1
|
Bayona-Feliu A, Aguilera A. Transcription-Replication Conflicts: Unlocking New Frontiers in Cancer. Bioessays 2025:e70025. [PMID: 40491064 DOI: 10.1002/bies.70025] [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: 01/25/2025] [Revised: 05/22/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025]
Abstract
Genome instability (GIN) is a cell pathology linked to cancer promotion and tumor evolution. Transcription is an essential cellular process but also a potential source of DNA damage and GIN. Transcription-replication conflicts (TRCs) are a predominant source of GIN, and defective TRC resolution may seriously compromise genome integrity. Importantly, chromatin dynamics helps orchestrate the response to TRCs to preserve genome integrity. Multiple epigenetic deficiencies have been shown to cause transcription-induced replication stress, resulting in DNA breaks and mutations. Consistently, chromatin alterations are frequent in cancer and correlate with increased mutation burden at TRC sites in tumors. Here, we review our current knowledge of TRC processing, the consequences of its dysfunction, and its relevance in cancer. We focus on the interplay between the DNA damage response (DDR) and chromatin dynamics and discuss the clinical potential of targeting TRCs as anticancer strategies and drugging the associated epigenetic signatures.
Collapse
Affiliation(s)
- Aleix Bayona-Feliu
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
| | - Andrés Aguilera
- Centro Andaluz de Biología Molecular y Medicina Regenerativa CABIMER, Universidad de Sevilla, Seville, Spain
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
2
|
Thakur V, Thakur VS, Wang D, de Freitas JT, Bianchi A, Nivelo LA, Umland O, Welford SM, Bedogni B. Co-inhibition of Notch1 and ChK1 triggers genomic instability and melanoma cell death increasing the lifespan of mice bearing melanoma brain metastasis. J Exp Clin Cancer Res 2025; 44:163. [PMID: 40437523 PMCID: PMC12117938 DOI: 10.1186/s13046-025-03411-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 05/06/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Melanoma brain metastases (MBM) are a leading cause of death in patients with advanced disease. MBM treatment relay on targeted and immunotherapy and on stereotactic radiosurgery as gold standard. Life expectancy has improved significantly with these therapies however, targeted therapy is short lived and only about half of the patients respond to immunotherapy, while radiation is limited by melanoma cells intrinsic resistance to DNA damage. New therapeutic approaches are therefore needed to treat MBM. Here we investigate a new role of Notch1 in genomic instability and demonstrate that blockade of both Notch1 and the DNA repair factor ChK1 causes extensive DNA damage and tumor cell death increasing survival in MBM bearing mice. METHODS Anti-Notch1 (anti-N1) was previously described. Prexaserib, a ChK1 inhibitor, is currently in clinical trials. K457 and A375 melanoma cells were used. RNA sequencing was performed in K457 cells treated with anti-N1 and Gene Set Enrichment Analysis performed. DNA damage was evaluated by a DNA fiber assay to assess replication fork speed; and γH2AX foci count and neutral comet assay to quantify double strand breaks. Cell survival was evaluated by trypan blue and a colony formation assay. Luciferase expressing A375 cells were orthotopically inoculated in the right cerebral cortex of athymic nude mice, for in vivo evaluation of a therapy with anti-N1 and prexasertib. Survival was assessed by Kaplan-Meyer survival curves and significance assessed by a Log-rank test. RESULTS Notch1 blockade caused genomic instability by reducing histone availability, leading to DNA replication stress and DNA damage. This in turn, resulted in the activation of the DNA Damage Response pathway ATR/ChK1 to counter the damage. Co-inhibition of Notch1, via anti-N1, and ChK1, via prexasertib (prex), exacerbated DNA damage increasing melanoma cell death. Importantly, combination anti-N1/prex significantly improved survival of mice bearing MBMs. CONCLUSIONS A therapy with anti-N1/prexasertib could represent a novel treatment strategy, alone or in combination with current treatment regimens, for melanoma brain metastases.
Collapse
Affiliation(s)
- Varsha Thakur
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Vijay S Thakur
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Dazhi Wang
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Juliano Tiburcio de Freitas
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Anna Bianchi
- Division of Surgical Oncology, Dewitt Daughtry Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Luis Alberto Nivelo
- Division of Surgical Oncology, Dewitt Daughtry Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Oliver Umland
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Scott M Welford
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Barbara Bedogni
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA.
| |
Collapse
|
3
|
Nesic K, Parker P, Swisher EM, Krais JJ. DNA repair and the contribution to chemotherapy resistance. Genome Med 2025; 17:62. [PMID: 40420317 PMCID: PMC12107761 DOI: 10.1186/s13073-025-01488-8] [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: 07/17/2024] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
The DNA damage response comprises a set of imperfect pathways that maintain cell survival following exposure to DNA damaging agents. Cancers frequently exhibit DNA repair pathway alterations that contribute to their intrinsic genome instability. This, in part, facilitates a therapeutic window for many chemotherapeutic agents whose mechanisms of action often converge at the generation of a double-strand DNA break. The development of therapy resistance occurs through countless molecular mechanisms that promote tolerance to DNA damage, often by preventing break formation or increasing repair capacity. This review broadly discusses the DNA damaging mechanisms of action for different classes of chemotherapeutics, how avoidance and repair of double-strand breaks can promote resistance, and strategic directions for counteracting therapy resistance.
Collapse
Affiliation(s)
- Ksenija Nesic
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Phoebe Parker
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | | | - John J Krais
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA.
| |
Collapse
|
4
|
Lago S, Poli V, Fol L, Botteon M, Busi F, Turdo A, Gaggianesi M, Ciani Y, D'Amato G, Fagnocchi L, Fasciani A, Demichelis F, Todaro M, Zippo A. ANP32E drives vulnerability to ATR inhibitors by inducing R-loops-dependent transcription replication conflicts in triple negative breast cancer. Nat Commun 2025; 16:4602. [PMID: 40382323 PMCID: PMC12085574 DOI: 10.1038/s41467-025-59804-0] [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: 01/29/2024] [Accepted: 05/02/2025] [Indexed: 05/20/2025] Open
Abstract
Oncogene-induced replicative stress (RS) drives tumor progression by disrupting genome stability, primarily through transcription-replication conflicts (TRCs), which promote R-loop accumulation and trigger the DNA damage response (DDR). In this study, we investigate the role of chromatin regulators in exacerbating TRCs and R-loop accumulation in cancer. We find that in breast cancer patients, the simultaneous upregulation of MYC and the H2A.Z-specific chaperone ANP32E correlates with increased genomic instability. Genome-wide analyses reveal that ANP32E-driven H2A.Z turnover alters RNA polymerase II processivity, leading to the accumulation of long R-loops at TRC sites. Furthermore, we show that ANP32E overexpression enhances TRC formation and activates an ATR-dependent DDR, predisposing cancer cells to R-loop-mediated genomic fragility. By exploiting the vulnerability of ANP32E-expressing cancer cells to ATR inhibitors, we find that tumors relied on this DDR pathway, whose inhibition halts their pro-metastatic capacity. These findings identify ANP32E as a key driver of TRC-induced genomic instability, indicating ATR inhibition as a potential therapeutic strategy for ANP32E-overexpressing tumors.
Collapse
Affiliation(s)
- Sara Lago
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Vittoria Poli
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Lisa Fol
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
- Institute of Molecular Biology (IMB), Mainz, Germany
| | - Mattia Botteon
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Federica Busi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Alice Turdo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Miriam Gaggianesi
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, 90127, Palermo, Italy
| | - Yari Ciani
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Giacomo D'Amato
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Luca Fagnocchi
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Alessandra Fasciani
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Francesca Demichelis
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy
| | - Matilde Todaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Alessio Zippo
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, 38123, Trento, Italy.
| |
Collapse
|
5
|
Wróblewski M, Krajewski K, Gocek N, Żabka A, Polit JT. Acorus calamus L. Essential Oil Induces Oxidative Stress and DNA Replication Disruptions in Root Meristem Cells of Two Fabaceae and Two Brassicaceae Species. Int J Mol Sci 2025; 26:4715. [PMID: 40429859 PMCID: PMC12111713 DOI: 10.3390/ijms26104715] [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: 04/03/2025] [Revised: 05/06/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Environmental concerns regarding synthetic herbicides have sparked interest in plant-derived bioactive compounds as eco-friendly alternatives. This study investigated the cellular targets of sweet flag essential oil (Acorus calamus L., SEO at IC50 concentration) in root meristem cells of Fabaceae (Vicia faba, Lupinus luteus) and Brassicaceae (Brassica napus, Arabidopsis thaliana), focusing on reactive oxygen species (ROS) accumulation (DAB, NBT staining), DNA replication dynamics (EdU labeling), and genome integrity (γ-H2AX immunocytochemistry, TUNEL assay, and DNA electrophoresis). SEO induced oxidative stress (200-250% of control depending on the species) and replication stress, causing DNA double-strand breaks in 50% of proliferating cells, confirmed by γ-H2AX/TUNEL. Consequently, cells were prolonged in the G1 phase, replication activity dropped to 70% of control in Fabaceae and 80% in Brassicaceae, and EdU incorporation intensity decreased to 80% and 70% of control, respectively. An increased proportion of cells replicating heterochromatin indicated slowed S-phase progression. Despite genotoxic effects, SEO did not trigger endoreplication, apoptotic DNA fragmentation, or extensive cell death. All species exhibited a uniform stress response, although sensitivity varied, which previously enabled the establishment of selective SEO doses between Fabaceae and Brassicaceae. These findings suggest that SEO exerts phytotoxicity by disrupting S-phase progression, supporting its potential as a selective bioherbicide.
Collapse
Affiliation(s)
- Mateusz Wróblewski
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (K.K.); (N.G.); (A.Ż.); (J.T.P.)
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Konrad Krajewski
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (K.K.); (N.G.); (A.Ż.); (J.T.P.)
| | - Natalia Gocek
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (K.K.); (N.G.); (A.Ż.); (J.T.P.)
- Doctoral School of Exact and Natural Sciences, University of Lodz, 90-237 Lodz, Poland
| | - Aneta Żabka
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (K.K.); (N.G.); (A.Ż.); (J.T.P.)
| | - Justyna T. Polit
- Department of Cytophysiology, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland; (K.K.); (N.G.); (A.Ż.); (J.T.P.)
| |
Collapse
|
6
|
Ahmad ST, Li Y, Garcia-Lopez J, Gudenas BL, Hadley J, Paul L, Wu SC, Refaat A, Kojic M, Batts M, Soliman T, Pitre A, Arnskötter F, Zindy F, Jones A, Twarog NR, Mayasundari A, Bianski B, Tinkle C, Shirinifard A, Janke L, Lu M, Lewis SA, Onar-Thomas A, Pfister SM, Gajjar A, Baker SJ, Roussel MF, Rankovic Z, Robinson GW, Orr BA, Wainwright B, Shelat AA, Waszak SM, Kutscher LM, Lin H, Northcott PA. Genetic modeling of ELP1-associated Sonic hedgehog medulloblastoma identifies MDM2 as a selective therapeutic target. Cancer Cell 2025:S1535-6108(25)00173-4. [PMID: 40378836 DOI: 10.1016/j.ccell.2025.04.014] [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: 03/10/2024] [Revised: 02/23/2025] [Accepted: 04/28/2025] [Indexed: 05/19/2025]
Abstract
Germline loss-of-function (LOF) variants in Elongator acetyltransferase complex subunit 1 (ELP1) are the most prevalent predisposing genetic events in childhood medulloblastoma (MB), accounting for ∼30% of the Sonic hedgehog (SHH) 3 subtype. The mechanism(s) by which germline ELP1 deficiency provokes SHH-MB pathogenesis remain unknown. Genetically engineered mice mimicking heterozygous Elp1 LOF (Elp1HET) seen in affected germline carriers exhibit hallmark features of premalignancy in cerebellar granule neuron progenitors (GNPs), including increased DNA replication stress, genomic instability, accelerated cell cycle, and stalled differentiation. Orthotopic transplantation of Elp1HET GNPs harboring somatic Ptch1 inactivation yields SHH-MB-like tumors with compromised p53 signaling, providing a plausible explanation for the exclusivity of ELP1-associated MBs in the SHH-3 subtype. Preclinical treatment of ELP1-mutant patient-derived xenografts with an FDA-approved MDM2 inhibitor reactivates p53-dependent apoptosis and extends survival. Our findings functionally substantiate the role of ELP1 deficiency in SHH-MB predisposition and nominate therapeutics targeting MDM2 as a rational treatment option.
Collapse
Affiliation(s)
- Shiekh Tanveer Ahmad
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Yiran Li
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jesus Garcia-Lopez
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brian L Gudenas
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jennifer Hadley
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Leena Paul
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stephanie C Wu
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alaa Refaat
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Marija Kojic
- Frazer Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Melissa Batts
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Taha Soliman
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Aaron Pitre
- Cell and Tissue Imaging Shared Resource, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Frederik Arnskötter
- Hopp Children's Cancer Center Heidelberg (KiTZ), JRG Developmental Origins of Pediatric Cancers, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Frederique Zindy
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Alun Jones
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4102, Australia
| | - Nathaniel R Twarog
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Anand Mayasundari
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brandon Bianski
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christopher Tinkle
- Department of Radiation Oncology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Abbas Shirinifard
- Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Laura Janke
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Meifen Lu
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sara A Lewis
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Arzu Onar-Thomas
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stefan M Pfister
- Hopp Children's Cancer Center Heidelberg (KiTZ), Division Pediatric Neurooncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Department of Pediatric Hematology and Oncology, Heidelberg University Hospital and National Center for Tumor Diseases (NCT), 69120 Heidelberg, Germany
| | - Amar Gajjar
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Suzanne J Baker
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Martine F Roussel
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Zoran Rankovic
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Giles W Robinson
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brent A Orr
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Brandon Wainwright
- Frazer Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Anang A Shelat
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Sebastian M Waszak
- Swiss Institute for Experimental Cancer Research, School of Life Sciences, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland; Department of Neurology, University of California, San Francisco (UCSF), San Francisco, CA 94143, USA
| | - Lena M Kutscher
- Hopp Children's Cancer Center Heidelberg (KiTZ), JRG Developmental Origins of Pediatric Cancers, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Hong Lin
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Paul A Northcott
- Center of Excellence in Neuro-Oncology Sciences (CENOS), St. Jude Children's Research Hospital, Memphis, TN 38105, USA; Department of Developmental Neurobiology, St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
7
|
Park J, Lee DH. Loss of protein phosphatase 4 inhibitory protein leads to genomic instability and heightens vulnerability to replication stress. Biochim Biophys Acta Gen Subj 2025; 1869:130797. [PMID: 40157551 DOI: 10.1016/j.bbagen.2025.130797] [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: 11/20/2024] [Revised: 03/09/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
Protein phosphatase 4 inhibitory protein (PP4IP) has recently emerged as a key player in cellular processes, particularly in DNA double-strand break repair and telomere maintenance, although research on its functions remains limited. To further investigate the cellular pathways involving PP4IP, we conducted transcriptomic analysis via RNA sequencing in PP4IP-knockout cells and observed an upregulation of p21 expression. This upregulation was linked to an increased population of p21-positive G1-phase cells in the absence of PP4IP. Prior studies have suggested that unresolved under-replicated DNA in mother cells, transmitted to daughter cells, can trigger a quiescent G1 phase characterized by p21 expression and the formation of p53-binding protein 1 (53BP1) nuclear bodies. Consistent with this, we found a higher proportion of 53BP1 nuclear bodies-positive G1 cells in PP4IP-knockout cells compared to controls. Additionally, PP4IP-deficient cells displayed an increased occurrence of anaphase bridges-indicative of incomplete DNA replication-without a corresponding increase in lagging chromosomes. Furthermore, PP4IP-knockout cells exhibited a heightened susceptibility to replication stress, as evidenced by an elevated frequency of replication stress-induced chromatid breaks and increased sensitivity to such stress. Collectively, these results suggest that PP4IP plays a critical role in safeguarding cells from replication stress and ensuring genomic stability.
Collapse
Affiliation(s)
- Jaehong Park
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea.
| | - Dong-Hyun Lee
- Department of Biological Sciences, College of Natural Sciences, Chonnam National University, Gwangju, Republic of Korea; Research Center of Ecomimetics, Chonnam National University, Gwangju, Republic of Korea; Institute of Sustainable Ecological Environment, Chonnam National Univesity, Gwangju, Republic of Korea.
| |
Collapse
|
8
|
Zou X, Zhou X, Guo X, Pan L, Li W. SMARCA4 deficiency: implications for non-small cell lung cancer and management strategies, with relevance to and distinctions from thoracic undifferentiated tumor. Transl Lung Cancer Res 2025; 14:1456-1470. [PMID: 40386720 PMCID: PMC12082232 DOI: 10.21037/tlcr-24-927] [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: 10/10/2024] [Accepted: 02/19/2025] [Indexed: 05/20/2025]
Abstract
In 2021, the fifth edition of the World Health Organization (WHO) classification of thoracic tumors introduced a new category, "Thoracic SMARCA4-deficient undifferentiated tumor", highlighting SMARCA4 deficiency as a key molecular marker for classifying as "other pulmonary epithelial tumors". SMARCA4 is a gene encoding a protein involved in chromatin remodeling, and approximately 8% of non-small cell lung cancer (NSCLC) patients exhibit SMARCA4 deletions. These patients are more prone to drug resistance, early recurrence, and unfavorable clinical outcomes. Moreover, NSCLC patients with concomitant SMARCA4 mutations may not benefit from currently available treatments, underscoring the distinctiveness of this subgroup. Thoracic SMARCA4-deficient undifferentiated tumors (SMARCA4-UT) represent distinct entities from SMARCA4-deficient non-small cell lung cancer (SMARCA4-dNSCLC). This distinction is supported by their divergent pathological characteristics, demographic profiles, and survival outcomes. NSCLC cases deficient in SMARCA4 exhibit high malignancy, yet the precise biological mechanisms underlying this phenomenon remain under intensive investigation. Pathological examination and immunohistochemistry can effectively differentiate SMARCA4-UT from SMARCA4-dNSCLC. SMARCA4-UT typically manifests as adenocarcinoma or, more rarely, as squamous cell carcinoma with undifferentiated rhabdomyoblastic morphology. Therefore, elucidating the mechanisms underlying SMARCA4 alterations in NSCLC and their regulatory roles in tumorigenesis and the microenvironment is crucial. This article aims to discuss the structure, biological functions, significance in NSCLC development, and emerging potential therapeutic strategies related to SMARCA4 while providing clinical practice guidance for NSCLC patients with SMARCA4 deletions.
Collapse
Affiliation(s)
- Xin Zou
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Air Force Medical University, Xi’an, China
- Institute for Lung Disease, Air Force Medical University, Xi’an, China
| | - Xiaoqing Zhou
- Department of Disease Control and Prevention, Xi’an Eighth Hospital, Xi’an, China
| | - Xian Guo
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Air Force Medical University, Xi’an, China
- Institute for Lung Disease, Air Force Medical University, Xi’an, China
| | - Lei Pan
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Air Force Medical University, Xi’an, China
- Institute for Lung Disease, Air Force Medical University, Xi’an, China
| | - Wangping Li
- Department of Respiratory and Critical Care Medicine, Tangdu Hospital, Air Force Medical University, Xi’an, China
- Institute for Lung Disease, Air Force Medical University, Xi’an, China
| |
Collapse
|
9
|
Zhou Y, Deng Z, Xiong S, Li W, Luo W, Luo M, Tang H, Wu W, Chak-Lui Wong C, Yin D, Hu K, Luo B. NUDT16 enhances the resistance of cancer cells to DNA-damaging agents by regulating replication fork stability via reversing HMGA1 ADP-ribosylation. J Biol Chem 2025; 301:108551. [PMID: 40288645 DOI: 10.1016/j.jbc.2025.108551] [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: 10/07/2024] [Revised: 03/14/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Precise DNA replication is the basis for maintaining cell proliferation and genome stability. Current chemotherapy drugs and radiotherapy induce cell death by aggravating replication stress, albeit with poor efficacy. The replication stress response has been shown to play fundamental roles in resistance to radiotherapy and chemotherapy. High mobility group A1 (HMGA1) promotes tumor progression by regulating autophagy, angiogenesis, and chemoresistance; however, its role in coordinating replication stress and cell cycle progression remains elusive. Our results indicated that HMGA1 recruited FANCD2 to promote DNA replication and cell cycle progression both by attenuating R-loop-induced replication stress and by protecting stalled replication forks from degradation, ultimately enhancing tumor resistance to chemotherapy and irradiation treatment. We also identified HMGA1 as a novel substrate for the dePARylase NUDT16. NUDT16 was found to suppress the binding of HMGA1 to the E3 ubiquitin ligase CHFR by removing its PARylation at Glu 50, thereby reducing its ubiquitin-proteasome pathway-mediated degradation and enhancing HMGA1 protein stability. NUDT16-HMGA1 inhibition can significantly improve the sensitivity of tumor cells to chemotherapy and irradiation treatment. Collectively, these data suggest that NUDT16 enhances the ability of tumor cells to cope with replication stress by reversing the PARylation and positively regulating the protein expression of HMGA1. Therefore, targeting the NUDT16-HMGA1 pathway may be a novel strategy to enhance the sensitivity of radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- Yingshi Zhou
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhihuai Deng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shiyu Xiong
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wenjia Li
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Wanrong Luo
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Man Luo
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Haifeng Tang
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Wenjing Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China; Department of Breast Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Carmen Chak-Lui Wong
- Li Ka Shing Faculty of Medicine, Department of Pathology, The University of Hong Kong, Hong Kong, China
| | - Dong Yin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Kaishun Hu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Baoming Luo
- Department of Ultrasound, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
10
|
Ortega P, Bournique E, Li J, Sanchez A, Santiago G, Harris BR, Striepen J, Maciejowski J, Green AM, Buisson R. Mechanism of DNA replication fork breakage and PARP1 hyperactivation during replication catastrophe. SCIENCE ADVANCES 2025; 11:eadu0437. [PMID: 40238882 DOI: 10.1126/sciadv.adu0437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/11/2025] [Indexed: 04/18/2025]
Abstract
Ataxia telangiectasia and Rad3-related (ATR) inhibition triggers a surge in origin firing, resulting in increased levels of single-stranded DNA (ssDNA) that rapidly deplete all available RPA. This leaves ssDNA unprotected and susceptible to breakage, a phenomenon known as replication catastrophe. However, the mechanism by which unprotected ssDNA breaks remains unclear. Here, we reveal that APOBEC3B is the key enzyme targeting unprotected ssDNA at replication forks, initiating a reaction cascade that induces fork collapse and poly(ADP-ribose) polymerase 1 (PARP1) hyperactivation. Mechanistically, we demonstrate that uracils generated by APOBEC3B at replication forks are removed by UNG2, resulting in abasic sites that are subsequently cleaved by APE1 endonuclease. Moreover, we show that APE1-mediated DNA cleavage is the critical enzymatic step for PARP1 hyperactivation in cells, regardless of how abasic sites are generated on DNA. Last, we demonstrate that APOBEC3B-induced PARP1 trapping and DNA double-strand breaks drive cell sensitivity to ATR inhibition, creating a context of synthetic lethality when coupled with PARP inhibitors.
Collapse
Affiliation(s)
- Pedro Ortega
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Virus Research, University of California Irvine, Irvine, CA, USA
| | - Elodie Bournique
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Virus Research, University of California Irvine, Irvine, CA, USA
| | - Junyi Li
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Virus Research, University of California Irvine, Irvine, CA, USA
| | - Ambrocio Sanchez
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Virus Research, University of California Irvine, Irvine, CA, USA
| | - Gisselle Santiago
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Virus Research, University of California Irvine, Irvine, CA, USA
| | - Brooke R Harris
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Center for Genome Integrity, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Josefine Striepen
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John Maciejowski
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Abby M Green
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
- Center for Genome Integrity, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Rémi Buisson
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, CA, USA
- Center for Virus Research, University of California Irvine, Irvine, CA, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
11
|
Kołodziejczak-Guglas I, Simões RLS, de Souza Santos E, Demicco EG, Lazcano Segura RN, Ma W, Wang P, Geffen Y, Storrs E, Petralia F, Colaprico A, da Veiga Leprevost F, Pugliese P, Ceccarelli M, Noushmehr H, Nesvizhskii AI, Kamińska B, Priebe W, Lubiński J, Zhang B, Lazar AJ, Kurzawa P, Mesri M, Robles AI, Ding L, Malta TM, Wiznerowicz M. Proteomic-based stemness score measures oncogenic dedifferentiation and enables the identification of druggable targets. CELL GENOMICS 2025:100851. [PMID: 40250426 DOI: 10.1016/j.xgen.2025.100851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/13/2024] [Accepted: 03/21/2025] [Indexed: 04/20/2025]
Abstract
Cancer progression and therapeutic resistance are closely linked to a stemness phenotype. Here, we introduce a protein-expression-based stemness index (PROTsi) to evaluate oncogenic dedifferentiation in relation to histopathology, molecular features, and clinical outcomes. Utilizing datasets from the Clinical Proteomic Tumor Analysis Consortium across 11 tumor types, we validate PROTsi's effectiveness in accurately quantifying stem-like features. Through integration of PROTsi with multi-omics, including protein post-translational modifications, we identify molecular features associated with stemness and proteins that act as active nodes within transcriptional networks, driving tumor aggressiveness. Proteins highly correlated with stemness were identified as potential drug targets, both shared and tumor specific. These stemness-associated proteins demonstrate predictive value for clinical outcomes, as confirmed by immunohistochemistry in multiple samples. The findings emphasize PROTsi's efficacy as a valuable tool for selecting predictive protein targets, a crucial step in customizing anti-cancer therapy and advancing the clinical development of cures for cancer patients.
Collapse
Affiliation(s)
- Iga Kołodziejczak-Guglas
- International Institute for Molecular Oncology, 60-203 Poznań, Poland; Postgraduate School of Molecular Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Renan L S Simões
- School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil
| | - Emerson de Souza Santos
- School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto 14040-900, Brazil
| | - Elizabeth G Demicco
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital and Laboratory Medicine and Pathobiology, University of Toronto, Toronto ON M5G 1X5, Canada
| | - Rossana N Lazcano Segura
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Weiping Ma
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yifat Geffen
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA 02115, USA
| | - Erik Storrs
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Antonio Colaprico
- Sylvester Comprehensive Cancer Center and Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | - Pietro Pugliese
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center and Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Houtan Noushmehr
- Hermelin Brain Tumor Center, Henry Ford Health System, Detroit, MI 48202, USA
| | - Alexey I Nesvizhskii
- Departments of Pathology and Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bożena Kamińska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Waldemar Priebe
- Department of Experimental Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Jan Lubiński
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, 70-204 Szczecin, Poland
| | - Bing Zhang
- Lester and Sue Smith Breast Center and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander J Lazar
- Departments of Pathology & Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paweł Kurzawa
- Department of Oncological Pathology, University Clinical Hospital in Poznan, Poznan University of Medical Sciences, 60-514 Poznań, Poland
| | - Mehdi Mesri
- Office of Cancer Clinical Proteomics Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD 20850, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD 20850, USA
| | - Li Ding
- Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tathiane M Malta
- School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto 14040-900, Brazil.
| | - Maciej Wiznerowicz
- International Institute for Molecular Oncology, 60-203 Poznań, Poland; Department of Oncology, Institute of Oncology, University Clinical Hospital in Poznan, Poznan University of Medical Sciences, 60-659 Poznań, Poland.
| |
Collapse
|
12
|
Merrick BA, Brooks AM, Foley JF, Martin NP, Fannin RD, Gladwell W, Gerrish KE. hTERT and SV40LgT Renal Cell Lines Adjust Their Transcriptional Responses After Copy Number Changes from the Parent Proximal Tubule Cells. Int J Mol Sci 2025; 26:3607. [PMID: 40332109 PMCID: PMC12027150 DOI: 10.3390/ijms26083607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
Primary mouse renal proximal tubule epithelial cells (moRPTECs) were immortalized by lentivirus transduction to create hTERT or SV40LgT (LgT) cell lines. Prior work showed a more pronounced injury and repair response in LgT versus hTERT cells after chemical challenge. We hypothesized that unique genomic changes occurred after immortalization, altering critical genes and pathways. RNA-seq profiling and whole-genome sequencing (WGS) of parent, hTERT, and LgT cells showed that 92.5% of the annotated transcripts were shared, suggesting a conserved proximal tubule expression pattern. However, the cell lines exhibited unique transcriptomic and genomic profiles different from the parent cells. Three transcript classes were quite relevant for chemical challenge response-Cyps, ion channels, and metabolic transporters-each important for renal function. A pathway analysis of the hTERT cells suggested alterations in intermediary and energy metabolism. LgT cells exhibited pathway activation in cell cycle and DNA repair that was consistent with replication stress. Genomic karyotyping by combining WGS and RNA-seq data showed increased gene copy numbers in chromosome 5 for LgT cells, while hTERT cells displayed gene copy losses in chromosomes 4 and 9. These data suggest that the exaggerated transcriptional responses of LgT cells versus hTERT cells result from differences in gene copy numbers, replication stress, and the unique selection processes underlying LgT or hTERT immortalization.
Collapse
Affiliation(s)
- Bruce Alex Merrick
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA;
| | - Ashley M. Brooks
- Biostatistics and Computational Biology Branch, Integrative Bioinformatics Support Group, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA;
| | - Julie F. Foley
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA;
| | - Negin P. Martin
- Viral Vector Core, Neurobiology Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA;
| | - Rick D. Fannin
- Molecular Genomics Core Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA; (R.D.F.); (W.G.); (K.E.G.)
| | - Wesley Gladwell
- Molecular Genomics Core Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA; (R.D.F.); (W.G.); (K.E.G.)
| | - Kevin E. Gerrish
- Molecular Genomics Core Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA; (R.D.F.); (W.G.); (K.E.G.)
| |
Collapse
|
13
|
Pugh K, Jones RDO, Powathil G, Hamis S. Simulations probe the role of space in the interplay between drug-sensitive and drug-resistant cancer cells. J Theor Biol 2025; 602-603:112048. [PMID: 39914489 DOI: 10.1016/j.jtbi.2025.112048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 12/22/2024] [Accepted: 01/09/2025] [Indexed: 02/14/2025]
Abstract
The interplay between drug-sensitive and drug-resistant cancer cells has been observed to impact cell-to-cell interactions in experimental settings. However, the role that space plays in these interactions remains unclear. In this study, we develop mathematical models to investigate how spatial factors affect cell-to-cell competition between drug-sensitive and drug-resistant cancer cells in silico. We develop two baseline models to study cells from the epithelial FaDu cell line subjected to two drugs, specifically the ATR inhibitor ceralasertib and the PARP inhibitor olaparib, that target DNA damage response pathways. Our baseline models are: (1) a temporally resolved ordinary differential equation (ODE) model, and (2) a spatio-temporally resolved agent-based model (ABM). The models simulate cells in well-mixed and spatially structured cell systems, respectively. The ODE model is calibrated against in vitro data and is thereafter mapped onto the baseline ABM which, in turn, is extended to enable a simulation-based investigation on how spatial factors impact cell-to-cell competition. Simulation results from the extended ABMs demonstrate that the in silico treatment responses are simultaneously affected by: (i) the initial spatial cell configurations, (ii) the initial fraction of drug-resistant cells, (iii) the drugs to which cells express resistance, (iv) drug combinations, (v) drug doses, and (vi) the doubling time of drug-resistant cells compared to the doubling time of drug-sensitive cells. These results reveal that spatial structures of the simulated cancer cells affect both cell-to-cell interactions, and the impact that these interactions have on the ensuing population dynamics. This leads us to suggest that the role that space plays in cell-to-cell interactions should be further investigated and quantified in experimental settings.
Collapse
Affiliation(s)
- Kira Pugh
- Department of Mathematics, College of Science, Swansea University, Swansea, United Kingdom.
| | - Rhys D O Jones
- The Oncology Drug Metabolism and Pharmacokinetics (DMPK) Department, Oncology Research and Development (R&D), AstraZeneca, Cambridge, United Kingdom
| | - Gibin Powathil
- Department of Mathematics, College of Science, Swansea University, Swansea, United Kingdom
| | - Sara Hamis
- Department of Information Technology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
14
|
Fernandez A, Zhou T, Esworthy S, Shen C, Liu H, Hess JD, Yuan H, Liu N, Shi G, Zhou M, Kosiyatrakul S, Gaur V, Sommers J, Edelman W, Li GM, Brosh R, Chai W, Lee MYWT, Zhang D, Schildkraut C, Zheng L, Shen B. DNA2 and MSH2 activity collectively mediate chemically stabilized G4 for efficient telomere replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647332. [PMID: 40330857 PMCID: PMC12051496 DOI: 10.1101/2025.04.04.647332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
G-quadruplexes (G4s) are widely existing stable DNA secondary structures in mammalian cells. A long-standing hypothesis is that timely resolution of G4s is needed for efficient and faithful DNA replication. In vitro , G4s may be unwound by helicases or alternatively resolved via DNA2 nuclease mediated G4 cleavage. However, little is known about the biological significance and regulatory mechanism of the DNA2-mediated G4 removal pathway. Here, we report that DNA2 deficiency or its chemical inhibition leads to a significant accumulation of G4s and stalled replication forks at telomeres, which is demonstrated by a high-resolution technology: Single molecular analysis of replicating DNA (SMARD). We further identify that the DNA repair complex MutSα (MSH2-MSH6) binds G4s and stimulates G4 resolution via DNA2-mediated G4 excision. MSH2 deficiency, like DNA2 deficiency or inhibition, causes G4 accumulation and defective telomere replication. Meanwhile, G4-stabilizing environmental compounds block G4 unwinding by helicases but not G4 cleavage by DNA2. Consequently, G4 stabilizers impair telomere replication and cause telomere instabilities, especially in cells deficient in DNA2 or MSH2.
Collapse
|
15
|
Zheng C, Niu G, Tan H, Huang X, Lu J, Mai Q, Yu T, Zhang C, Chen S, Wei M, Pan W, Guo Y, Wang J, Xu M, Yao S, Liu J, Li J, Pan C. A noncanonical role of SAT1 enables anchorage independence and peritoneal metastasis in ovarian cancer. Nat Commun 2025; 16:3174. [PMID: 40180916 PMCID: PMC11968987 DOI: 10.1038/s41467-025-58525-8] [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: 07/19/2024] [Accepted: 03/25/2025] [Indexed: 04/05/2025] Open
Abstract
Anchorage-independent survival of ovarian tumor cells in ascites is the initial and critical step for peritoneal metastasis. How ovarian tumor cells achieve anchorage independence remains unclear. Here we show that a noncanonical role of spermidine/spermine N1-acetyltransferase 1 (SAT1) dictates anchorage-independent cell survival and potentiates metastatic dissemination in ovarian cancer. SAT1-high cancer cells are prevalent in ascitic tumors, and high SAT1 expression in primary tumors is linked to increased peritoneal metastasis rates in ovarian cancer patients. Mechanistically, SAT1 noncanonically acetylates H3K27 domains in multiple mitosis-regulating genes, increasing their transcriptional levels and protecting disseminating cells from aberrant mitosis and mitotic cell death. Notably, the acetylation of H3K27 by SAT1 depends on the reductive carboxylation of glutamine to supply acetyl-CoA in the nucleus. SAT1 inhibition with the small-molecule inhibitor ginkgolide B attenuates the metastatic tumor burden in mouse models. We conclude that SAT1 inhibition is a promising therapeutic strategy for metastatic ovarian cancer.
Collapse
Affiliation(s)
- Cuimiao Zheng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Gang Niu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hao Tan
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xi Huang
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jingyi Lu
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qiuwen Mai
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Tiantian Yu
- Metabolomics Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China
| | - Chunyu Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, 510080, China
| | - Siqi Chen
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Mengxun Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Wenfeng Pan
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yu Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Manman Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shuzhong Yao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, 510080, China
| | - Junxiu Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, 510080, China.
| | - Jie Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Chaoyun Pan
- Department of Biochemistry and Molecular Biology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Metabolomics Research Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, Guangdong, China.
- Guangdong Provincial Clinical Research Center for Obstetrical and Gynecological Diseases, Guangzhou, 510080, China.
| |
Collapse
|
16
|
Zhou N, Zheng Q, Liu Y, Huang Z, Feng Y, Chen Y, Hu F, Zheng H. Strain diversity and host specificity of the gut symbiont Gilliamella in Apis mellifera, Apis cerana and Bombus terrestris. Microbiol Res 2025; 293:128048. [PMID: 39813751 DOI: 10.1016/j.micres.2025.128048] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/15/2024] [Accepted: 01/02/2025] [Indexed: 01/18/2025]
Abstract
Social bees, with their specialized gut microbiota and societal transmission between individuals, provide an ideal model for studying host-gut microbiota interactions. While the functional disparities arising from strain-level diversity of gut symbionts and their effects on host health have been studied in Apis mellifera and bumblebees, studies focusing on host-specific investigations of individual strains across different honeybee hosts remain relatively unexplored. In this study, the complete genomic sequences of 17 strains of Gilliamella from A. mellifera, Apis cerana and Bombus terrestris were analyzed. The analysis revealed that the strains of A. mellifera display a more expansive genomic and functional content compared to the strains of A. cerana and B. terrestris. Phylogenetic analysis showed a deep divergence among the Gilliamella strains from different hosts. Additionally, biochemistry tests and antibiotic susceptibility tests revealed that gut strains from A. mellifera exhibited a more extensive pathway for carbohydrate metabolism and a greater resistance to antibiotics than gut strains from A. cerana and B. terrestris. Strains from A. mellifera and A. cerana showed higher colonization efficiency and competitive ability whithin their respective host species, indicating a higher degree of host-specific adaptation of local gut microbiota. In addition, colonization by A. mellifera-derived strain triggers a stronger transcriptional response in the host than A. cerana-derived strain. The variation in the number of differentially expressed genes and the involvement of distinct signaling pathways across these two host species suggest species-specific adaptations to Gilliamella strains. These findings suggest that despite occupying similar niches in the bee gut, strain-level variations can influence microbial functions, and their impact on host physiological functions may vary across different strains.
Collapse
Affiliation(s)
- Nihong Zhou
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiulan Zheng
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yao Liu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhichu Huang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ye Feng
- Institute of Translational Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yanping Chen
- Bee Research Laboratory, USDA-ARS, Beltsville, MD, USA
| | - Fuliang Hu
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Huoqing Zheng
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
17
|
Gonzalez‐Rubio J, Kubiza H, Xu Y, Koenigs‐Werner H, Schmitz MS, Schedel M, Apel C, Jockenhoevel S, Cornelissen CG, Thiebes AL. Pericytes Promote More Vascularization than Stromal Cells via an Interleukin-6-Dependent Mechanism in Microfluidic Chips. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408131. [PMID: 39887579 PMCID: PMC11984840 DOI: 10.1002/advs.202408131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/10/2025] [Indexed: 02/01/2025]
Abstract
Pericytes are a key player in vascularization, protecting endothelial cells from external harm and promoting the formation of new vessels when necessary. However, pericytic identity and its relationship with other cell types, such as mesenchymal stromal/stem cells, is highly debated. This study compares the role of pericytes and unselected stromal cells in vascularization using multichannel microfluidic chips. In both angiogenesis and vasculogenesis, pericytes promote more vessel formation than stromal cells. Pericytes can wrap around endothelial vessels acting as mural cells, while stromal cells remain separated. Whole-transcriptome sequencing confirms an upregulation of pro-vascularization genes in endothelial cell-pericyte co-cultures, while metabolism increases and inflammation decreases in stromal cell co-cultures. Treatment of stromal-endothelial cell co-cultures with either conditioned media or isolated extracellular vesicles from pericytes replicates the increase in vasculogenesis of the direct co-cultures. Cytokine quantification reveals that interleukin 6 (IL-6) is significantly increased in pericyte conditions. Blocking it with siltuximab results in a reduction of pericyte vasculogenic potential comparable to stromal cell levels, revealing that pericyte pro-vascularization is mediated by IL-6. This study provides new insights into the relationship between pericytes and endothelial cells and the elusive identity of mesenchymal stromal cells. These findings are relevant for both vascular biology and tissue engineering.
Collapse
Affiliation(s)
- Julian Gonzalez‐Rubio
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
| | - Hannah Kubiza
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
| | - Yong Xu
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
| | - Hiltrud Koenigs‐Werner
- Institute of PathologyElectron Microscopy FacilityRWTH Aachen University HospitalAachen, Pauwelsstrasse 3052074AachenGermany
| | - Mona Sophie Schmitz
- Department of Pulmonary MedicineUniversity Medicine Essen‐RuhrlandklinikTueschener Weg 4045239EssenGermany
- Department of Pulmonary MedicineUniversity Medicine EssenEssen, Hufelandstraße 5545147EssenGermany
| | - Michaela Schedel
- Department of Pulmonary MedicineUniversity Medicine Essen‐RuhrlandklinikTueschener Weg 4045239EssenGermany
- Department of Pulmonary MedicineUniversity Medicine EssenEssen, Hufelandstraße 5545147EssenGermany
| | - Christian Apel
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
| | - Stefan Jockenhoevel
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
| | - Christian G. Cornelissen
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
- Clinic for Pneumology and Internal Intensive Care Medicine (Medical Clinic V)RWTH Aachen University HospitalPauwelsstrasse 3052074AachenGermany
| | - Anja Lena Thiebes
- Department of Biohybrid & Medical Textiles (BioTex)AME – Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstrasse 5552074AachenGermany
| |
Collapse
|
18
|
Catalano A, Haas LS, Zodel K, Adlesic M, Cuomo F, Peighambari A, Metzger P, Huang H, Haug S, Köttgen A, Köhler N, Boerries M, Frew IJ. Mutations in tumor suppressor genes Vhl and Rassf1a cause DNA damage, chromosomal instability and induce gene expression changes characteristic of clear cell renal cell carcinoma. Kidney Int 2025; 107:666-686. [PMID: 39725222 DOI: 10.1016/j.kint.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 11/06/2024] [Accepted: 12/04/2024] [Indexed: 12/28/2024]
Abstract
RASSF1A is frequently biallelically inactivated in clear cell renal cell carcinoma (ccRCC) due to loss of chromosome 3p and promoter hypermethylation. Here we investigated the cellular and molecular consequences of single and combined deletion of the Rassf1a and Vhl tumor suppressor genes to model the common ccRCC genotype of combined loss of function of RASSF1A and VHL. In mouse embryonic fibroblasts and in primary kidney epithelial cells, double deletion of Rassf1a and Vhl caused chromosomal segregation defects and increased formation of micronuclei, demonstrating that pVHL and RASSF1A function to maintain genomic integrity. Combined Rassf1a and Vhl deletion in kidney epithelial cells in vivo increased proliferation and caused mild tubular disorganization, but did not lead to the development of kidney tumors. Single cell RNA-sequencing unexpectedly revealed that Rassf1a or Vhl deletion both induce the expression of an overlapping set of genes in a sub-population of proximal tubule cells. Many of these genes are also upregulated in the Vhl/Trp53/Rb1 deficient mouse model of ccRCC. In other subsets of proximal tubule cells, combined Vhl/Rassf1a deletion induced the expression of additional genes that were not upregulated in each of the single knockouts. The expression of the human homologues of Rassf1a-regulated genes correlate negatively with RASSF1 expression levels in human ccRCC. Our results suggest that the loss of RASSF1A function establishes a ccRCC-characteristic gene expression pattern.
Collapse
Affiliation(s)
- Antonella Catalano
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Laura S Haas
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kyra Zodel
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mojca Adlesic
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Francesca Cuomo
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Asin Peighambari
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Patrick Metzger
- Institute of Medical Bioinformatics and Systems Medicine, Medical Centre-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hsin Huang
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Stefan Haug
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center-University of Freiburg, Freiburg, Germany
| | - Natalie Köhler
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Centre-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Comprehensive Cancer Center Freiburg (CCCF), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Ian J Frew
- Clinic of Internal Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Centre - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Comprehensive Cancer Center Freiburg (CCCF), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, Faculty of Biology University of Freiburg, Freiburg, Germany.
| |
Collapse
|
19
|
Zhou M, Yang Y, He S, Xu Q, Du C, Tian W, Chen H. Ingenane Diterpenoids from Euphorbia peplus as Potential New CHK1 Inhibitors That Sensitize Cancer Cells to Chemotherapy. JOURNAL OF NATURAL PRODUCTS 2025; 88:688-705. [PMID: 40056138 DOI: 10.1021/acs.jnatprod.4c01343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/10/2025]
Abstract
Phosphorylation of checkpoint kinase 1 at Ser-345 (p-CHK1(S345)) mediates the replication stress response in cancer cells, leading to chemotherapy resistance. Therefore, finding inhibitors of p-CHK1(S345) is currently a promising strategy to prevent acquired drug resistance. In this study, 14 ingenane diterpenoids (1-14), involving two undescribed compounds possessing an unprecedented exocyclic double bond Δ6(20), were identified from Euphorbia peplus. The inhibitory effects of the isolated compounds on p-CHK1(S345) and their structure-activity relationship (SAR) were investigated. Compounds 7 and 8 presented the strongest inhibitory effects, abrogated cell cycle arrest, and caused the accumulation of DNA damage, improving the sensitivity of cancer cells to chemotherapeutic drugs. An in vivo assay confirmed the enhancement of 8 on the anticancer effect of topotecan via blocking of p-CHK1(S345). Mechanistically, 8 increased CHK1 ubiquitination to inhibit p-CHK1(S345) via activation of protein kinase C (PKC). PKC activation was first found to enhance CHK1 ubiquitination to block p-CHK1(S345). Above all, this finding not only indicates that compound 8 could be developed as a novel CHK1 inhibitor but also reveals a previously unrecognized role of PKC in regulating cancer chemotherapy sensitivity.
Collapse
Affiliation(s)
- Mi Zhou
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
- Guangxi Key Laboratory of Marine Drugs, Institute of Marine Drugs, Guangxi University of Chinese Medicine, Nanning 530200, China
| | - Yanlan Yang
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
| | - Shoulun He
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
| | - Qiannan Xu
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
| | - Chunchun Du
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
| | - Wenjing Tian
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
| | - Haifeng Chen
- School of Pharmaceutical Science, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen University, Xiamen 361002, China
| |
Collapse
|
20
|
Musiani D, Yücel H, Vallette M, Angrisani A, El Botty R, Ouine B, Schintu N, Adams C, Chevalier M, Heloise D, El Marjou A, Nemazanyy I, Regairaz M, Marangoni E, Fachinetti D, Ceccaldi R. Uracil processing by SMUG1 in the absence of UNG triggers homologous recombination and selectively kills BRCA1/2-deficient tumors. Mol Cell 2025; 85:1072-1084.e10. [PMID: 40010343 DOI: 10.1016/j.molcel.2025.01.031] [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: 03/22/2024] [Revised: 01/10/2025] [Accepted: 01/30/2025] [Indexed: 02/28/2025]
Abstract
Resistance to poly (ADP-ribose) polymerase (PARP) inhibitors (PARPis) is the major obstacle to their effectiveness in the treatment of homologous recombination (HR)-deficient (HRD) tumors. Hence, developing alternative treatments for HRD tumors is critical. Here, we show that targeting the uracil excision pathway kills HRD tumors, including those with PARPi resistance. We found that the interplay between the two major uracil DNA glycosylases UNG and SMUG1 is regulated by nuclear nicotinamide adenine dinucleotide (NAD+), which maintains UNG at replication forks (RFs) and restrains SMUG1 chromatin binding. In the absence of UNG, SMUG1 retention on chromatin leads to persistent abasic sites, which incision by APE1 results in PARP1 hyperactivation, stalled RFs, and RAD51 foci. In HRD cells (i.e., BRCA1/2-deficient), this leads to under-replicated DNA that, when propagated throughout mitosis, results in chromosome fragmentation and cell death. Our findings open up unique possibilities for targeted therapies for HRD tumors based on UNG inhibition and uracil accumulation in the genome.
Collapse
Affiliation(s)
- Daniele Musiani
- INSERM U830, PSL Research University, Institut Curie, Paris, France
| | - Hatice Yücel
- INSERM U830, PSL Research University, Institut Curie, Paris, France
| | - Marie Vallette
- INSERM U830, PSL Research University, Institut Curie, Paris, France
| | - Annapaola Angrisani
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR144 and UMR3664, 26 rue d'Ulm, 75005 Paris, France
| | - Rania El Botty
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, Paris, France
| | - Bérengère Ouine
- Recombinant Protein Facility CNRS UMR144, Institut Curie, PSL Research University, Paris, France
| | - Niccolo Schintu
- INSERM U830, PSL Research University, Institut Curie, Paris, France
| | - Caroline Adams
- INSERM U830, PSL Research University, Institut Curie, Paris, France
| | - Manon Chevalier
- INSERM U830, PSL Research University, Institut Curie, Paris, France
| | - Derrien Heloise
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, Paris, France
| | - Ahmed El Marjou
- Recombinant Protein Facility CNRS UMR144, Institut Curie, PSL Research University, Paris, France
| | - Ivan Nemazanyy
- Platform for Metabolic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Marie Regairaz
- INSERM U830, PSL Research University, Institut Curie, Paris, France; Laboratoire de Biologie et Pharmacologie Appliquée, ENS-Paris-Saclay, CNRS UMR 8113, Université Paris-Saclay, 91190 Gif-sur-Yvette, France
| | - Elisabetta Marangoni
- Laboratory of Preclinical Investigation, Translational Research Department, Institut Curie, PSL Research University, Paris, France
| | - Daniele Fachinetti
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR144 and UMR3664, 26 rue d'Ulm, 75005 Paris, France
| | - Raphael Ceccaldi
- INSERM U830, PSL Research University, Institut Curie, Paris, France.
| |
Collapse
|
21
|
Wang YT, Hsiao WY, Pham TV, Huang BR, Yeh SD, Hsu EC, Wang SW. An enzymatic-independent function of palmitoyl hydrolase in cohesin loading onto chromosome. Nucleic Acids Res 2025; 53:gkaf257. [PMID: 40193710 PMCID: PMC11975282 DOI: 10.1093/nar/gkaf257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 02/25/2025] [Accepted: 03/21/2025] [Indexed: 04/09/2025] Open
Abstract
Sister chromatid cohesion is mediated by a conserved multiprotein complex called cohesin. The loading of cohesin onto chromosomes involves the RSC (remodels the structure of chromatin) chromatin remodeling complex. Here, we demonstrate that the fission yeast Phi1, a palmitoyl hydrolase inactive protein 1, serves to bridge the interaction between cohesin and the RSC complex. Phi1 interacts with Rad21 in cohesin and Snf21, the RSC complex ATPase, to promote chromosome loading of cohesin. The identified characteristic features of Phi1 are conserved in the human homologues Apt1 and Apt2, which interact with Rad21 and Brg1, the human homologue of Snf21, in an enzymatic-independent manner. Intriguingly, the cohesin-Apt1-Brg1 complex is upregulated in C4-2B prostate cancer cells, and co-depletion of Apt1 and Apt2 by small interfering RNA triggers mitotic catastrophe in these cells. In addition, Apt1 nuclear localization is associated with poor clinical outcomes in prostate cancer. These results suggest a pro-survival function against mitotic stress for the complex.
Collapse
Affiliation(s)
- Yi-Ting Wang
- Institute of Molecular & Genomic Medicine, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Wan-Yi Hsiao
- Institute of Molecular & Genomic Medicine, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Thanh-Vy Pham
- Institute of Molecular & Genomic Medicine, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
- Department of Life Sciences, National Central University, Taoyuan City 320, Taiwan
| | - Bo-Ru Huang
- Institute of Molecular & Genomic Medicine, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Shu-Dan Yeh
- Department of Life Sciences, National Central University, Taoyuan City 320, Taiwan
| | - En-Chi Hsu
- Institute of Molecular & Genomic Medicine, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| | - Shao-Win Wang
- Institute of Molecular & Genomic Medicine, National Health Research Institutes, Zhunan Town, Miaoli County 350, Taiwan
| |
Collapse
|
22
|
Frigerio C, Galli M, Castelli S, Da Prada A, Clerici M. Control of Replication Stress Response by Cytosolic Fe-S Cluster Assembly (CIA) Machinery. Cells 2025; 14:442. [PMID: 40136691 PMCID: PMC11941123 DOI: 10.3390/cells14060442] [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: 02/19/2025] [Revised: 03/11/2025] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
Accurate DNA replication is essential for the maintenance of genome stability and the generation of healthy offspring. When DNA replication is challenged, signals accumulate at blocked replication forks that elicit a multifaceted cellular response, orchestrating DNA replication, DNA repair and cell cycle progression. This replication stress response promotes the recovery of DNA replication, maintaining chromosome integrity and preventing mutations. Defects in this response are linked to heightened genetic instability, which contributes to tumorigenesis and genetic disorders. Iron-sulfur (Fe-S) clusters are emerging as important cofactors in supporting the response to replication stress. These clusters are assembled and delivered to target proteins that function in the cytosol and nucleus via the conserved cytosolic Fe-S cluster assembly (CIA) machinery and the CIA targeting complex. This review summarizes recent advances in understanding the structure and function of the CIA machinery in yeast and mammals, emphasizing the critical role of Fe-S clusters in the replication stress response.
Collapse
Affiliation(s)
| | | | | | | | - Michela Clerici
- Dipartimento di Biotecnologie e Bioscienze, Università degli Studi di Milano-Bicocca, 20126 Milano, Italy; (C.F.); (M.G.); (S.C.); (A.D.P.)
| |
Collapse
|
23
|
Molano-Fernández M, Hickson ID, Herranz H. Replication stress promotes cellular transformation in Drosophila epithelium. Cell Death Discov 2025; 11:96. [PMID: 40075075 PMCID: PMC11904189 DOI: 10.1038/s41420-025-02383-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/11/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
The accurate control of DNA replication is crucial for the maintenance of genomic stability and cell viability. In this study, we explore the consequences of depleting the replicative DNA Polymerase α (POLA) in the wing disc of Drosophila melanogaster. Our findings reveal that reduced POLA activity induces DNA replication stress and activates the replication checkpoint in vivo. Consistent with this, we demonstrate that dATR, a key component in DNA replication checkpoint signaling, is essential for the maintenance of tissue integrity under conditions of compromised POLA activity. We show that cells within the wing disc exhibiting reduced POLA activity arrest in the G2 phase and undergo p53-dependent apoptosis. We also reveal a critical role for DNA Ligase 4 in sustaining cell viability when POLA function is impaired. Most notably, we report the appearance of oncogenic traits in wing disc cells with diminished POLA activity when apoptosis is suppressed. In this context, the overexpression of the oncogene cdc25/string enhances the oncogenic phenotype. These results indicate that a combination of oncogenic activation, replication stress, and suppression of apoptosis is sufficient to promote the emergence of hallmarks of tumorigenesis, highlighting major implications for cancer development in humans.
Collapse
Affiliation(s)
- Maria Molano-Fernández
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Ian D Hickson
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.
- Department of Cellular and Molecular Medicine, Center for Chromosome Stability and Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark.
| | - Héctor Herranz
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
24
|
Nakatani T. Dynamics of replication timing during mammalian development. Trends Genet 2025:S0168-9525(25)00026-5. [PMID: 40082146 DOI: 10.1016/j.tig.2025.01.010] [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: 11/06/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 03/16/2025]
Abstract
Recent developments in low-input genomics techniques have greatly advanced the analysis of the order in which DNA is replicated in the genome - that is, replication timing (RT) - and its interrelationships with other processes. RT correlates or anticorrelates with genomic-specific parameters such as gene expression, chromatin accessibility, histone modifications, and the 3D structure of the genome, but the significance of how they influence each other and how they relate to biological processes remains unclear. In this review I discuss the results of recent analyses of RT, the time at which it is remodeled and consolidated during embryogenesis, how it influences development and differentiation, and the regulatory mechanisms and factors involved.
Collapse
Affiliation(s)
- Tsunetoshi Nakatani
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum München, D-81377, München, Germany.
| |
Collapse
|
25
|
Batra S, Allwein B, Kumar C, Devbhandari S, Brüning JG, Bahng S, Lee CM, Marians KJ, Hite RK, Remus D. G-quadruplex-stalled eukaryotic replisome structure reveals helical inchworm DNA translocation. Science 2025; 387:eadt1978. [PMID: 40048517 DOI: 10.1126/science.adt1978] [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] [Accepted: 12/15/2024] [Indexed: 03/15/2025]
Abstract
DNA G-quadruplexes (G4s) are non-B-form DNA secondary structures that threaten genome stability by impeding DNA replication. To elucidate how G4s induce replication fork arrest, we characterized fork collisions with preformed G4s in the parental DNA using reconstituted yeast and human replisomes. We demonstrate that a single G4 in the leading strand template is sufficient to stall replisomes by arresting the CMG helicase. Cryo-electron microscopy structures of stalled yeast and human CMG complexes reveal that the folded G4 is lodged inside the central CMG channel, arresting translocation. The G4 stabilizes the CMG at distinct translocation intermediates, suggesting an unprecedented helical inchworm mechanism for DNA translocation. These findings illuminate the eukaryotic replication fork mechanism under normal and perturbed conditions.
Collapse
Affiliation(s)
- Sahil Batra
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benjamin Allwein
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- BCMB Allied PhD Program, Weill Cornell Medical Graduate School, Weill Cornell Medicine, New York, NY, USA
| | - Charanya Kumar
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sujan Devbhandari
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jan-Gert Brüning
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Soon Bahng
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chong M Lee
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth J Marians
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard K Hite
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dirk Remus
- Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
26
|
Chen Z, Wu F, Li Y, Li L, Lei Y, Gao S, Chen T, Xie Y, Xiao J, Zeng H, Deng J, Zhao X, Hou Y. Inhibition of DEK restores hematopoietic stem cell function in Fanconi anemia. J Exp Med 2025; 222:e20241248. [PMID: 39836085 PMCID: PMC11748990 DOI: 10.1084/jem.20241248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/11/2024] [Accepted: 12/19/2024] [Indexed: 01/30/2025] Open
Abstract
Hematopoietic stem cells (HSCs) are susceptible to replication stress, which is a major contributor to HSC defects in Fanconi anemia (FA). Here, we report that HSCs relax the global chromatin by downregulating the expression of a chromatin architectural protein, DEK, in response to replication stress. DEK is abnormally accumulated in bone marrow (BM) CD34+ cells from patients with FA and in Fancd2-deficient HSCs. DEK haploinsufficiency promotes chromatin relaxation, replication stress relief, and function recovery of Fancd2-deficient HSCs. Furthermore, inhibition of DEK restores the proliferation of FA CD34+ cells in vitro and enhances their engraftment in vivo. Mechanistically, the activating transcription factor 2 (ATF2), specifically phosphorylated ATF2 at Thr69/71, was identified as a promoter of DEK transcription. Fancd2 deficiency results in p38 hyperphosphorylation, which in turn phosphorylates ATF2 at Thr69/71, leading to DEK accumulation in HSCs. In conclusion, our findings establish a functional link between chromatin relaxation and replication stress tolerance in HSCs and highlight DEK as a target for FA.
Collapse
Affiliation(s)
- Zhe Chen
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Feng Wu
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Yan Li
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Lei Li
- School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Yufei Lei
- School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Siwei Gao
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Tao Chen
- School of Medicine, Guizhou University, Guiyang, China
| | - Yuxin Xie
- School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Jianwen Xiao
- Department of Hematology and Oncology, Children’s Hospital, Chongqing Medical University, Chongqing, China
| | - Hanqing Zeng
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Jianchuan Deng
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Xueya Zhao
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Yu Hou
- Department of Hematology, The Second Affiliated Hospital of Chongqing Medical University, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing, China
| |
Collapse
|
27
|
Huang S, Soto AM, Sonnenschein C. The end of the genetic paradigm of cancer. PLoS Biol 2025; 23:e3003052. [PMID: 40100793 DOI: 10.1371/journal.pbio.3003052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
Genome sequencing of cancer and normal tissues, alongside single-cell transcriptomics, continues to produce findings that challenge the idea that cancer is a 'genetic disease', as posited by the somatic mutation theory (SMT). In this prevailing paradigm, tumorigenesis is caused by cancer-driving somatic mutations and clonal expansion. However, results from tumor sequencing, motivated by the genetic paradigm itself, create apparent 'paradoxes' that are not conducive to a pure SMT. But beyond genetic causation, the new results lend credence to old ideas from organismal biology. To resolve inconsistencies between the genetic paradigm of cancer and biological reality, we must complement deep sequencing with deep thinking: embrace formal theory and historicity of biological entities, and (re)consider non-genetic plasticity of cells and tissues. In this Essay, we discuss the concepts of cell state dynamics and tissue fields that emerge from the collective action of genes and of cells in their morphogenetic context, respectively, and how they help explain inconsistencies in the data in the context of SMT.
Collapse
Affiliation(s)
- Sui Huang
- Institute for Systems Biology, Seattle, Washington, United States of America
| | - Ana M Soto
- Tufts University School of Medicine, Immunology, Boston, Massachusetts, United States of America
| | - Carlos Sonnenschein
- Tufts University School of Medicine, Immunology, Boston, Massachusetts, United States of America
| |
Collapse
|
28
|
Françon A, Behar-Cohen F, Torriglia A. Wavelength-dependency of the impact of light on proliferation and DNA damage of corneal cells in vitro. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2025; 264:113118. [PMID: 39922040 DOI: 10.1016/j.jphotobiol.2025.113118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/20/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025]
Abstract
The wavelength-dependent impact of light has been mainly studied focusing on retina. In particular, an opposite effect of the two ends of the visible spectrum was observed, with blue wavelengths being harmful and red wavelengths being protective. However, few studies on the cornea indicate that the increasing exposition to artificial light due to digital devices is linked to an increase in computer vision syndrome affecting the cornea. In this study, we aim at deciphering the impact of blue and red LED light on a primary culture of corneal endothelial cells, by looking at cell death and proliferation, and at DNA replication and DNA breaks. Our results show that exposure to blue light at 5.35 J/cm2 (455 nm) induces the inhibition of DNA replication and cell proliferation, and the formation of DNA breaks, highlighted by the formation of γH2AX foci and DNA fragmentation. Addition of red light at 0.3 J/cm2 (630 nm) to blue light mitigates the formation of DNA damage and delays the kinetics of formation and repair of the damage. Interestingly, exposure of the corneal cells to red light alone induces the formation of γH2AX foci that do not correspond to DNA breaks, but to DNA replication forks in proliferative cells. Our results highlight the wavelength-dependent effect of light on the cornea, and point out that the formation of γH2AX foci is not always representative of DNA breaks. This emphasizes the importance of light spectrum in eye health, an important issue in today's changing light environment.
Collapse
Affiliation(s)
- Anaïs Françon
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université, Team: Physiopathology of Ocular Diseases: Therapeutic Innovations, 15, rue de l'école de Médecine, 75006 Paris, France
| | - Francine Behar-Cohen
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université, Team: Physiopathology of Ocular Diseases: Therapeutic Innovations, 15, rue de l'école de Médecine, 75006 Paris, France; Assistance Publique, Hôpitaux de Paris, Hôpital Cochin, Ophtalmopole, 27, rue du Faubourg Saint-Jacques, 75014 Paris, France
| | - Alicia Torriglia
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Université Paris Cité, Sorbonne Université, Team: Physiopathology of Ocular Diseases: Therapeutic Innovations, 15, rue de l'école de Médecine, 75006 Paris, France.
| |
Collapse
|
29
|
Letai A, de The H. Conventional chemotherapy: millions of cures, unresolved therapeutic index. Nat Rev Cancer 2025; 25:209-218. [PMID: 39681637 DOI: 10.1038/s41568-024-00778-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
In recent decades, millions of patients with cancer have been cured by chemotherapy alone. By 'cure', we mean that patients with cancers that would be fatal if left untreated receive a time-limited course of chemotherapy and their cancer disappears, never to return. In an era when hundreds of thousands of cancer genomes have been sequenced, a remarkable fact persists: in most patients who have been cured, we still do not fully understand the mechanisms underlying the therapeutic index by which the tumour cells are killed, but normal cells are somehow spared. In contrast, in more recent years, patients with cancer have benefited from targeted therapies that usually do not cure but whose mechanisms of therapeutic index are, at least superficially, understood. In this Perspective, we will explore the various and sometimes contradictory models that have attempted to explain why chemotherapy can cure some patients with cancer, and what gaps in our understanding of the therapeutic index of chemotherapy remain to be filled. We will summarize principles which have benefited curative conventional chemotherapy regimens in the past, principles which might be deployed in constructing combinations that include modern targeted therapies.
Collapse
Affiliation(s)
- Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| | - Hugues de The
- College de France, CIRB, INSERM, CNRS, Université PSL Paris, Paris, France.
- Hematology Laboratory, St Louis Hospital, Assistance Publique Hôpitaux de Paris, Paris, France.
- IRSL, INSERM, CNRS, Université Paris-Cité, Paris, France.
| |
Collapse
|
30
|
Glaviano A, Singh SK, Lee EHC, Okina E, Lam HY, Carbone D, Reddy EP, O'Connor MJ, Koff A, Singh G, Stebbing J, Sethi G, Crasta KC, Diana P, Keyomarsi K, Yaffe MB, Wander SA, Bardia A, Kumar AP. Cell cycle dysregulation in cancer. Pharmacol Rev 2025; 77:100030. [PMID: 40148026 DOI: 10.1016/j.pharmr.2024.100030] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/12/2024] [Indexed: 03/29/2025] Open
Abstract
Cancer is a systemic manifestation of aberrant cell cycle activity and dysregulated cell growth. Genetic mutations can determine tumor onset by either augmenting cell division rates or restraining normal controls such as cell cycle arrest or apoptosis. As a result, tumor cells not only undergo uncontrolled cell division but also become compromised in their ability to exit the cell cycle accurately. Regulation of cell cycle progression is enabled by specific surveillance mechanisms known as cell cycle checkpoints, and aberrations in these signaling pathways often culminate in cancer. For instance, DNA damage checkpoints, which preclude the generation and augmentation of DNA damage in the G1, S, and G2 cell cycle phases, are often defective in cancer cells, allowing cell division in spite of the accumulation of genetic errors. Notably, tumors have evolved to become dependent on checkpoints for their survival. For example, checkpoint pathways such as the DNA replication stress checkpoint and the mitotic checkpoint rarely undergo mutations and remain intact because any aberrant activity could result in irreparable damage or catastrophic chromosomal missegregation leading to cell death. In this review, we initially focus on cell cycle control pathways and specific functions of checkpoint signaling involved in normal and cancer cells and then proceed to examine how cell cycle control and checkpoint mechanisms can provide new therapeutic windows that can be exploited for cancer therapy. SIGNIFICANCE STATEMENT: DNA damage checkpoints are often defective in cancer cells, allowing cell division in spite of the accumulation of genetic errors. Conversely, DNA replication stress and mitotic checkpoints rarely undergo mutations because any aberrant activity could result in irreparable damage or catastrophic chromosomal missegregation, leading to cancer cell death. This review focuses on the checkpoint signaling mechanisms involved in cancer cells and how an emerging understanding of these pathways can provide new therapeutic opportunities for cancer therapy.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Samarendra K Singh
- School of Biotechnology, Institute of Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - E Premkumar Reddy
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mark J O'Connor
- Discovery Centre, AstraZeneca, Francis Crick Avenue, Cambridge CB2 0AA, United Kingdom
| | - Andrew Koff
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York
| | - Garima Singh
- School of Biotechnology, Institute of Sciences, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Justin Stebbing
- School of Life Sciences, Anglia Ruskin University, Cambridge, United Kingdom
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Karen Carmelina Crasta
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Centre for Healthy Longevity, National University Health System, Singapore, Singapore
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, Palermo, Italy
| | - Khandan Keyomarsi
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Michael B Yaffe
- MIT Center for Precision Cancer Medicine, Koch Institute for Integrative Cancer Research, Broad Institute, Massachusetts Institute of Technology, Cambridge, Boston, Massachusetts
| | - Seth A Wander
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aditya Bardia
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
31
|
Shao C, Zhang Y, Li H, Chen J, Huang T, Li J, Wen S, Wang S, Fan S, Zhao Y. Radiotherapy-resistant prostate cancer cells escape immune checkpoint blockade through the senescence-related ataxia telangiectasia and Rad3-related protein. Cancer Commun (Lond) 2025; 45:218-244. [PMID: 39698847 PMCID: PMC11947621 DOI: 10.1002/cac2.12636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 10/31/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND The majority of patients with prostate cancer (PCa) exhibit intrinsic resistance to immune checkpoint blockade (ICB) following radiotherapy (RT). This resistance is generally attributed to the limited antigen presentation of heterogeneous cells within tumors. Here, we aimed to isolate and characterize these diverse subgroups of tumor post-RT to understand the molecular mechanisms of their resistance to ICB. METHODS Single-cell RNA-sequencing (scRNA-seq) was used to profile senescent cancer cell clusters induced by RT in LNCaP cells. The expression and phosphorylation levels of ataxia telangiectasia and Rad3-related protein (ATR) were assessed by immunohistochemistry in clinical samples from patients with or without RT. Co-immunoprecipitation, mutagenesis, and Western blotting were used to measure the interactions between proteins. Xenograft experiments were performed to assess the tumor immune response in the mice. RESULTS We identified a subset of PCa cells that exhibited resistance to RT, characterized by a reduced antigen presentation capability, which enhanced their ability to evade immune detection and resist cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) blockade. scRNA-seq revealed that the senescent state was a transient phase of PCa cells post-RT, particularly in CTLA-4 blockade treatment-resistant cells. This state was marked by increased cytosolic ATR level. Cytosolic ATR phosphorylated CD86 in its cytosolic domain and enhanced the interaction between CD86 and its E3 ligase MARCH1 through electrostatic attraction. Depletion or inhibition of Atr increased the sensitivity to immune attack and improved responses to anti-Ctla-4 antibody treatment in a mouse model. CONCLUSIONS Our findings indicate that the activation of cytosolic ATR, which is associated with cellular senescence, impedes the effectiveness of combined RT and ICB treatments. This discovery may provide valuable insights for improving the efficacy of combined RT and ICB therapies in PCa.
Collapse
Affiliation(s)
- Chenyi Shao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinP. R. China
| | - Yingyi Zhang
- School of Disaster and Emergency MedicineTianjin UniversityTianjinP. R. China
- Department of Biochemistry and Molecular BiologyMayo Clinic College of MedicineRochesterMinnesotaUSA
| | - Hang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinP. R. China
| | - Jiajia Chen
- Department of OncologyShengjing Hospital of China Medical UniversityShenyangLiaoningP. R. China
| | - Ting Huang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinP. R. China
| | - Jiaze Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinP. R. China
| | - Simeng Wen
- Department of UrologyThe Second Hospital of Tianjin Medical UniversityTianjin Medical UniversityTianjinP. R. China
| | - Sen Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinP. R. China
| | - Saijun Fan
- Institute of Radiation MedicineChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinP. R. China
| | - Yu Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation MedicineChinese Academy of Medical Sciences and Peking Union Medical CollegeTianjinP. R. China
| |
Collapse
|
32
|
Jia X, Zhu X, Chen S, Qiu Q, Song W, Zhang S, Dong H, Li Z, Bian S, Wu H, Dai H, Jin C, Zhou M, Chen J, Xuan Z, Liu P, Zeng Q, Xie H, Zheng S, Song P. Comprehensive multi-omics analyses expose a precision therapy strategy that targets replication stress in hepatocellular carcinoma using WEE1 inhibition. J Adv Res 2025:S2090-1232(25)00114-6. [PMID: 39978541 DOI: 10.1016/j.jare.2025.02.016] [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: 07/31/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025] Open
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is an extremely heterogeneous malignancy with a poor prognosis, highlighting the need to target specific vulnerabilities within the tumor during treatment. OBJECTIVES This study employs multi-omics analysis techniques to provide novel insights into personalized therapeutic strategies for HCC patients. METHODS We performed proteomic and transcriptomic sequencing on 178 and 94 clinical samples of primary HCC without prior treatment, respectively. We employed an unbiased Kinome CRISPR-Cas9 library screening approach to systematically evaluate and identify novel therapeutic strategies that specifically target replication stress (RS). The synergy between oxaliplatin and adavosertib was verified using in vitro and in vivo models, including hydrodynamic injection, patient-derived organoids, and patient-derived xenografts. RESULTS In both proteomic- and transcriptomic-based subtyping analyses, subtypes characterized by hyperproliferative features demonstrated the poorest prognosis and the highest levels of RS. Among all first-line chemotherapeutic agents in these analyses, oxaliplatin accumulated the highest RS levels in HCC, while resistance remained a major challenge. With unbiased Kinome CRISPR loss-of-function gene screening, WEE1 was identified as a synthetic lethal target of oxaliplatin. The synergy between the WEE1 inhibitor adavosertib and oxaliplatin has been demonstrated in multiple in vitro and in vivo models. Mechanistically, adavosertib inhibits oxaliplatin-induced homologous recombination repair and G2/M checkpoint activation, leading to the accumulation of lethal DNA damage. Furthermore, patients with HCC showing high RS levels had poor prognoses and responded well to adavosertib and oxaliplatin combination treatments. This was validated by preclinical models and unsupervised clustering analysis. CONCLUSIONS Our findings provide promising insights into the precise therapeutic targeting of RS in HCC at both the proteomic and transcriptomic levels. Furthermore, our study highlights the potential of combining oxaliplatin with adavosertib as a treatment approach for HCC. In this study, we analyzed 178 and 94 pairs of clinical HCC samples using proteomic and transcriptomic sequencing, respectively. We discovered that the subtype characterized by high proliferation had the worst prognosis and highest RS level. Drug screening revealed that oxaliplatin promotes RS accumulation in HCC, but its resistance remains a challenge. Through unbiased CRISPR deletion-gene screening, WEE1 was identified as a lethal target of oxaliplatin. The WEE1 inhibitor adavosertib inhibits oxaliplatin-induced DNA repair, leading to lethal DNA damage accumulation. Furthermore, our clustering analysis based on RS levels demonstrated that HCC patients with high RS levels have poorer prognoses and be more beneficial from adavosertib and oxaliplatin combination therapy. These findings support an individualized treatment approach for HCC targeting RS based on WEE1 Inhibition.
Collapse
Affiliation(s)
- Xing Jia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Thyroid Surgery Department, Zhejiang Cancer Hospital, Hangzhou 310022 Zhejiang, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Xingxin Zhu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Shinuo Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Qiongzi Qiu
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Women's Reproductive Health Key Laboratory of Zhejiang Province, and Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou 310029 Zhejiang, China
| | - Wenfeng Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Shiyu Zhang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Haijiang Dong
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052 Henan, China
| | - Zequn Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Suchen Bian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Hao Wu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Haojiang Dai
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Cheng Jin
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Mengqiao Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Jun Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Zefeng Xuan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Pengfei Liu
- Shanghai Applied Protein Technology Co. Ltd, Shanghai 200000, China
| | - Qiufang Zeng
- Shanghai Applied Protein Technology Co. Ltd, Shanghai 200000, China
| | - Haiyang Xie
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China; Department of Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital, Hangzhou 310003 Zhejiang, China.
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; NHC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou 310003, China; Key Laboratory of the Diagnosis and Treatment of Organ Transplantation, Research Unit of Collaborative Diagnosis and Treatment for Hepatobiliary and Pancreatic Cancer, Chinese Academy of Medical Sciences (2019RU019), Hangzhou 310003, China; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou 310003, China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China.
| |
Collapse
|
33
|
Vasudevarao MD, Posadas Pena D, Ihle M, Bongiovanni C, Maity P, Geissler D, Mohammadi HF, Rall-Scharpf M, Niemann J, Mommersteeg MTM, Redaelli S, Happ K, Wu CC, Beisaw A, Scharffetter-Kochanek K, D'Uva G, Malek Mohammadi M, Wiesmüller L, Geiger H, Weidinger G. BMP signaling promotes zebrafish heart regeneration via alleviation of replication stress. Nat Commun 2025; 16:1708. [PMID: 39962064 PMCID: PMC11832743 DOI: 10.1038/s41467-025-56993-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
In contrast to mammals, adult zebrafish achieve complete heart regeneration via proliferation of cardiomyocytes. Surprisingly, we found that regenerating cardiomyocytes experience DNA replication stress, which represents one reason for declining tissue regeneration during aging in mammals. Pharmacological inhibition of ATM and ATR kinases revealed that DNA damage response signaling is essential for zebrafish heart regeneration. Manipulation of Bone Morphogenetic Protein (BMP)-Smad signaling using transgenics and mutants showed that BMP signaling alleviates cardiomyocyte replication stress. BMP signaling also rescues neonatal mouse cardiomyocytes, human fibroblasts and human hematopoietic stem and progenitor cells (HSPCs) from replication stress. DNA fiber spreading assays indicate that BMP signaling facilitates re-start of replication forks after replication stress-induced stalling. Our results identify the ability to overcome replication stress as key factor for the elevated zebrafish heart regeneration capacity and reveal a conserved role for BMP signaling in promotion of stress-free DNA replication.
Collapse
Affiliation(s)
| | - Denise Posadas Pena
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Michaela Ihle
- Department of Obstetrics and Gynecology, Ulm University, Prittwitzstraße 43, 89075, Ulm, Germany
| | - Chiara Bongiovanni
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, via Massarenti 9, 40138, Bologna, Italy
| | - Pallab Maity
- Department of Dermatology and Allergic Diseases, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Dominik Geissler
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Hossein Falah Mohammadi
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Melanie Rall-Scharpf
- Department of Obstetrics and Gynecology, Ulm University, Prittwitzstraße 43, 89075, Ulm, Germany
| | - Julian Niemann
- Institute of Molecular Medicine, Ulm University, Meyerhofstrasse N27, 89081, Ulm, Germany
| | - Mathilda T M Mommersteeg
- Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford, OX1 3PT, United Kingdom
| | - Simone Redaelli
- Institute of Biomedical Ethics and History of Medicine, University of Zurich, Winterthurerstrasse 30, 8006, Zurich, Switzerland
| | - Kathrin Happ
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Chi-Chung Wu
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Arica Beisaw
- Institute of Experimental Cardiology, Heidelberg University, Im Neuenheimer Feld 669, 69120, Heidelberg, Germany
| | - Karin Scharffetter-Kochanek
- Department of Dermatology and Allergic Diseases, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Gabriele D'Uva
- Department of Medical and Surgical Sciences, University of Bologna, via Massarenti 9, 40138, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, via Massarenti 9, 40138, Bologna, Italy
| | - Mona Malek Mohammadi
- Institute of Physiology I, Medical Faculty, University of Bonn, Venusberg-Campus 1, 53127, Bonn, Germany
| | - Lisa Wiesmüller
- Department of Obstetrics and Gynecology, Ulm University, Prittwitzstraße 43, 89075, Ulm, Germany
| | - Hartmut Geiger
- Institute of Molecular Medicine, Ulm University, Meyerhofstrasse N27, 89081, Ulm, Germany
| | - Gilbert Weidinger
- Institute of Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
34
|
Ziener J, Henao-Restrepo JA, Leonhardi J, Sturm MJ, Becker S, Morales-Prieto DM, Milde T, Beck JF, Sonnemann J. Combined inhibition of ribonucleotide reductase and WEE1 induces synergistic anticancer activity in Ewing's sarcoma cells. BMC Cancer 2025; 25:277. [PMID: 39962391 PMCID: PMC11831844 DOI: 10.1186/s12885-025-13691-2] [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: 08/09/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Ewing's sarcoma is a childhood bone and soft tissue cancer with poor prognosis. Treatment outcomes for Ewing's sarcoma patients have improved only modestly over the past decades, making the development of new treatment strategies paramount. In this study, the combined targeting of ribonucleotide reductase (RNR) and WEE1 was explored for its effectiveness against Ewing's sarcoma cells. METHODS The RNR inhibitor triapine and the WEE1 inhibitors adavosertib and ZN-c3 were tested in p53 wild-type and p53 mutant Ewing's sarcoma cells. The combination of adavosertib with the PARP inhibitors olaparib and veliparib was tested for comparison. Combinatorial effects were determined by flow cytometric analyses of cell death, loss of mitochondrial membrane potential and DNA fragmentation as well as by caspase 3/7 activity assay, immunoblotting and real-time RT-PCR. The drug interactions were assessed using combination index analysis. RESULTS RNR and WEE1 inhibitors were weakly to moderately effective on their own, but highly effective in combination. The combination treatments were similarly effective in p53 wild-type and p53 mutant cells. They synergistically induced cell death and cooperated to elicit mitochondrial membrane potential decay, to activate caspase 3/7 and to trigger DNA fragmentation, evidencing the induction of the apoptotic cell death cascade. They also cooperated to boost CHK1 phosphorylation, indicating augmented replication stress after combination treatment. In comparison, the combination of adavosertib with PARP inhibitors produced weaker synergistic effects. CONCLUSION Our findings show that combined inhibition of RNR and WEE1 was effective against Ewing's sarcoma in vitro. They thus provide a rationale for the evaluation of the potential of combined targeting of RNR and WEE1 in Ewing's sarcoma in vivo.
Collapse
Affiliation(s)
- Judy Ziener
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | | | - Johanna Leonhardi
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Max-Johann Sturm
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Sabine Becker
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Comprehensive Cancer Centre Central Germany (CCCG), Jena, Germany
| | | | - Till Milde
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
- Comprehensive Cancer Centre Central Germany (CCCG), Jena, Germany
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center Heidelberg (DKFZ), Heidelberg, Germany
| | - James F Beck
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany
| | - Jürgen Sonnemann
- Department of Paediatric and Adolescent Medicine, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.
- Research Centre Lobeda, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.
- Comprehensive Cancer Centre Central Germany (CCCG), Jena, Germany.
- Klinik für Kinder- und Jugendmedizin, Friedrich-Schiller-Universität Jena, Am Klinikum 1, D-07747, Jena, Germany.
| |
Collapse
|
35
|
Li Z, Zhang W, Zhang Z, Mao G, Qi L, Wang Y, Yang H, Ye H. PICH, A protein that maintains genomic stability, can promote tumor growth. Gene 2025; 935:149074. [PMID: 39491600 DOI: 10.1016/j.gene.2024.149074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 11/05/2024]
Abstract
Genomic instability is regardedas a hallmark of cancer cells. It can be presented in many ways, among which chromosome instability has received attention. Ultrafine anaphase bridges are a typeof chromatin bridges, the untimely resolution of which can also lead to chromosome instability. PICH can play a role in maintaining chromosome stability by regulating chromosome morphologyand resolving ultrafine anaphase bridges. Recently, PICH has been found to be overexpressed in various cancers. Overexpression of PICH is related to the proliferation of tumors and poor prognosis. In this article, we consider that PICH can maintain genomic stability by regulating appropriate chromosome structure, ensuring proper chromosome segregation, and facilitating replication fork reversal. We summarize how PICH regulates chromosome stability, how PICH resolves Ultrafine anaphase bridges with other proteins, and how PICH promotes tumor progression.
Collapse
Affiliation(s)
- Zeyuan Li
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Wentao Zhang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Zihan Zhang
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Guoming Mao
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Linping Qi
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China
| | - Yubin Wang
- Laboratory Medicine Center Gansu Provincial Natural Science, Lanzhou University Second Hospital, People's Republic of China
| | - Hanteng Yang
- The Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, People's Republic of China.
| | - Huili Ye
- The Second Clinical Medical School, Lanzhou University, Lanzhou, People's Republic of China; Cuiying Biomedical Research Center, Lanzhou University Second Hospital, Lanzhou, People's Republic of China; Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou, People's Republic of China.
| |
Collapse
|
36
|
Yano K, Kato M, Endo S, Igarashi T, Wada R, Kohno T, Zimmermann A, Dahmen H, Zenke FT, Shiotani B. PARP inhibition-associated heterochromatin confers increased DNA replication stress and vulnerability to ATR inhibition in SMARCA4-deficient cells. Cell Death Discov 2025; 11:31. [PMID: 39875375 PMCID: PMC11775187 DOI: 10.1038/s41420-025-02306-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/18/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
DNA replication stress (RS), a prevalent feature of various malignancies, arises from both genetic mutations and genotoxic exposure. Elevated RS levels increase the vulnerability of cancer cells to ataxia telangiectasia and Rad3-related kinase inhibitors (ATRis). Here, we screened for DNA damage response inhibitors that enhance ATRi-induced cytotoxicity using SWI/SNF complex-deficient cells and identified a potent synergy between ATRi and poly(ADP-ribose) polymerase inhibitor (PARPi), particularly in SMARCA4-deficient cells. PARP inhibition triggers chromatin changes, namely elevated histone H3 at lysine 9 di-methylation (H3K9me2), a hallmark of facultative heterochromatin, increasing dependence on ATR activity for replication fork progression and cell survival. Interestingly, SMARCA4 deficient cells, intrinsically vulnerable to replication stress, exhibited exacerbated DNA damage upon combined ATRi and PARPi treatment in a Mre11- and Mus81-mediated manner. In vivo, combined treatment with intermittent ATRi and continuous PARPi showed greater inhibition of tumor growth than ATRi alone in SMARCA4-deficient lung adenocarcinoma xenograft models. These findings demonstrate that PARPi-induced heterochromatin amplifies RS and ATRi susceptibility, providing a potential rationale for therapeutic strategies targeting SMARCA4-deficient tumors.
Collapse
Affiliation(s)
- Kimiyoshi Yano
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Cellular and Molecular Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima-city, Hiroshima, 734-8553, Japan
| | - Megumi Kato
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
- Department of NCC Cancer Science, Health Sciences and Biomedical Engineering, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Syoju Endo
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
- Department of NCC Cancer Science, Division of Integrative Molecular Biomedicine, Biomedical Sciences and Engineering, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, 113-8510, Japan
| | - Taichi Igarashi
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Biosciences, School of Science, Kitasato University, Minami-ku, Sagamihara-city, Kanagawa, 252-0373, Japan
| | - Ryoga Wada
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Pharmacology, Hoshi University School of Pharmacy and Pharmaceutical Sciences, Shinagawa-ku, Tokyo, 142-8501, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan
| | - Astrid Zimmermann
- Research Unit Oncology, The healthcare business of Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Heike Dahmen
- Research Unit Oncology, The healthcare business of Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Frank T Zenke
- Research Unit Oncology, The healthcare business of Merck KGaA, Frankfurter Str. 250, 64293, Darmstadt, Germany
| | - Bunsyo Shiotani
- Laboratory of Genome Stress Signaling, National Cancer Center Research Institute, Chuo-ku, Tokyo, 104-0045, Japan.
- Department of Genome Stress Signaling, Institute of Medical Science, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-0023, Japan.
| |
Collapse
|
37
|
Narozna M, Latham MC, Gorbsky GJ. Origin of Chromosome 12 Trisomy Surge in Human Induced Pluripotent Stem Cells (iPSCs). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.02.626470. [PMID: 39677655 PMCID: PMC11642788 DOI: 10.1101/2024.12.02.626470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Cultured pluripotent stem cells are unique in being the only fully diploid immortal human cell lines. However, during continued culture, they acquire significant chromosome abnormalities. Chromosome 12 trisomy is the most common whole-chromosome abnormality found during culture of human induced pluripotent stem cells (iPSCs). The conventional paradigm is that trisomy 12 occurs very rarely but provides a proliferative advantage, enabling these cells to outcompete the diploid. Here, we challenge this prevailing model by demonstrating that trisomy 12 arises simultaneously in a very high percentage of diploid cells. Using a single cell line that reproducibly undergoes transition from diploid to trisomy 12, we found that proliferation differences alone do not account for the rapid dominance of trisomic cells. Through careful mapping by fluorescent in-situ hybridization, we identified critical transition passages where trisomic cells first appeared and swiftly gained dominance. Remarkably, single trisomic cells repeatedly emerged de novo from diploid parents. Delving deeper, we discovered an extremely high incidence of chromosome 12 anaphase bridging exclusively during transition passages, along with overrepresentation of chromosome 12 chromatids in micronuclei. These micronuclei fail to replicate during S phase. Subsequently, when these micronucleated cells enter mitosis they contain an unreplicated chromosome 12 chromatids. We also found that nearly 20% of the shorter p arms of chromosome 12 but not the longer q arms exhibited loss of subtelomeric repeats during transition passages. Chromosome 12p arms were exclusively responsible for the bridging observed in anaphase cells. Our findings unveil a novel mechanism of whole-chromosome instability in human stem cells, where chromosome 12p arm-specific segregation errors occur simultaneously in a high percentage of cells. The slight yet significant growth advantage of trisomy 12 cells allows them to persist and eventually dominate the population. Our findings detailing this novel interpretation of the origin of chromosome instability in cultured of human stem cells may have broad implications for understanding the genesis of aneuploidy across diverse biological systems.
Collapse
Affiliation(s)
- Maria Narozna
- Program in Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Megan C. Latham
- Program in Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Gary J. Gorbsky
- Program in Cell Cycle and Cancer Biology, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
38
|
Sishc BJ, Saha J, Alves EM, Ding L, Lu H, Wang SY, Swancutt KL, Nicholson JH, Facoetti A, Pompos A, Ciocca M, Aguilera TA, Story MD, Davis AJ. Defective homologous recombination and genomic instability predict increased responsiveness to carbon ion radiotherapy in pancreatic cancer. NPJ Precis Oncol 2025; 9:20. [PMID: 39824957 PMCID: PMC11742413 DOI: 10.1038/s41698-025-00800-4] [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: 04/09/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is notably resistant to conventional chemotherapy and radiation treatment. However, clinical trials indicate that carbon ion radiotherapy (CIRT) with concurrent gemcitabine is effective for unresectable locally advanced PDAC. This study aimed to identify patient characteristics predictive of CIRT response. We utilized a panel of human PDAC cell lines with diverse genetic profiles to determine their sensitivity to CIRT compared to γ-rays, assessing relative biological effectiveness (RBE) at 10% survival, which ranged from 1.96 to 3.04. Increased radiosensitivity was linked to impaired DNA double-strand break (DSB) repair, particularly in cell lines with deficiencies in the homologous recombination (HR) repair pathway and/or elevated genomic instability from replication stress. Furthermore, pretreatment with the HR inhibitor B02 significantly enhanced CIRT sensitivity in a radioresistant PDAC cell line when irradiated in the spread-out Bragg peak but not at the entry position of the beam. These findings suggest that PDAC tumors with HR pathway mutations or high replication stress are more likely to benefit from CIRT while minimizing normal tissue toxicity.
Collapse
Affiliation(s)
- Brock J Sishc
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Mayo Clinic Florida, Jacksonville, FL, USA
| | - Janapriya Saha
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elizabeth M Alves
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lianghao Ding
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Huiming Lu
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shih-Ya Wang
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Katy L Swancutt
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - James H Nicholson
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Angelica Facoetti
- Medical Physics Unit & Research Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Arnold Pompos
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mario Ciocca
- Medical Physics Unit & Research Department, CNAO National Center for Oncological Hadrontherapy, Pavia, Italy
| | - Todd A Aguilera
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Michael D Story
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Mayo Clinic Florida, Jacksonville, FL, USA.
| | - Anthony J Davis
- Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
39
|
Dong R, Wang J, Guan R, Sun J, Jin P, Shen J. Role of Oxidative Stress in the Occurrence, Development, and Treatment of Breast Cancer. Antioxidants (Basel) 2025; 14:104. [PMID: 39857438 PMCID: PMC11760893 DOI: 10.3390/antiox14010104] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Breast cancer is one of the most prevalent cancers worldwide. Recent studies have increasingly emphasized the role of oxidative stress in the initiation and progression of breast cancer. This article reviews how oxidative stress imbalance influences the occurrence and advancement of breast cancer, elucidating the intricate mechanisms through which reactive oxygen species (ROS) operate in this context and their potential therapeutic applications. By highlighting these critical insights, this review aims to enhance our understanding of oxidative stress as a potential target for innovative therapeutic strategies in the management of breast cancer.
Collapse
Affiliation(s)
- Rui Dong
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China; (R.D.); (J.W.); (R.G.); (J.S.)
| | - Jing Wang
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China; (R.D.); (J.W.); (R.G.); (J.S.)
| | - Ruiqi Guan
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China; (R.D.); (J.W.); (R.G.); (J.S.)
| | - Jianwei Sun
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China; (R.D.); (J.W.); (R.G.); (J.S.)
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, China
| | - Ping Jin
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China; (R.D.); (J.W.); (R.G.); (J.S.)
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650091, China
| | - Junling Shen
- Yunnan Key Laboratory of Cell Metabolism and Diseases, Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650091, China; (R.D.); (J.W.); (R.G.); (J.S.)
- Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, China
| |
Collapse
|
40
|
Li Z, Zhang Z. A tale of two strands: Decoding chromatin replication through strand-specific sequencing. Mol Cell 2025; 85:238-261. [PMID: 39824166 PMCID: PMC11750172 DOI: 10.1016/j.molcel.2024.10.035] [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: 08/15/2024] [Revised: 10/03/2024] [Accepted: 10/25/2024] [Indexed: 01/20/2025]
Abstract
DNA replication, a fundamental process in all living organisms, proceeds with continuous synthesis of the leading strand by DNA polymerase ε (Pol ε) and discontinuous synthesis of the lagging strand by polymerase δ (Pol δ). This inherent asymmetry at each replication fork necessitates the development of methods to distinguish between these two nascent strands in vivo. Over the past decade, strand-specific sequencing strategies, such as enrichment and sequencing of protein-associated nascent DNA (eSPAN) and Okazaki fragment sequencing (OK-seq), have become essential tools for studying chromatin replication in eukaryotic cells. In this review, we outline the foundational principles underlying these methodologies and summarize key mechanistic insights into DNA replication, parental histone transfer, epigenetic inheritance, and beyond, gained through their applications. Finally, we discuss the limitations and challenges of current techniques, highlighting the need for further technological innovations to better understand the dynamics and regulation of chromatin replication in eukaryotic cells.
Collapse
Affiliation(s)
- Zhiming Li
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA; West China School of Public Health and West China Fourth Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Zhiguo Zhang
- Institute for Cancer Genetics and Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY 10032, USA; Department of Pediatrics and Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA.
| |
Collapse
|
41
|
Xie H, Song L, Mao G, Han J, Pu J, Wu Z, Chen J, Zhou J, Huang J, Fang D, Liu T. Synergistic protection of nascent DNA at stalled forks by MSANTD4 and BRCA1/2-RAD51. Nat Chem Biol 2025:10.1038/s41589-024-01833-9. [PMID: 39809895 DOI: 10.1038/s41589-024-01833-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025]
Abstract
The regressed arms of reversed replication forks exhibit structural similarities to one-ended double-stranded breaks and need to be protected against uncontrolled nucleolytic degradation. Here, we identify MSANTD4 (Myb/SANT-like DNA-binding domain-containing protein 4), a functionally uncharacterized protein that uniquely counters the replication protein A (RPA)-Bloom (BLM)/Werner syndrome helicase (WRN)-DNA replication helicase/nuclease 2 (DNA2) complex to safeguard reversed replication forks from detrimental degradation, independently of the breast cancer susceptibility proteins (BRCA1/2)-DNA repair protein RAD51 pathway. MSANTD4 specifically interacts with the junctions between single-stranded DNA (ssDNA) and double-stranded DNA (dsDNA) in DNA substrates harboring a 3' overhang, which resemble the structural features of regressed arms processed by WRN-DNA2. This DNA-binding capability allows MSANTD4 to accumulate at reversed forks, strategically antagonizing the RPA-BLM/WRN-DNA2 complex by impeding its access to the ssDNA-dsDNA junction of the regressed arms. Loss of MSANTD4 exacerbates genome instability induced by replication stress in BRCA1/2-deficient cells. Our findings unveil a collaborative defense mechanism orchestrated by MSANTD4 and BRCA1/2-RAD51, effectively counteracting nucleolytic attacks on the regressed arms and synergistically preserving the integrity of reversed forks.
Collapse
Affiliation(s)
- Haihua Xie
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lizhi Song
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China
| | - Genxiang Mao
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinhua Han
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiali Pu
- Department of Neurology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhibing Wu
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Chen
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Huang
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- Center for Life Sciences, Shaoxing Institute, Zhejiang University, Shaoxing, China.
| | - Dong Fang
- MOE Laboratory of Biosystems Homeostasis and Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
| | - Ting Liu
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Affiliated Zhejiang Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
42
|
Zhao XH, Han MM, Yan QQ, Yue YM, Ye K, Zhang YY, Teng L, Xu L, Shi XJ, La T, Feng YC, Xu R, Narayana VK, De Souza DP, Quek LE, Holst J, Liu T, Baker MA, Thorne RF, Zhang XD, Jin L. DNA replication stress underpins the vulnerability to oxidative phosphorylation inhibition in colorectal cancer. Cell Death Dis 2025; 16:16. [PMID: 39809754 PMCID: PMC11733219 DOI: 10.1038/s41419-025-07334-4] [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: 07/09/2024] [Revised: 12/07/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Mitochondrial oxidative phosphorylation (OXPHOS) is a therapeutic vulnerability in glycolysis-deficient cancers. Here we show that inhibiting OXPHOS similarly suppresses the proliferation and tumorigenicity of glycolytically competent colorectal cancer (CRC) cells in vitro and in patient-derived CRC xenografts. While the increased glycolytic activity rapidly replenished the ATP pool, it did not restore the reduced production of aspartate upon OXPHOS inhibition. This shortage in aspartate, in turn, caused nucleotide deficiencies, leading to S phase cell cycle arrest, replication fork stalling, and enrichment of the p53 pathway, manifestations of replication stress. The addition of purine nucleobases adenine and guanine along with the pyrimidine nucleoside uridine restored replication fork progression and cell proliferation, whereas the supplementation of exogenous aspartate recovered the nucleotide pool, demonstrating a causal role of the aspartate shortage in OXPHOS inhibition-induced nucleotide deficiencies and consequently replication stress and reductions in proliferation. Moreover, we demonstrate that glutamic-oxaloacetic transaminase 1 (GOT1) is critical for maintaining the minimum aspartate pool when OXPHOS is inhibited, as knockdown of GOT1 further reduced aspartate levels and rendered CRC cells more sensitive to OXPHOS inhibition both in vitro and in vivo. These results propose GOT1 targeting as a potential avenue to sensitize cancer cells to OXPHOS inhibitors, thus lowering the necessary doses to efficiently inhibit cancer growth while alleviating their adverse effects.
Collapse
Affiliation(s)
- Xiao Hong Zhao
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
| | - Man Man Han
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Qian Qian Yan
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Meng Yue
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Kaihong Ye
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuan Yuan Zhang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
| | - Liu Teng
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Liang Xu
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
| | - Xiao-Jing Shi
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ting La
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
- National-Local Joint Engineering Research Center of Biodiagnosis & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yu Chen Feng
- School of Medicine and Public Health, The University of Newcastle, Newcastle, NSW, Australia
| | - Ran Xu
- School of Medicine and Public Health, The University of Newcastle, Newcastle, NSW, Australia
| | - Vinod K Narayana
- Bio21 Institute and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia
- Metabolomics Australia, University of Melbourne, Parkville, VIC, Australia
| | - David P De Souza
- Bio21 Institute and Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, VIC, Australia
- Metabolomics Australia, University of Melbourne, Parkville, VIC, Australia
| | - Lake-Ee Quek
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, Australia
| | - Jeff Holst
- School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Tao Liu
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, NSW, Australia
| | - Mark A Baker
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
| | - Rick F Thorne
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xu Dong Zhang
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, Australia.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China.
| | - Lei Jin
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
- Translational Research Institute, Henan Provincial and Zhengzhou City Key Laboratory of Noncoding RNA and Cancer Metabolism, Henan International Join Laboratory of Noncoding RNA and Metabolism in Cancer, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, Henan, China.
- School of Medicine and Public Health, The University of Newcastle, Newcastle, NSW, Australia.
| |
Collapse
|
43
|
Chiou LF, Jayaprakash D, Droby GN, Zhang X, Yang Y, Mills CA, Webb TS, Barker NK, Wu D, Herring LE, Bowser J, Vaziri C. The RING Finger E3 Ligase RNF25 Protects DNA Replication Forks Independently of its Canonical Roles in Ubiquitin Signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.09.632184. [PMID: 39829812 PMCID: PMC11741350 DOI: 10.1101/2025.01.09.632184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The DNA damage response (DDR) mechanisms that allow cells to tolerate DNA replication stress are critically important for genome stability and cell viability. Using an unbiased genetic screen we identify a role for the RING finger E3 ubiquitin ligase RNF25 in promoting DNA replication stress tolerance. In response to DNA replication stress, RNF25-deficient cells generate aberrantly high levels of single-stranded DNA (ssDNA), accumulate in S-phase and show reduced mitotic entry. Using single-molecule DNA fiber analysis, we show that RNF25 protects reversed DNA replication forks generated by the fork remodeler HLTF from nucleolytic degradation by MRE11 and CtIP. Mechanistically, RNF25 interacts with the replication fork protection factor REV7 and recruits REV7 to nascent DNA after replication stress. The role of RNF25 in protecting replication forks is fully separable from its canonical functions in ubiquitin conjugation. This work reveals the RNF25-REV7 signaling axis as an important protective mechanism in cells experiencing replication stress.
Collapse
Affiliation(s)
- Lilly F. Chiou
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Deepika Jayaprakash
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Oral and Craniofacial Biomedicine Program, Adams School of Dentistry, University of North Carolina at Chapel Hill, NC 27599, USA
- Present address: Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37237, USA
| | - Gaith N. Droby
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xingyuan Zhang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Present address: Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Yang Yang
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Present address: In Vivo Neurobiology Group, Neurobiology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - C. Allie Mills
- UNC Metabolomics & Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Thomas S. Webb
- UNC Metabolomics & Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Natalie K. Barker
- UNC Metabolomics & Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Di Wu
- Division of Oral and Craniofacial Health Science, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Laura E. Herring
- UNC Metabolomics & Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jessica Bowser
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Cyrus Vaziri
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lead Contact
| |
Collapse
|
44
|
Yin H, Zheng Y, Chen M, Ding M, Zhang L, Wang R, Wang C, Jia J, Liu X. Pesticide avermectin B1a exerts cytotoxicity by blocking the interaction between mini-chromosome maintenance 6 protein (MCM6) and chromatin licensing and DNA replication factor 1 (CDT1). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 364:125377. [PMID: 39579921 DOI: 10.1016/j.envpol.2024.125377] [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: 09/22/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 11/25/2024]
Abstract
Avermectin B1a, a widely used pesticide, has recently raised safety concerns since it possesses potential cytotoxicity toward mammalian cells. Nevertheless, the exact mechanisms that underlie the cytotoxicity induced by avermectin B1a remain elusive. The loading of the mini-chromosome maintenance 6 protein (MCM6) onto chromatin at replication origins by chromatin licensing and DNA replication factor 1 (CDT1) is an essential step for licensing DNA for replication. Here, we first report that avermectin B1a occupies the CDT1-binding domain (CBD) of MCM6 to block the interaction between MCM6 and CDT1 and thus inhibits the licensing for DNA replication. Avermectin B1a inhibits the proliferation with IC50 being 15.1 μM and induces cell cycle arrest at the G0/G1 phase in MEF cells. Moreover, abnormal replication licensing induced by avermectin B1a causes replication stress and DNA double strand breaks, which in turn leads to apoptosis in MEF cells. Further molecular docking uncovers that four residues Glu763, Ile760, Arg771, and Glu774 are vital for the formation of hydrogen bonds in avermectin B1a-CBD interaction. Furthermore, the upregulation of MCM6 or/and CDT1 reverses the avermectin B1a-induced decrease in cell viability and normalizes the cell cycle, indicating that the blockage of MCM6-CDT1 interaction is one of the mechanisms underlying avermectin B1a-induced cytotoxicity. This study not only provides new insights into the mechanism of avermectin B1a-induced cytotoxicity but also offers a useful molecular tool for the investigation of MCM6-CDT1 interaction.
Collapse
Affiliation(s)
- Hao Yin
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Laboratory for Molecular Identification and Biological Evaluation of Chinese Herbal Pieces, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Yaoyao Zheng
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Laboratory for Molecular Identification and Biological Evaluation of Chinese Herbal Pieces, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Menghan Chen
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Laboratory for Molecular Identification and Biological Evaluation of Chinese Herbal Pieces, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Ming Ding
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China; Laboratory for Molecular Identification and Biological Evaluation of Chinese Herbal Pieces, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Lin Zhang
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Roujia Wang
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Chunyu Wang
- Level Three Laboratory of Traditional Chinese Medicine Preparation of National Administration of Traditional Chinese Medicine, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China
| | - Jing Jia
- Laboratory for Molecular Identification and Biological Evaluation of Chinese Herbal Pieces, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China.
| | - Xiaoqian Liu
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, China.
| |
Collapse
|
45
|
Guo XL, Wang YD, Liu YJ, Chu L, Zhu H, Hu Y, Wu RY, Xie HY, Yu J, Li SP, Xiong CY, Li RY, Ke F, Chen L, Chen GQ, Chen L, Bai F, Enver T, Li GH, Li HF, Hong DL. Fetal hepatocytes protect the HSPC genome via fetuin-A. Nature 2025; 637:402-411. [PMID: 39633051 PMCID: PMC11711094 DOI: 10.1038/s41586-024-08307-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 10/30/2024] [Indexed: 12/07/2024]
Abstract
The maintenance of genomic integrity in rapidly proliferating cells is a substantial challenge during embryonic development1-3. Although numerous cell-intrinsic mechanisms have been revealed4-7, little is known about genome-protective effects and influences of developmental tissue microenvironments on tissue-forming cells. Here we show that fetal liver hepatocytes provide protection to haematopoietic stem and progenitor cell (HSPC) genomes. Lineage tracing and depletion in mice demonstrated that delayed hepatocyte development in early fetal livers increased the chromosomal instability of newly colonizing HSPCs. In addition, HSPCs developed tolerance to genotoxins in hepatocyte-conditioned medium, suggesting that hepatocytes protect the HSPC genome in a paracrine manner. Proteomic analyses demonstrated the enrichment of fetuin-A in hepatocyte-conditioned medium but not in early fetal livers. Fetuin-A activates a Toll-like receptor pathway to prevent pathogenic R-loop accumulation in HSPCs undergoing DNA replication and gene transcription in the fetal liver. Numerous haematopoietic regulatory genes frequently involved in leukaemogenic mutations are associated with R-loop-enriched regions. In Fetua-knockout mice, HSPCs showed increased genome instability and susceptibility to malignancy induction. Moreover, low concentrations of fetuin-A correlated with the oncogenesis of childhood leukaemia. Therefore, we uncover a mechanism operating in developmental tissues that offers tissue-forming cell genome protection and is implicated in developmental-related diseases.
Collapse
Affiliation(s)
- Xiao-Lin Guo
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Ding Wang
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-Jun Liu
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Chu
- Department of Obstetrics and Gynecology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hua Zhu
- Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ye Hu
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ren-Yan Wu
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Yu Xie
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan Yu
- National laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Shui-Ping Li
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao-Yang Xiong
- National laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Ruo-Yan Li
- Biomedical Pioneering Innovation Centre (BIOPIC) and Translational Cancer Research Centre, School of Life Sciences, First Hospital, Peking University, Beijing, China
| | - Fang Ke
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Chen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guo-Qiang Chen
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Research Units of Stress and Tumor, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Hainan Academy of Medical Sciences, School of Basic Medicine and Life Science, Hainan Medical University, Haikou, China
| | - Liang Chen
- RNA Institute, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Fan Bai
- Biomedical Pioneering Innovation Centre (BIOPIC) and Translational Cancer Research Centre, School of Life Sciences, First Hospital, Peking University, Beijing, China
| | - Tariq Enver
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, UK
| | - Guo-Hong Li
- National laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Huai-Fang Li
- Department of Obstetrics and Gynecology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Deng-Li Hong
- Key Laboratory of Cell Differentiation and Apoptosis of Ministry of Education, Department of Pathophysiology, Shanghai Institute of Haematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Research Units of Stress and Tumor, Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Innovative Research Team of High-level Local Universities in Shanghai, Shanghai, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
46
|
Yang QL, Xie Y, Qiao K, Lim JYS, Wu S. Modern biology of extrachromosomal DNA: A decade-long voyage of discovery. Cell Res 2025; 35:11-22. [PMID: 39748050 PMCID: PMC11701097 DOI: 10.1038/s41422-024-01054-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 11/07/2024] [Indexed: 01/04/2025] Open
Abstract
Genomic instability is a hallmark of cancer and is a major driving force of tumorigenesis. A key manifestation of genomic instability is the formation of extrachromosomal DNAs (ecDNAs) - acentric, circular DNA molecules ranging from 50 kb to 5 Mb in size, distinct from chromosomes. Ontological studies have revealed that ecDNA serves as a carrier of oncogenes, immunoregulatory genes, and enhancers, capable of driving elevated transcription of its cargo genes and cancer heterogeneity, leading to rapid tumor evolution and therapy resistance. Although ecDNA was documented over half a century ago, the past decade has witnessed a surge in breakthrough discoveries about its biological functions. Here, we systematically review the modern biology of ecDNA uncovered over the last ten years, focusing on how discoveries during this pioneering stage have illuminated our understanding of ecDNA-driven transcription, heterogeneity, and cancer progression. Furthermore, we discuss ongoing efforts to target ecDNA as a novel approach to cancer therapy. This burgeoning field is entering a new phase, poised to reshape our knowledge of cancer biology and therapeutic strategies.
Collapse
Affiliation(s)
- Qing-Lin Yang
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yipeng Xie
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kailiang Qiao
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jun Yi Stanley Lim
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sihan Wu
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
47
|
Brown N, Luniewski A, Yu X, Warthan M, Liu S, Zulawinska J, Ahmad S, Congdon M, Santos W, Xiao F, Guler JL. Replication stress increases de novo CNVs across the malaria parasite genome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.19.629492. [PMID: 39803504 PMCID: PMC11722320 DOI: 10.1101/2024.12.19.629492] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Changes in the copy number of large genomic regions, termed copy number variations (CNVs), contribute to important phenotypes in many organisms. CNVs are readily identified using conventional approaches when present in a large fraction of the cell population. However, CNVs that are present in only a few genomes across a population are often overlooked but important; if beneficial under specific conditions, a de novo CNV that arises in a single genome can expand during selection to create a larger population of cells with novel characteristics. While the reach of single cell methods to study de novo CNVs is increasing, we continue to lack information about CNV dynamics in rapidly evolving microbial populations. Here, we investigated de novo CNVs in the genome of the Plasmodium parasite that causes human malaria. The highly AT-rich P. falciparum genome readily accumulates CNVs that facilitate rapid adaptation to new drugs and host environments. We employed a low-input genomics approach optimized for this unique genome as well as specialized computational tools to evaluate the de novo CNV rate both before and after the application of stress. We observed a significant increase in genomewide de novo CNVs following treatment with a replication inhibitor. These stress-induced de novo CNVs encompassed genes that contribute to various cellular pathways and tended to be altered in clinical parasite genomes. This snapshot of CNV dynamics emphasizes the connection between replication stress, DNA repair, and CNV generation in this important microbial pathogen.
Collapse
Affiliation(s)
- Noah Brown
- University of Virginia, Department of Biology, Charlottesville, VA, USA
| | | | - Xuanxuan Yu
- Unifersity of Florida, Department of Biostatistics, Gainesville, FL, USA
- Unifersity of Florida, Department of Surgery, College of Medicine, Gainesville, FL, USA
| | - Michelle Warthan
- University of Virginia, Department of Biology, Charlottesville, VA, USA
| | - Shiwei Liu
- University of Virginia, Department of Biology, Charlottesville, VA, USA
- Current affiliation: Indiana University School of Medicine, Indianapolis, IN, USA
| | - Julia Zulawinska
- University of Virginia, Department of Biology, Charlottesville, VA, USA
| | - Syed Ahmad
- University of Virginia, Department of Biology, Charlottesville, VA, USA
| | - Molly Congdon
- Virginia Tech, Department of Chemistry, Blacksburg, VA, USA
| | - Webster Santos
- Virginia Tech, Department of Chemistry, Blacksburg, VA, USA
| | - Feifei Xiao
- Unifersity of Florida, Department of Biostatistics, Gainesville, FL, USA
| | - Jennifer L Guler
- University of Virginia, Department of Biology, Charlottesville, VA, USA
| |
Collapse
|
48
|
Zhai Z, Cui Z, Zhang Y, Song P, Wu J, Tan Z, Lin S, Ma X, Guan F, Kang H. Integrated pan-cancer analysis and experimental verification of the roles of meiotic nuclear divisions 1 in breast cancer. Biochem Biophys Res Commun 2024; 739:150600. [PMID: 39191147 DOI: 10.1016/j.bbrc.2024.150600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/08/2024] [Accepted: 08/22/2024] [Indexed: 08/29/2024]
Abstract
INTRODUCTION The aberrant up-regulation of meiotic nuclear division 1 (MND1) in somatic cells is considered as one of the driving factors of oncogenesis, whereas its expression and role in breast invasive cancer (BRCA) remain unclear. Hence, this study embarked on a comprehensive evaluation of MND1 across various cancers and identified its roles in BRCA. METHODS Based on publicly available databases, including but not limited to UCSC Xena, TCGA, GTEx, GEO, STRING, GeneMANIA, and CancerSEA, we evaluated the expression patterns, genomic features, and biological functions of MND1 from a pan-cancer viewpoint and delved into the implications of MND1 in the prognosis and treatment of BRCA. Further molecular biology experiments were undertaken to identify the role of MND1 in proliferation, migration, and apoptosis in BRCA cells. RESULTS Elevated levels of MND1 were notably observed in a wide array of tumor types, especially in BRCA, COAD, HNSC, LIHC, LUAD, LUSC, STAD, and UCEC. Elevated MND1 expression was markedly associated with shortened OS in several tumors, including BRCA (HR = 1.52 [95%CI, 1.10-2.09], P = 0.011). The up-regulation of MND1 in BRCA was validated in external cohorts and clinical samples. Survival analyses demonstrated that elevated MND1 expression was associated with decreased survival for patients with BRCA. Co-expressed genes of MND1 were identified, and subsequent pathway analyses based on significantly associated genes indicated that MND1 plays key roles in DNA replication, cell cycle regulation, and DNA damage repair. The observed abnormal elevation and activation of MND1 led to increased proliferation and migration, along with decreased apoptosis in BRCA cells. CONCLUSIONS MND1 emerges as a promising biomarker for diagnostic and therapeutic targeting in various cancers, including BRCA. The abnormal up-regulation and activation of MND1 are linked to carcinogenesis and poor prognosis among BRCA patients, which may be attributed to its involvement in HR-dependent ALT, warranting further scrutiny.
Collapse
Affiliation(s)
- Zhen Zhai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China; Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China
| | - Zhiwei Cui
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, Yanta West Road, Xi' an, China
| | - Yu Zhang
- Department of Infectious Diseases, Honghui-hospital, Xi'an Jiaotong University, Shanghua Road, Xi'an, China
| | - Ping Song
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, No. 157, West Fifth Road, Xi'an, China
| | - Jinpeng Wu
- College of Life Sciences, Northwest University, No. 229, Taibai North Road, Xi'an, China
| | - Zengqi Tan
- Institute of Hematology, Provincial Key Laboratory of Biotechnology, School of Medicine, Northwest University, No. 229, Taibai North Road, Xi'an, China
| | - Shuai Lin
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China; Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China
| | - Xiaobin Ma
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China; Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China
| | - Feng Guan
- College of Life Sciences, Northwest University, No. 229, Taibai North Road, Xi'an, China.
| | - Huafeng Kang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China; Comprehensive Breast Care Center, The Second Affiliated Hospital of Xi'an Jiaotong University, No. 157, West Fifth Road, Xi'an, China.
| |
Collapse
|
49
|
Ghorai A, Singh B, Dutt S. Biphasic DNA damage and non-canonical replication stress response govern radiation-induced senescence in glioblastoma. J Cell Sci 2024; 137:jcs261844. [PMID: 39568404 DOI: 10.1242/jcs.261844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 10/28/2024] [Indexed: 11/22/2024] Open
Abstract
Therapy-induced senescence (TIS) in glioblastoma (GBM) residual disease and escape from TIS account for resistance and recurrence, but the mechanism of TIS manifestation remains obscure. Here, we demonstrate that replication stress (RS) is critical for the induction of TIS in residual cells by employing an in vitro GBM therapy-resistance cellular model. Interestingly, we found a 'biphasic' mode of DNA damage after radiation treatment and reveal that the second phase of DNA damage arises majorly in the S phase of residual cells due to RS. Mechanistically, we show that persistent phosphorylated ATR is a safeguard for radiation resilience, whereas the other canonical RS molecules remain unaltered during the second phase of DNA damage. Importantly, RS preceded the induction of senescence, and ATR inhibition resulted in TIS reduction, leading to apoptosis. Moreover, ATR inhibition sensitized PARP-1 inhibitor-induced enhanced TIS-mediated resistance, leading to cell death. Our study demonstrates the crucial role of RS in TIS induction and maintenance in GBM residual cells, and targeting ATR alone or in combination with a PARP-1 inhibitor will be an effective strategy to eliminate TIS for better treatment outcomes.
Collapse
Affiliation(s)
- Atanu Ghorai
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
| | - Bhawna Singh
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
- Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai 400085, India
| | - Shilpee Dutt
- Shilpee Dutt Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai 410210, India
- Shilpee Dutt Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi 110067, India
| |
Collapse
|
50
|
Kutz J, Schmietendorf H, Rahman SA, Opel F, Pospiech H. HROB Is Implicated in DNA Replication. Genes (Basel) 2024; 15:1587. [PMID: 39766854 PMCID: PMC11675949 DOI: 10.3390/genes15121587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/29/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
DNA replication represents a series of precisely regulated events performed by a complex protein machinery that guarantees accurate duplication of the genetic information. Since DNA replication is permanently faced by a variety of exogenous and endogenous stressors, DNA damage response, repair and replication must be closely coordinated to maintain genomic integrity. HROB has been identified recently as a binding partner and activator of the Mcm8/9 helicase involved in DNA interstrand crosslink (ICL) repair. We identified HROB independently as a nuclear protein whose expression is co-regulated with various DNA replication factors. Accordingly, the HROB protein level showed a maximum in S phase and a downregulation in quiescence. Structural prediction and homology searches revealed that HROB is a largely intrinsically disordered protein bearing a helix-rich region and a canonical oligonucleotide/oligosaccharide-binding-fold motif that originated early in eukaryotic evolution. Employing a flow cytometry Förster resonance energy transfer (FRET) assay, we detected associations between HROB and proteins of the DNA replication machinery. Moreover, ectopic expression of HROB protein led to an almost complete shutdown of DNA replication. The available data imply a function for HROB during DNA replication across barriers such as ICLs.
Collapse
Affiliation(s)
- Julia Kutz
- Project Group Biochemistry, Leibniz Institute on Aging—Fritz Lipmann Institute, D-07745 Jena, Germany; (J.K.); (H.S.); (S.A.R.); (F.O.)
- Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich Schiller University, D-07745 Jena, Germany
| | - Hannes Schmietendorf
- Project Group Biochemistry, Leibniz Institute on Aging—Fritz Lipmann Institute, D-07745 Jena, Germany; (J.K.); (H.S.); (S.A.R.); (F.O.)
- Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich Schiller University, D-07745 Jena, Germany
| | - Sheikh Anika Rahman
- Project Group Biochemistry, Leibniz Institute on Aging—Fritz Lipmann Institute, D-07745 Jena, Germany; (J.K.); (H.S.); (S.A.R.); (F.O.)
- Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich Schiller University, D-07745 Jena, Germany
| | - Franz Opel
- Project Group Biochemistry, Leibniz Institute on Aging—Fritz Lipmann Institute, D-07745 Jena, Germany; (J.K.); (H.S.); (S.A.R.); (F.O.)
- Department of Medical Engineering and Biotechnology, Ernst-Abbe University of Applied Sciences, D-07745 Jena, Germany
| | - Helmut Pospiech
- Project Group Biochemistry, Leibniz Institute on Aging—Fritz Lipmann Institute, D-07745 Jena, Germany; (J.K.); (H.S.); (S.A.R.); (F.O.)
- Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich Schiller University, D-07745 Jena, Germany
- Department of Obstetrics and Gynecology, University Hospital Düsseldorf and Heinrich-Heine University, D-40225 Düsseldorf, Germany
| |
Collapse
|