1
|
Wallisch E, Tomita-Mitchell A, Liang HL, Szabo A, Lenarczyk M, Kwitek A, Smith JR, Tutaj M, Baker JE. Advancing cell-free DNA as a biomarker of damage to heart caused by ionizing radiation. JOURNAL OF RADIATION RESEARCH 2025; 66:329-340. [PMID: 40304705 PMCID: PMC12100473 DOI: 10.1093/jrr/rraf022] [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] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/17/2025] [Indexed: 05/02/2025]
Abstract
Exposure to diagnostic and therapeutic radiation introduces risks for development of diseases later in life by causing DNA damage in cells. Currently, there is no clinical method for determining exposure risk caused by radiation toxicity to DNA. Cell-free DNA (cfDNA), a marker of DNA damage, is currently used to assess risk for long-term effects following organ transplantation, surgery and inflammation. The goal of our proposed study is to develop cfDNA as an early biomarker for assessing risk for cardiovascular disease and cancer from radiation exposure so that strategies to mitigate the damaging effects of medical radiation can be assessed. Hearts from male and female WAG/RijCmcr rats (n = 6-10/group) were exposed to increasing doses of X-radiation (50 mGy and 3.5 Gy). Blood was collected prior to and after (15 minutes-96 hours) irradiation, and cell-free plasma was prepared. Primers and probes were designed for quantitative analysis of sequences of mitochondria (12S rRNA) and nuclear (Gapdh) cfDNA present in rat plasma using quantitative reverse transcription polymerase chain reaction (RT-qPCR). Exposure of hearts to radiation increased nuclear and mitochondrial cfDNA in a dose-dependent manner. Three point five grays from X-radiation increase cfDNA for Gapdh in plasma after 1 hour with a 15.8-fold increase (P < 0.001) after 6 hours. The earliest time nuclear and mitochondrial cfDNA increases were detected in plasma was at 60 minutes following exposure to 3.5 Gy. cfDNA has potential to advance as a biomarker of exposure to medical doses of radiation in patients.
Collapse
Affiliation(s)
- Erin Wallisch
- Division of Congenital Heart Surgery, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Aoy Tomita-Mitchell
- Division of Congenital Heart Surgery, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Huan-Ling Liang
- Division of Congenital Heart Surgery, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Aniko Szabo
- Data Science Institute, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Marek Lenarczyk
- Division of Congenital Heart Surgery, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Radiation Biosciences Laboratory, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Anne Kwitek
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Jennifer R Smith
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Monika Tutaj
- Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - John E Baker
- Division of Congenital Heart Surgery, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Radiation Biosciences Laboratory, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
2
|
Hildebrand LS, Jost T, Schindler M, Derer A, Fuhrmann G, Fietkau R, Distel LV. Inhibiting NHEJ in HNSCC cell lines by the ligase IV inhibitor SCR130 has limited radiosensitizing effects. Sci Rep 2025; 15:17871. [PMID: 40404928 PMCID: PMC12098888 DOI: 10.1038/s41598-025-03159-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025] Open
Abstract
Radiotherapy (RT) is a relevant treatment for head and neck squamous cell carcinoma (HNSCC) patients but radioresistance, which depends on DNA damage response (DDR), restrains outcome. Therefore, manipulating DDR by small molecule inhibitors (SMI) is a promising treatment option. The main DNA double strand break (DSB) repair mechanisms in healthy mammalian cells are homologous recombination (HR) and non-homologous end joining (NHEJ). It is known that HR is already often impaired in tumors because of cancerous transitions. Therefore, additionally inhibiting NHEJ is a possibility to specifically target tumor cells and spare healthy tissue, which has the alternative DSB repair mechanism available. We treated HNSCC and healthy fibroblast cell lines with 30 µM of the ligase IV inhibitor SCR130 and a single dose of 2 Gy (Gy) ionizing radiation (IR) to investigate the inhibitor's radiosensitizing effect. In short, the effect of SCR130 in combination with IR on cell death, clonogenicity, and DNA damage is limited and highly cell line specific. Nevertheless, SCR130 increases the number of cells in G0/G1 phase concomitant with gained p21 expression consistently. We suggest that SCR130 in combination with IR has anti-proliferative effects, but an escape of the cells by upregulation of ligase IV resulting from the treatment is possible.
Collapse
Affiliation(s)
- Laura S Hildebrand
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Tina Jost
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Department of Radiation Oncology, Translational Radiobiology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
| | - Marion Schindler
- Department of Biology, Pharmaceutical Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstraße 5, 91058, Erlangen, Germany
| | - Anja Derer
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
- Department of Radiation Oncology, Translational Radiobiology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
| | - Gregor Fuhrmann
- Department of Biology, Pharmaceutical Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstraße 5, 91058, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany
| | - Luitpold V Distel
- Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Universitätsstraße 27, 91054, Erlangen, Germany.
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Erlangen, Germany.
| |
Collapse
|
3
|
Ostalé CM, Azpiazu N, Peropadre A, Martín M, Ruiz-Losada M, López-Varea A, Viales RR, Girardot C, Furlong EEM, de Celis JF. A function of Spalt proteins in heterochromatin organization and maintenance of genomic DNA integrity. Development 2025; 152:dev204258. [PMID: 40326666 PMCID: PMC12091872 DOI: 10.1242/dev.204258] [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: 07/18/2024] [Accepted: 04/15/2025] [Indexed: 05/07/2025]
Abstract
The conserved Spalt proteins regulate gene expression and cell fate choices during multicellular development, generally acting as transcriptional repressors in different gene regulatory networks. In addition to their roles as DNA sequence-specific transcription factors, Spalt proteins show a consistent localization to heterochromatic regions. Vertebrate Spalt-like proteins can act through the nucleosome remodeling and deacetylase complex to promote closing of open chromatin domains, but their activities also rely on interactions with DNA methyltransferases or with the lysine-specific histone demethylase LSD1, suggesting that they participate in multiple regulatory mechanisms. Here, we describe several consequences of loss of Spalt function in Drosophila cells, including changes in chromatin accessibility, generation of DNA damage, alterations in the localization of chromosomes within the nucleus in the salivary glands and misexpression of transposable elements. We suggest that these effects are related to roles of Spalt proteins in the regulation of heterochromatin formation and chromatin organization. We propose that Drosophila Spalt proteins have two complementary functions, acting as sequence-specific transcriptional repressors on specific target genes and regulating more global gene silencing through the generation or maintenance of heterochromatic domains.
Collapse
Affiliation(s)
- Cristina M. Ostalé
- Centro de Biología Molecular ‘Severo Ochoa’, Department of Tissue and Organ Homeostasis, CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Natalia Azpiazu
- Centro de Biología Molecular ‘Severo Ochoa’, Department of Tissue and Organ Homeostasis, CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Ana Peropadre
- Department of Biology, Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Mercedes Martín
- Centro de Biología Molecular ‘Severo Ochoa’, Department of Tissue and Organ Homeostasis, CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Mireya Ruiz-Losada
- Centro de Biología Molecular ‘Severo Ochoa’, Department of Tissue and Organ Homeostasis, CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Ana López-Varea
- Centro de Biología Molecular ‘Severo Ochoa’, Department of Tissue and Organ Homeostasis, CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Rebecca R. Viales
- European Molecular Biology Laboratory, Genome Biology Department, Heidelberg 69117, Germany
| | - Charles Girardot
- European Molecular Biology Laboratory, Genome Biology Department, Heidelberg 69117, Germany
| | - Eileen E. M. Furlong
- European Molecular Biology Laboratory, Genome Biology Department, Heidelberg 69117, Germany
| | - Jose F. de Celis
- Centro de Biología Molecular ‘Severo Ochoa’, Department of Tissue and Organ Homeostasis, CSIC and Universidad Autónoma de Madrid, Madrid 28049, Spain
| |
Collapse
|
4
|
Song L, Xu L, Zhang P, Li S, Qu Y, Zhao Y, Shi Z, Ma R, Li Y, Chen Y, Wang Y, Jiang Z, Wei G, Shen X. A Dual-Targeting T6SS DNase Drives Bacterial Antagonism and Eukaryotic Apoptosis via the cGAS-STING-TNF Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2504086. [PMID: 40365777 DOI: 10.1002/advs.202504086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/27/2025] [Indexed: 05/15/2025]
Abstract
The Type VI secretion system (T6SS) is a key virulence mechanism utilized by many Gram-negative bacteria to mediate the microbial competition and host pathogenesis. Despite the identification of diverse T6SS effectors targeting eukaryotic or prokaryotic cells, the trans-kingdom T6SS effectors that simultaneously target both eukaryotic and prokaryotic cells remain rarely reported. In this study, it is demonstrated that Yersinia pseudotuberculosis (Yptb) T6SS secretes a DNase effector, TkeA, which induces apoptosis in host cells. The translocation of TkeA into host cells causes nuclear DNA damage. This, in turn, activates the DNA-sensing cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway. The activation of the cGAS-STING pathway by TkeA subsequently triggers apoptosis in host cells via extrinsic pathways, with tumor necrosis factor (TNF) signaling playing a critical role. Additionally, TkeA enhances bacterial competition by targeting rival bacteria, thereby promoting host colonization. These findings reveal that the transkingdom T6SS effector TkeA executes a "one weapon, two battlefields" strategy, acting as a trans-kingdom effector that enhances interbacterial competition while inducing apoptosis in host cells through the activation of the cGAS-STING-TNF axis. This highlights a previously unrecognized dimension of bacterial virulence strategies and expands the understanding of host-pathogen interactions involving T6SS effectors.
Collapse
Affiliation(s)
- Li Song
- Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi, 12100, P. R. China
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Lei Xu
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Pengfei Zhang
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Shuying Li
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Yichen Qu
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Yixin Zhao
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Zhenkun Shi
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Ruiqi Ma
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Yongdong Li
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo, Zhejiang, 315010, P. R. China
| | - Yi Chen
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo, Zhejiang, 315010, P. R. China
| | - Yao Wang
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Zhengfan Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, 100871, P. R. China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, P. R. China
| | - Gehong Wei
- Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi, 12100, P. R. China
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Xihui Shen
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| |
Collapse
|
5
|
Abraham JE, O'Connor LO, Grybowicz L, Alba KP, Dayimu A, Demiris N, Harvey C, Drewett LM, Lucey R, Fulton A, Roberts AN, Worley JR, Chhabra MA, Qian W, Brown J, Hardy R, Vallier AL, Chan S, Cidon MEU, Sherwin E, Chakrabarti A, Sadler C, Barnes J, Persic M, Smith S, Raj S, Borley A, Braybrooke JP, Staples E, Scott LC, Palmer CA, Moody M, Churn MJ, Pilger D, Zagnoli-Vieira G, Wijnhoven PWG, Mukesh MB, Roylance RR, Schouten PC, Levitt NC, McAdam K, Armstrong AC, Copson ER, McMurtry E, Galbraith S, Tischkowitz M, Provenzano E, O'Connor MJ, Earl HM. Neoadjuvant PARP inhibitor scheduling in BRCA1 and BRCA2 related breast cancer: PARTNER, a randomized phase II/III trial. Nat Commun 2025; 16:4269. [PMID: 40360463 PMCID: PMC12075821 DOI: 10.1038/s41467-025-59151-0] [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: 10/25/2024] [Accepted: 04/11/2025] [Indexed: 05/15/2025] Open
Abstract
Poly (ADP-ribose) polymerase inhibitors (PARPi) exploit DNA repair deficiency in germline BRCA1 and BRCA2 pathogenic variant (gBRCAm) cancers. Haematological toxicity limits chemotherapy-PARPi treatment combinations. In preclinical models we identified a schedule combining olaparib and carboplatin that avoids enhanced toxicity but maintains anti-tumour activity. We investigated this schedule in a neoadjuvant, phase II-III, randomised controlled trial for gBRCAm breast cancers (ClinicalTrials.gov ID:NCT03150576; PARTNER). The research arm included carboplatin (Area Under the Curve 5, 3-weekly); paclitaxel (80 mg/m2, weekly) day 1, plus olaparib (150 mg twice daily) day 3-14 (4 cycles), followed by anthracycline-containing chemotherapy (3 cycles); control arm gave chemotherapy alone. The primary endpoint, pathological complete response rate, showed no statistical difference between research 64.1% (25/39); control 69.8% (30/43) (p = 0.59). However, estimated survival outcomes at 36-months demonstrated improved event-free survival: research 96.4%, control 80.1% (p = 0.04); overall survival: research 100%, control 88.2% (p = 0.04) and breast cancer specific survival: research 100%, control 88.2% (p = 0.04). There were no statistical differences in relapse-free survival and distant disease-free survival, both were: research 96.4%, control 87.9% (p = 0.20). Similarly, local recurrence-free survival and time to second cancer were both: research 96.4%, control 87.8% (p = 0.20). The PARTNER trial identified a safe, tolerable schedule combining neoadjuvant chemotherapy with olaparib. This combination demonstrated schedule-dependent overall survival benefit in early-stage gBRCAm breast cancer. This result needs confirmation in larger trials.
Collapse
Affiliation(s)
- Jean E Abraham
- Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, United Kingdom.
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom.
| | | | - Louise Grybowicz
- Cambridge Cancer Trials Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Karen Pinilla Alba
- Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - Alimu Dayimu
- Cambridge Clinical Trials Centre, Cancer Theme, University of Cambridge, Cambridge, United Kingdom
| | - Nikolaos Demiris
- Department of Statistics, Athens University of Economics and Business, Athens, Greece
| | - Caron Harvey
- Cambridge Cancer Trials Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Lynsey M Drewett
- Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom
| | - Rebecca Lucey
- Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - Alexander Fulton
- Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - Anne N Roberts
- Cambridge Cancer Trials Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Joanna R Worley
- Precision Breast Cancer Institute, Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, University of Cambridge, Cambridge, United Kingdom
| | - Ms Anita Chhabra
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Wendi Qian
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | | | - Richard Hardy
- Cambridge Clinical Trials Centre, Cancer Theme, University of Cambridge, Cambridge, United Kingdom
| | - Anne-Laure Vallier
- Cambridge Cancer Trials Centre, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Steve Chan
- The City Hospital, Nottingham University Hospitals NHS Trust, Nottingham, United Kingdom
- Nottingham Breast Cancer Research Centre, University of Nottingham, Nottingham, United Kingdom
| | - Maria Esther Una Cidon
- Royal Bournemouth General Hospital, University Hospitals Dorset NHS Foundation Trust, Bournemouth, United Kingdom
| | - Elizabeth Sherwin
- Ipswich Hospital, East Suffolk and North Essex NHS Foundation Trust, Ipswich, United Kingdom
| | | | - Claire Sadler
- Apconix Ltd, Alderley Edge, Cheshire, United Kingdom
| | | | - Mojca Persic
- University Hospital of Derby and Burton, Derby, United Kingdom
| | - Sarah Smith
- Bedford Hospital, Bedfordshire Hospitals NHS Foundation Trust, Bedford, United Kingdom
| | - Sanjay Raj
- University Hospital Southampton NHS Foundation Trust, Southampton, United Kingdom
- Hampshire Hospitals NHS Foundation Trust, Hampshire, United Kingdom
| | | | - Jeremy P Braybrooke
- University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Emma Staples
- Queens Hospital, Barking, Havering and Redbridge University Hospitals NHS Trust, Romford, United Kingdom
| | - Lucy C Scott
- Beatson West Of Scotland Cancer Centre, Glasgow, Scotland, United Kingdom
| | - Cheryl A Palmer
- Hinchingbrooke Hospital, North West Anglia NHS Foundation Trust, Huntingdon, United Kingdom
| | - Margaret Moody
- Macmillan Unit, West Suffolk Hospital NHS Foundation Trust, Bury St Edmunds, United Kingdom
| | - Mark J Churn
- Worcestershire Acute Hospitals NHS Trust, Worcester, United Kingdom
| | | | | | | | - Mukesh B Mukesh
- Colchester General Hospital, East Suffolk & North Essex NHS Trust, Colchester, United Kingdom
| | - Rebecca R Roylance
- University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Philip C Schouten
- Department of Histopathology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Nicola C Levitt
- Oxford University Hospital NHS Foundation Trust, Oxford, United Kingdom
| | - Karen McAdam
- Peterborough City Hospital, North West Anglia NHS Foundation Trust, Peterborough, United Kingdom
| | | | - Ellen R Copson
- Cancer Sciences Academic Unit, University of Southampton, Southampton, United Kingdom
| | | | | | - Marc Tischkowitz
- Department of Genomic Medicine, National Institute for Health Research Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| | - Elena Provenzano
- Department of Histopathology, Addenbrooke's Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | | | - Helena M Earl
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
6
|
Kashkinbayev Y, Kazhiyakhmetova B, Altaeva N, Bakhtin M, Tarlykov P, Saifulina E, Aumalikova M, Ibrayeva D, Bolatov A. Radon Exposure and Cancer Risk: Assessing Genetic and Protein Markers in Affected Populations. BIOLOGY 2025; 14:506. [PMID: 40427695 DOI: 10.3390/biology14050506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025]
Abstract
Radon is an inert gas produced by the radioactive decay of uranium-238, commonly found in the environment. Radon and its decay products are the main sources of human exposure to radiation from natural sources. When inhaled, radon's alpha particles impact lung tissue, potentially causing lung cancer by damaging DNA and altering oxidative processes. This review article addresses the need for a deeper understanding of the genetic and molecular changes associated with radon-induced lung cancer, aiming to clarify key genetic mutations and protein markers linked to carcinogenesis. Particular attention in recent studies has been given to mutations in tumor suppressor genes (RASSF1, TP53), oncogenes (KRAS, EGFR), and changes in the expression levels of protein biomarkers associated with inflammation, stress, and apoptosis. Identifying these markers is critical for developing effective screening methods for radon-induced lung cancer, enabling timely identification of high-risk patients and supporting effective preventive strategies. Summarizing current genetic and protein biomarkers, this review highlights the importance of a comprehensive approach to studying radon-induced carcinogenesis. Understanding these molecular mechanisms could ultimately improve early diagnostic methods and enhance therapy for cancers associated with radon exposure.
Collapse
Affiliation(s)
- Yerlan Kashkinbayev
- Institute of Radiobiology and Radiation Protection, Astana Medical University, Astana 010000, Kazakhstan
| | - Baglan Kazhiyakhmetova
- Institute of Radiobiology and Radiation Protection, Astana Medical University, Astana 010000, Kazakhstan
| | - Nursulu Altaeva
- Medical Genetics and Molecular Biology Department, Astana Medical University, Astana 010000, Kazakhstan
| | - Meirat Bakhtin
- Institute of Radiobiology and Radiation Protection, Astana Medical University, Astana 010000, Kazakhstan
| | - Pavel Tarlykov
- National Center for the Biotechnology, Astana 010000, Kazakhstan
| | - Elena Saifulina
- Institute of Radiobiology and Radiation Protection, Astana Medical University, Astana 010000, Kazakhstan
| | - Moldir Aumalikova
- Institute of Radiobiology and Radiation Protection, Astana Medical University, Astana 010000, Kazakhstan
| | - Danara Ibrayeva
- Institute of Radiobiology and Radiation Protection, Astana Medical University, Astana 010000, Kazakhstan
| | - Aidos Bolatov
- School of Medicine, Shenzhen University, Shenzhen 518060, China
- School of Medicine, Astana Medical University, Astana 010000, Kazakhstan
| |
Collapse
|
7
|
Prieto-Dominguez N, Goel P, Ojo OA, Moretto K, Holtzhausen A, Humphryes A, Zhou X, Kuznetsova V, Dempsey F, Pittman K, Lu R, Green TJ, Shi LZ, Welner RS, Earp HS, Ubil E. PTP Inhibition Improves the Macrophage Antitumor Immune Response and the Efficacy of Chemo- and Radiotherapy. Cancer Immunol Res 2025; 13:749-766. [PMID: 39937118 PMCID: PMC12048275 DOI: 10.1158/2326-6066.cir-24-0335] [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: 04/12/2024] [Revised: 10/15/2024] [Accepted: 02/11/2025] [Indexed: 02/13/2025]
Abstract
Traditional anticancer therapies induce tumor cell death and subsequent release of damage-associated molecular patterns (DAMPs) that activate the innate inflammatory response. Paradoxically, after treatment, macrophages often adopt a pro-wound healing, rather than proinflammatory, phenotype and contribute to cancer progression. We found that in areas proximal to DAMP release, tumor cells upregulate the expression of Pros1. Tumor-secreted Pros1 binds to the macrophage Mer receptor, consequently limiting responsiveness to DAMPs by preventing Toll-like receptor signal transduction. Pharmacological inhibition of PTP1b signaling downstream of Mer rescued the proinflammatory response, even in the presence of Pros1. Combining protein tyrosine phosphatase (PTP) inhibition with traditional therapeutics, such as chemo- or radiotherapy, rescued the innate immune response to DAMPs, increased immune infiltration, and resulted in a 40% to 90% reduction in tumor growth in multiple treatment-refractory preclinical models. Our findings suggest using PTP1b inhibitors may be a tumor agnostic means of improving the efficacy of some of the most widely used anticancer therapeutic agents.
Collapse
Affiliation(s)
| | - Paran Goel
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Oluwagbemiga A. Ojo
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Katarina Moretto
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Alisha Holtzhausen
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Departments of Medicine and Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Angel Humphryes
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Xinyue Zhou
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Valeriya Kuznetsova
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Francesca Dempsey
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kelly Pittman
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Rui Lu
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Todd J. Green
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lewis Z. Shi
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Robert S. Welner
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - H. Shelton Earp
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Departments of Medicine and Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Eric Ubil
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, Alabama, USA
- O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
8
|
Taniguchi R, Orniacki C, Kreysing JP, Zila V, Zimmerli CE, Böhm S, Turoňová B, Kräusslich HG, Doye V, Beck M. Nuclear pores safeguard the integrity of the nuclear envelope. Nat Cell Biol 2025; 27:762-775. [PMID: 40205196 DOI: 10.1038/s41556-025-01648-3] [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: 02/28/2024] [Accepted: 02/25/2025] [Indexed: 04/11/2025]
Abstract
Nuclear pore complexes (NPCs) mediate nucleocytoplasmic exchange, which is essential for eukaryotes. Mutations in the central scaffolding components of NPCs are associated with genetic diseases, but how they manifest only in specific tissues remains unclear. This is exemplified in Nup133-deficient mouse embryonic stem cells, which grow normally during pluripotency, but differentiate poorly into neurons. Here, using an innovative in situ structural biology approach, we show that Nup133-/- mouse embryonic stem cells have heterogeneous NPCs with non-canonical symmetries and missing subunits. During neuronal differentiation, Nup133-deficient NPCs frequently disintegrate, resulting in abnormally large nuclear envelope openings. We propose that the elasticity of the NPC scaffold has a protective function for the nuclear envelope and that its perturbation becomes critical under conditions that impose an increased mechanical load onto nuclei.
Collapse
Affiliation(s)
- Reiya Taniguchi
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Japan
| | - Clarisse Orniacki
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France
- The Neuro - Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Jan Philipp Kreysing
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- IMPRS on Cellular Biophysics, Frankfurt am Main, Germany
| | - Vojtech Zila
- Department of Infectious Diseases, Virology, Heidelberg University, Heidelberg, Germany
- AskBio GmbH, Heidelberg, Germany
| | - Christian E Zimmerli
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
- Institute of Physics, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Stefanie Böhm
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Beata Turoňová
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Hans-Georg Kräusslich
- Department of Infectious Diseases, Virology, Heidelberg University, Heidelberg, Germany
- German Centre for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
| | - Valérie Doye
- Université Paris Cité, CNRS, Institut Jacques Monod, Paris, France.
| | - Martin Beck
- Department of Molecular Sociology, Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
- Institute of Biochemistry, Goethe University Frankfurt, Frankfurt am Main, Germany.
| |
Collapse
|
9
|
Pray B, Baiocchi E, Leon S, Barta B, Koirala S, Tsyba L, Hinterschied C, Carvajal-Moreno J, Hout I, Nishat S, Jindal U, Jain N, Singh S, Sehgal L, Chan WK, Hanel W, Yalowich J, Baiocchi RA, Alinari L. Targeting the DNA damage response through TBL1X in mantle cell lymphoma. Blood Adv 2025; 9:2006-2018. [PMID: 40009753 PMCID: PMC12034073 DOI: 10.1182/bloodadvances.2024015769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
ABSTRACT Mantle cell lymphoma (MCL) is an incurable B-cell lymphoma characterized by significant genomic instability. Patients with MCL who progress on targeted therapies have a short survival; thus, novel therapeutic strategies are urgently needed. Overexpression of transducin β-like protein 1 X-linked (TBL1X) has been documented in several types of cancer and associated with poor prognosis. TBL1X is a critical regulator of multiple oncogenic networks; however, its function in MCL has not been explored. Our data show that, unlike normal B cells, MCL cells express abundant levels of TBL1X and that genetic knockdown of TBL1X and treatment with tegavivint (Iterion), a first-in-class small molecule targeting TBL1X, promote MCL cell death in vitro and in vivo. Moreover, TBL1X controls the stability of key MCL oncogenic drivers, cyclin D1 and RAD51; and targeting TBL1X results in significant DNA damage, cell cycle arrest, and ultimately cell death. Combining tegavivint with poly(adenosine 5'-diphosphate-ribose) polymerase-1/2 inhibitor talazoparib results in synergistic MCL cell death in vitro, and in vivo this combination significantly prolongs the survival of a patient-derived MCL xenograft. Together, our results define the role of TBL1X in maintaining genomic stability in MCL and establish targeting TBL1X as a novel therapeutic strategy for patients with this incurable disease.
Collapse
Affiliation(s)
- Betsy Pray
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH
| | - Ethan Baiocchi
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Sydney Leon
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Brian Barta
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Shirsha Koirala
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Liudmyla Tsyba
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Claire Hinterschied
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Jessika Carvajal-Moreno
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Ian Hout
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Shamama Nishat
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Udita Jindal
- Academy of Scientific and Innovative Research, Ghaziabad, India
| | - Neeraj Jain
- Division of Cancer Biology, Council of Scientific and Industrial Research-Central Drug Research Institute, Lucknow, India
| | - Satishkumar Singh
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Lalit Sehgal
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Wing Keung Chan
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Walter Hanel
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Jack Yalowich
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Robert A. Baiocchi
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| | - Lapo Alinari
- Division of Hematology, Department of Internal Medicine, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH
| |
Collapse
|
10
|
Lukasak BJ, Korb E. Histone variants: expanding the epigenetic potential of neurons one amino acid at a time. Trends Biochem Sci 2025:S0968-0004(25)00079-9. [PMID: 40268580 DOI: 10.1016/j.tibs.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 04/25/2025]
Abstract
Replication-independent histone variants play an essential role in postmitotic neurons. Here, we review how the subtle sequence differences of histone variants compared to their canonical counterparts underly neuronal function. We focus on variants H3.3, H2A.Z, H2A.X, macroH2A, and H2BE; all of which contain divergent sequences that coordinate a diverse set of outcomes. In particular, we highlight their role in neuronal development, plasticity, and memory, with an emphasis on how single amino acid changes can mediate these complex functions. Lastly, we comment on an emerging field of study evaluating the link between histone variants and neurological disorders. Future studies of histone variants will be important to furthering our understanding of neuronal function.
Collapse
Affiliation(s)
- Bradley J Lukasak
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Erica Korb
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Epigenetics Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Yang L, Wei W, Yuan X, Guo E, Peng P, Wang J, Sun W. Targeting DNA Damage Repair to Enhance Antitumor Immunity in Radiotherapy: Mechanisms and Opportunities. Int J Mol Sci 2025; 26:3743. [PMID: 40332379 PMCID: PMC12027993 DOI: 10.3390/ijms26083743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/07/2025] [Accepted: 04/13/2025] [Indexed: 05/08/2025] Open
Abstract
Radiotherapy is a standard cancer treatment that involves the induction of DNA damage. DNA damage repair (DDR) pathways maintain genomic integrity and make tumors resistant to radiotherapy and certain chemotherapies. In turn, DDR dysfunction results in cumulative DNA damage, leading to increased sensitivity for antitumor treatment. Moreover, radiotherapy has been shown to trigger antitumor immunity. Currently, immunotherapy has become a new and widely used standard strategy for treating a broad spectrum of tumor types. Notably, recent studies have demonstrated that DDR pathways play important roles in driving the response to immunotherapy. Herein, we review and discuss how DDR affects antitumor immunity induced by radiotherapy. Furthermore, we summarize the development of strategies for combining DDR inhibitors with radiotherapy and/or immunotherapy to enhance their efficacy against cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Wei Sun
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (L.Y.); (W.W.); (X.Y.); (E.G.); (P.P.); (J.W.)
| |
Collapse
|
12
|
Santos JF, Laere CV, Silva CD, Cassells I, Fernandes C, Raposinho P, Belchior A, Pinto CIG, Mendes F, Cawthorne C, Ooms M, Voorde MVD, Cleeren F, Paulo A. Mitochondria-tropic radioconjugates to enhance the therapeutic potential of terbium-161. EJNMMI Radiopharm Chem 2025; 10:18. [PMID: 40214871 PMCID: PMC11992321 DOI: 10.1186/s41181-025-00339-6] [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: 01/07/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Strategies that focus on delivering Auger electron emitters to highly radiosensitive intracellular targets-such as the nucleus, cell membrane, or mitochondria-are gaining attention. Targeting these organelles could enhance therapeutic efficacy while minimizing off-target toxicity by allowing lower administered doses. In this context, this study explores the therapeutic potential of 161Tb-labeled radiocomplexes that integrate the mitochondria-targeting triphenylphosphonium (TPP) moiety with a prostate-specific membrane antigen (PSMA) targeting vector. The goal is to assess these dual-targeted radiocomplexes for their ability to deliver conversion electrons (CE) and Auger electrons (AEs) to prostate cancer (PCa) cells, specifically targeting the mitochondria to enhance therapeutic efficacy. RESULTS Two novel radiocomplexes, [161Tb]Tb-TPP-PSMA and [161Tb]Tb-TPP-G3-PSMA, were synthesized with high radiochemical yield and purity. The proposed structures were validated using HPLC and ESI-MS analysis, with their natTb counterparts serving as reference compounds. In vitro experiments included cellular uptake, internalization, mitochondrial uptake, and DNA damage assays in PSMA-positive PCa cell lines. Clonogenic assays were performed to evaluate cell survival post-treatment. In vivo studies were conducted using SCID/Beige mice bearing PCa xenografts and involved µSPECT/CT imaging and radiometabolite analysis to evaluate biodistribution, pharmacokinetics, tumor uptake and in vivo stability of the radiocomplexes. Both [161Tb]Tb-TPP-PSMA and [161Tb]Tb-TPP-G3-PSMA showed high radiochemical stability and were efficiently internalized by PSMA-positive cells, while showing minimal uptake in PSMA-negative cells. These dual-targeted radiocomplexes demonstrated significantly higher mitochondrial uptake compared to the non-TPP-containing [161Tb]Tb-PSMA-617, leading to increased DNA damage and enhanced radiocytotoxicity. In vivo, the dual-targeted complexes demonstrated PSMA-specific tumor uptake and pharmacokinetics comparable to [161Tb]Tb-PSMA-617, with effective clearance from non-target tissues. CONCLUSIONS The TPP-modified 161Tb-radiocomplexes effectively targeted the mitochondria of PSMA-positive PCa cells, leading to increased DNA damage and reduced cell viability compared to single-targeted radiocomplexes. These findings suggest that dual-targeting strategies, which combine PSMA and mitochondrial targeting, can enhance the therapeutic potential of radiopharmaceuticals for prostate cancer treatment.
Collapse
Affiliation(s)
- Joana F Santos
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Camille Van Laere
- Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, B-3000, Belgium
| | - Catarina D Silva
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Irwin Cassells
- Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, B-3000, Belgium
| | - Célia Fernandes
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Paula Raposinho
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Ana Belchior
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Catarina I G Pinto
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Filipa Mendes
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Christopher Cawthorne
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University of Leuven, Leuven, Belgium
| | - Maarten Ooms
- Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Michiel Van de Voorde
- Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, B-3000, Belgium.
| | - António Paulo
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal.
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal.
| |
Collapse
|
13
|
Bril M, Boesveld JN, Coelho-Rato LS, Sahlgren CM, Bouten CVC, Kurniawan NA. Dynamic substrate topographies drive actin- and vimentin-mediated nuclear mechanoprotection events in human fibroblasts. BMC Biol 2025; 23:94. [PMID: 40189524 PMCID: PMC11974106 DOI: 10.1186/s12915-025-02199-7] [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: 01/27/2025] [Accepted: 03/27/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Dynamic physical changes in the extracellular environment of living tissues present a mechanical challenge for resident cells that can lead to damage to the nucleus, genome, and DNA. Recent studies have started to uncover nuclear mechanoprotection mechanisms that prevent excessive mechanical deformations of the nucleus. Here, we hypothesized that dynamic topographical changes in the cellular environment can be mechanically transmitted to the nucleus and trigger nuclear mechanoprotection events. We tested this using a photoresponsive hydrogel whose surface topography can be reversibly changed on demand upon light illumination, allowing us to subject cells to recurring microscale topographical changes. RESULTS With each recurring topographical change, fibroblasts were found to increasingly compact and relocate their nuclei away from the dynamic regions of the hydrogel. These cell-scale reorganization events were accompanied by an increase of global histone acetylation and decreased methylation in cells on the dynamic topographies, resulting in a minimization of DNA strand breakage. We further found that these nuclear mechanoprotection events were mediated by both vimentin intermediate filaments and the actin cytoskeleton. CONCLUSIONS Together, these data reveal that fibroblasts actively protect their nuclei in the presence of dynamic topographical changes through cytoskeleton-mediated mechanisms. Broadly, these results stress the importance of gaining a deeper fundamental understanding of the cellular mechanoresponse under dynamically changing conditions.
Collapse
Affiliation(s)
- Maaike Bril
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Jules N Boesveld
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Leila S Coelho-Rato
- Faculty of Science and Engineering, Åbo Akademi University, 20520, Turku, Finland
| | - Cecilia M Sahlgren
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
- Faculty of Science and Engineering, Åbo Akademi University, 20520, Turku, Finland
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands
| | - Nicholas A Kurniawan
- Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, PO Box 513, 5600 MB, Eindhoven, The Netherlands.
| |
Collapse
|
14
|
Skamagki M, Zhang C, Hacisuleyman E, Galleti G, Wu C, Vinagolu RK, Cha H, Ata D, Kim J, Weiskittel T, Diop M, Aung T, Del Latto M, Kim AS, Li Z, Miele M, Zhao R, Tang LH, Hendrickson RC, Romesser PB, Smith JJ, Giannakakou P, Darnell RB, Bott MJ, Li H, Kim K. Aging-dependent dysregulation of EXOSC2 is maintained in cancer as a dependency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647279. [PMID: 40236131 PMCID: PMC11996493 DOI: 10.1101/2025.04.04.647279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Reprogramming of aged donor tissue cells into induced pluripotent stem cells (A-iPSC) preserved the epigenetic memory of aged-donor tissue, defined as genomic instability and poor tissue differentiation in our previous study. The unbalanced expression of RNA exosome subunits affects the RNA degradation complex function and is associated with geriatric diseases including premature aging and cancer progression. We hypothesized that the age-dependent progressive subtle dysregulation of EXOSC2 (exosome component 2) causes the aging traits (abnormal cell cycle and poor tissue differentiation). We used embryonic stem cells as a tool to study EXOSC2 function as the aging trait epigenetic memory determined in A-iPSC because these aging traits could not be studied in senesced aged cells or immortalized cancer cells. We found that the regulatory subunit of PP2A phosphatase, PPP2R5E, is a key target of EXOSC2 and this regulation is preserved in stem cells and cancer.
Collapse
|
15
|
Marian AJ. Causes and consequences of DNA double-stranded breaks in cardiovascular disease. Mol Cell Biochem 2025; 480:2043-2064. [PMID: 39404936 DOI: 10.1007/s11010-024-05131-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 09/29/2024] [Indexed: 04/02/2025]
Abstract
The genome, whose stability is essential for survival, is incessantly exposed to internal and external stressors, which introduce an estimated 104 to 105 lesions, such as oxidation, in the nuclear genome of each mammalian cell each day. A delicate homeostatic balance between the generation and repair of DNA lesions maintains genomic stability. To initiate transcription, DNA strands unwind to form a transcription bubble and provide a template for the RNA polymerase II (RNAPII) complex to synthesize nascent RNA. The process generates DNA supercoils and introduces torsional stress. To enable RNAPII processing, the supercoils are released by topoisomerases by introducing strand breaks, including double-stranded breaks (DSBs). Thus, DSBs are intrinsic genomic features of gene expression. The breaks are quickly repaired upon processing of the transcription. DNA lesions and damaged proteins involved in transcription could impede the integrity and efficiency of RNAPII processing. The impediment, which is referred to as transcription stress, not only could lead to the generation of aberrant RNA species but also the accumulation of DSBs. The latter is particularly the case when topoisomerase processing and/or the repair mechanisms are compromised. The DSBs activate the DNA damage response (DDR) pathways to repair the damaged DNA and/or impose cell cycle arrest and cell death. In addition, the release of DSBs into the cytosol activates the cytosolic DNA-sensing proteins (CDSPs), which along with the nuclear DDR pathways induce the expression of senescence-associated secretory phenotype (SASP), cell cycle arrest, senescence, cell death, inflammation, and aging. The primary stimulus in hereditary cardiomyopathies is a mutation(s) in genes encoding the protein constituents of cardiac myocytes; however, the phenotype is the consequence of intertwined complex interactions among numerous stressors and the causal mutation(s). Increased internal DNA stressors, such as oxidation, alkylation, and cross-linking, are expected to be common in pathological conditions, including in hereditary cardiomyopathies. In addition, dysregulation of gene expression also imposes transcriptional stress and collectively with other stressors provokes the generation of DSBs. In addition, the depletion of nicotinamide adenine dinucleotide (NAD), which occurs in pathological conditions, impairs the repair mechanism and further facilitates the accumulation of DSBs. Because DSBs activate the DDR pathways, they are expected to contribute to the pathogenesis of cardiomyopathies. Thus, interventions to reduce the generation of DSBs, enhance their repair, and block the deleterious DDR pathways would be expected to impart salubrious effects not only in pathological states, as in hereditary cardiomyopathies but also aging.
Collapse
Affiliation(s)
- A J Marian
- Center for Cardiovascular Genetic Studies, Institute of Molecular Medicine, The University of Texas Health Science Center, 6770 Bertner Street, Suite C900A, Houston, TX, 77030, USA.
| |
Collapse
|
16
|
Meng Y, Xu Y, Qiu A, Lin M, Yang L, Huang Y. USP28-mediated deubiquitination of FOXK1 activates the Hippo signaling pathway to regulate cell proliferation and radiosensitivity in lung cancer. Life Sci 2025; 366-367:123483. [PMID: 39983825 DOI: 10.1016/j.lfs.2025.123483] [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: 11/08/2024] [Revised: 01/17/2025] [Accepted: 02/16/2025] [Indexed: 02/23/2025]
Abstract
AIMS Radioresistance remains a significant challenge for lung cancer therapeutics. Forkhead box K1 (FOXK1) plays a role in regulating various biological processes and the progression of multiple cancers. However, the role of FOXK1 in lung cancer progression and radioresistance are not fully understood. MAIN METHODS Functional analyses were conducted on lung cancer cells transfected with specified siRNAs or plasmids. The ubiquitination of FOXK1 was evaluated by in vitro ubiquitination assays. RNA sequencing analysis was conducted to identify the downstream signaling pathway regulated by FOXK1. Mouse xenograft models were constructed using lung cancer cells with stable expression of either sh-NC or sh-FOXK1. Immunohistochemistry was used to assess FOXK1 and USP28 expression levels in lung cancer and paired normal lung tissues. KEY FINDINGS We found that elevated FOXK1 expression markedly enhances radioresistance and tumorigenesis in lung cancer. Furthermore, we demonstrated that ubiquitin specific peptidase 28 (USP28) interacts with and targets FOXK1 for deubiquitination and stabilization. Moreover, we showed that FOXK1 exerts its biological function via activating the Hippo pathway. SIGNIFICANCE Our research showed that FOXK1 is deubiquitinated by USP28 and facilitates cell proloferation and radioresistance by activating the Hippo pathway, suggesting that FOXK1 may act as a potential radiosensitizing target for lung cancer.
Collapse
Affiliation(s)
- Yimei Meng
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Yunhong Xu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Anchen Qiu
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Ming Lin
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Liu Yang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Zhejiang Province Key Disciplines in Traditional Chinese Medicine-Integrated Traditional Chinese and Western Medicine Clinical Oncology, Hangzhou, Zhejiang, 310014, China.
| | - Yumei Huang
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Zhejiang Province Key Disciplines in Traditional Chinese Medicine-Integrated Traditional Chinese and Western Medicine Clinical Oncology, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
17
|
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
|
18
|
Lee J, Kim D, Cha SJ, Lee JW, Lee EY, Kim HJ, Kim K. Tau reduction impairs nephrocyte function in Drosophila. BMB Rep 2025; 58:169-174. [PMID: 39757203 PMCID: PMC12041923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 05/14/2024] [Accepted: 08/02/2024] [Indexed: 01/07/2025] Open
Abstract
Tau, a microtubule-associated protein, is known for its significant involvement in neurodegenerative diseases. While various molecular and immunohistochemical techniques have confirmed the presence of Tau in podocytes, its precise function within these cells remains elusive. In this study, we investigate the role of Tau in kidney podocytes using Drosophila pericardial nephrocytes as a model. We found that knockdown of Drosophila Tau in nephrocytes resulted in apoptotic cell death and the disruption of nephrocyte structure. Furthermore, we observed that decreased Tau levels induced genomic damage and abnormal distribution of γ-H2Av, altering nuclei architecture in nephrocytes, and affecting the nuclear membrane structure by interfering with lamin with aging. Additionally, Tau knockdown led to a reduction in lipid droplets in Drosophila fat body tissues, suggesting a potential role of Tau in inter-organ communication. These findings underscore the importance of Tau in the nephrocytes of Drosophila, and advocate further research to broaden our understanding of podocyte biology in kidney diseases. [BMB Reports 2025; 58(4): 169-174].
Collapse
Affiliation(s)
- Jiyoung Lee
- Department of Medical Science, Soonchunhyang University, Asan 31538, Korea
| | - Dayoung Kim
- Department of Medical Biotechnology, Soonchunhyang University, Asan 31538, Korea
| | - Sun Joo Cha
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu 41068, Korea
| | - Jang-Won Lee
- Department of Integrated Bio-Industry, Sejong University, Seoul 05006, Korea
| | - Eun-Young Lee
- Division of Nephrology, Department of Internal Medicine, Cheonan Hospital, Soonchunhyang University, Cheonan 31151, Korea
| | - Hyung-Jun Kim
- Dementia Research Group, Korea Brain Research Institute (KBRI), Daegu 41068, Korea
| | - Kiyoung Kim
- Department of Medical Science, Soonchunhyang University, Asan 31538, Korea
| |
Collapse
|
19
|
Abrahams RR, Majumder K. Small Genomes, Big Disruptions: Parvoviruses and the DNA Damage Response. Viruses 2025; 17:494. [PMID: 40284937 PMCID: PMC12031541 DOI: 10.3390/v17040494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
Parvoviruses are small, single-stranded DNA viruses that have evolved sophisticated mechanisms to hijack host cell machinery for replication and persistence. One critical aspect of this interaction involves the manipulation of the host's DNA Damage Response (DDR) pathways. While the viral genome is comparatively simple, parvoviruses have developed strategies that cause significant DNA damage, activate DDR pathways, and disrupt the host cell cycle. This review will explore the impact of parvovirus infections on host genome stability, focusing on key viral species such as Adeno-Associated Virus (AAV), Minute Virus of Mice (MVM), and Human Bocavirus (HBoV), and their interactions with DDR proteins. Since parvoviruses are used as oncolytic agents and gene therapy vectors, a better understanding of cellular DDR pathways will aid in engineering potent anti-cancer agents and gene therapies for chronic diseases.
Collapse
Affiliation(s)
| | - Kinjal Majumder
- Institute for Molecular Virology and McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, WI 53707, USA;
| |
Collapse
|
20
|
Zhang L, Wang J, Tang Z, Lin Z, Su R, Hu N, Tang Y, Ge G, Fan J, Tong MH, Xue Y, Zhou Y, Cheng H. The nuclear exosome co-factor MTR4 shapes the transcriptome for meiotic initiation. Nat Commun 2025; 16:2605. [PMID: 40097464 PMCID: PMC11914058 DOI: 10.1038/s41467-025-57898-0] [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: 02/08/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
Nuclear RNA decay has emerged as a mechanism for post-transcriptional gene regulation in cultured cells. However, whether this process occurs in animals and holds biological relevance remains largely unexplored. Here, we demonstrate that MTR4, the central cofactor of the nuclear RNA exosome, is essential for embryogenesis and spermatogenesis. Embryonic development of Mtr4 knockout mice arrests at 6.5 day. Germ cell-specific knockout of Mtr4 results in male infertility with a specific and severe defect in meiotic initiation. During the pre-meiotic stage, MTR4/exosome represses meiotic genes, which are typically shorter in size and possess fewer introns, through RNA degradation. Concurrently, it ensures the expression of mitotic genes generally exhibiting the opposite features. Consistent with these regulation rules, mature replication-dependent histone mRNAs and polyadenylated retrotransposon RNAs were identified as MTR4/exosome targets in germ cells. In addition, MTR4 regulates alternative splicing of many meiotic genes. Together, our work underscores the importance of nuclear RNA degradation in regulating germline transcriptome, ensuring the appropriate gene expression program for the transition from mitosis to meiosis during spermatogenesis.
Collapse
Affiliation(s)
- Li Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jianshu Wang
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhidong Tang
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, RNA Institute, Wuhan University, Wuhan, China
- Key Laboratory of Molecular Biology on Infectious Diseases, Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Zhen Lin
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ruibao Su
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Naijing Hu
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yao Tang
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, RNA Institute, Wuhan University, Wuhan, China
| | - Gaoxiang Ge
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jing Fan
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ming-Han Tong
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yuanchao Xue
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
| | - Yu Zhou
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Hubei Key Laboratory of Cell Homeostasis, RNA Institute, Wuhan University, Wuhan, China.
| | - Hong Cheng
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Key Laboratory of Molecular Andrology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
21
|
Ocampo J, Carena S, López MDR, Vela VS, Zambrano Siri RT, Balestra SA, Alonso GD. Trypanosomatid histones: the building blocks of the epigenetic code of highly divergent eukaryotes. Biochem J 2025; 482:BCJ20240543. [PMID: 40094426 DOI: 10.1042/bcj20240543] [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: 11/05/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/19/2025]
Abstract
Histones play a fundamental role in eukaryotic organisms not only as scaffolding proteins in DNA packaging but also in regulating gene expression. They constitute the protein reel around which DNA wraps forming nucleosomes. This initial packing gives rise to the chromatin fiber which is next folded into three-dimensional arrangements. Additionally, histones have expanded their functions through the emergence of histone variants which have specialized purposes and can deeply affect chromatin organization and dynamics. Moreover, both canonical histones and histone variants comprise the building blocks of the histone code by being targets of different post-translational modifications (PTMs) that occur in a highly regulated manner both in place and time. Most of the above-mentioned about chromatin organization is conserved among eukaryotes. However, trypanosomatid histones have many peculiarities that entail a special description. In this review, we compile the current knowledge of canonical core histones, histone variants, and their PTMs in trypanosomatids. We highlight the similarities and differences between histone variants and their canonical counterparts in trypanosomatids, and we compare them with those from model organisms. Finally, we discuss the crosstalk between different histone marks and their genomic distribution underlying the uniqueness of trypanosomatids.
Collapse
Affiliation(s)
- Josefina Ocampo
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Santiago Carena
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - María Del Rosario López
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Valentina Sol Vela
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Romina Trinidad Zambrano Siri
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Sofia Antonella Balestra
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Guillermo Daniel Alonso
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular "Dr. Héctor N. Torres" (INGEBI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
- Departamento de Fisiología, Biología Molecular y Celular, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
22
|
Asami N, Yano S, Tsuruta F. Potential for micronuclear turnover through autophagy secretion pathway. MICROPUBLICATION BIOLOGY 2025; 2025:10.17912/micropub.biology.001545. [PMID: 40134942 PMCID: PMC11933925 DOI: 10.17912/micropub.biology.001545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Revised: 03/01/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025]
Abstract
Micronuclei (MN) serve as well-established markers of genomic instability. MN arise from various stresses, such as segregation errors and mechanical stress, and are subsequently eliminated by the autophagy pathway. It has been suggested that MN are traditionally considered markers of cancer cells, often without recognized functional significance. Meanwhile, we recently discovered that MN act as mediators in regulating microglial characteristics. Neurons produce MN in response to migrating stress during the developmental stage and release them to the extracellular space, subsequently transferring them to microglia. In this study, we report the potential mechanisms underlying MN release through the autophagic secretion pathway. Our data show a possibility by which damaged MN are recognized autophagy regulatory factors, resulting in the propagation of MN to microglia.
Collapse
Affiliation(s)
- Natsu Asami
- Master's Program in Biology, Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Sarasa Yano
- Graduate School of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| | - Fuminori Tsuruta
- Master's and Doctoral Program in Biology, Institute of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
- Master's and Doctoral Program in Neuroscience, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
- Doctoral Program in Human Biology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
- Doctoral Program in Humanics, School of Integrative and Global Majors, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
- Center for Quantum and Information Life Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki 305-8577, Japan
| |
Collapse
|
23
|
Alexandrova E, Smal M, Di Rosa D, Brancaccio RN, Parisi R, Russo F, Tarallo R, Nassa G, Giurato G, Weisz A, Rizzo F. BRPF1 inhibition reduces migration and invasion of metastatic ovarian cancer cells, representing a potential therapeutic target. Sci Rep 2025; 15:7602. [PMID: 40038391 PMCID: PMC11880521 DOI: 10.1038/s41598-025-92438-2] [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: 05/10/2024] [Accepted: 02/27/2025] [Indexed: 03/06/2025] Open
Abstract
Ovarian Cancer (OC) is the most lethal gynecological malignancy, characterized by peritoneal metastasis, directly linked to most OC-related deaths. Here, by interrogating CRISPR-Cas9 loss-of-function genetic screen data, we identified a list of genes essential for metastatic OC, including several well-known oncogenes (PAX8, CCNE1, WWTR1, WT1, KAT6A, MECOM, and SOX17) and others whose roles in OC have not yet been explored. Protein-protein interaction analysis of the selected genes revealed the presence of a protein network participating in the epigenetic regulation of gene expression. For one of the network components, BRPF1, we found that its increased expression correlates with OC progression and a poor prognosis for OC patients. Functional assays demonstrated that BRPF1 inhibition significantly reduces cellular migration and invasion, supporting its role in metastatic progression. Pharmacological blockade of BRPF1 using small molecule inhibitors resulted in reduced proliferation of high-grade serous OC cells through mechanisms involving the activation of programmed cell death, cell cycle deregulation, and enhanced DNA damage. Furthermore, analysis of transcriptional changes induced by BRPF1 targeting showed that the growth inhibitory effects may be mediated by the deregulation of PPARα signaling. The obtained results indicate that BRPF1 represents a novel potential therapeutic target for metastatic OC treatment.
Collapse
Affiliation(s)
- Elena Alexandrova
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica, Salernitana" University of Salerno, via S. Allende, 1, Baronissi, 84081, SA, Italy.
- Medical Genomics Program, Division of Oncology, AOU "S. Giovanni di Dio e Ruggi d'Aragona", University of Salerno, Salerno, Italy.
| | - Marharyta Smal
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica, Salernitana" University of Salerno, via S. Allende, 1, Baronissi, 84081, SA, Italy
| | - Domenico Di Rosa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica, Salernitana" University of Salerno, via S. Allende, 1, Baronissi, 84081, SA, Italy
| | - Rosario Nicola Brancaccio
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica, Salernitana" University of Salerno, via S. Allende, 1, Baronissi, 84081, SA, Italy
| | - Roberto Parisi
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica, Salernitana" University of Salerno, via S. Allende, 1, Baronissi, 84081, SA, Italy
| | - Fabio Russo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica, Salernitana" University of Salerno, via S. Allende, 1, Baronissi, 84081, SA, Italy
- Medical Genomics Program, Division of Oncology, AOU "S. Giovanni di Dio e Ruggi d'Aragona", University of Salerno, Salerno, Italy
| | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica, Salernitana" University of Salerno, via S. Allende, 1, Baronissi, 84081, SA, Italy
- Medical Genomics Program, Division of Oncology, AOU "S. Giovanni di Dio e Ruggi d'Aragona", University of Salerno, Salerno, Italy
- Genome Research Center for Health-CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica, Salernitana" University of Salerno, via S. Allende, 1, Baronissi, 84081, SA, Italy
- Medical Genomics Program, Division of Oncology, AOU "S. Giovanni di Dio e Ruggi d'Aragona", University of Salerno, Salerno, Italy
- Genome Research Center for Health-CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
| | - Giorgio Giurato
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica, Salernitana" University of Salerno, via S. Allende, 1, Baronissi, 84081, SA, Italy
- Genome Research Center for Health-CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica, Salernitana" University of Salerno, via S. Allende, 1, Baronissi, 84081, SA, Italy
- Medical Genomics Program, Division of Oncology, AOU "S. Giovanni di Dio e Ruggi d'Aragona", University of Salerno, Salerno, Italy
- Genome Research Center for Health-CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica, Salernitana" University of Salerno, via S. Allende, 1, Baronissi, 84081, SA, Italy.
- Medical Genomics Program, Division of Oncology, AOU "S. Giovanni di Dio e Ruggi d'Aragona", University of Salerno, Salerno, Italy.
- Genome Research Center for Health-CRGS, Campus of Medicine of the University of Salerno, Baronissi, Italy.
| |
Collapse
|
24
|
Höfer S, Frasch L, Brajkovic S, Putzker K, Lewis J, Schürmann H, Leone V, Sakhteman A, The M, Bayer FP, Müller J, Hamood F, Siveke JT, Reichert M, Kuster B. Gemcitabine and ATR inhibitors synergize to kill PDAC cells by blocking DNA damage response. Mol Syst Biol 2025; 21:231-253. [PMID: 39838187 PMCID: PMC11876601 DOI: 10.1038/s44320-025-00085-6] [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: 04/02/2024] [Revised: 12/22/2024] [Accepted: 01/03/2025] [Indexed: 01/23/2025] Open
Abstract
The DNA-damaging agent Gemcitabine (GEM) is a first-line treatment for pancreatic cancer, but chemoresistance is frequently observed. Several clinical trials investigate the efficacy of GEM in combination with targeted drugs, including kinase inhibitors, but the experimental evidence for such rationale is often unclear. Here, we phenotypically screened 13 human pancreatic adenocarcinoma (PDAC) cell lines against GEM in combination with 146 clinical inhibitors and observed strong synergy for the ATR kinase inhibitor Elimusertib in most cell lines. Dose-dependent phosphoproteome profiling of four ATR inhibitors following DNA damage induction by GEM revealed a strong block of the DNA damage response pathway, including phosphorylated pS468 of CHEK1, as the underlying mechanism of drug synergy. The current work provides a strong rationale for why the combination of GEM and ATR inhibition may be useful for the treatment of PDAC patients and constitutes a rich phenotypic and molecular resource for further investigating effective drug combinations.
Collapse
Affiliation(s)
- Stefanie Höfer
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Larissa Frasch
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Sarah Brajkovic
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Kerstin Putzker
- Chemical Biology Core Facility, EMBL Heidelberg, Heidelberg, Germany
| | - Joe Lewis
- Chemical Biology Core Facility, EMBL Heidelberg, Heidelberg, Germany
| | - Hendrik Schürmann
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Valentina Leone
- Department of Internal Medicine II, University Hospital Rechts der Isar, Technical University Munich, Munich, Germany
| | - Amirhossein Sakhteman
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Matthew The
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Florian P Bayer
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Julian Müller
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Firas Hamood
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany
| | - Jens T Siveke
- Bridge Institute of Experimental Tumor Therapy (BIT) and Division of Solid Tumor Translational Oncology (DKTK), West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), partner site Essen, a partnership between German Cancer Research Center (DKFZ) and University Hospital Essen, Essen, Germany
| | - Maximilian Reichert
- Department of Internal Medicine II, University Hospital Rechts der Isar, Technical University Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.
| |
Collapse
|
25
|
Bhat A, Bhan S, Kabiraj A, Pandita RK, Ramos KS, Nandi S, Sopori S, Sarkar PS, Dhar A, Pandita S, Kumar R, Das C, Tainer JA, Pandita TK. A predictive chromatin architecture nexus regulates transcription and DNA damage repair. J Biol Chem 2025; 301:108300. [PMID: 39947477 PMCID: PMC11931391 DOI: 10.1016/j.jbc.2025.108300] [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: 09/02/2024] [Revised: 12/16/2024] [Accepted: 01/16/2025] [Indexed: 03/28/2025] Open
Abstract
Genomes are blueprints of life essential for an organism's survival, propagation, and evolutionary adaptation. Eukaryotic genomes comprise of DNA, core histones, and several other nonhistone proteins, packaged into chromatin in the tiny confines of nucleus. Chromatin structural organization restricts transcription factors to access DNA, permitting binding only after specific chromatin remodeling events. The fundamental processes in living cells, including transcription, replication, repair, and recombination, are thus regulated by chromatin structure through ATP-dependent remodeling, histone variant incorporation, and various covalent histone modifications including phosphorylation, acetylation, and ubiquitination. These modifications, particularly involving histone variant H2AX, furthermore play crucial roles in DNA damage responses by enabling repair protein's access to damaged DNA. Chromatin also stabilizes the genome by regulating DNA repair mechanisms while suppressing damage from endogenous and exogenous sources. Environmental factors such as ionizing radiations induce DNA damage, and if repair is compromised, can lead to chromosomal abnormalities and gene amplifications as observed in several tumor types. Consequently, chromatin architecture controls the genome fidelity and activity: it orchestrates correct gene expression, genomic integrity, DNA repair, transcription, replication, and recombination. This review considers connecting chromatin organization to functional outcomes impacting transcription, DNA repair and genomic integrity as an emerging grand challenge for predictive molecular cell biology.
Collapse
Affiliation(s)
- Audesh Bhat
- Centre for Molecular Biology, Central University of Jammu, Jammu and Kashmir, India.
| | - Sonali Bhan
- Centre for Molecular Biology, Central University of Jammu, Jammu and Kashmir, India
| | - Aindrila Kabiraj
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai, Maharashtra, India
| | - Raj K Pandita
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, Texas, USA
| | - Keneth S Ramos
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, Texas, USA
| | - Sandhik Nandi
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai, Maharashtra, India
| | - Shreya Sopori
- Centre for Molecular Biology, Central University of Jammu, Jammu and Kashmir, India
| | - Parthas S Sarkar
- Department of Neurobiology and Neurology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Arti Dhar
- Department of Pharmacy, Birla Institute of Technology and Sciences Pilani, Hyderabad Campus, Telangana, India
| | | | - Rakesh Kumar
- Department of Biotechnology, Shri Mata Vaishnav Devi University, Katra, India
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India; Homi Bhabha National Institute, BARC Training School Complex, Mumbai, Maharashtra, India.
| | - John A Tainer
- Department of Molecular & Cellular Oncology and Department of Cancer Biology, UT MD Anderson Cancer Center, Houston, Texas, USA
| | - Tej K Pandita
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, Texas, USA.
| |
Collapse
|
26
|
Bíró P, H. Kovács BB, Novák T, Erdélyi M. Cluster parameter-based DBSCAN maps for image characterization. Comput Struct Biotechnol J 2025; 27:920-927. [PMID: 40123797 PMCID: PMC11930167 DOI: 10.1016/j.csbj.2025.02.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025] Open
Abstract
Single-molecule localization microscopy techniques are one of the most powerful methods in biological studies, allowing the visualization of nanoclusters. Cluster analysis algorithms are used for quantitative evaluation, with DBSCAN being one of the most widely used. Clustering results are extremely sensitive to the initial parameters; thus, several methods including DBSCAN maps, have been developed for parameter optimization. Here, we introduce cluster parameter-based DBSCAN maps, which are directly applicable to measured datasets. These maps can be used for image characterization and parameter optimization through sensitivity studies. We show the applicability of these maps to simulated and measured datasets and compare our results with the recently implemented lacunarity analysis for SMLM measurements.
Collapse
Affiliation(s)
- Péter Bíró
- Department of Optics and Quantum Electronics, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Bálint Barna H. Kovács
- Department of Optics and Quantum Electronics, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Tibor Novák
- Department of Optics and Quantum Electronics, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| | - Miklós Erdélyi
- Department of Optics and Quantum Electronics, University of Szeged, Dóm tér 9, Szeged, 6720, Hungary
| |
Collapse
|
27
|
Senwitz C, Vogel M, Drobot B, Stumpf T, Heller A. Impact of DTPA and 3,4,3-LI(1,2-HOPO) on Eu III interactions with renal cells in vitro. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 966:178736. [PMID: 39923481 DOI: 10.1016/j.scitotenv.2025.178736] [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: 11/29/2024] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
This study represents a first comprehensive investigation on how the decorporation agents CaNa3-DTPA (DTPA) and 3,4,3-LI(1,2-HOPO) (LIHOPO) affect EuIII interactions with human and rat kidney cells in vitro. Cell biological investigations were complemented with physicochemical measurements to correlate cytotoxic impairments with intracellular metal uptake and EuIII speciation. Upon exposure to sole DTPA or LIHOPO, cell viability and morphology are affected in a time- and concentration-dependent manner. For both decorporation agents, detailed EC50 values for renal cells in vitro are reported. Simultaneous application of EuIII + DTPA in the medium leads to formation of the soluble and largely cell impermeable EuDTPA2- complex. At ligand excess, this significantly reduces intracellular EuIII uptake. However, EuDTPA2- was spectroscopically detected also inside cells indicating that small fractions of this complex are able to pass the plasma membrane. When EuIII + LIHOPO is applied to the medium, the soluble EuLIHOPO- complex is formed. In contrast to DTPA, this drastically enhances intracellular EuIII uptake even at ligand deficit demonstrating that EuLIHOPO- is highly cell permeable. Concomitantly, this complex was spectroscopically detected inside cells confirming its plasma membrane passage and intracellular stability. Nevertheless, due to stable EuIII binding, the cell viability is not influenced by the increased intracellular EuIII content. In fact, the applied ligand concentration is much more critical in this regard, emphasizing the need for cytotoxic investigations. Our results improve the knowledge of the cellular interactions of lanthanides ± decorporation agents and demonstrate the combination of in vitro cell culture and spectroscopy being a sophisticated toolbox for this.
Collapse
Affiliation(s)
- Christian Senwitz
- Technische Universität Dresden, Faculty of Chemistry, Institute of Analytical Chemistry, Professorship of Radiochemistry/Radioecology, 01062 Dresden, Germany; Technische Universität Dresden, Radiation Protection, Central Radionuclide Laboratory, 01062 Dresden, Germany
| | - Manja Vogel
- VKTA - Radiation Protection, Analytics & Disposal Rossendorf e.V., 01328 Dresden, Germany
| | - Björn Drobot
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Resource Ecology, 01328 Dresden, Germany
| | - Thorsten Stumpf
- Technische Universität Dresden, Faculty of Chemistry, Institute of Analytical Chemistry, Professorship of Radiochemistry/Radioecology, 01062 Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Institute of Resource Ecology, 01328 Dresden, Germany
| | - Anne Heller
- Technische Universität Dresden, Faculty of Chemistry, Institute of Analytical Chemistry, Professorship of Radiochemistry/Radioecology, 01062 Dresden, Germany; Technische Universität Dresden, Radiation Protection, Central Radionuclide Laboratory, 01062 Dresden, Germany.
| |
Collapse
|
28
|
Su R, Wang Q, Hu Q, Wendurige, Li K, Wang C, Tao L. HDGF Knockout Suppresses Colorectal Cancer Progression and Drug Resistance by Modulating the DNA Damage Response. Biomolecules 2025; 15:282. [PMID: 40001585 PMCID: PMC11853149 DOI: 10.3390/biom15020282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Colorectal cancer (CRC) is a highly heterogeneous gastrointestinal malignancy. Despite significant advances in molecular targeted therapies for CRC in recent years, the increase in the overall survival rates for CRC patients remains limited. Therefore, there is an urgent need to explore novel drug targets. Herein, we show that heparin binding growth factor (HDGF) is highly expressed in CRC, and that its overexpression is associated with a poor disease-free interval. Additionally, we reveal that HDGF knockout reduces proliferation, migration, and invasion, while enhancing apoptosis in CRC cells, thereby validating HDGF as a potential therapeutic target for CRC. Mechanistically, we found that HDGF modulates DNA damage response and, by recruiting C-terminal binding protein-interacting protein (CtIP), it facilitates homologous recombination repair to influence CRC drug sensitivity. Furthermore, we propose that HDGF may serve as a recognition protein for H3K36me3, participating in the repair of damaged transcriptionally active genes, thus maintaining genomic stability in CRC.
Collapse
Affiliation(s)
- Riya Su
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Qin Wang
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Qun Hu
- Department of Oncology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010030, China
| | - Wendurige
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010020, China
| | - Kexin Li
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010020, China
| | - Changshan Wang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot 010020, China
| | - Liang Tao
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
29
|
Li S, Hu Z, Pan W, Wu H, Peng W, Wu Y, Jiang F, Peng X. Discovery of Highly Potent and Orally Bioavailable Histone Deacetylase 3 Inhibitors as Immunomodulators and Enhancers of DNA-Damage Response in Cancer Therapy. J Med Chem 2025; 68:3212-3237. [PMID: 39873221 DOI: 10.1021/acs.jmedchem.4c02445] [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: 01/30/2025]
Abstract
Histone deacetylase 3 (HDAC3) is a well-established target for cancer therapy. Herein, we developed LSQ-28 as a novel HDAC3 inhibitor, which exhibited high HDAC3 inhibitory activity (IC50 = 42 nM, SI > 161) and displayed potent antiproliferative activity against four cancer cells and further demonstrated excellent antimigratory, anti-invasive, and antiwound healing activities. Further studies revealed that LSQ-28 induced a dose-dependent increase in Ac-H3 expression and promoted the degradation of PD-L1. Additionally, LSQ-28 enhanced the DNA damage response induced by PARP inhibitor, as evidenced by regulated expression of PARP1 and γ-H2AX. Notably, LSQ-28 also possessed favorable pharmacokinetic properties with significant oral bioavailability (F = 95.34%). Importantly, the combination of LSQ-28 with the PD-L1 inhibitor NP-19 could enhance antitumor immune response (TGI = 80%). When combined with olaparib, LSQ-28 significantly enhanced the in vivo tumor-suppression activity (TGI = 91%). Collectively, LSQ-28 represents a promising HDAC3 inhibitor for further exploration in cancer therapeutic strategies.
Collapse
Affiliation(s)
- Shuqing Li
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Zhihao Hu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Wanyi Pan
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Haiyan Wu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Weijie Peng
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Yi Wu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Feng Jiang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| | - Xiaopeng Peng
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Disease, Ministry of Education; Jiangxi Provincal Key Laboratory of Tissue Engineering; College of Pharmacy, Gannan Medical University, Ganzhou 314000, China
| |
Collapse
|
30
|
Nadin SB, Cuello-Carrión FD, Cayado-Gutiérrez N, Fanelli MA. Overview of Wnt/β-Catenin Pathway and DNA Damage/Repair in Cancer. BIOLOGY 2025; 14:185. [PMID: 40001953 PMCID: PMC11851563 DOI: 10.3390/biology14020185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025]
Abstract
The Wnt/β-catenin pathway takes part in important cellular processes in tumor cells, such as gene expression, adhesion, and survival. The canonical pathway is activated in several tumors, and β-catenin is its major effector. The union of Wnt to the co-receptor complex causes the inhibition of GSK3β activity, thus preventing the phosphorylation and degradation of β-catenin, which accumulates in the cytoplasm, to subsequently be transported to the nucleus to associate with transcription factors. The relationship between Wnt/β-catenin and DNA damage/repair mechanisms has been a focus for the last few years. Studying the Wnt/β-catenin network interactions with DNA damage/repair proteins has become a successful research field. This review provides an overview of the participation of Wnt/β-catenin in DNA damage/repair mechanisms and their future implications as targets for cancer therapy.
Collapse
Affiliation(s)
- Silvina B. Nadin
- Laboratorio de Biología Tumoral, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Universidad Nacional de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Centro Científico Tecnológico (CCT), Mendoza 5500, Argentina
| | - F. Darío Cuello-Carrión
- Laboratorio de Oncología, IMBECU, CONICET, CCT, Mendoza 5500, Argentina; (F.D.C.-C.); (N.C.-G.); (M.A.F.)
| | - Niubys Cayado-Gutiérrez
- Laboratorio de Oncología, IMBECU, CONICET, CCT, Mendoza 5500, Argentina; (F.D.C.-C.); (N.C.-G.); (M.A.F.)
- Cátedra de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza 5500, Argentina
| | - Mariel A. Fanelli
- Laboratorio de Oncología, IMBECU, CONICET, CCT, Mendoza 5500, Argentina; (F.D.C.-C.); (N.C.-G.); (M.A.F.)
| |
Collapse
|
31
|
Li Y, Liu H, Fang R, Jin J, Yang F, Chen J, Zhang J. Designing novel Au(III) complexes based on the structure of diazepam: Achieving a multiaction mechanism against glioma. Eur J Med Chem 2025; 283:117171. [PMID: 39705733 DOI: 10.1016/j.ejmech.2024.117171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/01/2024] [Accepted: 12/12/2024] [Indexed: 12/22/2024]
Abstract
Metal-based drugs have been used in the clinical treatment of tumors for over 30 years. However, no metal-based drugs have been clinically approved to treat glioma. Although metal complexes have excellent cytotoxicity, their most critical problem is crossing the blood-brain barrier. Therefore, to enable metal complexes to cross blood-brain barrier and target glioma therapy, herein, we propose to rationally used the basic structure of diazepam (5-chlorobenzophenone) and thiosemicarbazide to synthesize gold (Au) complexes C1, C2 and C3 with antiglioma activity. The C3 complex with two methyl groups attached to the N3 of thiosemicarbazone exhibited excellent cytotoxicity to glioma cells through its multiaction mechanism against glioma, inducing apoptosis, autophagy death, and deoxyribonucleic acid damage. In addition, the synthesized C3 complex can effectively cross the blood-brain barrier and accumulate in glioma, considerably decreasing the untoward reaction in vivo. Our findings provide a novel strategy for designing metal-based complexes for the treatment of glioma.
Collapse
Affiliation(s)
- Yanping Li
- Mental Health Education Center of College Student, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Haoran Liu
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Ronghao Fang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Jiamin Jin
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi, 541004, PR China
| | - Jian Chen
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China.
| | - Juzheng Zhang
- Guangxi Key Laboratory of Tumor Immunology and Microenvironmental Regulation, Guangxi Health Commission Key Laboratory of Tumor Immunology and Receptor-Targeted Drug Basic Research, Guilin Medical University, Huan Cheng North 2nd Road 109, Guilin, 541004, PR China.
| |
Collapse
|
32
|
Herrero Y, Velazquez C, Pascuali N, Hauk V, de Zúñiga I, Martínez G, Lavolpe M, Neuspiller F, Veiga MF, Scotti L, Abramovich D, Parborell F. Ovarian function after COVID-19: long-term effects and vaccine safety in ART patients. J Assist Reprod Genet 2025; 42:563-576. [PMID: 39883303 PMCID: PMC11871259 DOI: 10.1007/s10815-025-03403-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
PURPOSE This study aimed to evaluate the long-term impact of mild COVID-19 infection and COVID-19 vaccination on ovarian function in patients undergoing assisted reproductive technology (ART). Specifically, we assessed ovarian outcomes between 9 and 18 months post-infection and investigated the effects of COVID-19 vaccines (inactivated virus and adenovirus) on reproductive parameters. METHODS The study included two objectives: (a) examining ovarian function in post-COVID-19 patients (9-18 months post-infection) compared to a control group and (b) comparing reproductive outcomes in vaccinated versus unvaccinated patients. According to the study objectives, ART patients were divided into the following groups: a control group (n = 30), a post-COVID-19 group (n = 55), an unvaccinated group (n = 70), and a vaccinated group (n = 55). Findings revealed a reduction in the number of retrieved and mature oocytes in patients over 36 years in the post-COVID-19 group. Lower IL-1β levels were found in follicular fluid (FF) of post-COVID-19 patients, while VEGF levels were reestablished between 9 and 18 months post-infection. Although cell migration was reduced in endothelial cells incubated with post-COVID-19 FF, angiogenic factor levels and DNA integrity remained stable. No significant differences in retrieved or mature oocytes were observed between vaccinated and unvaccinated patients. CONCLUSIONS VEGF levels and DNA integrity in FF from post-COVID-19 patients were normalized between 9 and 18 months post-infection. Additionally, COVID-19 vaccination did not negatively impact ovarian response in ART patients, supporting vaccine safety in reproductive contexts.
Collapse
Affiliation(s)
- Yamila Herrero
- Ovarian Physiopathology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
| | - Candela Velazquez
- Ovarian Physiopathology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
| | - Natalia Pascuali
- Ovarian Physiopathology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
- Department of Pathology, College of Medicine, University of Illinois at Chicago (UIC), Chicago, IL, USA
| | - Vanesa Hauk
- Immunopharmacology Laboratory, Institute of Biological Chemistry (IQUIBICEN), School of Exact and Natural Sciences, University of Buenos Aires-CONICET, Buenos Aires, Argentina
| | | | | | | | | | | | - Leopoldina Scotti
- Ovarian Physiopathology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
- Centro de Investigaciones y Transferencia del Noroeste de La Provincia de Buenos Aires (CITNOBA-UNSADA-CONICET), San Antonio de Areco, Argentina
| | - Dalhia Abramovich
- Ovarian Physiopathology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina
| | - Fernanda Parborell
- Ovarian Physiopathology Studies Laboratory, Institute of Experimental Biology and Medicine (IByME) - CONICET, Buenos Aires, Argentina.
| |
Collapse
|
33
|
Lee JH. ATM in immunobiology: From lymphocyte development to cancer immunotherapy. Transl Oncol 2025; 52:102268. [PMID: 39752906 PMCID: PMC11754496 DOI: 10.1016/j.tranon.2024.102268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/14/2024] [Accepted: 12/30/2024] [Indexed: 01/25/2025] Open
Abstract
Ataxia Telangiectasia Mutated (ATM) is a protein kinase traditionally known for its role in DNA damage response and cell cycle regulation. However, emerging research has revealed its multifaceted and crucial functions in the immune system. This comprehensive review explores the diverse roles of ATM in immune regulation, from lymphocyte development to its involvement in cancer immunotherapy. The review describes ATM's critical functions in V(D)J recombination and class switch recombination, highlighting its importance in adaptive immunity. It examines ATM's role in innate immunity, particularly in NF-κB signaling and cytokine production. Furthermore, the review analyzes the impact of ATM deficiency on oxidative stress and mitochondrial function in immune cells, providing insights into the immunological defects observed in Ataxia Telangiectasia (A-T). The article explores ATM's significance in maintaining hematopoietic stem cell function and its implications for bone marrow transplantation and gene therapy. Additionally, it addresses ATM's involvement in inflammation and immune senescence, linking DNA damage response to age-related immune decline. Finally, this review highlights the emerging role of ATM in cancer immunotherapy, where its inhibition shows promise in enhancing immune checkpoint blockade therapy. This review synthesizes current knowledge on ATM's functions in the immune system, offering insights into the pathophysiology of ATM-related disorders and potential therapeutic strategies for immune-related conditions and cancer immunotherapy.
Collapse
Affiliation(s)
- Ji-Hoon Lee
- Department of Biological Sciences, Research Center of Ecomimetics, Chonnam National University, Gwangju 61186, South Korea.
| |
Collapse
|
34
|
Nuijens AC, Oei AL, Franken NAP, Rasch CRN, Stalpers LJA. Towards Personalized Radiotherapy in Pelvic Cancer: Patient-Related Risk Factors for Late Radiation Toxicity. Curr Oncol 2025; 32:47. [PMID: 39851963 PMCID: PMC11763857 DOI: 10.3390/curroncol32010047] [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: 11/11/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
Normal tissue reactions vary significantly among patients receiving the same radiation treatment regimen, reflecting the multifactorial etiology of late radiation toxicity. Predicting late radiation toxicity is crucial, as it aids in the initial decision-making process regarding the treatment modalities. For patients undergoing radiotherapy, anticipating late toxicity allows for planning adjustments to optimize individualized care. Various dosimetric parameters have been shown to influence the incidence of late toxicity, and the literature available on this topic is extensive. This narrative review examines patient-related determinants of late toxicity following external beam radiotherapy for pelvic tumors, with a focus on prostate and cervical cancer patients. In Part I, we address various methods for quantifying radiation toxicity, providing context for interpreting toxicity data. Part II examines the current insights into the clinical risk factors for late toxicity. While certain factors-such as previous abdominal surgery, smoking behavior, and severe acute toxicity-have consistently been reported, most of the others show inconsistent associations. In Part III, we explore the influence of genetic factors and discuss promising predictive assays. Single-nucleotide polymorphisms (SNPs) likely elevate the risk in specific combinations. Advances in artificial intelligence now allow for the identification of SNP patterns from large datasets, supporting the development of polygenic risk scores. These innovations hold promise for improving personalized treatment strategies and reducing the burden of late toxicity in cancer survivors.
Collapse
Affiliation(s)
- Anna C. Nuijens
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands (L.J.A.S.)
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC Location University of Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Meibergdreef, 1105 AZ Amsterdam, The Netherlands
| | - Arlene L. Oei
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands (L.J.A.S.)
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC Location University of Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands
| | - Nicolaas A. P. Franken
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands (L.J.A.S.)
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental and Molecular Medicine (CEMM), Amsterdam UMC Location University of Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands
| | - Coen R. N. Rasch
- Department of Radiation Oncology, Leiden University Medical Center, Albinusdreef, 2333 ZA Leiden, The Netherlands
| | - Lukas J. A. Stalpers
- Department of Radiation Oncology, Amsterdam UMC Location University of Amsterdam, Meibergdreef, 1105 AZ Amsterdam, The Netherlands (L.J.A.S.)
| |
Collapse
|
35
|
Zheng H, Yang X, Zhong H, Song C, Wu Z, Yang H. HDAC6 Facilitates PRV and VSV Infection by Inhibiting Type I Interferon Production. Viruses 2025; 17:90. [PMID: 39861880 PMCID: PMC11768819 DOI: 10.3390/v17010090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/27/2025] Open
Abstract
HDAC6 modulates viral infection through diverse mechanisms. Here, we investigated the role of HDAC6 in influencing viral infection in pig cells with the aim of exploiting the potential antiviral gene targets in pigs. Using gene knockout and overexpression strategies, we found that HDAC6 knockout greatly reduced PRV and VSV infectivity, whereas HDAC6 overexpression increased their infectivity in PK15 cells. Mechanistic studies identified HDAC6 as a DNA damage inhibitor in PK15 cells. HDAC6 overexpression attenuated DNA damage levels, which can further reduce type I IFN production to promote viral infection. Conversely, HDAC6 deficiency can limit viral infection by increasing DNA damage-mediated type I IFN production. This work demonstrates that HDAC6 affects the infection process of multiple viruses by modulating type I IFN production, highlighting a regulatory role of HDAC6 linking host immune response and viral infection levels in pig cells.
Collapse
Affiliation(s)
- Hu Zheng
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Xiaohui Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Haiwen Zhong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Changxu Song
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
| | - Zhenfang Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
- Yunfu Branch Center of Guangdong Laboratory of Lingnan Modern Agricultural Science and Technology, Yunfu 527400, China
| | - Huaqiang Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou 510642, China; (H.Z.); (X.Y.); (H.Z.); (C.S.)
- Yunfu Branch Center of Guangdong Laboratory of Lingnan Modern Agricultural Science and Technology, Yunfu 527400, China
| |
Collapse
|
36
|
Lee C, Oh JS. Novel BRCA1-PLK1-CIP2A axis orchestrates homologous recombination-mediated DNA repair to maintain chromosome integrity during oocyte meiosis. Nucleic Acids Res 2025; 53:gkae1207. [PMID: 39657788 PMCID: PMC11754672 DOI: 10.1093/nar/gkae1207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/18/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024] Open
Abstract
Double-strand breaks (DSBs) are a formidable threat to genome integrity, potentially leading to cancer and various genetic diseases. The prolonged lifespan of mammalian oocytes increases their susceptibility to DNA damage over time. While somatic cells suppress DSB repair during mitosis, oocytes exhibit a remarkable capacity to repair DSBs during meiotic maturation. However, the precise mechanisms underlying DSB repair in oocytes remain poorly understood. Here, we describe the pivotal role of the BRCA1-PLK1-CIP2A axis in safeguarding genomic integrity during meiotic maturation in oocytes. We found that inhibition of homologous recombination (HR) severely impaired chromosome integrity by generating chromosome fragments during meiotic maturation. Notably, HR inhibition impaired the recruitment of CIP2A to damaged chromosomes, and the depletion of CIP2A led to chromosome fragmentation following DSB induction. Moreover, BRCA1 depletion impaired chromosomal recruitment of CIP2A, but not vice versa. Importantly, the impaired chromosomal recruitment of CIP2A could be rescued by PLK1 inhibition. Consequently, our findings not only underscore the importance of the chromosomal recruitment of CIP2A in preventing chromosome fragmentation, but also demonstrate the regulatory role of the BRCA1-PLK1-CIP2A axis in this process during oocyte meiotic maturation.
Collapse
Affiliation(s)
- Crystal Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, South Korea
| | - Jeong Su Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, South Korea
| |
Collapse
|
37
|
Delint-Ramirez I, Madabhushi R. DNA damage and its links to neuronal aging and degeneration. Neuron 2025; 113:7-28. [PMID: 39788088 PMCID: PMC11832075 DOI: 10.1016/j.neuron.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/07/2024] [Accepted: 12/02/2024] [Indexed: 01/12/2025]
Abstract
DNA damage is a major risk factor for the decline of neuronal functions with age and in neurodegenerative diseases. While how DNA damage causes neurodegeneration is still being investigated, innovations over the past decade have provided significant insights into this issue. Breakthroughs in next-generation sequencing methods have begun to reveal the characteristics of neuronal DNA damage hotspots and the causes of DNA damage. Chromosome conformation capture-based approaches have shown that, while DNA damage and the ensuing cellular response alter chromatin topology, chromatin organization at damage sites also affects DNA repair outcomes in neurons. Additionally, neuronal activity results in the formation of programmed DNA breaks, which could burden DNA repair mechanisms and promote neuronal dysfunction. Finally, emerging evidence implicates DNA damage-induced inflammation as an important contributor to the age-related decline in neuronal functions. Together, these discoveries have ushered in a new understanding of the significance of genome maintenance for neuronal function.
Collapse
Affiliation(s)
- Ilse Delint-Ramirez
- Departments of Psychiatry, Neuroscience, and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Peter O' Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ram Madabhushi
- Departments of Psychiatry, Neuroscience, and Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Peter O' Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
38
|
Maggs LR, McVey M. REV7: a small but mighty regulator of genome maintenance and cancer development. Front Oncol 2025; 14:1516165. [PMID: 39839778 PMCID: PMC11747621 DOI: 10.3389/fonc.2024.1516165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
REV7, also known as MAD2B, MAD2L2, and FANCV, is a HORMA-domain family protein crucial to multiple genome stability pathways. REV7's canonical role is as a member of polymerase ζ, a specialized translesion synthesis polymerase essential for DNA damage tolerance. REV7 also ensures accurate cell cycle progression and prevents premature mitotic progression by sequestering an anaphase-promoting complex/cyclosome activator. Additionally, REV7 supports genome integrity by directing double-strand break repair pathway choice as part of the recently characterized mammalian shieldin complex. Given that genome instability is a hallmark of cancer, it is unsurprising that REV7, with its numerous genome maintenance roles, is implicated in multiple malignancies, including ovarian cancer, glioma, breast cancer, malignant melanoma, and small-cell lung cancer. Moreover, high REV7 expression is associated with poor prognoses and treatment resistance in these and other cancers. Promisingly, early studies indicate that REV7 suppression enhances sensitivity to chemotherapeutics, including cisplatin. This review aims to provide a comprehensive overview of REV7's myriad roles in genome maintenance and other functions as well as offer an updated summary of its connections to cancer and treatment resistance.
Collapse
Affiliation(s)
- Lara R. Maggs
- Department of Biology, Tufts University, Medford, MA, United States
| | - Mitch McVey
- Department of Biology, Tufts University, Medford, MA, United States
| |
Collapse
|
39
|
Buonanno M, Hashmi R, Petersen CE, Tang Z, Welch D, Shuryak I, Brenner DJ. Wavelength-dependent DNA damage induced by single wavelengths of UV-C radiation (215 to 255 nm) in a human cornea model. Sci Rep 2025; 15:252. [PMID: 39747969 PMCID: PMC11696903 DOI: 10.1038/s41598-024-84196-4] [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: 09/21/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
Scientific bodies overseeing UV radiation protection recommend safety limits for exposure to ultraviolet radiation in the workplace based on published peer-reviewed data. To support this goal, a 3D model of the human cornea was used to assess the wavelength dependence of corneal damage induced by UV-C radiation. In the first set of experiments the models were exposed with or without simulated tears; at each wavelength (215-255 nm) cells with DNA dimers and their distribution within the epithelium were measured. Simulated tears reduced the fraction of damaged cells to an extent dependent on the wavelength and tissue layer. Subsequent experiments were performed with models exposed without simulated tears; yields of DNA-damaged cells and their distribution within the corneal epithelium were evaluated at each wavelength, together with other markers of cell and tissue integrity. Unlike relatively longer wavelengths, the range of wavelengths commonly referred to as far-UV-C (215-235 nm) only induced dimers in the uppermost layers of the epithelium and did not result in lasting damage or halt proliferation of the germinative cells. These results provide evidence for the recommended exposure limits for far-UV-C wavelengths, which have been proposed as a practical technology to reduce the risk of transmission of airborne diseases in occupied locations.
Collapse
Affiliation(s)
- Manuela Buonanno
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA.
| | - Raabia Hashmi
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA
| | - Camryn E Petersen
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA
| | - Zheng Tang
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA
| | - David Welch
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA
| | - Igor Shuryak
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA
| | - David J Brenner
- Center for Radiological Research, Columbia University Irving Medical Center, 630 West 168th Street, New York, 10032, USA
| |
Collapse
|
40
|
Parsons AM, Byrne S, Kooistra J, Dewey J, Zebolsky AL, Alvarado G, Bouma GJ, Vanden Heuvel GB, Larson ED. G-quadruplex stabilization provokes DNA breaks in human PKD1, revealing a second hit mechanism for ADPKD. Nat Commun 2025; 16:121. [PMID: 39747084 PMCID: PMC11696556 DOI: 10.1038/s41467-024-55684-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
The "secondhit" pathway is responsible for biallelic inactivation of many tumor suppressors, where a pathogenic germline allele is joined by somatic mutation of the remaining functional allele. The mechanisms are unresolved, but the human PKD1 tumor suppressor is a good experimental model for identifying the molecular determinants. Inactivation of PKD1 results in autosomal dominant polycystic kidney disease, a very common disorder characterized by the accumulation of fluid-filled cysts and end-stage renal disease. Since human PKD1 follows second hit and mouse Pkd1 heterozygotes do not, we reasoned that there is likely a molecular difference that explains the elevated mutagenesis of the human gene. Here we demonstrate that guanine quadruplex DNA structures are abundant throughout human, but not mouse, PKD1 where they activate the DNA damage response. Our results suggest that guanine quadruplex DNAs provoke DNA breaks in PKD1, providing a potential mechanism for cystogenesis in autosomal dominant polycystic kidney disease specifically and for the inactivation of guanine quadruplex-rich tumor suppressors generally.
Collapse
Affiliation(s)
- Agata M Parsons
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Seth Byrne
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Jesse Kooistra
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - John Dewey
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Aaron L Zebolsky
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Gloria Alvarado
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Gerrit J Bouma
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Gregory B Vanden Heuvel
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Erik D Larson
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA.
| |
Collapse
|
41
|
Hegazy YA, Dhahri H, El Osmani N, George S, Chandler DP, Fondufe-Mittendorf YN. Histone variants: The bricks that fit differently. J Biol Chem 2025; 301:108048. [PMID: 39638247 PMCID: PMC11742582 DOI: 10.1016/j.jbc.2024.108048] [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: 08/06/2024] [Revised: 10/28/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Histone proteins organize nuclear DNA in eukaryotic cells and play crucial roles in regulating chromatin structure and function. Histone variants are produced by distinct histone genes and are produced independently of their canonical counterparts throughout the cell cycle. Even though histone variants may differ by only one or a few amino acids relative to their canonical counterparts, these minor variations can profoundly alter chromatin structure, accessibility, dynamics, and gene expression. Histone variants often interact with dedicated chaperones and remodelers and can have unique post-translational modifications that shape unique gene expression landscapes. Histone variants also play essential roles in DNA replication, damage repair, and histone-protamine transition during spermatogenesis. Importantly, aberrant histone variant expression and DNA mutations in histone variants are linked to various human diseases, including cancer, developmental disorders, and neurodegenerative diseases. In this review, we explore how core histone variants impact nucleosome structure and DNA accessibility, the significance of variant-specific post-translational modifications, how variant-specific chaperones and remodelers contribute to a regulatory network governing chromatin behavior, and discuss current knowledge about the association of histone variants with human diseases.
Collapse
Affiliation(s)
- Youssef A Hegazy
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Hejer Dhahri
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, Michigan, USA; Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky, USA
| | - Nour El Osmani
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Smitha George
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - Darrell P Chandler
- Department of Epigenetics, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | | |
Collapse
|
42
|
Koch M, Ferrarese L, Ben-Yehuda Greenwald M, Werner S. Dose-dependent effects of Nrf2 on the epidermis in chronic skin inflammation. Dis Model Mech 2025; 18:dmm052126. [PMID: 39744884 PMCID: PMC11708820 DOI: 10.1242/dmm.052126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease, characterized by an impaired epidermal barrier and immunological alterations. The activity of the cytoprotective NRF2 transcription factor is reduced in the epidermis of AD patients. To determine the functional relevance of this deficiency, we used mice lacking fibroblast growth factor receptors 1 and 2 in keratinocytes (K5-R1/R2 mice), which exhibit several AD-like symptoms. Proteomics analysis of their epidermis revealed reduced Nrf2 activity. This was accompanied by an increase in DNA damage and in the number of senescent cells. Genetic deletion of Nrf2 in keratinocytes of these mice further promoted DNA damage and senescence, but time-limited pharmacological activation of Nrf2 in the skin had a mild protective effect. Surprisingly, long-term genetic activation of Nrf2 in keratinocytes of K5-R1/R2 mice caused strong hyperkeratosis, keratinocyte hyperproliferation, epidermal thickening, increased keratinocyte apoptosis and DNA damage, and altered immune cell composition. These results reveal a complex role of Nrf2 in the epidermis and show the necessity to optimize the duration and intensity of NRF2 activation for the treatment of epidermal alterations in patients with AD.
Collapse
Affiliation(s)
- Michael Koch
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Luca Ferrarese
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | | | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| |
Collapse
|
43
|
Zhao X, Wei T, Hou Y, Wu Y, Zhou H, Meng J, Wang Q, Liu Y. ATM/ATR-Mediated DNA Damage Response Facilitates SARS-CoV-2 Spike Protein-Induced Syncytium Formation. J Med Virol 2025; 97:e70137. [PMID: 39740081 DOI: 10.1002/jmv.70137] [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: 07/12/2024] [Revised: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 01/02/2025]
Abstract
Multinucleated cells are present in lung tissues of patients infected by SARS-CoV-2. Although the spike protein can cause the fusion of infected cells and ACE2-expressing cells to form syncytia and induce damage, how host cell responses to this damage and the role of DNA damage response (DDR) signals in cell fusion are still unclear. Therefore, we investigated the effect of SARS-CoV-2 spike protein on the fusion of homologous and heterologous cells expressing ACE2 in vitro models, focusing on the protein levels of ATR and ATM, the major kinases responding to DNA damage, and their substrates CHK1 and CHK2. We found that both homologous and heterologous cell fusion activated the ATR-CHK1 and ATM-CHK2 signaling axis and induced the aggregation of γH2AX, 53BP1 and RAD51 in syncytia. In addition, siRNA or inhibitors of ATM and ATR suppressed syncytia formation by decreasing the level of S protein. These results showed the important role of DDR in stabilizing the S protein and in favoring its induction of cell fusion and syncytium formation, suggesting that the virus exploits the host DDR to facilitate its spread among infected cells.
Collapse
Affiliation(s)
- Xiaotong Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Tingting Wei
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yujia Hou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yanjin Wu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Haitao Zhou
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiahui Meng
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Qin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yang Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, State Key Laboratory of Advanced Medical Materials and Devices, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
44
|
Sekikawa Y, Kato TA. Radiation-Induced Gamma-H2AX Foci Staining and Analysis. Methods Mol Biol 2025; 2933:25-29. [PMID: 40418470 DOI: 10.1007/978-1-0716-4574-1_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
DNA damage induces activation of DNA damage-associated proteins. In the case of DNA double-strand breaks (DSBs), H2A histone family member X (H2AX) plays a crucial role in DNA repair. Upon DSBs, H2AX is phosphorylated at serine 139, forming γH2AX, which creates characteristic "foci" at the site of damage. These foci can be visualized through fluorescent immunostaining under a microscope, allowing for the counting of individual DSBs within nuclei. This technique is superior to previously established DSB detection assays, such as neutral elution, pulse field gel electrophoresis, and neutral comet assays, in terms of sensitivity for detecting single DSBs. Further research supports the idea that this assay provides valuable insight into the dynamics of DSB responses and repair mechanisms. In this chapter, we present a technique to target and visualize the foci of γH2AX by fluorescent immunocytochemical methods.
Collapse
Affiliation(s)
- Yuya Sekikawa
- Department of Radiological Technology, Faculty of Fukuoka Medical Technology, Teikyo University, Fukuoka, Japan.
| | - Takamitsu A Kato
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| |
Collapse
|
45
|
Djerir B, Maréchal A. Detection of γ-H2A.X for Rapid Assessment of Genotoxic Agent-induced Double-strand DNA Breaks by Immunofluorescence. Methods Mol Biol 2025; 2919:83-89. [PMID: 40257558 DOI: 10.1007/978-1-0716-4486-7_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Cells are constantly exposed to a range of intrinsic and environmental factors, such as reactive oxygen species, ionizing radiation, and chemical agents, which threaten DNA integrity and can induce double-strand breaks (DSBs). DSBs are highly cytotoxic and must be rapidly repaired to maintain genome stability. One of the earliest chromatin marks associated with DSBs is the phosphorylation of histone variant H2A.X on serine 139, which leads to the formation of γ-H2A.X foci detectable by immunofluorescence. Immunocytochemical detection of γ-H2A.X is a widely used method to indirectly assess DSB formation and repair. In this chapter, we describe a simple and efficient immunofluorescence (IF) procedure for detecting DSB formation in U-2 OS cells treated with genotoxic agents. This technique does not require an expensive laboratory setup and can also be used to detect the formation of foci of additional repair factors using appropriate labeling.
Collapse
Affiliation(s)
- Billel Djerir
- Faculty of Sciences, Department of Biology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Alexandre Maréchal
- Faculty of Sciences, Department of Biology, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
46
|
Black EM, Ramírez Parrado CA, Trier I, Li W, Joo YK, Pichurin J, Liu Y, Kabeche L. Chk2 sustains PLK1 activity in mitosis to ensure proper chromosome segregation. Nat Commun 2024; 15:10782. [PMID: 39737931 PMCID: PMC11685634 DOI: 10.1038/s41467-024-54922-7] [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: 03/19/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Polo-like kinase 1 (PLK1) protects against genome instability by ensuring timely and accurate mitotic cell division, and its activity is tightly regulated throughout the cell cycle. Although the pathways that initially activate PLK1 in G2 are well-characterized, the factors that directly regulate mitotic PLK1 remain poorly understood. Here, we identify that human PLK1 activity is sustained by the DNA damage response kinase Checkpoint kinase 2 (Chk2) in mitosis. Chk2 directly phosphorylates PLK1 T210, a residue on its T-loop whose phosphorylation is essential for full PLK1 kinase activity. Loss of Chk2-dependent PLK1 activity causes increased mitotic errors, including chromosome misalignment, chromosome missegregation, and cytokinetic defects. Moreover, Chk2 deficiency increases sensitivity to PLK1 inhibitors, suggesting that Chk2 status may be an informative biomarker for PLK1 inhibitor efficacy. This work demonstrates that Chk2 sustains mitotic PLK1 activity and protects genome stability through discrete functions in interphase DNA damage repair and mitotic chromosome segregation.
Collapse
Affiliation(s)
- Elizabeth M Black
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Carlos Andrés Ramírez Parrado
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Isabelle Trier
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Wenxue Li
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
- Department of Pharmacology, Yale University, New Haven, CT, 06511, USA
| | - Yoon Ki Joo
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Jennifer Pichurin
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Yansheng Liu
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
- Department of Pharmacology, Yale University, New Haven, CT, 06511, USA
| | - Lilian Kabeche
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA.
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA.
| |
Collapse
|
47
|
Farmakis D, Stravopodis DJ, Prombona A. TH301 Emerges as a Novel Anti-Oncogenic Agent for Human Pancreatic Cancer Cells: The Dispensable Roles of p53, CRY2 and BMAL1 in TH301-Induced CDKN1A/p21 CIP1/WAF1 Upregulation. Int J Mol Sci 2024; 26:178. [PMID: 39796036 PMCID: PMC11720130 DOI: 10.3390/ijms26010178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/21/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Pancreatic Ductal Adeno-Carcinoma (PDAC) is a highly aggressive cancer, with limited treatment options. Disruption of the circadian clock, which regulates key cellular processes, has been implicated in PDAC initiation and progression. Hence, targeting circadian clock components may offer new therapeutic opportunities for the disease. This study investigates the cytopathic effects of TH301, a novel CRY2 stabilizer, on PDAC cells, aiming to evaluate its potential as a novel therapeutic agent. Methods: PDAC cell lines (AsPC-1, BxPC-3 and PANC-1) were treated with TH301, and cell viability, cell cycle progression, apoptosis, autophagy, circadian gene, and protein expression profiles were analyzed, using MTT assay, flow cytometry, Western blotting, and RT-qPCR technologies. Results: TH301 proved to significantly decrease cell viability and to induce cell cycle arrest at the G1-phase across all PDAC cell lines herein examined, especially the AsPC-1 and BxPC-3 ones. It caused dose-dependent apoptosis and autophagy, and it synergized with Chloroquine and Oxaliplatin to enhance anti-oncogenicity. The remarkable induction of p21 by TH301 was shown to follow clock- and p53-independent patterns, thereby indicating the critical engagement of alternative mechanisms. Conclusions: TH301 demonstrates significant anti-cancer activities in PDAC cells, thus serving as a promising new therapeutic agent, which can also synergize with approved treatment schemes by targeting pathways beyond circadian clock regulation. Altogether, TH301 likely opens new therapeutic windows for the successful management of pancreatic cancer in clinical practice.
Collapse
Affiliation(s)
- Danae Farmakis
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, Zografou, 157 01 Athens, Greece;
- Laboratory of Chronobiology, Institute of Biosciences and Applications (IBA), National Centre for Scientific Research (NCSR) “Demokritos”, 153 41 Aghia Paraskevi, Greece;
| | - Dimitrios J. Stravopodis
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens (NKUA), Panepistimiopolis, Zografou, 157 01 Athens, Greece;
| | - Anastasia Prombona
- Laboratory of Chronobiology, Institute of Biosciences and Applications (IBA), National Centre for Scientific Research (NCSR) “Demokritos”, 153 41 Aghia Paraskevi, Greece;
| |
Collapse
|
48
|
Ben Diouf O, Gilbert A, Bernay B, Syljuåsen RG, Tudor M, Temelie M, Savu DI, Soumboundou M, Sall C, Chevalier F. Phospho-Proteomics Analysis of Early Response to X-Ray Irradiation Reveals Molecular Mechanism Potentially Related to U251 Cell Radioresistance. Proteomes 2024; 13:1. [PMID: 39846632 PMCID: PMC11755531 DOI: 10.3390/proteomes13010001] [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: 08/31/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Glioblastoma (GBM) is a devastating malignant brain tumor with a poor prognosis. GBM is associated with radioresistance. Post-translational modifications (PTMs) such as protein phosphorylation can play an important role in the cellular response to radiation. To better understand the early cellular activities after radiation in GBM, we carried out a phospho-proteomic study on the U251 cell line 3 h after X-ray irradiation (6Gy) and on non-irradiated cells. Our study showed a strong modification of proteoform phosphorylation in response to radiation. We found 453 differentially expressed phosphopeptides (DEPs), with 211 being upregulated and 242 being downregulated. A GO enrichment analysis of DEPs showed a strong enrichment of the signaling pathways involved in DNA damage response after irradiation and categorized them into biological processes (BPs), cellular components (CCs) and molecular functions (MFs). Certain accessions such as BRCA1, MDC1, H2AX, MDC1, TP53BP1 were dynamically altered in our fraction and are highly associated with the signaling pathways enriched after radiation.
Collapse
Affiliation(s)
- Ousseynou Ben Diouf
- Mixed Research Exploration and Diagnosis (UMRED), UFR-Healthy, Iba Der THIAM University of Thies, Thies BP A967, Senegal; (O.B.D.)
| | - Antoine Gilbert
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN, Université de Caen Normandie, 14000 Caen, France
| | - Benoit Bernay
- Proteogen Platform, US EMerode, CAEN Normandie University, 14032 Caen, France
| | - Randi G. Syljuåsen
- Department of Radiation Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Mihaela Tudor
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania (D.I.S.)
| | - Mihaela Temelie
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania (D.I.S.)
| | - Diana I. Savu
- Department of Life and Environmental Physics, Horia Hulubei National Institute of Physics and Nuclear Engineering, 077125 Magurele, Romania (D.I.S.)
| | - Mamadou Soumboundou
- Mixed Research Exploration and Diagnosis (UMRED), UFR-Healthy, Iba Der THIAM University of Thies, Thies BP A967, Senegal; (O.B.D.)
| | - Cheikh Sall
- Mixed Research Exploration and Diagnosis (UMRED), UFR-Healthy, Iba Der THIAM University of Thies, Thies BP A967, Senegal; (O.B.D.)
| | - François Chevalier
- UMR6252 CIMAP, Team Applications in Radiobiology with Accelerated Ions, CEA-CNRS-ENSICAEN, Université de Caen Normandie, 14000 Caen, France
| |
Collapse
|
49
|
Zhu L, Dluzewska J, Fernández-Jiménez N, Ranjan R, Pelé A, Dziegielewski W, Szymanska-Lejman M, Hus K, Górna J, Pradillo M, Ziolkowski PA. The kinase ATR controls meiotic crossover distribution at the genome scale in Arabidopsis. THE PLANT CELL 2024; 37:koae292. [PMID: 39471331 DOI: 10.1093/plcell/koae292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/07/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024]
Abstract
Meiotic crossover, i.e. the reciprocal exchange of chromosome fragments during meiosis, is a key driver of genetic diversity. Crossover is initiated by the formation of programmed DNA double-strand breaks (DSBs). While the role of ATAXIA-TELANGIECTASIA AND RAD3-RELATED (ATR) kinase in DNA damage signaling is well-known, its impact on crossover formation remains understudied. Here, using measurements of recombination at chromosomal intervals and genome-wide crossover mapping, we showed that ATR inactivation in Arabidopsis (Arabidopsis thaliana) leads to dramatic crossover redistribution, with an increase in crossover frequency in chromosome arms and a decrease in pericentromeres. These global changes in crossover placement were not caused by alterations in DSB numbers, which we demonstrated by analyzing phosphorylated H2A.X foci in zygonema. Using the seed-typing technique, we found that hotspot usage remains mainly unchanged in atr mutants compared with wild-type individuals. Moreover, atr showed no change in the number of crossovers caused by two independent pathways, which implies no effect on crossover pathway choice. Analyses of genetic interaction indicate that while the effects of atr are independent of MMS AND UV SENSITIVE81 (MUS81), ZIPPER1 (ZYP1), FANCONI ANEMIA COMPLEMENTATION GROUP M (FANCM), and D2 (FANCD2), the underlying mechanism may be similar between ATR and FANCD2. This study extends our understanding of ATR's role in meiosis, uncovering functions in regulating crossover distribution.
Collapse
Affiliation(s)
- Longfei Zhu
- Laboratory of Genome Biology, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| | - Julia Dluzewska
- Laboratory of Genome Biology, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| | - Nadia Fernández-Jiménez
- Departamento de Genética, Fisiología y Microbiología, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Rajeev Ranjan
- Laboratory of Genome Biology, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| | - Alexandre Pelé
- Laboratory of Genome Biology, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| | - Wojciech Dziegielewski
- Laboratory of Genome Biology, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| | - Maja Szymanska-Lejman
- Laboratory of Genome Biology, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| | - Karolina Hus
- Laboratory of Genome Biology, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| | - Julia Górna
- Laboratory of Genome Biology, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| | - Mónica Pradillo
- Departamento de Genética, Fisiología y Microbiología, Facultad de Ciencias Biológicas, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Piotr A Ziolkowski
- Laboratory of Genome Biology, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland
| |
Collapse
|
50
|
Nair JR, Huang TT, Sunkara A, Pruitt MR, Ibanez KR, Chiang CY, Cheng KCC, Wilson K, Cardillo TM, Hofsess S, Lee JM. Distinct effects of sacituzumab govitecan and berzosertib on DNA damage response in ovarian cancer. iScience 2024; 27:111283. [PMID: 39628575 PMCID: PMC11613210 DOI: 10.1016/j.isci.2024.111283] [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: 03/19/2024] [Revised: 09/10/2024] [Accepted: 10/25/2024] [Indexed: 12/06/2024] Open
Abstract
Antibody-drug conjugates (ADCs) have become an important class of anticancer drugs in solid tumors including drug-resistant gynecologic malignancies. TROP2 is a cell surface antigen that is highly expressed in ovarian carcinoma (OC) but minimally expressed in normal ovarian tissues. In this study, we aimed to identify how TROP2-specific ADC, sacituzumab govitecan (SG), modulates DNA damage response pathways in drug-resistant OC. We found that SG induces G2/M arrest, increases RPA1 foci, and decreases replication fork speed, resulting in replication stress in TROP2-positive cells while these were less evident in TROP2-negative cells. In OC in vitro and in vivo models, SN-38 sensitivity and TROP2 expression play key roles in response to either ATR inhibitor or SG alone, or in combination. Additionally, inhibition of translesion DNA synthesis enhances SG and PARP inhibitor (PARPi) sensitivity in PARPi-resistant OC cells. These findings provide mechanistic insights for clinical development of SG in drug-resistant OC.
Collapse
Affiliation(s)
- Jayakumar R. Nair
- Women’s Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Tzu-Ting Huang
- Women’s Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Anu Sunkara
- Women’s Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Margaret R. Pruitt
- Women’s Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Kristen R. Ibanez
- Women’s Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Chih-Yuan Chiang
- Functional Genomics Laboratory, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Ken Chih-Chien Cheng
- Functional Genomics Laboratory, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Kelli Wilson
- Functional Genomics Laboratory, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | | | - Scott Hofsess
- Gilead Sciences, Inc., 333 Lakeside Dr., Foster City, CA 94404, USA
| | - Jung-Min Lee
- Women’s Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|