBEC3A (A3A) and/or APOBEC3B (A3B). However, how APOBEC3A/B enzymes affect tumor evolution in the presence of exogenous mutagenic processes is largely unknown. Here, multi-omics profiling of 309 lung cancers from smokers identifies two subtypes defined by low (LAS) and high (HAS) APOBEC mutagenesis. LAS are enriched for A3B-like mutagenesis and KRAS mutations; HAS for A3A-like mutagenesis and TP53 mutations. Compared to LAS, HAS have older age at onset and high proportions of newly generated progenitor-like cells likely due to the combined tobacco smoking- and APOBEC3A-associated DNA damage and apoptosis. Consistently, HAS exhibit high expression of pulmonary healing signaling pathway, stemness markers, distal cell-of-origin, more neoantigens, slower clonal expansion, but no smoking-associated genomic/epigenomic changes. With validation in 184 lung tumor samples, these findings show how heterogeneity in mutational burden across co-occurring mutational processes and cell types contributes to tumor development.
Collapse
Affiliation(s)
- Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jian Sang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Phuc H Hoang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Wei Zhao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | | | | | - Leszek J Klimczak
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - John McElderry
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Alyssa Klein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Christopher Wirth
- Manchester Cancer Research Centre, The University of Manchester, Manchester, UK
| | - Erik N Bergstrom
- Department of Cellular and Molecular Medicine and Department of Bioengineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Marcos Díaz-Gay
- Department of Cellular and Molecular Medicine and Department of Bioengineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Raviteja Vangara
- Department of Cellular and Molecular Medicine and Department of Bioengineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Frank Colon-Matos
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Amy Hutchinson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Scott M Lawrence
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Nathan Cole
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Teresa M Przytycka
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Neil E Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Robert Homer
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Angela C Pesatori
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Dario Consonni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - David C Wedge
- Manchester Cancer Research Centre, The University of Manchester, Manchester, UK
| | - Dmitry A Gordenin
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine and Department of Bioengineering and Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Reuben S Harris
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, USA
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
6
|
Sentís I, Melero JL, Cebria-Xart A, Grzelak M, Soto M, Michel A, Rovira Q, Rodriguez-Hernandez CJ, Caratù G, Urpi A, Sauvage C, Mendizabal-Sasieta A, Maspero D, Lavarino CE, Pascual-Reguant A, Castañeda Heredia A, Muñoz Perez JP, Mora J, Harari A, Nieto JC, Avgustinova A, Heyn H. Spatio-temporal T cell tracking for personalized TCR-T designs in childhood cancer. Ann Oncol 2025:S0923-7534(25)00733-1. [PMID: 40403847 DOI: 10.1016/j.annonc.2025.05.530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 04/21/2025] [Accepted: 05/08/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibition (ICI) has revolutionized oncology, offering extended survival and long-term remission in previously incurable cancers. While highly effective in tumors with high mutational burden, lowly mutated cancers, including pediatric malignancies, present low response rate and limited predictive biomarkers. PATIENTS AND METHODS We present a framework for the identification and validation of tumor-reactive T cells as a biomarker to quantify ICI efficacy and as candidates for a personalized TCR-T cell therapy. Therefore, we profiled a pediatric malignant rhabdoid tumor patient with complete remission after ICI therapy using deep single-cell T cell receptor (TCR) repertoire sequencing of the tumor microenvironment (TME) and the peripheral blood. RESULTS Tracking T cell dynamics longitudinally from the tumor to cells in circulation over a time course of 12 months revealed a systemic response and durable clonal expansion of tumor-resident and ICI-induced TCR clonotypes. We functionally validated tumor reactivity of TCRs identified from the TME and the blood by co-culturing patient-derived tumor cells with TCR-engineered autologous T cells. Here, we observed unexpectedly high frequencies of tumor-reactive TCR clonotypes in the TME and confirmed T cell dynamics in the blood post-ICI to predict tumor-reactivity. CONCLUSION These findings strongly support spatio-temporal tracking of T cell activity in response to ICI to inform therapy efficacy and to serve as a source of tumor-reactive TCRs for personalized TCR-T designs.
Collapse
Affiliation(s)
- I Sentís
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues de Llobregat, Spain
| | | | - A Cebria-Xart
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues de Llobregat, Spain
| | | | - M Soto
- Omniscope, Barcelona, Spain
| | - A Michel
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Agora Cancer Research Center, Lausanne, Switzerland; Center for Cell Therapy, CHUV-Ludwig Institute, Lausanne, Switzerland
| | - Q Rovira
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues de Llobregat, Spain
| | | | - G Caratù
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
| | - A Urpi
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues de Llobregat, Spain
| | - C Sauvage
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Agora Cancer Research Center, Lausanne, Switzerland; Center for Cell Therapy, CHUV-Ludwig Institute, Lausanne, Switzerland
| | | | - D Maspero
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
| | - C E Lavarino
- Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu, Barcelona, Spain
| | | | - A Castañeda Heredia
- Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu, Barcelona, Spain
| | - J P Muñoz Perez
- Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu, Barcelona, Spain
| | - J Mora
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues de Llobregat, Spain; Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu, Barcelona, Spain
| | - A Harari
- Ludwig Institute for Cancer Research, Lausanne Branch, Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Agora Cancer Research Center, Lausanne, Switzerland; Center for Cell Therapy, CHUV-Ludwig Institute, Lausanne, Switzerland
| | - J C Nieto
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
| | - A Avgustinova
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues de Llobregat, Spain.
| | - H Heyn
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain; Omniscope, Barcelona, Spain; Universitat de Barcelona (UB), Barcelona, Spain; ICREA, Barcelona, Spain.
| |
Collapse
|
7
|
Thakur R, Xu M, Sowards H, Yon J, Jessop L, Myers T, Zhang T, Chari R, Long E, Rehling T, Hennessey R, Funderburk K, Yin J, Machiela MJ, Johnson ME, Wells AD, Chesi A, Grant SFA, Iles MM, Landi MT, Law MH, Melanoma Meta-Analysis Consortium, Choi J, Brown KM. Mapping chromatin interactions at melanoma susceptibility loci uncovers distant cis-regulatory gene targets. Am J Hum Genet 2025:S0002-9297(25)00178-8. [PMID: 40409268 DOI: 10.1016/j.ajhg.2025.04.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/25/2025] Open
Abstract
Genome-wide association studies (GWASs) of melanoma risk have identified 68 independent signals at 54 loci. For most loci, specific functional variants and their respective target genes remain to be established. Capture-HiC is an assay that links fine-mapped risk variants to candidate target genes by comprehensively mapping chromatin interactions. We performed a melanoma GWAS region-focused capture-HiC assay in human primary melanocytes to identify physical interactions between fine-mapped risk variants and potential causal melanoma-susceptibility genes. Overall, chromatin-interaction data alone nominated potential causal genes for 61 of the 68 melanoma risk signals, identifying many candidates beyond those reported by previous studies. We further integrated these data with epigenomic (chromatin state, accessibility), gene expression (expression quantitative trait locus [eQTL]/transcriptome-wide association study [TWAS]), DNA methylation (methylation QTL [meQTL]/methylome-wide association study [MWAS]), and massively parallel reporter assay (MPRA) data generated from melanoma-relevant cell types to prioritize potentially cis-regulatory variants and their respective candidate gene targets. From the set of fine-mapped variants across these loci, we identified 140 prioritized credible causal variants linked to 195 candidate genes at 42 risk signals. In addition, we developed an integrative scoring system to facilitate candidate gene prioritization, integrating melanocyte and melanoma datasets. Notably, at several GWAS risk signals, we observed long-range chromatin connections (500 kb to >1 Mb) with distant candidate target genes. We validated several such cis-regulatory interactions using CRISPR inhibition, providing evidence for known cancer driver genes MDM4 and CBL, as well as the SRY-box transcription factor SOX4, as likely melanoma risk genes.
Collapse
Affiliation(s)
- Rohit Thakur
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mai Xu
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Hayley Sowards
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Joshuah Yon
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Lea Jessop
- Laboratory of Genetic Susceptibility, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Timothy Myers
- Laboratory of Genetic Susceptibility, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Tongwu Zhang
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Raj Chari
- Genome Modification Core, Frederick National Lab for Cancer Research, Frederick, MD, USA
| | - Erping Long
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA; Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Thomas Rehling
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Rebecca Hennessey
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Karen Funderburk
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jinhu Yin
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mitchell J Machiela
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Matthew E Johnson
- Division of Human Genetics, Children's Hospital of Philadelphia Research Institute, Philadelphia, PA, USA
| | - Andrew D Wells
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Mark M Iles
- Leeds Institute for Data Analytics, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Maria Teresa Landi
- Integrative Tumor Epidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Matthew H Law
- Population Health Department, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia; School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | | | - Jiyeon Choi
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kevin M Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
8
|
Wang H, Mei Q, Mei P. Comprehensive analysis of the role of Caspases in glioma. Brain Res 2025; 1855:149529. [PMID: 40032044 DOI: 10.1016/j.brainres.2025.149529] [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: 09/15/2024] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/05/2025]
Abstract
Caspases (CASPs) are attractive targets for cancer therapy. Many prognostic models based on gene signatures include genes from the CASPs family in diffuse glioma. CASP3, CASP4 and CASP6 in glioma have been studied individually. However, specialized comprehensive analysis of the roles of CASPs family in glioma is lacking. Therefore, this study utilized bioinformatics methods to investigate this issue. CASP1-10 expressionlevels were significantly up-regulated in LGG and GBM and glioma, and varied significantly across different clinical subgroups of glioma and LGG and various cell types, and most of CASP1-10 members showed significant differences in recurrence status of LGG. 10 signatures (CASP1-10) were associated with poor overall survival (OS) in glioma and LGG and GBM. However, pan-cancer survival analysis showed that CASP1-10 were associated with the prognosis of LGG, but not GBM. CASP1-10 were related to poor prognosis of glioma and LGG, except for CASP9, which was the opposite of a protective factor. CASP1-10 were independent prognostic factors for OS in glioma and LGG, except for CASP5, and also for recurrence-free survival (RFS) in LGG. Most of CASP1-10 were also independent prognostic factors for disease-specific survival (DSS) and progression-free interval (PFI) and had diagnostic value in glioma and LGG. Genetic alterations of CASP1-10 genes set were associated with poor prognosis in LGG. CASP1-10 were involved in immune infiltration and programmed cell death in glioma and LGG and GBM, and might promote the apoptosis of immune cells. Compared to GBM, CASP1-10 had a more significant impact on the prognosis, cancer-related pathways, and immune infiltration in LGG, indicating that CASP1-10 might play important roles in the recurrence and progression of LGG, and might be promising therapeutic targets for LGG. Therefore, it is speculated that natural caspase inhibitor p35 may be a promising drug for the treatment of glioma, especially for LGG.
Collapse
Affiliation(s)
- Heming Wang
- Hainan Key Laboratory for Sustainable Utilization of Tropical Bioresource, Hainan University, Haikou 570228, China
| | - Qunfang Mei
- Fujian Provincial Key Laboratory of Plant Functional Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Pengying Mei
- Fujian Provincial Key Laboratory of Plant Functional Biology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
9
|
Garcia-Salinas OI, Hwang S, Huang QQ, Sanghvi R, Malawsky DS, Kaplanis J, Neville MDC, Day FR, Rahbari R, Scally A, Martin HC. The impact of ancestral, genetic, and environmental influences on germline de novo mutation rates and spectra. Nat Commun 2025; 16:4527. [PMID: 40374658 DOI: 10.1038/s41467-025-59750-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 05/02/2025] [Indexed: 05/17/2025] Open
Abstract
De novo germline mutation is an important factor in the evolution of allelic diversity and disease predisposition in a population. Here, we study the influence of genetically-inferred ancestry and environmental factors on de novo mutation rates and spectra. Using a genetically diverse sample of ~10 K whole-genome sequenced trios, one of the largest de novo mutation catalogues to date, we found that genetically-inferred ancestry is associated with modest but significant changes in both germline mutation rate and spectra across continental populations. These effects may be due to genetic or environmental factors correlated with ancestry. We find epidemiological evidence that cigarette smoking is significantly associated with increased de novo mutation rate, but it does not mediate the observed ancestry effects. Investigation of several other potential mutagenic factors using Mendelian randomisation showed no consistent effects, except for age at menopause, where factors increasing this corresponded to a reduction in de novo mutation rate. Overall, our study sheds light on factors influencing de novo mutation rates and spectra.
Collapse
Affiliation(s)
| | - Seongwon Hwang
- Department of Genetics, University of Cambridge, Cambridge, UK
- MRC Biostatistics Unit, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Qin Qin Huang
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Rashesh Sanghvi
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | | | | | | | - Felix R Day
- MRC Epidemiology Unit, Box 285 Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Raheleh Rahbari
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
| | - Aylwyn Scally
- Department of Genetics, University of Cambridge, Cambridge, UK.
| | - Hilary C Martin
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
| |
Collapse
|
10
|
Wu X, Li C. Development of a prognostic model for breast cancer patients based on intratumoral tumor-infiltrating lymphocytes using machine learning algorithms. Discov Oncol 2025; 16:762. [PMID: 40366513 PMCID: PMC12078914 DOI: 10.1007/s12672-025-02585-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 05/06/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Breast cancer remains a formidable global health challenge, with tumor-infiltrating lymphocytes (TILs) serving as pivotal biomarkers associated with disease progression, therapeutic response, and survival. While research typically focused on stromal TILs (sTILs), we hypothesize that intratumoral TILs (iTILs), which are in direct contact with tumor cells, have a more profound role in the immune-tumor interactions. In light of this, we have developed an iTIL-centric model for breast cancer patient stratification and prognostic prediction. METHODS We sourced RNA-seq data and clinical profiles of breast cancer patients from The Cancer Genome Atlas (TCGA) and the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) to form our training dataset. Testing datasets, including GSE20685, GSE42568, GSE48390, and GSE88770, were retrieved from Gene Expression Omnibus (GEO). Employing consensus clustering and Weighted Correlation Network Analysis (WGCNA), we identified iTIL-associated hub genes. Our iTIL-centric signature was developed using a machine learning framework integrating 101 algorithms, validated across independent testing sets. Kaplan-Meier analysis and a nomogram model were utilized to evaluate the prognostic accuracy and clinical correlation of our model. GO and KEGG analyses elucidated the biological processes and pathways related to the iTIL signature. The immune profiling provided a comprehensive assessment of the immunological landscape. Moreover, potential drugs for high-risk patients were identified using CTRP v.2.0 and PRISM databases. RESULTS Our study constructed a pioneering prognostic model based on iTIL-centric signature via a machine learning framework that evaluated 101 algorithm combinations. This model revealed significant differences in the immune landscape among stratified patient cohorts, and demonstrated robust predictive capabilities across multiple datasets. The model showed excellent predictive performance with area under the curve (AUC) values of 0.940, 0.959, and 0.973 for 3-, 5-, and 10-year survival predictions, respectively. Additionally, it was identified as a significant risk factor for overall survival (OS) in the univariate analysis, with a hazard ratio (HR) > 1 and a p-value < 0.001. CONCLUSIONS Our prognostic model, founded on machining learning algorithms and anchored by an iTIL-centric signature, stands out as an invaluable tool for breast cancer patients, offering advanced prognostic insights and facilitating the development of personalized therapeutic strategies.
Collapse
Affiliation(s)
- Xinyi Wu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, Putuo District, Shanghai, 200065, China
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Chun Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Tongji Hospital, School of Medicine, Tongji University, 389 Xincun Road, Putuo District, Shanghai, 200065, China.
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
11
|
Wang Z, Dai Z, Gao Y, Zhao Z, Li Z, Wang L, Gao X, Qiu Q, Qiu X, Liu Z. Development of a machine learning-based predictive risk model combining fatty acid metabolism and ferroptosis for immunotherapy response and prognosis in prostate cancer. Discov Oncol 2025; 16:744. [PMID: 40355680 PMCID: PMC12069205 DOI: 10.1007/s12672-025-02484-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 04/24/2025] [Indexed: 05/14/2025] Open
Abstract
Prostate cancer (PCa) remains a leading cause of cancer-related mortality, necessitating robust prognostic models and personalized therapeutic strategies. This study integrated bulk RNA sequencing, single-cell RNA sequencing (scRNA-seq), and spatial transcriptomics to construct a prognostic model based on genes shared between ferroptosis and fatty acid metabolism (FAM). Using the TCGA-PRAD dataset, we identified 73 differentially expressed genes (DEGs) at the intersection of ferroptosis and FAM, of which 19 were significantly associated with progression-free survival (PFS). A machine learning-based prognostic model, optimized using the Lasso + Random Survival Forest (RSF) algorithm, achieved a high C-index of 0.876 and demonstrated strong predictive accuracy (1-, 2-, and 3-year AUCs: 0.77, 0.75, and 0.78, respectively). The model, validated in the DFKZ cohort, stratified patients into high- and low-risk groups, with the high-risk group exhibiting worse PFS and higher tumor mutation burden (TMB). Functional enrichment analysis revealed distinct pathway activities, with high-risk patients showing enrichment in immune-related and proliferative pathways, while low-risk patients were enriched in metabolic pathways. Immune microenvironment analysis revealed heightened immune activity in high-risk patients, characterized by increased infiltration of CD8 + T cells, regulatory T cells, and M2 macrophages, alongside elevated TIDE scores, suggesting immune evasion and resistance to immunotherapy. In contrast, low-risk patients exhibited higher infiltration of plasma cells and neutrophils and demonstrated better responses to immune checkpoint inhibitors (ICIs). Spatial transcriptomics and scRNA-seq further elucidated the spatial distribution of model genes, highlighting the central role of macrophages in mediating risk stratification. Additionally, chemotherapy sensitivity analysis identified potential therapeutic agents, such as Erlotinib and Picolinic acid, for low-risk patients. In vitro experiments showed that overexpression of CD38 in the PC-3 cell line led to elevated lipid peroxidation (C11-BODIPY) and reactive oxygen species (ROS), suggesting increased cell ferroptosis. These findings provide a comprehensive framework for risk stratification and personalized treatment in PCa, bridging molecular mechanisms with clinical outcomes.
Collapse
Affiliation(s)
- Zhenwei Wang
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Zhihong Dai
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Yuren Gao
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Zhongxiang Zhao
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China
| | - Zhen Li
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Liang Wang
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Xiang Gao
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China
| | - Qiuqiu Qiu
- Department of Urology, Gaohou People's Hospital, Maoming, 525200, China.
| | - Xiaofu Qiu
- Department of Urology, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510317, China.
| | - Zhiyu Liu
- Department of Urology, Second Hospital of Dalian Medical University, Dalian, 116023, China.
| |
Collapse
|
12
|
Hauseman ZJ, Stauffer F, Beyer KS, Mollé S, Cavicchioli E, Marchand JR, Fodor M, Viscomi J, Dhembi A, Katz S, Faggion B, Lanter M, Kerr G, Schildknecht D, Handl C, Maddalo D, Pissot Soldermann C, Brady J, Shrestha O, Nguyen Z, Leder L, Cremosnik G, Lopez Romero S, Hassiepen U, Stams T, Linder M, Galli GG, Guthy DA, King DA, Maira SM, Thoma CR, Ehmke V, Tordella L. Targeting the SHOC2-RAS interaction in RAS-mutant cancers. Nature 2025:10.1038/s41586-025-08931-1. [PMID: 40335703 DOI: 10.1038/s41586-025-08931-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 03/24/2025] [Indexed: 05/09/2025]
Abstract
Activating mutations in the rat sarcoma (RAS) genes HRAS, NRAS and KRAS collectively represent the most frequent oncogenic driver in human cancer1. They have previously been considered undruggable, but advances in the past few years have led to the clinical development of agents that target KRAS(G12C) and KRAS(G12D) mutants, yielding promises of therapeutic responses at tolerated doses2. However, clinical agents that selectively target NRAS(Q61*) mutants (* represents 'any'), the second-most-frequent oncogenic driver in melanoma, are still lacking. Here we identify SHOC2, a component of the SHOC2-MRAS-PP1C complex, as a dependency of RAS(Q61*) tumours in a nucleotide-state-dependent and isoform-agnostic manner. Mechanistically, we found that oncogenic NRAS(Q61R) forms a direct interaction with SHOC2, evidenced by X-ray co-crystal structure. In vitro high-throughput screening enabled the discovery of small molecules that bind to SHOC2 and disrupt the interaction with NRAS(Q61*). Structure-based optimization led to a cellularly active tool compound that shows inhibition of mitogen-activated protein kinase (MAPK) signalling and proliferation in RAS-mutant cancer models, most notably in NRAS(Q61*) settings. These findings provide evidence for a neomorph SHOC2-(canonical)RAS protein interaction that is pharmacologically actionable and relevant to cancer sustenance. Overall, this work provides the concept validation and foundation for developing new therapies at the core of the RAS signalling pathway.
Collapse
Affiliation(s)
| | | | - Kim S Beyer
- Novartis BioMedical Research, Basel, Switzerland
| | - Sandra Mollé
- Novartis BioMedical Research, Basel, Switzerland
| | | | | | | | | | | | | | | | | | - Grainne Kerr
- Novartis BioMedical Research, Basel, Switzerland
| | | | | | | | | | - Jacob Brady
- Novartis BioMedical Research, Cambridge, MA, USA
| | - Om Shrestha
- Novartis BioMedical Research, Cambridge, MA, USA
| | | | - Lukas Leder
- Novartis BioMedical Research, Basel, Switzerland
| | | | | | | | - Travis Stams
- Novartis BioMedical Research, Cambridge, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Kinnersley B, Jung J, Cornish AJ, Chubb D, Laxton R, Frangou A, Gruber AJ, Sud A, Caravagna G, Sottoriva A, Wedge DC, Booth T, Al-Sarraj S, Lawrence SED, Albanese E, Anichini G, Baxter D, Boukas A, Chowdhury YA, D'Urso P, Corns R, Dapaah A, Edlmann E, Greenway F, Grundy P, Hill CS, Jenkinson MD, Trichinopoly Krishna S, Smith S, Manivannan S, Martin AJ, Matloob S, Mukherjee S, O'Neill K, Plaha P, Pollock J, Price S, Rominiyi O, Sachdev B, Saeed F, Sinha S, Thorne L, Ughratdar I, Whitfield P, Youshani AS, Bulbeck H, Arumugam P, Houlston R, Ashkan K. Genomic landscape of diffuse glioma revealed by whole genome sequencing. Nat Commun 2025; 16:4233. [PMID: 40335506 PMCID: PMC12059081 DOI: 10.1038/s41467-025-59156-9] [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: 10/27/2023] [Accepted: 04/11/2025] [Indexed: 05/09/2025] Open
Abstract
Diffuse gliomas are the commonest malignant primary brain tumour in adults. Herein, we present analysis of the genomic landscape of adult glioma, by whole genome sequencing of 403 tumours (256 glioblastoma, 89 astrocytoma, 58 oligodendroglioma; 338 primary, 65 recurrence). We identify an extended catalogue of recurrent coding and non-coding genetic mutations that represents a source for future studies and provides a high-resolution map of structural variants, copy number changes and global genome features including telomere length, mutational signatures and extrachromosomal DNA. Finally, we relate these to clinical outcome. As well as identifying drug targets for treatment of glioma our findings offer the prospect of improving treatment allocation with established targeted therapies.
Collapse
Affiliation(s)
- Ben Kinnersley
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK.
- UCL Cancer Institute, 72 Huntley St, WC1E 6DD, London, UK.
| | - Josephine Jung
- Institute of Psychiatry, Psychology and Neurosciences, Kings College London, Strand, WC2R 2LS, London, UK.
- Department of Neurosurgery, King's College Hospital NHS Foundation Trust, Denmark Hill, SE5 9RS, London, UK.
| | - Alex J Cornish
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Daniel Chubb
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Ross Laxton
- Department of Clinical Neuropathology, King's College Hospital NHS Foundation Trust, Denmark Hill, SE5 9RS, London, UK
| | - Anna Frangou
- Cancer Genomics, Big Data Institute, Nuffield Department of Medicine, Old Road Campus, OX3 7LF, Oxford, UK
| | - Andreas J Gruber
- Department of Biology, University of Konstanz, Konstanz, 78464, Germany
| | - Amit Sud
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Giulio Caravagna
- Evolutionary Genomics and Modelling Lab, Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Andrea Sottoriva
- Evolutionary Genomics and Modelling Lab, Centre for Evolution and Cancer, The Institute of Cancer Research, London, SM2 5NG, UK
| | - David C Wedge
- Manchester Cancer Research Centre, University of Manchester, 555 Wilmslow Road, Manchester, M20 4GJ, UK
| | - Thomas Booth
- School of Biomedical Engineering & Imaging Sciences, King's College London, St Thomas's Hospital, London, UK
- Department of Neuroradiology, King's College Hospital NHS Foundation Trust, Denmark Hill, SE5 9RS, London, UK
| | - Safa Al-Sarraj
- Department of Clinical Neuropathology, King's College Hospital NHS Foundation Trust, Denmark Hill, SE5 9RS, London, UK
| | - Samuel E D Lawrence
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK
| | - Erminia Albanese
- Department of Neurosurgery, Royal Stoke University Hospital, Newcastle Road, ST4 6QG, Stoke-on-Trent, UK
| | - Giulio Anichini
- Imperial College Healthcare NHS Trust, Charing Cross Hospital, 3S corridor, Fulham Palace Road, London, W6 8RF, UK
| | - David Baxter
- Department of Neurosurgery, Royal National Orthopaedic Hospital, Brockley Hill, Stanmore, HA7 4LP, UK
| | - Alexandros Boukas
- Department of Neurosurgery, John Radcliffe Hospital, Headley Way, Headington, OX3 9DU, Oxford, UK
| | - Yasir A Chowdhury
- Department of Neurosurgery, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, B15 2GW, Birmingham, UK
| | - Pietro D'Urso
- Department of Neurosurgery, Manchester Royal Infirmary, Oxford Rd, M13 9WL, Manchester, UK
| | - Robert Corns
- Department of Neurosurgery, Leeds General Infirmary, Great George St, LS1 3EX, Leeds, UK
| | - Andrew Dapaah
- Department of Neurosurgery, Queen's Medical Centre NHS Trust, Derby Road, Lenton, NG7 2UH, Nottingham, UK
| | - Ellie Edlmann
- South West Neurosurgery Unit, University Hospitals Plymouth NHS Trust, Derriford Road, Crownhill, PL6 8DH, Plymouth, UK
| | - Fay Greenway
- Department of Neurosurgery, St. George's University Hospitals NHS Foundation Trust, Blackshaw Rd, SW17 0QT, London, UK
| | - Paul Grundy
- Department of Neurosurgery, Southampton General Hospital, Tremona Road, SO16 6YD, Southampton, UK
| | - Ciaran S Hill
- UCL Cancer Institute, 72 Huntley St, WC1E 6DD, London, UK
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, WC1N 3BG, London, UK
| | - Michael D Jenkinson
- Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Lower Lane, Fazakerley, L9 7LJ, Liverpool, UK
| | - Sandhya Trichinopoly Krishna
- Department of Neurosurgery, The Walton Centre NHS Foundation Trust, Lower Lane, Fazakerley, L9 7LJ, Liverpool, UK
| | - Stuart Smith
- Department of Neurosurgery, Queen's Medical Centre NHS Trust, Derby Road, Lenton, NG7 2UH, Nottingham, UK
| | - Susruta Manivannan
- Department of Neurosurgery, Southampton General Hospital, Tremona Road, SO16 6YD, Southampton, UK
| | - Andrew J Martin
- Department of Neurosurgery, St. George's University Hospitals NHS Foundation Trust, Blackshaw Rd, SW17 0QT, London, UK
| | - Samir Matloob
- Department of Neurosurgery, Queen's Hospital Romford, Rom Valley Way, RM7 0AG, Romford, UK
| | - Soumya Mukherjee
- Department of Neurosurgery, Addenbrookes Hospital, Hills Rd, CB2 0QQ, Cambridge, UK
| | - Kevin O'Neill
- Imperial College Healthcare NHS Trust, Charing Cross Hospital, 3S corridor, Fulham Palace Road, London, W6 8RF, UK
| | - Puneet Plaha
- Department of Neurosurgery, John Radcliffe Hospital, Headley Way, Headington, OX3 9DU, Oxford, UK
| | - Jonathan Pollock
- Department of Neurosurgery, Queen's Hospital Romford, Rom Valley Way, RM7 0AG, Romford, UK
| | - Stephen Price
- Department of Neurosurgery, Addenbrookes Hospital, Hills Rd, CB2 0QQ, Cambridge, UK
| | - Ola Rominiyi
- Department of Neurosurgery, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Glossop Rd, Broomhall, S10 2JF, Sheffield, UK
| | - Bobby Sachdev
- Department of Neurosurgery, Royal Stoke University Hospital, Newcastle Road, ST4 6QG, Stoke-on-Trent, UK
| | - Fozia Saeed
- Department of Neurosurgery, Leeds General Infirmary, Great George St, LS1 3EX, Leeds, UK
| | - Saurabh Sinha
- Department of Neurosurgery, Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Glossop Rd, Broomhall, S10 2JF, Sheffield, UK
| | - Lewis Thorne
- Department of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, WC1N 3BG, London, UK
| | - Ismail Ughratdar
- Department of Neurosurgery, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, B15 2GW, Birmingham, UK
| | - Peter Whitfield
- South West Neurosurgery Unit, University Hospitals Plymouth NHS Trust, Derriford Road, Crownhill, PL6 8DH, Plymouth, UK
| | - Amir Saam Youshani
- Department of Neurosurgery, Manchester Royal Infirmary, Oxford Rd, M13 9WL, Manchester, UK
| | - Helen Bulbeck
- Brainstrust, 4 Yvery Court, Castle Road, PO31 7QG, Cowes, Isle of Wight, UK
| | | | - Richard Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, SM2 5NG, UK.
| | - Keyoumars Ashkan
- Institute of Psychiatry, Psychology and Neurosciences, Kings College London, Strand, WC2R 2LS, London, UK.
- Department of Neurosurgery, King's College Hospital NHS Foundation Trust, Denmark Hill, SE5 9RS, London, UK.
| |
Collapse
|
14
|
Okonechnikov K, Joshi P, Körber V, Rademacher A, Bortolomeazzi M, Mallm JP, Vaillant J, da Silva PBG, Statz B, Sepp M, Sarropoulos I, Yamada T, Wittmann A, Schramm K, Blattner-Johnson M, Fiesel P, Jones B, Jäger N, Milde T, Pajtler KW, van Tilburg CM, Witt O, Bochennek K, Weber KJ, Nonnenmacher L, Reimann C, Ghasemi DR, Schüller U, Mynarek M, Rutkowski S, Jones DTW, Korshunov A, Rippe K, Westermann F, Thongjuea S, Höfer T, Kaessmann H, Kutscher LM, Pfister SM. Oncogene aberrations drive medulloblastoma progression, not initiation. Nature 2025:10.1038/s41586-025-08973-5. [PMID: 40335697 DOI: 10.1038/s41586-025-08973-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/03/2025] [Indexed: 05/09/2025]
Abstract
Despite recent advances in understanding disease biology, treatment of group 3/4 medulloblastoma remains a therapeutic challenge in paediatric neuro-oncology1. Bulk-omics approaches have identified considerable intertumoural heterogeneity in group 3/4 medulloblastoma, including the presence of clear single-gene oncogenic drivers in only a subset of cases, whereas in most cases, large-scale copy number aberrations prevail2,3. However, intratumoural heterogeneity, the role of oncogene aberrations, and broad copy number variation in tumour evolution and treatment resistance remain poorly understood. To dissect this interplay, we used single-cell technologies (single-nucleus RNA sequencing (snRNA-seq), single-nucleus assay for transposase-accessible chromatin with high-throughput sequencing (snATAC-seq) and spatial transcriptomics) on a cohort of group 3/4 medulloblastoma with known alterations in the oncogenes MYC, MYCN and PRDM6. We show that large-scale chromosomal aberrations are early tumour-initiating events, whereas the single-gene oncogenic events arise late and are typically subclonal, but MYC can become clonal upon disease progression to drive further tumour development and therapy resistance. Spatial transcriptomics shows that the subclones are mostly interspersed across tumour tissue, but clear segregation is also present. Using a population genetics model, we estimate medulloblastoma initiation in the cerebellar unipolar brush cell lineage starting from the first gestational trimester. Our findings demonstrate how single-cell technologies can be applied for early detection and diagnosis of this fatal disease.
Collapse
Affiliation(s)
- Konstantin Okonechnikov
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Piyush Joshi
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Verena Körber
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Anne Rademacher
- Division of Chromatin Networks, German Cancer Research Center (DKFZ) and Bioquant, Heidelberg, Germany
| | | | - Jan-Philipp Mallm
- Single-cell Open Lab, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jan Vaillant
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Patricia Benites Goncalves da Silva
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Britta Statz
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Mari Sepp
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Ioannis Sarropoulos
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Tetsuya Yamada
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Andrea Wittmann
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kathrin Schramm
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mirjam Blattner-Johnson
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Petra Fiesel
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- CCU Neuropathology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Barbara Jones
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Natalie Jäger
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Till Milde
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Pediatric Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Kristian W Pajtler
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Cornelis M van Tilburg
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Pediatric Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Olaf Witt
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany
- CCU Pediatric Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Konrad Bochennek
- Frankfurt University Hospital, Goethe University, Frankfurt, Germany
| | - Katharina Johanna Weber
- Goethe University Frankfurt, University Hospital, Neurological Institute (Edinger Institute), Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Frankfurt Cancer Institute (FCI), Frankfurt am Main, Germany
- Goethe University Frankfurt, University Hospital, University Cancer Center (UCT) Frankfurt, Frankfurt am Main, Germany
| | | | | | - David R Ghasemi
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
| | - Ulrich Schüller
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Research Institute Children's Cancer Center Hamburg, Hamburg, Germany
- Department of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Martin Mynarek
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Rutkowski
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Andrey Korshunov
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- CCU Neuropathology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Karsten Rippe
- Division of Chromatin Networks, German Cancer Research Center (DKFZ) and Bioquant, Heidelberg, Germany
| | - Frank Westermann
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Neuroblastoma Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Supat Thongjuea
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Thomas Höfer
- Division of Theoretical Systems Biology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Henrik Kaessmann
- Division of Pediatric Glioma Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lena M Kutscher
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany.
- Developmental Origins of Pediatric Cancer Junior Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Stefan M Pfister
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT) Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany.
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany.
- Department of Pediatric Oncology, Hematology & Immunology, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
15
|
Xu Y, Lou D, Chen P, Li G, Usoskin D, Pan J, Li F, Huang S, Hess C, Tang R, Hu X, Yu J, Arceo M, de Krijger RR, Tischler AS, Schlisio S, Ernfors P, Hu Y, Wang J. Single-cell MultiOmics and spatial transcriptomics demonstrate neuroblastoma developmental plasticity. Dev Cell 2025:S1534-5807(25)00251-5. [PMID: 40347947 DOI: 10.1016/j.devcel.2025.04.013] [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: 06/07/2024] [Revised: 10/27/2024] [Accepted: 04/17/2025] [Indexed: 05/14/2025]
Abstract
Neuroblastoma, the most prevalent extracranial pediatric solid tumor, arises from neural crest progeny cells. It exhibits substantial developmental plasticity and intratumoral heterogeneity, leading to survival rates below 50% in high-risk cases. The regulatory mechanisms underlying this plasticity remain largely elusive. In this integrative study, we used single-cell MultiOmics from a mouse spontaneous tumor model and spatial transcriptomics from human patient samples to dissect the transcriptional and epigenetic landscapes that govern developmental states in neuroblastoma. We identified developmental intermediate states in high-risk neuroblastomas critical for malignant transitions and uncovered extensive epigenetic priming with latent capacity for diverse state transitions. Furthermore, we mapped enhancer gene regulatory networks (eGRNs) and tumor microenvironments sustaining these aggressive states. State transitions and malignancy could be interfered with by targeting transcription factors controlling the eGRNs.
Collapse
Affiliation(s)
- Yunyun Xu
- Pediatric Clinical Research Institute, Children's Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215000, China
| | - Daohua Lou
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17165, Sweden
| | - Ping Chen
- Department of Laboratory Medicine, Karolinska Institute, Huddinge 14157, Sweden; Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsingfors 00014, Finland
| | - Gang Li
- Pediatric Clinical Research Institute, Children's Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215000, China
| | - Dimtry Usoskin
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17165, Sweden
| | - Jian Pan
- Pediatric Clinical Research Institute, Children's Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215000, China
| | - Fang Li
- Department of Human Anatomy, Histology and Embryology, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, Jiangsu 215000, China
| | - Shungen Huang
- Department of General Surgery, Children's Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215000, China
| | - Caroline Hess
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17165, Sweden
| | - Ruze Tang
- Department of General Surgery, Children's Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215000, China
| | - Xiaohan Hu
- Pediatric Clinical Research Institute, Children's Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215000, China
| | - Juanjuan Yu
- Pediatric Clinical Research Institute, Children's Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215000, China
| | - Maria Arceo
- Department of Oncology-Pathology, Karolinska Institute, Stockholm 17165, Sweden
| | - Ronald R de Krijger
- Princess Máxima Center for Pediatric Oncology, Utrecht 3511AB, the Netherlands; Department of Pathology, University Medical Center Utrecht, Utrecht 3511AB, the Netherlands
| | - Arthur S Tischler
- Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | - Susanne Schlisio
- Department of Oncology-Pathology, Karolinska Institute, Stockholm 17165, Sweden
| | - Patrik Ernfors
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17165, Sweden.
| | - Yizhou Hu
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm 17165, Sweden; Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsingfors 00014, Finland.
| | - Jian Wang
- Pediatric Clinical Research Institute, Children's Hospital Affiliated to Soochow University, Suzhou, Jiangsu 215000, China.
| |
Collapse
|
16
|
Huang J, Wang J, Wang G, Zhao Y. Allele frequency in thyroid cancer: mechanisms, challenges, and applications in cancer therapy. Thyroid Res 2025; 18:19. [PMID: 40325461 PMCID: PMC12054298 DOI: 10.1186/s13044-025-00237-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/24/2025] [Indexed: 05/07/2025] Open
Abstract
Allele Frequency (AF) is the percentage of sequence reads with a specific mutation relative to the read depth at that locus, reflecting the proportion of gene mutation. This review explores the AF characteristics of different mutations in thyroid cancer, investigating their connection with tumor features and clinical characteristics. BRAF mutation AF is associated with tumour malignancy and prognosis, exhibiting a relatively low peak value. TERT mutations in AF are associated with invasive characteristics, and the combination between BRAF and TERT mutations AF improved the diagnostic value in identifying patients' risk of recurrence and tumour malignancy. RET mutation is frequently observed in medullary carcinoma, and RET mutation AF is associated with partial tumour characteristics. RAS mutation is prevalent in follicular tumors, but the association between RAS mutation AF and tumour characteristics is relatively weak. TP53 mutation is more frequently occurred in poorly differentiated and anaplastic carcinoma, and its AF might be associated with the dedifferentiation process. We also concentrated on the mutually exclusive and synergistic effect between different mutations. The mutation rate of TERT increases with the elevation of BRAF mutation AF. Finally, the detection and assessment of AF by NGS in clinical practice helps to provide a reference for individualised targeted therapy plans.
Collapse
Affiliation(s)
- Jiayu Huang
- Department of Thyroid Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Jiazhi Wang
- Department of Thyroid Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Guangzhi Wang
- Department of Thyroid Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China.
| | - Yongfu Zhao
- Department of Thyroid Surgery, The Second Hospital of Dalian Medical University, Dalian, Liaoning Province, China.
| |
Collapse
|
17
|
Struys I, Velázquez C, Ubels J, LeJeune CL, van Roosmalen MJ, Rosendahl Huber AK, van Leeuwen AJ, Bossuyt W, Thienpont B, Voet T, Van Calsteren K, Lenaerts L, van Boxtel R, Amant F. Prenatal Exposure to Chemotherapy Increases the Mutation Burden in Human Neonatal Hematopoietic Stem Cells. Cancer Discov 2025; 15:903-912. [PMID: 39852764 PMCID: PMC12046327 DOI: 10.1158/2159-8290.cd-24-1368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/17/2024] [Accepted: 01/22/2025] [Indexed: 01/26/2025]
Abstract
SIGNIFICANCE This study demonstrates that environmental mutagenic exposure during pregnancy can increase somatic mutation accumulation in the fetus. Given that detrimental early life exposures can adversely affect health outcomes later in life, our study highlights the need for further research into the impact of environmentally induced genomic insults during the perinatal period. See related commentary by Furudate and Takahashi, p. 870.
Collapse
Affiliation(s)
- Ilana Struys
- Department of Oncology, University of Leuven, KU Leuven, Leuven, Belgium
| | - Carolina Velázquez
- Department of Oncology, University of Leuven, KU Leuven, Leuven, Belgium
| | - Joske Ubels
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | | | - Markus J. van Roosmalen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Axel K.M. Rosendahl Huber
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Anais J.C.N. van Leeuwen
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Wouter Bossuyt
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
| | - Bernard Thienpont
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
| | - Thierry Voet
- Department of Human Genetics, University of Leuven, KU Leuven, Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), University of Leuven, KU Leuven, Leuven, Belgium
| | - Kristel Van Calsteren
- Department of Obstetrics and Gynaecology, University Hospitals Leuven, UZ Leuven, Leuven, Belgium
| | - Liesbeth Lenaerts
- Department of Oncology, University of Leuven, KU Leuven, Leuven, Belgium
| | - Ruben van Boxtel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Frédéric Amant
- Department of Oncology, University of Leuven, KU Leuven, Leuven, Belgium
- Department of Obstetrics and Gynaecology, University Hospitals Leuven, UZ Leuven, Leuven, Belgium
- Gynecologic Oncology, Netherlands Cancer Institute, Antoni Van Leeuwenhoek, Amsterdam, the Netherlands
| |
Collapse
|
18
|
Krasowska J, Imaoka T, Fornalski KW. Application of the Avrami-Dobrzyński model for mammary tumorigenesis in irradiated rats indicates new candidates for parametric cancer risk assessment. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2025; 64:229-239. [PMID: 40192782 DOI: 10.1007/s00411-025-01125-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 03/29/2025] [Indexed: 05/04/2025]
Abstract
The two-parametric Avrami-Dobrzyński model, originally based on the condensed matter physics for phase transitions, was applied to the cumulative populational mammary cancer data of laboratory rats. The joint effect of parity, irradiation and BRCA1 mutation on breast cancer incidence was analysed. The study showed that the proposed model fits well with the data points, however, the values of parameters differ regarding the investigated group of animals. It was concluded that both model's parameters, which relate to the dimension of carcinogenesis dynamics and the age distribution, are good candidates for cancer risk assessment regarding different risk factors.
Collapse
Affiliation(s)
- Julianna Krasowska
- Faculty of Physics, Warsaw University of Technology, Warsaw, 00-662, Poland.
| | - Tatsuhiko Imaoka
- Department of Radiation Effects Research, Institute for Radiological Science, National Institutes for Quantum Science and Technology, Chiba, 263-8555, Japan
- Institute for Quantum Life Science, National Institutes for Quantum Science and Technology, Chiba, 263- 8555, Japan
| | | |
Collapse
|
19
|
Martins Rodrigues F, Terekhanova NV, Imbach KJ, Clauser KR, Esai Selvan M, Mendizabal I, Geffen Y, Akiyama Y, Maynard M, Yaron TM, Li Y, Cao S, Storrs EP, Gonda OS, Gaite-Reguero A, Govindan A, Kawaler EA, Wyczalkowski MA, Klein RJ, Turhan B, Krug K, Mani DR, Leprevost FDV, Nesvizhskii AI, Carr SA, Fenyö D, Gillette MA, Colaprico A, Iavarone A, Robles AI, Huang KL, Kumar-Sinha C, Aguet F, Lazar AJ, Cantley LC, Marigorta UM, Gümüş ZH, Bailey MH, Getz G, Porta-Pardo E, Ding L. Precision proteogenomics reveals pan-cancer impact of germline variants. Cell 2025; 188:2312-2335.e26. [PMID: 40233739 DOI: 10.1016/j.cell.2025.03.026] [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: 10/09/2023] [Revised: 04/29/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025]
Abstract
We investigate the impact of germline variants on cancer patients' proteomes, encompassing 1,064 individuals across 10 cancer types. We introduced an approach, "precision peptidomics," mapping 337,469 coding germline variants onto peptides from patients' mass spectrometry data, revealing their potential impact on post-translational modifications, protein stability, allele-specific expression, and protein structure by leveraging the relevant protein databases. We identified rare pathogenic and common germline variants in cancer genes potentially affecting proteomic features, including variants altering protein abundance and structure and variants in kinases (ERBB2 and MAP2K2) impacting phosphorylation. Precision peptidome analysis predicted destabilizing events in signal-regulatory protein alpha (SIRPA) and glial fibrillary acid protein (GFAP), relevant to immunomodulation and glioblastoma diagnostics, respectively. Genome-wide association studies identified quantitative trait loci for gene expression and protein levels, spanning millions of SNPs and thousands of proteins. Polygenic risk scores correlated with distal effects from risk variants. Our findings emphasize the contribution of germline genetics to cancer heterogeneity and high-throughput precision peptidomics.
Collapse
Affiliation(s)
- Fernanda Martins Rodrigues
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Nadezhda V Terekhanova
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Kathleen J Imbach
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain; Universitat Autonoma de Barcelona, Barcelona, Spain
| | | | - Myvizhi Esai Selvan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Isabel Mendizabal
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain; Translational Prostate Cancer Research Lab, CIC bioGUNE-Basurto, Biocruces Bizkaia Health Research Institute, Derio, Spain
| | - Yifat Geffen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Yo Akiyama
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Tomer M Yaron
- Meyer Cancer Center, Department of Medicine, Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Yize Li
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Song Cao
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Erik P Storrs
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Olivia S Gonda
- Department of Biology, Brigham Young University, Salt Lake City, UT, USA
| | - Adrian Gaite-Reguero
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Akshay Govindan
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Emily A Kawaler
- Applied Bioinformatics Laboratories, New York University Langone Health, New York City, NY, USA
| | - Matthew A Wyczalkowski
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Robert J Klein
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Berk Turhan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Karsten Krug
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - D R Mani
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Steven A Carr
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David Fenyö
- Institute for Systems Genetics, NYU Grossman School of Medicine, New York, NY, USA
| | | | - Antonio Colaprico
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Antonio Iavarone
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA; Department of Neurological Surgery, Department of Biochemistry and Molecular Biology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, National Cancer Institute, Rockville, MD, USA
| | - Kuan-Lin Huang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Transformative Disease Modeling, Tisch Cancer Institute, Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chandan Kumar-Sinha
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Alexander J Lazar
- Departments of Pathology and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Urko M Marigorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Zeynep H Gümüş
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Matthew H Bailey
- Department of Biology, Brigham Young University, Salt Lake City, UT, USA.
| | - Gad Getz
- Broad Institute of MIT and Harvard, Cambridge, MA, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| | - Eduard Porta-Pardo
- Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Spain; Barcelona Supercomputing Center (BSC), Barcelona, Spain.
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, Saint Louis, MO, USA; McDonnell Genome Institute, Washington University in St. Louis, Saint Louis, MO, USA; Department of Genetics, Washington University in St. Louis, St. Louis, MO 63110, USA; Siteman Cancer Center, Washington University in St. Louis, Saint Louis, MO, USA.
| |
Collapse
|
20
|
Grossi E, Nguyen CB, Carcamo S, Kirigin Callaú V, Moran S, Filipescu D, Tagore S, Firestone TM, Keogh MC, Sun L, Izar B, Hasson D, Bernstein E. The SWI/SNF PBAF complex facilitates REST occupancy at repressive chromatin. Mol Cell 2025; 85:1714-1729.e7. [PMID: 40252649 PMCID: PMC12048221 DOI: 10.1016/j.molcel.2025.03.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/30/2025] [Accepted: 03/31/2025] [Indexed: 04/21/2025]
Abstract
SWI/SNF (switch/sucrose non-fermentable) chromatin remodelers possess unique functionalities difficult to dissect. Distinct cancers harbor mutations in specific subunits, such as the polybromo-associated BAF (PBAF)-specific component ARID2 in melanoma. Here, we perform epigenomic profiling of SWI/SNF complexes and their associated chromatin states in melanocytes and melanoma. Time-resolved approaches reveal that PBAF regions are generally less sensitive to ATPase inhibition than BAF sites. We further uncover a subset of PBAF-exclusive regions within Polycomb-repressed chromatin that are enriched for REST (RE1 silencing transcription factor), a transcription factor that represses neuronal genes. In turn, PBAF complex disruption via ARID2 loss hinders REST's ability to bind and inactivate its targets, leading to upregulation of synaptic transcripts. Remarkably, this gene signature is conserved in melanoma patients with ARID2 mutations and correlates with an expression program enriched in melanoma brain metastases. Overall, we demonstrate a unique role for PBAF in generating accessibility for a silencing transcription factor at repressed chromatin, with important implications for disease.
Collapse
Affiliation(s)
- Elena Grossi
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Christie B Nguyen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Saul Carcamo
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Bioinformatics for Next Generation Sequencing (BiNGS) Shared Resource Facility, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Valentina Kirigin Callaú
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shannon Moran
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Dan Filipescu
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Somnath Tagore
- Department of Medicine, Division of Hematology/Oncology, and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | | | | | - Lu Sun
- EpiCypher Inc., Durham, NC 27709, USA
| | - Benjamin Izar
- Department of Medicine, Division of Hematology/Oncology, and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Dan Hasson
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Bioinformatics for Next Generation Sequencing (BiNGS) Shared Resource Facility, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily Bernstein
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
21
|
Wang X, Feng H, Zhang Y, Lin F. SGCLMD: Signed graph-based contrastive learning model for predicting somatic mutation-drug association. Comput Biol Med 2025; 190:110067. [PMID: 40147185 DOI: 10.1016/j.compbiomed.2025.110067] [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: 03/12/2024] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
Somatic mutations could influence critical cellular processes, leading to uncontrolled cell growth and tumor formation. Understanding the intricate interactions between somatic mutations and drugs was crucial for advancing our knowledge of the underlying biological mechanisms of cancer. This knowledge, in turn, could drive advancements in cancer detection, diagnosis, and treatment. Exploring the relationships between specific somatic mutations and drug responses held the potential to identify targeted therapeutic interventions and improve personalized treatment strategies for cancer patients. In this study, we introduced a computational model, the signed graph comparison learning for mutation-drug associations (SGCLMD), designed to predict signs of somatic mutation-drug associations. Initially, we leveraged clinical data to construct a benchmark dataset encompassing somatic mutation-drug associations. We proposed a graph enhancement method, employing a random perturbation strategy, to expand the signed graph. This approach not only preserved interaction information across the two perspectives of the signed graph but also retained implicit relationships between these perspectives. Furthermore, we devised a multi-view comparison loss algorithm to learn node representations for the graph generated post-random perturbation. Through parameter optimization using 5-fold cross-validation, our SGCLMD model achieves optimal area under the curve (AUC) and area under the precision-recall curve (AUPR) values of 0.8306 and 0.8751, respectively, representing improvements of 3 % and 3.1 % over the state-of-the-art method. Through ablation experiments and case studies, we validated the importance of graph enhancement methods and multi-view contrast learning modules, demonstrating SGCLMD's potential in predicting somatic mutation-drug associations. The code and dataset for SGCLMD are available at https://github.com/wangxiaosong96/SGCLMD.
Collapse
Affiliation(s)
- Xiaosong Wang
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Haisong Feng
- School of Informatics, Xiamen University, Xiamen, Fujian, 361105, China
| | - Yilei Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, Anhui, 230036, China
| | - Fan Lin
- School of Informatics, Xiamen University, Xiamen, Fujian, 361105, China.
| |
Collapse
|
22
|
Sinberger LA, Zahavi T, Keren-Khadmy N, Dugach Y, Sonnenblick A, Salmon-Divon M. Refining prognostic tools for luminal breast cancer: genetic insights and comprehensive analysis. ESMO Open 2025; 10:105080. [PMID: 40305907 PMCID: PMC12088756 DOI: 10.1016/j.esmoop.2025.105080] [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: 09/02/2024] [Revised: 03/30/2025] [Accepted: 04/03/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Luminal breast cancer (BC) is generally associated with a lower risk of recurrence compared with other subtypes. However, patients with luminal BC can still experience recurrence, which remains a significant concern and contributes to BC-related mortality. Current clinical practice for recurrence risk prognosis relies on prognostic tests based on tumor gene expression profiles. MATERIALS AND METHODS In this study, we aimed to investigate the association between different genetic alterations with the likelihood of recurrence and gene expression prognostic prediction (Oncotype DX®, MammaPrint®, and PAM50-ROR) in luminal BC patients. We constructed three transcriptome-based predictive models, based on these widely used clinical tests, to evaluate the recurrence risk of patients with luminal BC, using RNA-seq data from 1527 samples across 11 datasets. We further classified 1780 patients from the TCGA and METABRIC datasets into risk groups and detected distinct recurrence risk patterns. RESULTS Our analysis revealed that low-risk groups had higher frequencies of mutations in PIK3CA, MAP3K1, CDH1, KMT2C, and CBFB, as well as co-mutations in PIK3CA-MAP3K1, PIK3CA-CBFB, and KMT2C-MAP3K1. In contrast, high-risk groups showed enrichment of TP53, RB1, and PTPN22 mutations compared with the whole cohort, with notable co-mutations in TP53-PIK3CA and TP53-KMT2C. Furthermore, mutations in TP53 and BRCA2, and deletions in the 7p22.3 region were at least threefold more frequent in high-risk patients compared with low-risk patients. Using an independent dataset, we validated our finding of higher frequency of BRCA2 mutations in Oncotype DX® high-risk patients. Notably, PIK3CA mutations had an unexpected negative impact on recurrence and survival among high-risk patients. CONCLUSION Our study reveals key genetic factors associated with recurrence risk in luminal BC. Identifying these mutations and copy number alterations provides a basis for refined prognostic models and suggests avenues for further research, potentially improving treatment strategies and follow-up care for patients with luminal BC.
Collapse
Affiliation(s)
- L A Sinberger
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - T Zahavi
- Department of Molecular Biology, Ariel University, Ariel, Israel
| | - N Keren-Khadmy
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Y Dugach
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - A Sonnenblick
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - M Salmon-Divon
- Department of Molecular Biology, Ariel University, Ariel, Israel; Adelson School of Medicine, Ariel University, Ariel, Israel.
| |
Collapse
|
23
|
García-Vázquez N, González-Robles TJ, Lane E, Spasskaya D, Zhang Q, Kerzhnerman MA, Jeong Y, Collu M, Simoneschi D, Ruggles KV, Róna G, Kaisari S, Pagano M. Stabilization of GTSE1 by cyclin D1-CDK4/6-mediated phosphorylation promotes cell proliferation with implications for cancer prognosis. eLife 2025; 13:RP101075. [PMID: 40272409 PMCID: PMC12021411 DOI: 10.7554/elife.101075] [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: 04/25/2025] Open
Abstract
In healthy cells, cyclin D1 is expressed during the G1 phase of the cell cycle, where it activates CDK4 and CDK6. Its dysregulation is a well-established oncogenic driver in numerous human cancers. The cancer-related function of cyclin D1 has been primarily studied by focusing on the phosphorylation of the retinoblastoma (RB) gene product. Here, using an integrative approach combining bioinformatic analyses and biochemical experiments, we show that GTSE1 (G-Two and S phases expressed protein 1), a protein positively regulating cell cycle progression, is a previously unrecognized substrate of cyclin D1-CDK4/6 in tumor cells overexpressing cyclin D1 during G1 and subsequent phases. The phosphorylation of GTSE1 mediated by cyclin D1-CDK4/6 inhibits GTSE1 degradation, leading to high levels of GTSE1 across all cell cycle phases. Functionally, the phosphorylation of GTSE1 promotes cellular proliferation and is associated with poor prognosis within a pan-cancer cohort. Our findings provide insights into cyclin D1's role in cell cycle control and oncogenesis beyond RB phosphorylation.
Collapse
Affiliation(s)
- Nelson García-Vázquez
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Tania J González-Robles
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
- Department of Medicine, New York University Grossman School of MedicineNew YorkUnited States
- Howard Hughes Medical Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Ethan Lane
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Daria Spasskaya
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Qingyue Zhang
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Marc A Kerzhnerman
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - YeonTae Jeong
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Marta Collu
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Daniele Simoneschi
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
| | - Kelly V Ruggles
- Department of Medicine, New York University Grossman School of MedicineNew YorkUnited States
| | - Gergely Róna
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
- Howard Hughes Medical Institute, New York University Grossman School of MedicineNew YorkUnited States
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural SciencesBudapestHungary
| | - Sharon Kaisari
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
- Howard Hughes Medical Institute, New York University Grossman School of MedicineNew YorkUnited States
| | - Michele Pagano
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of MedicineNew YorkUnited States
- Howard Hughes Medical Institute, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
24
|
Bayó C, Castellano G, Marín F, Castillo-Iturra J, Ocaña T, Kumari H, Pellisé M, Moreira L, Rivero L, Daca-Alvarez M, Ortiz O, Carballal S, Moreira R, Canet-Hermida J, Pineda M, Gabriel C, Flórez-Grau G, Juan M, Benitez-Ribas D, Balaguer F. Discovery and validation of frameshift-derived neopeptides in Lynch syndrome: paving the way for novel cancer prevention strategies. J Immunother Cancer 2025; 13:e011177. [PMID: 40254392 PMCID: PMC12010338 DOI: 10.1136/jitc-2024-011177] [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: 12/05/2024] [Accepted: 03/23/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Lynch syndrome (LS), caused by germline pathogenic variants in the mismatch repair genes, leads to high rates of frameshift-derived neopeptide (FSDN) expression due to microsatellite instability (MSI). While colorectal cancer (CRC) prevention is effective, most LS-related tumors lack such strategies. Cancer vaccines targeting FSDNs offer a promising approach for immune interception in LS. This study aimed to identify and validate LS-related FSDNs to develop vaccines for cancer prevention. METHODS We identified LS-related coding MS mutations and predicted FSDN with high coverage on common Human Leukocyte Antigen (HLA)-I and II alleles. We validated FSDN-associated mutations in colorectal adenomas (CrAD), endometrial cancers (EC), and CRC samples from patients with LS, non-LS tumors, and cell lines. Immunogenicity was assessed through interferon (IFN)-γ enzyme-linked immunospot and flow cytometry analysis of tissue-infiltrating lymphocytes (TILs) from LS carriers. RESULTS We prioritized 53 HLA-I and 45 HLA-II FSDNs in MSI tumors using in silico predictions. Validation revealed 86.7% of FSDN-associated mutations present in LS-CRC samples, with a median of 7.67 (6.5-9) mutations in CrADs and 6.02 (2-10) in CRCs. Sequencing of CrAD and EC samples showed 95% and 77.5% of predicted FSDN-associated mutations, respectively. MSI cancer cell lines transcribed 69.8% of FSDNs. Immunogenicity assays showed that 71% of potential FSDNs elicited IFN-γ responses, with a median of 7.37 (1-10.75) HLA-I and 6 (2-5.75) HLA-II FSDNs per patient. After prioritizing 24 FSDN, in a cohort of 19 LS-derived samples (4 CrAD and 15 normal mucosa), 52% (10/19) demonstrated T-cell reactivity to an HLA-I neoantigen pool. CD8+CD137+ activation markers increased significantly (p=0.037) over time and peptide-specific cells were detected by pentamer staining. CONCLUSIONS Our predicted FSDN set has optimal coverage among LS carriers and can induce IFN-γ inflammatory responses in LS-derived TILs, offering an opportunity for vaccine development.
Collapse
Affiliation(s)
- Cristina Bayó
- Immunology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Giancarlo Castellano
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Fátima Marín
- Hereditary Cancer Program, Catalan institute of oncology, IDIBELL, Badalona, Catalunya, Spain
- Consortium for Biomedical Research in Cancer, Carlos III Institute of Health, CIBERONC, Madrid, Comunidad de Madrid, Spain
| | - Joaquín Castillo-Iturra
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Teresa Ocaña
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Hardeep Kumari
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Maria Pellisé
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Leticia Moreira
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Liseth Rivero
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Maria Daca-Alvarez
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Oswaldo Ortiz
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Sabela Carballal
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Rebeca Moreira
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Julia Canet-Hermida
- Hereditary Cancer Program, Catalan institute of oncology, IDIBELL, Badalona, Catalunya, Spain
- Consortium for Biomedical Research in Cancer, Carlos III Institute of Health, CIBERONC, Madrid, Comunidad de Madrid, Spain
| | - Marta Pineda
- Hereditary Cancer Program, Catalan institute of oncology, IDIBELL, Badalona, Catalunya, Spain
- Consortium for Biomedical Research in Cancer, Carlos III Institute of Health, CIBERONC, Madrid, Comunidad de Madrid, Spain
| | - Capella Gabriel
- Hereditary Cancer Program, Catalan institute of oncology, IDIBELL, Badalona, Catalunya, Spain
- Consortium for Biomedical Research in Cancer, Carlos III Institute of Health, CIBERONC, Madrid, Comunidad de Madrid, Spain
| | - Georgina Flórez-Grau
- Immunology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
| | - Manel Juan
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Immunology, Servei d'Immunologia. Hospital Clínic de Barcelona, Barcelona, Barcelona, Spain
| | - Daniel Benitez-Ribas
- Immunology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
- Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Francesc Balaguer
- Institut d'Investigacions Biomediques August Pi i Sunyer, Barcelona, Catalunya, Spain
- Gastroenterology, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Barcelona, Spain
- Facultat de Medicina i Ciències de la Salud, Universitat de Barcelona (UB), Barcelona, Spain
| |
Collapse
|
25
|
Rios-Doria E, Parker EU, Kohrn BF, Pike M, Coombes C, Latorre-Esteves E, Reiter DJ, Fredrickson J, Katz R, Swisher EM, Doll KM, Risques RA. TP53 somatic evolution in the normal endometrium of Black and White individuals. Gynecol Oncol 2025; 197:1-10. [PMID: 40250028 DOI: 10.1016/j.ygyno.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/20/2025]
Abstract
BACKGROUND TP53 mutations are the main drivers of aggressive, high-risk endometrial carcinomas commonly diagnosed in Black individuals. However, TP53 mutations have also been identified in benign, non-cancerous tissues. We sought to understand the TP53 mutational landscape in benign endometrium throughout the lifespan of Black and White individuals, accounting for structural socioeconomic context. METHODS Ultra-sensitive TP53 mutation detection was performed with high-depth duplex sequencing (∼13,000×) in DNA extracted from histologically normal endometrium collected at autopsy (69 % of cases) or surgery (31 % of cases) from 83 individuals ages 0 to 81 (31 Black and 52 White, median age 35 years) without endometrial cancer. Histologically normal endometrium was also collected from 10 White individuals with endometrial cancer. RESULTS We identified 266 coding TP53 mutations in the normal endometrium of individuals without endometrial cancer, 57 % of which were pathogenic. The number, pathogenicity, and size of TP53 mutant clones in normal endometrium increased with age. Multivariable models showed no significant association between race or socioeconomic metrics and TP53 mutation frequency in normal endometrium. An exploratory analysis on the histologically normal endometrium of White individuals with endometrial cancer identified the tumor mutations at low levels in the normal biopsy of 5 out of 6 cases. CONCLUSIONS Our study revealed prevalent TP53 somatic evolution in benign endometrium across human lifespan and no racial differences in this cohort of predominantly younger individuals. Future studies should consider the analysis of larger cohorts with older individuals to detect potential effects of racial disparities on TP53 somatic evolution later in life.
Collapse
Affiliation(s)
- Eric Rios-Doria
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Elizabeth U Parker
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Brendan F Kohrn
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Mindy Pike
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Coohleen Coombes
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Elena Latorre-Esteves
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Daniel J Reiter
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Jeanne Fredrickson
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States
| | - Ronit Katz
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Elizabeth M Swisher
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Kemi M Doll
- Department of Obstetrics and Gynecology, University of Washington, Seattle, WA, United States
| | - Rosa Ana Risques
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States.
| |
Collapse
|
26
|
Kaneko R, Kishimoto Y, Ishikawa O, Funahashi N, Koshikawa N. Laminin-γ2-NR6A1 Fusion Protein Promotes Metastatic Potential in Non-Small-Cell Lung Carcinoma Cells without Epidermal Growth Factor Receptor Mutation. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00113-0. [PMID: 40252971 DOI: 10.1016/j.ajpath.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/11/2025] [Indexed: 04/21/2025]
Abstract
Laminin-γ2 fusion gene (Lm-γ2F), formed by translocation between LAMC2 and NR6A1, functions as an epidermal growth factor receptor (EGFR) ligand. However, its expression and impact on cancers beyond the initially studied contexts remain unclear. This study focused on Lm-γ2F protein secretion and its role in non-small-cell lung carcinoma (NSCLC), where EGFR signaling plays a pivotal role in malignancy progression. Lm-γ2F secretion was confirmed in serum-free conditioned medium from six NSCLC cell lines by Western blot analysis and further validated in NCI-H1650 cells. Hypothesizing that Lm-γ2F functions as an EGFR ligand, its effects in NSCLC cells lacking EGFR mutations were explored. In EKVX and RERF-LC-KJ cell lines, Lm-γ2F overexpression significantly enhanced cell growth, survival, motility, and invasiveness through EGFR signaling activation compared with controls. Conversely, no effects were observed in VMRC-LCD cells lacking EGFR expression. Additionally, increased membrane-type 1 matrix metalloproteinase expression was detected in Lm-γ2F-expressing EKVX cells. In vivo, these cells exhibited elevated metastatic activity in a lung metastasis model. These findings suggested that ectopic Lm-γ2F expression contributes to malignant progression in NSCLC cells without EGFR mutations. Furthermore, EGFR tyrosine kinase inhibitors may suppress metastasis in these contexts. This study provides novel insights into the oncogenic role of Lm-γ2F in NSCLC, highlighting its potential as a therapeutic target to mitigate tumor progression and metastasis.
Collapse
Affiliation(s)
- Ryo Kaneko
- Department of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan
| | - Yuri Kishimoto
- Department of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan
| | - Ozora Ishikawa
- Department of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan
| | - Nobuaki Funahashi
- Department of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan.
| | - Naohiko Koshikawa
- Department of Life Science and Technology, Institute of Science Tokyo, Yokohama, Japan; Clinical Cancer Proteomics Laboratory, Kanagawa Cancer Center Research Institute, Yokohama, Japan.
| |
Collapse
|
27
|
Lu D, Zheng Y, Yi X, Hao J, Zeng X, Han L, Li Z, Jiao S, Jiang B, Ai J, Peng J. Identifying potential risk genes for clear cell renal cell carcinoma with deep reinforcement learning. Nat Commun 2025; 16:3591. [PMID: 40234405 PMCID: PMC12000451 DOI: 10.1038/s41467-025-58439-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 03/18/2025] [Indexed: 04/17/2025] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most prevalent type of renal cell carcinoma. However, our understanding of ccRCC risk genes remains limited. This gap in knowledge poses challenges to the effective diagnosis and treatment of ccRCC. To address this problem, we propose a deep reinforcement learning-based computational approach named RL-GenRisk to identify ccRCC risk genes. Distinct from traditional supervised models, RL-GenRisk frames the identification of ccRCC risk genes as a Markov Decision Process, combining the graph convolutional network and Deep Q-Network for risk gene identification. Moreover, a well-designed data-driven reward is proposed for mitigating the limitation of scant known risk genes. The evaluation demonstrates that RL-GenRisk outperforms existing methods in ccRCC risk gene identification. Additionally, RL-GenRisk identifies eight potential ccRCC risk genes. We successfully validated epidermal growth factor receptor (EGFR) and piccolo presynaptic cytomatrix protein (PCLO), corroborated through independent datasets and biological experimentation. This approach may also be used for other diseases in the future.
Collapse
Affiliation(s)
- Dazhi Lu
- AI for Science Interdisciplinary Research Center, School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Yan Zheng
- College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Xianyanling Yi
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Jianye Hao
- College of Intelligence and Computing, Tianjin University, Tianjin, China.
| | - Xi Zeng
- AI for Science Interdisciplinary Research Center, School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Lu Han
- AI for Science Interdisciplinary Research Center, School of Computer Science, Northwestern Polytechnical University, Xi'an, China
| | - Zhigang Li
- College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Shaoqing Jiao
- School of Software, Northwestern Polytechnical University, Xi'an, China
| | - Bei Jiang
- Tianjin Second People's Hospital, Tianjin, China
| | - Jianzhong Ai
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, China.
| | - Jiajie Peng
- AI for Science Interdisciplinary Research Center, School of Computer Science, Northwestern Polytechnical University, Xi'an, China.
- Key Laboratory of Big Data Storage and Management, Northwestern Polytechnical University, Ministry of Industry and Information Technology, Xi'an, China.
| |
Collapse
|
28
|
Espinoza-Ferrao S, Echeverría-Garcés G, Rivera-Orellana S, Bueno-Miño J, Castellanos-Molina E, Benítez-Núñez M, López-Cortés A. Global analysis of actionable genomic alterations in thyroid cancer and precision-based pharmacogenomic strategies. Front Pharmacol 2025; 16:1524623. [PMID: 40297138 PMCID: PMC12034932 DOI: 10.3389/fphar.2025.1524623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Thyroid cancer, a prevalent endocrine malignancy, has an age-standardized incidence rate of 9.1 per 100,000 people and a mortality rate of 0.44 per 100,000 as of 2024. Despite significant advances in precision oncology driven by large-scale international consortia, gaps persist in understanding the genomic landscape of thyroid cancer and its impact on therapeutic efficacy across diverse populations. Methods To address this gap, we performed comprehensive data mining and in silico analyses to identify pathogenic variants in thyroid cancer driver genes, calculate allele frequencies, and assess deleteriousness scores across global populations, including African, Amish, Ashkenazi Jewish, East and South Asian, Finnish and non-Finnish European, Latino, and Middle Eastern groups. Additionally, pharmacogenomic profiling, in silico drug prescription, and clinical trial data were analyzed to prioritize targeted therapeutic strategies. Results Our analysis examined 56,622 variants in 40 thyroid cancer-driver genes across 76,156 human genomes, identifying 5,001 known and predicted oncogenic variants. Enrichment analysis revealed critical pathways such as MAPK, PI3K-AKT-mTOR, and p53 signaling, underscoring their roles in thyroid cancer pathogenesis. High-throughput validation strategies confirmed actionable genomic alterations in RET, BRAF, NRAS, KRAS, and EPHA7. Ligandability assessments identified these proteins as promising therapeutic targets. Furthermore, our findings highlight the clinical potential of targeted drug inhibitors, including vandetanib, dabrafenib, and selumetinib, for improving treatment outcomes. Discussion This study underscores the significance of integrating genomic insights with pharmacogenomic strategies to address disparities in thyroid cancer treatment. The identification of population-specific oncogenic variants and actionable therapeutic targets provides a foundation for advancing precision oncology. Future efforts should focus on including underrepresented populations, developing population-specific prevention strategies, and fostering global collaboration to ensure equitable access to pharmacogenomic testing and innovative therapies. These initiatives have the potential to transform thyroid cancer care and align with the broader goals of personalized medicine.
Collapse
Affiliation(s)
| | - Gabriela Echeverría-Garcés
- Centro de Referencia Nacional de Genómica, Secuenciación y Bioinformática, Instituto Nacional de Investigación en Salud Pública “Leopoldo Izquieta Pérez”, Quito, Ecuador
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Santiago, Chile
| | | | - José Bueno-Miño
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | | | - Melanie Benítez-Núñez
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | - Andrés López-Cortés
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| |
Collapse
|
29
|
Seyfried TN, Lee DC, Duraj T, Ta NL, Mukherjee P, Kiebish M, Arismendi-Morillo G, Chinopoulos C. The Warburg hypothesis and the emergence of the mitochondrial metabolic theory of cancer. J Bioenerg Biomembr 2025:10.1007/s10863-025-10059-w. [PMID: 40199815 DOI: 10.1007/s10863-025-10059-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
Otto Warburg originally proposed that cancer arose from a two-step process. The first step involved a chronic insufficiency of mitochondrial oxidative phosphorylation (OxPhos), while the second step involved a protracted compensatory energy synthesis through lactic acid fermentation. His extensive findings showed that oxygen consumption was lower while lactate production was higher in cancerous tissues than in non-cancerous tissues. Warburg considered both oxygen consumption and extracellular lactate as accurate markers for ATP production through OxPhos and glycolysis, respectively. Warburg's hypothesis was challenged from findings showing that oxygen consumption remained high in some cancer cells despite the elevated production of lactate suggesting that OxPhos was largely unimpaired. New information indicates that neither oxygen consumption nor lactate production are accurate surrogates for quantification of ATP production in cancer cells. Warburg also did not know that a significant amount of ATP could come from glutamine-driven mitochondrial substrate level phosphorylation in the glutaminolysis pathway with succinate produced as end product, thus confounding the linkage of oxygen consumption to the origin of ATP production within mitochondria. Moreover, new information shows that cytoplasmic lipid droplets and elevated aerobic lactic acid fermentation are both biomarkers for OxPhos insufficiency. Warburg's original hypothesis can now be linked to a more complete understanding of how OxPhos insufficiency underlies dysregulated cancer cell growth. These findings can also address several questionable assumptions regarding the origin of cancer thus allowing the field to advance with more effective therapeutic strategies for a less toxic metabolic management and prevention of cancer.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA.
| | - Derek C Lee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Tomas Duraj
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Nathan L Ta
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | - Purna Mukherjee
- Biology Department, Boston College, 140 Commonwealth Ave, Chestnut Hill, Boston, MA, 02467, USA
| | | | - Gabriel Arismendi-Morillo
- Facultad de Medicina, Instituto de Investigaciones Biológicas, Universidad del Zulia, Maracaibo, Venezuela
- Department of Medicine, Faculty of Health Sciences, University of Deusto, Bilbao (Bizkaia), Spain
| | - Christos Chinopoulos
- Department of Medical Biochemistry, Semmelweis University, Budapest, 1094, Hungary
| |
Collapse
|
30
|
Wu Z, Li Y, Dong J, Qin JJ. An updated review on the role of small molecules in mediating protein degradation. Eur J Med Chem 2025; 287:117370. [PMID: 39933402 DOI: 10.1016/j.ejmech.2025.117370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Targeted protein degradation (TPD) technologies, inspired by physiological processes, have recently provided new directions for drug development. Unlike conventional drug development focusing on targeting the active sites of disease-related proteins, TPD can utilize any nook or cranny of a protein to drive degradation through the cell's inherent destruction mechanism. It offers various advantages such as stronger pharmacological effects, an expanded range of drug targets, and higher selectivity. Based on the ubiquitin-proteasome system and the lysosomal degradation pathway, a variety of TPD strategies have been developed including PROTAC, PROTAB, and AUTOTAC. These TPD strategies have continuously enriched the toolbox for targeted protein degradation and expanded the scope of application, providing new ideas for biological research and drug discovery. This review attempts to introduce up-to-date research progress in the TPD strategies, focusing mainly on their design concepts, advantages, potential applications, and challenges, which may provide some inspiration for drug design, drug discovery, and clinical application for biologists and chemists.
Collapse
Affiliation(s)
- Zumei Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yulong Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jinyun Dong
- Center for Innovative Drug Research, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| | - Jiang-Jiang Qin
- Center for Innovative Drug Research, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| |
Collapse
|
31
|
Jenkins JW, Peña A, Castro SA, Hansen MJ, Van Keulen VP, Foster ST, Rios-Cruz PE, Yakubov J, Hinson DT, Olivier SM, Pavelko KD, Felts SJ, Johnson AJ, Pease LR. MHC class II-mediated spontaneous rejection of breast carcinomas expressing model neoantigens. J Immunother Cancer 2025; 13:e010434. [PMID: 40187751 PMCID: PMC11973762 DOI: 10.1136/jitc-2024-010434] [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: 08/23/2024] [Accepted: 03/06/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Cancers persist despite expression of immunogenic neoantigens and ongoing antitumor immune responses. While some occult tumors likely are cleared by effective antitumor immune responses, the targeted antigens are not easily identifiable as those tumors spontaneously disappear. METHODS We used mouse models with a defined antigenic protein mimicking tumor-specific neoantigens to address the nature of these spontaneous anti-tumor immune responses. RESULTS BALB/c (H-2d ) mice challenged with BALB/c breast tumors expressing the rat-erbB2 oncoprotein succumb to their tumors despite ongoing immune responses targeting tumor-specific model antigens. Meanwhile, congenic BALB.B (H-2b ) and H-2d/H-2b F1 hybrid mice spontaneously eliminate genetically matched tumors in a major histocompatibility complex (MHC)-II dependent manner. Adoptive transfer and immune cell depletion strategies revealed CD4+ T cells and CD20+ B cells are crucial mediators of the protective response in H-2b mice. Furthermore, passive transfer of immune serum from mice rejecting their tumors confers resistance in tumor antigen-tolerant animals with an inversely proportional relationship between tumor outgrowth and the amount of rat-erbB2 specific antibody present in tumor-bearing mice. Introduction of the rat-erb2 ectodomain into other H-2b tumor models also promotes their spontaneous tumor rejection. Notably, the tumor microenvironments differ in rat-erbB2+ tumor-bearing BALB.B and BALB/c mice at the time of fate decision in the models reflecting the differences between effective and ineffective tumor immune responses. CONCLUSIONS We find that the effective antitumor immunity targeting neoantigens in these breast cancer models is determined by MHC-II-restricted presentation of optimal cancer-associated antigens. These responses are dependent on CD4+ T cells, B cells, and antigen-specific antibodies.
Collapse
Affiliation(s)
| | - Alvaro Peña
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sarah A Castro
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael J Hansen
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Sean T Foster
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Joshua Yakubov
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Destin T Hinson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Samuel M Olivier
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kevin D Pavelko
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Sara J Felts
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Larry R Pease
- Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
32
|
Lan Y, Xia Z, Shao Q, Lin P, Lu J, Xiao X, Zheng M, Chen D, Dou Y, Xie Q. Synonymous mutations promote tumorigenesis by disrupting m 6A-dependent mRNA metabolism. Cell 2025; 188:1828-1841.e15. [PMID: 39952247 DOI: 10.1016/j.cell.2025.01.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 11/04/2024] [Accepted: 01/17/2025] [Indexed: 02/17/2025]
Abstract
Cancer cells acquire numerous mutations during tumorigenesis, including synonymous mutations that do not change the amino acid sequence of a protein. RNA N6-methyladenosine (m6A) is a post-transcriptional modification that plays critical roles in oncogenesis. Herein, we identified 12,849 mutations in the cancer genome with the potential to perturb m6A modification patterns, which we refer to as "m6A disruption mutations (m6A-DMs)." These are either synonymous m6A-DMs (sm6A-DMs) or missense m6A-DMs (mm6A-DMs) mutations, and the former is enriched within tumor suppressor genes, such as CDKN2A and BRCA2. Using epitranscriptomic editing, we demonstrate that manipulating m6A levels at specific sm6A-DM sites influences mRNA stability. Furthermore, introducing CDKN2A sm6A-DMs into cancer cells promotes tumor growth while BRCA2 sm6A-DMs sensitize tumors to the poly (ADP-ribose) polymerase inhibitor (PARPi) treatment. Our findings demonstrate sm6A-DMs as potential oncogenic drivers, unveiling implications for synonymous mutations in tumorigenesis and beyond.
Collapse
Affiliation(s)
- Yiheng Lan
- Westlake Disease Modeling Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Zhen Xia
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Qizhe Shao
- Center for Regeneration and Cell Therapy of Zhejiang University, University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Peng Lin
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Jinhong Lu
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Fudan University, Shanghai 200433, China
| | - Xiaoying Xiao
- Center for Regeneration and Cell Therapy of Zhejiang University, University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China
| | - Mengyue Zheng
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China
| | - Di Chen
- Center for Regeneration and Cell Therapy of Zhejiang University, University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China.
| | - Yanmei Dou
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China.
| | - Qi Xie
- Westlake Disease Modeling Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang 310024, China; School of Life Sciences, Westlake University, Hangzhou, Zhejiang 310024, China.
| |
Collapse
|
33
|
Li Y, Xu T, Ma H, Yue D, Lamao Q, Liu Y, Zhou Z, Wei W. Functional profiling of serine, threonine and tyrosine sites. Nat Chem Biol 2025; 21:532-543. [PMID: 39313591 DOI: 10.1038/s41589-024-01731-0] [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: 12/04/2023] [Accepted: 08/14/2024] [Indexed: 09/25/2024]
Abstract
Systematic perturbation of amino acids at endogenous loci provides diverse insights into protein function. Here, we performed a genome-wide screen to globally assess the cell fitness dependency of serine, threonine and tyrosine residues. Using an adenine base editor, we designed a whole-genome library comprising 817,089 single guide RNAs to perturb 584,337 S, T and Y sites. We identified 3,467 functional substitutions affecting cell fitness and 677 of them involving phosphorylation, including numerous phosphorylation-mediated gain-of-function substitutions that regulate phosphorylation levels of itself or downstream factors. Furthermore, our findings highlight that specific substitution types, notably serine to proline, are crucial for maintaining domain structure broadly. Lastly, we demonstrate that 309 enriched hits capable of initiating cell overproliferation might be potential cancer driver mutations. This study represents an extensive functional profiling of S, T and Y residues and provides insights into the distinctive roles of these amino acids in biological mechanisms and tumor progression.
Collapse
Affiliation(s)
- Yizhou Li
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Tao Xu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Huazheng Ma
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, China
| | - Di Yue
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Qiezhong Lamao
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Ying Liu
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- Changping Laboratory, Beijing, China
| | - Zhuo Zhou
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Suzhou, China
| | - Wensheng Wei
- Biomedical Pioneering Innovation Center, Beijing Advanced Innovation Center for Genomics, Peking-Tsinghua Center for Life Sciences, Peking University Genome Editing Research Center, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China.
- Changping Laboratory, Beijing, China.
| |
Collapse
|
34
|
Dinh KN, Vázquez-García I, Chan A, Malhotra R, Weiner A, McPherson AW, Tavaré S. CINner: Modeling and simulation of chromosomal instability in cancer at single-cell resolution. PLoS Comput Biol 2025; 21:e1012902. [PMID: 40179124 PMCID: PMC11990800 DOI: 10.1371/journal.pcbi.1012902] [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: 06/03/2024] [Revised: 04/11/2025] [Accepted: 02/24/2025] [Indexed: 04/05/2025] Open
Abstract
Cancer development is characterized by chromosomal instability, manifesting in frequent occurrences of different genomic alteration mechanisms ranging in extent and impact. Mathematical modeling can help evaluate the role of each mutational process during tumor progression, however existing frameworks can only capture certain aspects of chromosomal instability (CIN). We present CINner, a mathematical framework for modeling genomic diversity and selection during tumor evolution. The main advantage of CINner is its flexibility to incorporate many genomic events that directly impact cellular fitness, from driver gene mutations to copy number alterations (CNAs), including focal amplifications and deletions, missegregations and whole-genome duplication (WGD). We apply CINner to find chromosome-arm selection parameters that drive tumorigenesis in the absence of WGD in chromosomally stable cancer types from the Pan-Cancer Analysis of Whole Genomes (PCAWG, [Formula: see text]). We found that the selection parameters predict WGD prevalence among different chromosomally unstable tumors, hinting that the selective advantage of WGD cells hinges on their tolerance for aneuploidy and escape from nullisomy. Analysis of inference results using CINner across cancer types in The Cancer Genome Atlas ([Formula: see text]) further reveals that the inferred selection parameters reflect the bias between tumor suppressor genes and oncogenes on specific genomic regions. Direct application of CINner to model the WGD proportion and fraction of genome altered (FGA) in PCAWG uncovers the increase in CNA probabilities associated with WGD in each cancer type. CINner can also be utilized to study chromosomally stable cancer types, by applying a selection model based on driver gene mutations and focal amplifications or deletions (chronic lymphocytic leukemia in PCAWG, [Formula: see text]). Finally, we used CINner to analyze the impact of CNA probabilities, chromosome selection parameters, tumor growth dynamics and population size on cancer fitness and heterogeneity. We expect that CINner will provide a powerful modeling tool for the oncology community to quantify the impact of newly uncovered genomic alteration mechanisms on shaping tumor progression and adaptation.
Collapse
Affiliation(s)
- Khanh N. Dinh
- Irving Institute for Cancer Dynamics, Columbia University, New York, New York, United States of America
- Department of Statistics, Columbia University, New York, New York, United States of America
| | - Ignacio Vázquez-García
- Irving Institute for Cancer Dynamics, Columbia University, New York, New York, United States of America
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Department of Pathology, Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, United States of America
| | - Andrew Chan
- Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Rhea Malhotra
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Stanford University, Palo Alto, California, United States of America
| | - Adam Weiner
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
- Tri-Institutional PhD Program in Computational Biology and Medicine, Weill Cornell Medicine, New York, New York, United States of America
| | - Andrew W. McPherson
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, United States of America
| | - Simon Tavaré
- Irving Institute for Cancer Dynamics, Columbia University, New York, New York, United States of America
- Department of Statistics, Columbia University, New York, New York, United States of America
| |
Collapse
|
35
|
Al-Marrawi M, Petreaca RC, Bouley RA. In silico protein structural analysis of PRMT5 and RUVBL1 mutations arising in human cancers. Cancer Genet 2025; 292-293:49-56. [PMID: 39874873 DOI: 10.1016/j.cancergen.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 01/30/2025]
Abstract
DNA double strand breaks (DSBs) can be generated spontaneously during DNA replication and are repaired primarily by Homologous Recombination (HR). However, efficient repair requires chromatin remodeling to allow the recombination machinery access to the break. TIP60 is a complex conserved from yeast to humans that is required for histone acetylation and modulation of HR activity at DSBs. Two enzymatic activities within the TIP60 complex, KAT5 (a histone acetyltransferase) and RUVBL1 (an AAA+ ATPase) are required for efficient HR repair. Post-translational modification of RUVBL1 by the PRMT5 methyltransferase activates the complex acetyltransferase activity and facilitates error free HR repair. In S. pombe a direct interaction between PRMT5 and the acetyltransferase subunit of the TIP60 complex (KAT5) was also identified. The TIP60 complex has been partially solved experimentally in both humans and S. cerevisiae, but not S. pombe. Here, we used in silico protein structure analysis to investigate structural conservation between S. pombe and human PRMT5 and RUVBL1. We found that there is more similarity in structure conservation between S. pombe and human proteins than between S. cerevisiae and human. Next, we queried the COSMIC database to analyze how mutations occurring in human cancers affect the structure and function of these proteins. Artificial intelligence algorithms that predict how likely mutations are to promote cellular transformation and immortalization show that RUVBL1 mutations should have a more drastic effect than PRMT5. Indeed, in silico protein structural analysis shows that PRMT5 mutations are less likely to destabilize enzyme function. Conversely, most RUVBL1 mutations occur in a region required for interaction with its partner (RUVBL2). These data suggests that cancer mutations could destabilize the TIP60 complex. Sequence conservation analysis between S. pombe and humans shows that the residues identified in cancer cells are highly conserved, suggesting that this may be an essential process in eukaryotic DSB repair. These results shed light on mechanisms of DSB repair and also highlight how S. pombe remains a great model system for analyzing DSB repair processes that are tractable in human cells.
Collapse
Affiliation(s)
- Majd Al-Marrawi
- Neuroscience Undergraduate Program, The Ohio State University, USA
| | - Ruben C Petreaca
- Department of Molecular Genetics, The Ohio State University, Marion, USA; Cancer Biology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, USA
| | - Renee A Bouley
- Department of Chemistry and Biochemistry, The Ohio State University, Marion, USA.
| |
Collapse
|
36
|
An M, Davis JR, Levy JM, Serack FE, Harvey JW, Brauer PP, Pirtle CP, Berríos KN, Newby GA, Yeh WH, Kamath N, Mortberg M, Lian Y, Howard M, DeSouza-Lenz K, Guzman K, Thai A, Graffam S, Laversenne V, Coffey AA, Frei J, Pierce SE, Safar JG, Deverman BE, Minikel EV, Vallabh SM, Liu DR. In vivo base editing extends lifespan of a humanized mouse model of prion disease. Nat Med 2025; 31:1319-1328. [PMID: 39810005 PMCID: PMC12003183 DOI: 10.1038/s41591-024-03466-w] [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: 08/20/2024] [Accepted: 12/12/2024] [Indexed: 01/16/2025]
Abstract
Prion disease is a fatal neurodegenerative disease caused by the misfolding of prion protein (PrP) encoded by the PRNP gene. While there is currently no cure for the disease, depleting PrP in the brain is an established strategy to prevent or stall templated misfolding of PrP. Here we developed in vivo cytosine and adenine base strategies delivered by adeno-associated viruses to permanently modify the PRNP locus to achieve PrP knockdown in the mouse brain. Systemic injection of dual-adeno-associated virus PHP.eB encoding BE3.9max and single guide RNA installing PRNP R37X resulted in 37% average installation of the desired edit, 50% reduction of PrP in the mouse brain and 52% extension of lifespan in transgenic human PRNP mice inoculated with pathogenic human prion isolates representing the most common sporadic and genetic subtypes of prion disease. We further engineered base editing systems to achieve improved in vivo potency and reduced base editor expression in nontargeting tissues, resulting in 63% average PrP reduction in the mouse brain from a 6.7-fold lower viral dose, with no detected off-target editing of anticipated clinical significance observed in either human cells or mouse tissues. These findings support the potential of in vivo base editing as one-time treatment for prion disease.
Collapse
Affiliation(s)
- Meirui An
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Jessie R Davis
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Jonathan M Levy
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Fiona E Serack
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - John W Harvey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Pamela P Brauer
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Catherine P Pirtle
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kiara N Berríos
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Gregory A Newby
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Wei-Hsi Yeh
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Nikita Kamath
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Meredith Mortberg
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yuan Lian
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Michael Howard
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Kenia Guzman
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Aaron Thai
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Samantha Graffam
- Comparative Medicine, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vanessa Laversenne
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Alissa A Coffey
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jeannine Frei
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sarah E Pierce
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA
| | - Jiri G Safar
- Case Western Reserve University, Cleveland, OH, USA
| | - Benjamin E Deverman
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Eric Vallabh Minikel
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- McCance Center for Brain Health and Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Prion Alliance, Cambridge, MA, USA.
| | - Sonia M Vallabh
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- McCance Center for Brain Health and Department of Neurology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Prion Alliance, Cambridge, MA, USA.
| | - David R Liu
- Merkin Institute of Transformative Technologies in Healthcare, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
37
|
Su X, Lin Q, Liu B, Zhou C, Lu L, Lin Z, Si J, Ding Y, Duan S. The promising role of nanopore sequencing in cancer diagnostics and treatment. CELL INSIGHT 2025; 4:100229. [PMID: 39995512 PMCID: PMC11849079 DOI: 10.1016/j.cellin.2025.100229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 02/26/2025]
Abstract
Cancer arises from genetic alterations that impact both the genome and transcriptome. The utilization of nanopore sequencing offers a powerful means of detecting these alterations due to its unique capacity for long single-molecule sequencing. In the context of DNA analysis, nanopore sequencing excels in identifying structural variations (SVs), copy number variations (CNVs), gene fusions within SVs, and mutations in specific genes, including those involving DNA modifications and DNA adducts. In the field of RNA research, nanopore sequencing proves invaluable in discerning differentially expressed transcripts, uncovering novel elements linked to transcriptional regulation, and identifying alternative splicing events and RNA modifications at the single-molecule level. Furthermore, nanopore sequencing extends its reach to detecting microorganisms, encompassing bacteria and viruses, that are intricately associated with tumorigenesis and the development of cancer. Consequently, the application prospects of nanopore sequencing in tumor diagnosis and personalized treatment are expansive, encompassing tasks such as tumor identification and classification, the tailoring of treatment strategies, and the screening of prospective patients. In essence, this technology stands poised to unearth novel mechanisms underlying tumorigenesis while providing dependable support for the diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Xinming Su
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Qingyuan Lin
- The Second Clinical Medical College, Zhejiang Chinese Medicine University BinJiang College, Hangzhou 310053, Zhejiang, China
| | - Bin Liu
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Chuntao Zhou
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Liuyi Lu
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Zihao Lin
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Jiahua Si
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Yuemin Ding
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
- Institute of Translational Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| | - Shiwei Duan
- Department of Clinical Medicine, School of Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
- Institute of Translational Medicine, Hangzhou City University, Hangzhou 310015, Zhejiang, China
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Hangzhou City University, Hangzhou 310015, Zhejiang, China
| |
Collapse
|
38
|
Li J, Wang C, Yang C, Bao H, Li N, Huang X, Gong W, Hong X, Yin JC, Pang J, Gan M, Yuan D. Identification of clinicopathological-specific driver gene and genetic subtyping of colorectal cancer. Cancer Sci 2025; 116:1068-1081. [PMID: 39797621 PMCID: PMC11967266 DOI: 10.1111/cas.16432] [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: 08/26/2024] [Revised: 11/18/2024] [Accepted: 11/30/2024] [Indexed: 01/13/2025] Open
Abstract
This study analyzed targeted sequencing data from 6530 tissue samples from patients with metastatic Chinese colorectal cancer (CRC) to identify low mutation frequency and subgroup-specific driver genes, using three algorithms for overall CRC as well as across different clinicopathological subgroups. We analyzed 425 cancer-related genes, identifying 101 potential driver genes, including 36 novel to CRC. Notably, some genes demonstrated subgroup specificity; for instance, ERBB4 was found as a male-specific driver gene and mutations of ERBB4 only influenced the prognosis of male patients with CRC. This sex disparity of ERBB4 was validated in an independent large-scale Memorial Sloan Kettering Cancer Center CRC cohort with 2444 samples. Furthermore, using network-based stratification based on protein-protein interaction, we classified the microsatellite stable (MSS) and unstable (MSI) CRCs into six and three major subtypes, respectively, each showing unique phenotypes and prognoses. In MSS CRC, cluster 5 (APCAMER1-KRAS) and cluster 2 (RNF43-BRAF-PIK3CA) were predominant, and cluster 5 showed a superior overall survival compared with cluster 2. This extensive heterogeneity in driver gene mutations underscores the complexity of CRC and suggests significant implications for treatment and prognostic assessments.
Collapse
Affiliation(s)
- Jianjiong Li
- Department of Colorectal and Anal SurgeryNingbo No. 2 HospitalNingboChina
| | - Chunnian Wang
- Department of PathologyNingbo Diagnostic Pathology CenterNingboChina
| | - Changshun Yang
- Department of Surgical OncologyShengli Clinical Medical College of Fujian Medical UniversityFuzhouChina
| | - Hua Bao
- Nanjing Geneseeq Technology Inc.NanjingChina
| | - Ningyou Li
- Nanjing Geneseeq Technology Inc.NanjingChina
| | - Xianqiang Huang
- Department of SurgeryQuanzhou Guangqian HospitalQuanzhouChina
| | - Wei Gong
- Department of Radiation OncologyQuanzhou Guangqian HospitalQuanzhouChina
| | - Xinyue Hong
- Nanjing Geneseeq Technology Inc.NanjingChina
| | | | | | - Meifu Gan
- Department of PathologyTaizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical UniversityWenzhouChina
| | - Danping Yuan
- Department of colorectal surgeryThe First Affiliated Hospital of Ningbo UniversityNingboChina
| |
Collapse
|
39
|
Zaidh SM, Vengateswaran HT, Habeeb M, Aher KB, Bhavar GB, Irfan N, Lakshmi KNVC. Network pharmacology and AI in cancer research uncovering biomarkers and therapeutic targets for RALGDS mutations. Sci Rep 2025; 15:10938. [PMID: 40157967 PMCID: PMC11954960 DOI: 10.1038/s41598-025-91568-x] [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: 09/13/2024] [Accepted: 02/21/2025] [Indexed: 04/01/2025] Open
Abstract
The lack of target therapies is accountable for the higher mortality of various types of cancer. To address this issue, we selected a target mutated Kirsten rat sarcoma virus oncogene homologue, which plays a significant role in various cancers. Our study aims to identify selective biomarkers and develop diagnostic and therapeutic strategies for KRAS-associated genes using artificial intelligence. Initially, Genomic data, cancer epidemiology, proteomics network interactions, and omics enrichment were analyzed. Structured E-pharmacophore model aided in capturing the binding cavity using eraser algorithms and fabricating a new selective lead compound for the KRSA. The selective molecule was abridged inside the binding cavity and stability was validated through 100 ns molecular dynamics simulations. Epidemiological-neural network studies indicated KRAS mutations leads 40 types of cancer, exclusively pancreatic and colorectal cancers, with diploid and missense mutations as primary factors. Pathway analysis highlighted the involvement of the MAPK and RAS signaling pathways in cancer development and proteomics analysis identified RALGDS as a key protein. Protein-based pharmacophore analysis mapped the biologically active features such as donor, acceptor and aromatic ring with the designed ligands. The results of interaction interpretation illustrate that the amino acid Tyr566 formed an H-bond interaction with the amine group of the octyl ring system and 20 amino acids crafted to properly orient the molecule to fit inside the polar cavity of KRAS protein. The MMGBSA score of - 53.33 kcal/mol conformed to the well-configured binding with KRSA and realistic model simulation exposed the π-π, π-cationic and hydrophobic interactions stabilised the molecule inside the KRSA protein throughout 100 ns simulation. The study demonstrates the vitality of AI and network pharmacology to identify potential-target biomarkers for KRAS-associated genes, paving the way for improved cancer diagnostics and therapeutics.
Collapse
Affiliation(s)
- S Mohammed Zaidh
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India
- D3 Drug Tech Lab Pvt Ltd, Chennai, 600048, India
| | | | - Mohammad Habeeb
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India
| | - Kiran Balasaheb Aher
- Department of Pharmaceutical Quality Assurance, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra, 424001, India
| | - Girija Balasaheb Bhavar
- Department of Pharmaceutical Chemistry, Shri Vile Parle Kelavani Mandal's Institute of Pharmacy, Dhule, Maharashtra, 424001, India
| | - N Irfan
- Crescent School of Pharmacy, BS Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600048, India.
| | - K N V Chenchu Lakshmi
- Department of Pharmaceutical Chemistry, KL College of Pharmacy, Koneru Lakshmaiah Education Foundation, Green Fields, Vaddeswaram, A.P, 522302, India
| |
Collapse
|
40
|
Bahari F, Ahangari Cohan R, Montazeri H. Element-specific estimation of background mutation rates in whole cancer genomes through transfer learning. NPJ Precis Oncol 2025; 9:92. [PMID: 40155429 PMCID: PMC11953285 DOI: 10.1038/s41698-025-00871-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
Mutational burden tests are essential for detecting signals of positive selection in cancer driver discovery by comparing observed mutation rates with background mutation rates (BMRs). However, accurate BMR estimation is challenging due to the diversity of mutational processes across genomes, complicating driver discovery efforts. Existing methods rely on various genomic regions and features for BMR estimation but lack a model that integrates both intergenic intervals and functional genomic elements on a comprehensive set of genomic features. Here, we introduce eMET (element-specific Mutation Estimator with boosted Trees), which employs 1372 (epi)genomic features from intergenic data and fine-tunes it with element-specific data through transfer learning. Applied to PCAWG somatic mutations, eMET significantly improves BMR accuracy and has potential to enhance driver discovery. Additionally, we provide an extensive analysis of BMR estimation, examining different machine learning models, genomic interval strategies, feature categories, and dimensionality reduction techniques.
Collapse
Affiliation(s)
- Farideh Bahari
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Reza Ahangari Cohan
- Department of Nanobiotechnology, New Technologies Research Group, Pasteur Institute of Iran, Tehran, Iran.
| | - Hesam Montazeri
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
41
|
Cui R, Wang G, Hu R, Wang Y, Mu H, Song Y, Chen B, Jiang X. Prognostic and immunotherapeutic potential of disulfidptosis-associated signature in pancreatic cancer. Front Immunol 2025; 16:1568976. [PMID: 40207217 PMCID: PMC11979277 DOI: 10.3389/fimmu.2025.1568976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 03/10/2025] [Indexed: 04/11/2025] Open
Abstract
Disulfidptosis is a newly discovered formation of programmed cell death. However, the significance of disulfidptosis in pancreatic adenocarcinoma remains unclear. Our investigation aims to elucidate the significance of disulfidptosis in pancreatic ductal adenocarcinoma by integrating diverse datasets, including bulk RNA sequencing data, microarray profiles, single-cell transcriptome profiles, spatial transcriptome data, and biospecimens. Utilizing various bioinformatics tools, we screened disulfidptosis-related genes based on single-cell RNA sequencing profiles, subsequently validating them through enrichment analysis. An 8-gene disulfidptosis-related prognostic signature was established by constructing massive LASSO-Cox regression models and validated by multiple external PDAC cohorts. Evaluation methods, such as Kaplan-Meier curves, ROC curves, time-dependent ROC curves, and decision curve analysis, were employed to assess the prognostic signature's reliability. High disulfidptosis-related scores were associated with a poorer prognosis and diminished sensitivity to immune checkpoint blockade. Further investigation uncovered that the potential components of elevated DPS involve malignant tumor hallmarks, extensive interactions between myCAFs and tumor cells, and the exclusion of immune cells. Cell-cell communication analysis highlighted myCAFs' role in signaling, potentially influencing tumor cells towards increased malignancy through collagen, laminin, and FN1 signaling networks. Spatial transcriptome analysis confirmed the crosstalk between myCAFs and tumor cells. Biospecimens including 20 pairs of PDAC samples and adjacent normal tissues further demonstrated the robustness of DPS and its correlation with CAF markers. In conclusion, our study introduces a novel disulfidptosis-related signature with high efficacy in patient risk stratification, which has the ability to predict the sensitivity to immune checkpoint blockade.
Collapse
Affiliation(s)
- Ran Cui
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Gaoming Wang
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Renhao Hu
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yongkun Wang
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Huiling Mu
- Department of Biobank, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yanxiang Song
- Department of Biobank, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Bo Chen
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xiaohua Jiang
- Department of Hepatopancreatobiliary Surgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
42
|
Pinto RJ, Ferreira D, Salamanca P, Miguel F, Borges P, Barbosa C, Costa V, Lopes C, Santos LL, Pereira L. Coding and regulatory somatic profiling of triple-negative breast cancer in Sub-Saharan African patients. Sci Rep 2025; 15:10325. [PMID: 40133516 PMCID: PMC11937512 DOI: 10.1038/s41598-025-94707-6] [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: 09/20/2024] [Accepted: 03/17/2025] [Indexed: 03/27/2025] Open
Abstract
The burden of triple-negative breast cancer (TNBC) may be shaped by genetic factors, particularly inherited and somatic mutation profiles. However, data on this topic remain limited, especially for the African continent, where a higher TNBC incidence is observed. In the age of precision medicine, cataloguing TNBC diversity in African patients becomes imperative. We performed whole exome sequencing, including untranslated regions, on 30 samples from Angola and Cape Verde, which allowed to ascertain on potential regulatory mutations in TNBC for the first time. A high somatic burden was observed for the African cohort, with 86% of variants being so far unreported. Recurring to predictive functional algorithms, 17% of the somatic single nucleotide variants were predicted to be deleterious at the protein level, and 20% overlapped with candidate cis-regulatory elements controlling gene expression. Several of these somatic functionally-impactful mutations and copy number variation (mainly in 1q, 8q, 6 and 10p) occur in known BC- and all cancer-driver genes, enriched for several cancer mechanisms, including response to radiation and related DNA repair mechanisms. TP53 is the top of these known BC-driver genes, but our results identified possible novel TNBC driver genes that may play a main role in the African context, as TTN, CEACAM7, DEFB132, COPZ2 and GAS1. These findings emphasize the need to expand cancer omics screenings across the African continent, the region of the globe with highest genomic diversity, accelerating the discovery of new somatic mutations and cancer-related pathways.
Collapse
Affiliation(s)
- Ricardo J Pinto
- i3S, Instituto de Investigação e Inovação Em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP, Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Dylan Ferreira
- Research Center of IPO-Porto (CI-IPOP) / RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center (P.CCC) Raquel Seruca, Porto, Portugal
| | | | | | - Pamela Borges
- Hospital Universitário Agostinho Neto, Praia, Cabo Verde
| | - Carla Barbosa
- Hospital Universitário Agostinho Neto, Praia, Cabo Verde
| | - Vitor Costa
- Hospital Universitário Agostinho Neto, Praia, Cabo Verde
| | - Carlos Lopes
- Unilabs | Laboratório Anatomia Patológica, Porto, Portugal
| | - Lúcio Lara Santos
- Research Center of IPO-Porto (CI-IPOP) / RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO-Porto) / Porto Comprehensive Cancer Center (P.CCC) Raquel Seruca, Porto, Portugal
- FP-I3ID, University Fernando Pessoa, Porto, Portugal
- Department of Surgical Oncology, Portuguese Oncology Institute of Porto, Porto, Portugal
- School of Medicine and Biomedical Sciences, University Fernando Pessoa, Gondomar, Portugal
| | - Luisa Pereira
- i3S, Instituto de Investigação e Inovação Em Saúde, Universidade do Porto, Porto, Portugal.
- IPATIMUP, Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal.
| |
Collapse
|
43
|
Cen X, Lan Y, Zou J, Chen R, Hu C, Tong Y, Zhang C, Chen J, Wang Y, Zhou R, He W, Lu T, Dubee F, Jovic D, Dong W, Gao Q, Ma M, Lu Y, Xue Y, Cheng X, Li Y, Yang H. Pan-cancer analysis shapes the understanding of cancer biology and medicine. Cancer Commun (Lond) 2025. [PMID: 40120098 DOI: 10.1002/cac2.70008] [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: 09/10/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in multi-omics datasets and analytical methods have revolutionized cancer research, offering a comprehensive, pan-cancer perspective. Pan-cancer studies identify shared mechanisms and unique traits across different cancer types, which are reshaping diagnostic and treatment strategies. However, continued innovation is required to refine these approaches and deepen our understanding of cancer biology and medicine. This review summarized key findings from pan-cancer research and explored their potential to drive future advancements in oncology.
Collapse
Affiliation(s)
- Xiaoping Cen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
| | - Yuanyuan Lan
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Jiansheng Zou
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Ruilin Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, P. R. China
| | - Can Hu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yahan Tong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Chen Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Jingyue Chen
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yuanmei Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Run Zhou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Weiwei He
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Tianyu Lu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Fred Dubee
- BGI Research, Shenzhen, Guangdong, P. R. China
| | | | - Wei Dong
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- Clin Lab, BGI Genomics, Beijing, P. R. China
| | - Qingqing Gao
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
- BGI Research, Shenzhen, Guangdong, P. R. China
| | - Man Ma
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences (CAS), Hangzhou, Zhejiang, P. R. China
| | - Youyong Lu
- Laboratory of Molecular Oncology, Peking University Cancer Hospital and Institute, Beijing, P. R. China
| | - Yu Xue
- MOE Key Laboratory of Molecular Biophysics, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Center for Artificial Intelligence Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, P. R. China
| | - Xiangdong Cheng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, P. R. China
| | - Yixue Li
- Guangzhou National Laboratory, Guangzhou, Guangdong, P. R. China
- GZMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, Guangdong, P. R. China
| | - Huanming Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, P. R. China
- BGI, Shenzhen, Guangdong, P. R. China
- James D. Watson Institute of Genome Sciences, Hangzhou, Zhejiang, P. R. China
| |
Collapse
|
44
|
Martín R, Gaitán N, Torrents D. Protocol for the assessment, improvement, and harmonization of somatic variant calling using ONCOLINER. STAR Protoc 2025; 6:103533. [PMID: 39708326 PMCID: PMC11731501 DOI: 10.1016/j.xpro.2024.103533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 10/25/2024] [Accepted: 11/28/2024] [Indexed: 12/23/2024] Open
Abstract
The interoperability of variant identification pipelines is fundamental for achieving consistent clinical care across oncology research centers and hospitals. Here, we present a protocol for using ONCOLINER, a platform for the assessment, improvement, and harmonization of somatic variant discovery of multiple pipelines. We describe steps for acquiring benchmarking datasets and executing the user variant calling pipeline. We then detail the procedures for performing analyses to produce user-friendly reports showing the quality, scope, and applicable improvements for each tumor genome analysis. For complete details on the use and execution of this protocol, please refer to Martín et al.1.
Collapse
Affiliation(s)
- Rodrigo Martín
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Spain.
| | - Nicolás Gaitán
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Spain
| | - David Torrents
- Life Sciences Department, Barcelona Supercomputing Center (BSC), Barcelona, Spain; Institució Catalana per la Recerca i Estudis Avançats (ICREA), Barcelona, Spain.
| |
Collapse
|
45
|
Zhang H, Chen L, Li L, Liu Y, Das B, Zhai S, Tan J, Jiang Y, Turco S, Yao Y, Frishman D. Prediction and analysis of tumor infiltrating lymphocytes across 28 cancers by TILScout using deep learning. NPJ Precis Oncol 2025; 9:76. [PMID: 40108446 PMCID: PMC11923303 DOI: 10.1038/s41698-025-00866-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
The density of tumor-infiltrating lymphocytes (TILs) serves as a valuable indicator for predicting anti-tumor responses, but its broad impact across various types of cancers remains underexplored. We introduce TILScout, a pan-cancer deep-learning approach to compute patch-level TIL scores from whole slide images (WSIs). TILScout achieved accuracies of 0.9787 and 0.9628, and AUCs of 0.9988 and 0.9934 in classifying WSI patches into three categories-TIL-positive, TIL-negative, and other/necrotic-on validation and independent test sets, respectively, surpassing previous studies. The biological significance of TILScout-derived TIL scores across 28 cancers was validated through comprehensive functional and correlational analyses. A consistent decrease in TIL scores with an increase in cancer stage provides direct evidence that the lower TIL content may stimulate cancer progression. Additionally, TIL scores correlated with immune checkpoint gene expression and genomic variation in common cancer driver genes. Our comprehensive pan-cancer survey highlights the critical prognostic significance of TILs within the tumor microenvironment.
Collapse
Affiliation(s)
- Huibo Zhang
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lulu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Liu
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Barnali Das
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Shuang Zhai
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Juan Tan
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yan Jiang
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Simona Turco
- Electrical Engineering, Eindhoven University of Technology, Den Dolech 12, Eindhoven, 5612AZ, the Netherlands
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Dmitrij Frishman
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany.
| |
Collapse
|
46
|
Li J, Zhang S, Wang B, Dai Y, Wu J, Liu D, Liang Y, Xiao S, Wang Z, Wu J, Zheng D, Chen X, Shi F, Tan K, Ding X, Song H, Zhang S, Lu M. Pharmacological rescue of mutant p53 triggers spontaneous tumor regression via immune responses. Cell Rep Med 2025; 6:101976. [PMID: 39986271 PMCID: PMC11970324 DOI: 10.1016/j.xcrm.2025.101976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/05/2024] [Accepted: 01/28/2025] [Indexed: 02/24/2025]
Abstract
Tumor suppressor p53 is the most frequently mutated protein in cancer, possessing untapped immune-modulating capabilities in anticancer treatment. Here, we investigate the efficacy and underlying mechanisms of pharmacological reactivation of mutant p53 in treating spontaneous tumors in mice. In the p53 R279W (equivalent to the human hotspot R282W) mouse model developing spontaneous tumors, arsenic trioxide (ATO) treatment through drinking water significantly prolongs the survival of mice, dependent on p53-R279W reactivation. Transient regressions of spontaneous T-lymphomas are observed in 70% of the ATO-treated mice, accompanied by interferon (IFN) response. In allograft models, the tumor-suppressive effect of reactivated p53-R279W is detectably reduced in both immunodeficient Rag1-/- and CD8+ T cell-depleted mice. ATO also activates the IFN pathway in human cancer cells harboring various p53 mutations, as well as in primary samples derived from the p53-mutant patient treated with ATO. Together, p53 could serve as an alternative therapeutic target for the development of immunotherapies.
Collapse
Affiliation(s)
- Jiabing Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shuang Zhang
- State Key Laboratory of Systems Medicine for Cancer, Institute for Personalized Medicine and Med-X, Institute School of Biomedical Engineering Research, Shanghai Jiao Tong University, Shanghai, China
| | - Baohui Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Yuting Dai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiale Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Dianjia Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ying Liang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shujun Xiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zhengyuan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jiaqi Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Derun Zheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xueqin Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fangfang Shi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kai Tan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xianting Ding
- State Key Laboratory of Systems Medicine for Cancer, Institute for Personalized Medicine and Med-X, Institute School of Biomedical Engineering Research, Shanghai Jiao Tong University, Shanghai, China.
| | - Huaxin Song
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Sujiang Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Min Lu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
47
|
Zhang T, Zhao W, Wirth C, Díaz-Gay M, Yin J, Cecati M, Marchegiani F, Hoang PH, Leduc C, Baine MK, Travis WD, Sholl LM, Joubert P, Sang J, McElderry JP, Klein A, Khandekar A, Hartman C, Rosenbaum J, Colón-Matos FJ, Miraftab M, Saha M, Lee OW, Jones KM, Caporaso NE, Wong MP, Leung KC, Agnes Hsiung C, Chen CY, Edell ES, Martínez Santamaría J, Schabath MB, Yendamuri SS, Manczuk M, Lissowska J, Świątkowska B, Mukeria A, Shangina O, Zaridze D, Holcatova I, Mates D, Milosavljevic S, Savic M, Bossé Y, Gould Rothberg BE, Christiani DC, Gaborieau V, Brennan P, Liu G, Hofman P, Homer R, Yang SR, Pesatori AC, Consonni D, Yang L, Zhu B, Shi J, Brown K, Rothman N, Chanock SJ, Alexandrov LB, Choi J, Cardelli M, Lan Q, Nowak MA, Wedge DC, Landi MT. Deciphering lung adenocarcinoma evolution and the role of LINE-1 retrotransposition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.14.643063. [PMID: 40161734 PMCID: PMC11952568 DOI: 10.1101/2025.03.14.643063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Understanding lung cancer evolution can identify tools for intercepting its growth. In a landscape analysis of 1024 lung adenocarcinomas (LUAD) with deep whole-genome sequencing integrated with multiomic data, we identified 542 LUAD that displayed diverse clonal architecture. In this group, we observed an interplay between mobile elements, endogenous and exogenous mutational processes, distinct driver genes, and epidemiological features. Our results revealed divergent evolutionary trajectories based on tobacco smoking exposure, ancestry, and sex. LUAD from smokers showed an abundance of tobacco-related C:G>A:T driver mutations in KRAS plus short subclonal diversification. LUAD in never smokers showed early occurrence of copy number alterations and EGFR mutations associated with SBS5 and SBS40a mutational signatures. Tumors harboring EGFR mutations exhibited long latency, particularly in females of European-ancestry (EU_N). In EU_N, EGFR mutations preceded the occurrence of other driver genes, including TP53 and RBM10. Tumors from Asian never smokers showed a short clonal evolution and presented with heterogeneous repetitive patterns for the inferred mutational order. Importantly, we found that the mutational signature ID2 is a marker of a previously unrecognized mechanism for LUAD evolution. Tumors with ID2 showed short latency and high L1 retrotransposon activity linked to L1 promoter demethylation. These tumors exhibited an aggressive phenotype, characterized by increased genomic instability, elevated hypoxia scores, low burden of neoantigens, propensity to develop metastasis, and poor overall survival. Reactivated L1 retrotransposition-induced mutagenesis can contribute to the origin of the mutational signature ID2, including through the regulation of the transcriptional factor ZNF695, a member of the KZFP family. The complex nature of LUAD evolution creates both challenges and opportunities for screening and treatment plans.
Collapse
Affiliation(s)
- Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Wei Zhao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Christopher Wirth
- Manchester Cancer Research Centre, The University of Manchester, Manchester, UK
| | - Marcos Díaz-Gay
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Digital Genomics Group, Structural Biology Program, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Jinhu Yin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Monia Cecati
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
| | | | - Phuc H Hoang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Charles Leduc
- Department of Pathology, Centre Hospitalier de l'Université de Montréal, Montreal, Canada
| | - Marina K Baine
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - William D Travis
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Philippe Joubert
- Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec City, Canada
| | - Jian Sang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - John P McElderry
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Alyssa Klein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Azhar Khandekar
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Caleb Hartman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | | | - Frank J Colón-Matos
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mona Miraftab
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Monjoy Saha
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Olivia W Lee
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kristine M Jones
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Neil E Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Maria Pik Wong
- Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| | - Kin Chung Leung
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | - Chao Agnes Hsiung
- Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Chih-Yi Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Surgery, Division of Thoracic Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Eric S Edell
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Matthew B Schabath
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Sai S Yendamuri
- Thoracic Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Marta Manczuk
- Department of Cancer Epidemiology and Primary Prevention, Maria Skłodowska-Curie National Research Institute of Oncology, Warshaw, Poland
| | - Jolanta Lissowska
- Department of Cancer Epidemiology and Primary Prevention, Maria Skłodowska-Curie National Research Institute of Oncology, Warshaw, Poland
| | - Beata Świątkowska
- Department of Environmental Epidemiology, Nofer Institute of Occupational Medicine, Łódź, Poland
| | - Anush Mukeria
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - Oxana Shangina
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - David Zaridze
- Department of Clinical Epidemiology, N.N. Blokhin National Medical Research Centre of Oncology, Moscow, Russia
| | - Ivana Holcatova
- Institute of Hygiene and Epidemiology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
- Department of Oncology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Dana Mates
- Department of Occupational Health and Toxicology, National Center for Environmental Risk Monitoring, National Institute of Public Health, Bucharest, Romania
| | - Sasa Milosavljevic
- International Organisation for Cancer Prevention and Research (IOCPR), Belgrade, Serbia
| | - Milan Savic
- Department of Thoracic Surgery, Clinical Center of Serbia, Belgrade, Serbia
| | - Yohan Bossé
- Institut universitaire de cardiologie et de pneumologie de Québec, Laval University, Quebec City, Canada
| | - Bonnie E Gould Rothberg
- Sylvester Comprehensive Cancer Center, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL, USA
| | - David C Christiani
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Valerie Gaborieau
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Paul Brennan
- Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France
| | - Geoffrey Liu
- Princess Margaret Cancer Center, University of Toronto, Toronto, Ontario, Canada
| | - Paul Hofman
- IHU RespirERA, Biobank-BB-0033-0025, Côte d'Azur University, Nice, France
| | - Robert Homer
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Soo-Ryum Yang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Angela C Pesatori
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Dario Consonni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lixing Yang
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
- Department of Human Genetics, The University of Chicago, Chicago, IL, USA
- The University of Chicago Medicine Comprehensive Cancer Center, The University of Chicago, Chicago, IL, USA
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kevin Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Nathaniel Rothman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Ludmil B Alexandrov
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA, USA
| | - Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Maurizio Cardelli
- Advanced Technology Center for Aging Research, IRCCS INRCA, Ancona, Italy
| | - Qing Lan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Martin A Nowak
- Department of Mathematics, Harvard University, Cambridge, MA, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - David C Wedge
- Manchester Cancer Research Centre, The University of Manchester, Manchester, UK
- Manchester NIHR Biomedical Research Centre, Manchester, UK
| | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
48
|
Liu Z, Huang S, Luo R, Shi X, Xiu M, Wang Y, Wang R, Zhang W, Lv M, Tang X. EXO1's pan-cancer roles: diagnostic, prognostic, and immunological analyses through bioinformatics. Discov Oncol 2025; 16:310. [PMID: 40074873 PMCID: PMC11903978 DOI: 10.1007/s12672-025-02045-w] [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: 11/15/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a leading cause of mortality worldwide, with human exonuclease 1 (EXO1) emerging as a key player in DNA repair and damage response pathways, critical for genomic stability and tumor evolution. The aim of this study was to conduct a comprehensive pan-cancer analysis to elucidate the multifaceted roles of EXO1 in various malignancies. Leveraging public databases including TCGA, GTEx, HPA, cBioPortal, UALCAN, STRING, CancerSEA and TISIDB database, we examined EXO1's expression, diagnostic potential, prognostic significance, mutational characteristics, functional roles, and immunological effects across different cancer types. EXO1 was found to be upregulated in multiple cancers, with significant diagnostic potential as indicated by high AUC values in ROC analyses. Elevated EXO1 expression correlated with adverse prognosis in several cancer types, including breast, lung, and pancreatic cancers. Epigenetic alterations, including DNA methylation and mRNA modifications, were also associated with EXO1 expression. Enrichment analyses identified EXO1-related genes involved in DNA recombination, replication, and repair, with GSEA implicating EXO1 in cell cycle regulation and DNA processing pathways. Importantly, immunogenomic analyses revealed EXO1's significant role in modulating the tumor microenvironment, as it is associated with immune cell infiltration and cytokine expression, suggesting its involvement in tumor immunology and immune response regulation. These results implied that EXO1 as a significant biomarker with prognostic and diagnostic potential across various malignancies, suggesting its potential as a therapeutic target and its involvement in immunomodulatory processes within the tumor microenvironment.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Shu Huang
- Department of Gastroenterology, Lianshui County People' Hospital, Huaian, China
- Department of Gastroenterology, Lianshui People' Hospital of Kangda College Affiliated to Nanjing Medical University, Huaian, China
| | - Rui Luo
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Xiaomin Shi
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Mingzhu Xiu
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Yizhou Wang
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Ruiyu Wang
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Wei Zhang
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China
| | - Muhan Lv
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China.
| | - Xiaowei Tang
- Department of Gastroenterology, the Affiliated Hospital of Southwest Medical University, Taiping No.25, Jiangyang, Luzhou, Sichuan, China.
| |
Collapse
|
49
|
LaFlam TN, Billesbølle CB, Dinh T, Wolfreys FD, Lu E, Matteson T, An J, Xu Y, Singhal A, Brandes N, Ntranos V, Manglik A, Cyster JG, Ye CJ. Phenotypic pleiotropy of missense variants in human B cell-confinement receptor P2RY8. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640567. [PMID: 40093123 PMCID: PMC11908195 DOI: 10.1101/2025.02.28.640567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Missense variants can have pleiotropic effects on protein function and predicting these effects can be difficult. We performed near-saturation deep mutational scanning of P2RY8, a G-protein-coupled receptor that promotes germinal center B cell confinement. We assayed the effect of each variant on surface expression, migration, and proliferation. We delineated variants that affected both expression and function, affected function independently of expression, and discrepantly affected migration and proliferation. We also used cryo-electron microscopy to determine the structure of activated, ligand-bound P2RY8, providing structural insights into the effects of variants on ligand binding and signal transmission. We applied the deep mutational scanning results to both improve computational variant effect predictions and to characterize the phenotype of germline variants and lymphoma-associated variants. Together, our results demonstrate the power of integrating deep mutational scanning, structure determination, and in silico prediction to advance the understanding of a receptor important in human health.
Collapse
Affiliation(s)
- Taylor N. LaFlam
- Division of Pediatric Rheumatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Christian B. Billesbølle
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
| | - Tuan Dinh
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Finn D. Wolfreys
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Erick Lu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Tomas Matteson
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
| | - Jinping An
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Ying Xu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Arushi Singhal
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
| | - Nadav Brandes
- Department of Biochemistry and Molecular Pharmacology, New York University, New York, NY, USA
| | - Vasilis Ntranos
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Diabetes Center, University of California, San Francisco, CA, USA
| | - Aashish Manglik
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Quantitative Biosciences Institute, San Francisco, CA, USA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA, USA
| | - Jason G. Cyster
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, USA
- Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Chun Jimmie Ye
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
- Division of Rheumatology, Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, USA
- Arc Institute, Palo Alto, CA, USA
| |
Collapse
|
50
|
Strobl EV, Gamazon E. Discovering root causal genes with high-throughput perturbations. eLife 2025; 13:RP100949. [PMID: 40042510 PMCID: PMC11882141 DOI: 10.7554/elife.100949] [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] [Indexed: 05/13/2025] Open
Abstract
Root causal gene expression levels - or root causal genes for short - correspond to the initial changes to gene expression that generate patient symptoms as a downstream effect. Identifying root causal genes is critical towards developing treatments that modify disease near its onset, but no existing algorithms attempt to identify root causal genes from data. RNA-sequencing (RNA-seq) data introduces challenges such as measurement error, high dimensionality and non-linearity that compromise accurate estimation of root causal effects even with state-of-the-art approaches. We therefore instead leverage Perturb-seq, or high-throughput perturbations with single-cell RNA-seq readout, to learn the causal order between the genes. We then transfer the causal order to bulk RNA-seq and identify root causal genes specific to a given patient for the first time using a novel statistic. Experiments demonstrate large improvements in performance. Applications to macular degeneration and multiple sclerosis also reveal root causal genes that lie on known pathogenic pathways, delineate patient subgroups and implicate a newly defined omnigenic root causal model.
Collapse
Affiliation(s)
| | - Eric Gamazon
- Vanderbilt University Medical CenterNashvilleUnited States
| |
Collapse
|