1
|
Vidi PA, Liu J, Bonin K, Bloom K. Closing the loops: chromatin loop dynamics after DNA damage. Nucleus 2025; 16:2438633. [PMID: 39720924 DOI: 10.1080/19491034.2024.2438633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/26/2024] Open
Abstract
Chromatin is a dynamic polymer in constant motion. These motions are heterogeneous between cells and within individual cell nuclei and are profoundly altered in response to DNA damage. The shifts in chromatin motions following genomic insults depend on the temporal and physical scales considered. They are also distinct in damaged and undamaged regions. In this review, we emphasize the role of chromatin tethering and loop formation in chromatin dynamics, with the view that pulsing loops are key contributors to chromatin motions. Chromatin tethers likely mediate micron-scale chromatin coherence predicted by polymer models and measured experimentally, and we propose that remodeling of the tethers in response to DNA breaks enables uncoupling of damaged and undamaged chromatin regions.
Collapse
Affiliation(s)
| | - Jing Liu
- Department of Physics and Astronomy, Purdue University, West Lafayette, IN, USA
| | - Keith Bonin
- Department of Physics, Wake Forest University, Winston-Salem, NC, USA
| | - Kerry Bloom
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
2
|
Kim J, Wang H, Ercan S. Cohesin organizes 3D DNA contacts surrounding active enhancers in C. elegans. Genome Res 2025; 35:1108-1123. [PMID: 40210441 PMCID: PMC12047539 DOI: 10.1101/gr.279365.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 02/15/2025] [Indexed: 04/12/2025]
Abstract
In mammals, cohesin and CTCF organize the 3D genome into topologically associating domains (TADs) to regulate communication between cis-regulatory elements. Many organisms, including S. cerevisiae, C. elegans, and A. thaliana contain cohesin but lack CTCF. Here, we used C. elegans to investigate the function of cohesin in 3D genome organization in the absence of CTCF. Using Hi-C data, we observe cohesin-dependent features called "fountains," which have also been reported in zebrafish and mice. These are population average reflections of DNA loops originating from distinct genomic regions and are ∼20-40 kb in C. elegans Hi-C analysis upon cohesin and WAPL-1 depletion supports the idea that cohesin is preferentially loaded at sites bound by the C. elegans ortholog of NIPBL and loop extrudes in an effectively two-sided manner. ChIP-seq analyses show that cohesin translocation along the fountain trajectory depends on a fully intact complex and is extended upon WAPL-1 depletion. Hi-C contact patterns at individual fountains suggest that cohesin processivity is unequal on each side, possibly owing to collision with cohesin loaded from surrounding sites. The putative cohesin loading sites are closest to active enhancers, and fountain strength is associated with transcription. Compared with mammals, the average processivity of C. elegans cohesin is about 10-fold shorter, and the binding of NIPBL ortholog does not depend on cohesin. We propose that preferential loading and loop extrusion by cohesin is an evolutionarily conserved mechanism that regulates the 3D interactions of enhancers in animal genomes.
Collapse
Affiliation(s)
- Jun Kim
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, New York 10003, USA
| | - Haoyu Wang
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, New York 10003, USA
| | - Sevinç Ercan
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, New York 10003, USA
| |
Collapse
|
3
|
Liu Y, Zhangding Z, Liu X, Hu J. Chromatin-centric insights into DNA replication. Trends Genet 2025; 41:412-424. [PMID: 39765445 DOI: 10.1016/j.tig.2024.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 05/08/2025]
Abstract
DNA replication ensures the precise transmission of genetic information from parent to daughter cells. In eukaryotes, this process involves the replication of every base pair within a highly complex chromatin environment, encompassing multiple levels of chromatin structure and various chromatin metabolic processes. Recent evidence has demonstrated that DNA replication is strictly regulated in both temporal and spatial dimensions by factors such as 3D genome structure and transcription, which is crucial for maintaining genomic stability in each cell cycle. In this review, we discuss the diverse mechanisms that govern eukaryotic DNA replication, emphasizing the roles of chromatin architecture and transcriptional activity within the mammalian chromatin landscape. These insights provide a foundation for future investigations in this field.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, PKU-THU Center for Life Sciences, Peking University, Beijing 100871, China; Department of Medical Genetics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Zhengrong Zhangding
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, PKU-THU Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xuhao Liu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, PKU-THU Center for Life Sciences, Peking University, Beijing 100871, China
| | - Jiazhi Hu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, PKU-THU Center for Life Sciences, Peking University, Beijing 100871, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, Sichuan 610213, China.
| |
Collapse
|
4
|
Delvaux de Fenffe CM, Govers J, Mattiroli F. Always on the Move: Overview on Chromatin Dynamics within Nuclear Processes. Biochemistry 2025. [PMID: 40312022 DOI: 10.1021/acs.biochem.5c00114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Our genome is organized into chromatin, a dynamic and modular structure made of nucleosomes. Chromatin organization controls access to the DNA sequence, playing a fundamental role in cell identity and function. How nucleosomes enable these processes is an active area of study. In this review, we provide an overview of chromatin dynamics, its properties, mechanisms, and functions. We highlight the diverse ways by which chromatin dynamics is controlled during transcription, DNA replication, and repair. Recent technological developments have promoted discoveries in this area, to which we provide an outlook on future research directions.
Collapse
Affiliation(s)
| | - Jolijn Govers
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Francesca Mattiroli
- Hubrecht Institute-KNAW & University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| |
Collapse
|
5
|
Llerena Schiffmacher DA, Pai YJ, Pines A, Vermeulen W. Transcription-coupled repair: tangled up in convoluted repair. FEBS J 2025. [PMID: 40272095 DOI: 10.1111/febs.70104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/08/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025]
Abstract
Significant progress has been made in understanding the mechanism of transcription-coupled nucleotide excision repair (TC-NER); however, numerous aspects remain elusive, including TC-NER regulation, lesion-specific and cell type-specific complex composition, structural insights, and lesion removal dynamics in living cells. This review summarizes and discusses recent advancements in TC-NER, focusing on newly identified interactors, mechanistic insights from cryo-electron microscopy (Cryo-EM) studies and live cell imaging, and the contribution of post-translational modifications (PTMs), such as ubiquitin, in regulating TC-NER. Furthermore, we elaborate on the consequences of TC-NER deficiencies and address the role of accumulated damage and persistent lesion-stalled RNA polymerase II (Pol II) as major drivers of the disease phenotype of Cockayne syndrome (CS) and its related disorders. In this context, we also discuss the severe effects of transcription-blocking lesions (TBLs) on neurons, highlighting their susceptibility to damage. Lastly, we explore the potential of investigating three-dimensional (3D) chromatin structure and phase separation to uncover further insights into this essential DNA repair pathway.
Collapse
Affiliation(s)
- Diana A Llerena Schiffmacher
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Yun Jin Pai
- Master Scientific Illustrations, Department of Anatomy and Embryology, Faculty of Health, Medicine and Life Sciences, Maastricht University, The Netherlands
| | - Alex Pines
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Wim Vermeulen
- Department of Molecular Genetics, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
6
|
Chi Y, Lu Y, Wu J, Li G, Li X, Wu Y, Zhao X, Wen J, Sha X, Lu GL, Hu K, Zhang Z. Rational Design of Bioinspired Lipoprotein System to Improve Penetration in Colorectal Peritoneal Metastases. NANO LETTERS 2025; 25:5231-5240. [PMID: 40106687 DOI: 10.1021/acs.nanolett.4c06402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Chemotherapy of lethal colorectal peritoneal metastases (PM) is notoriously challenged by poor drug delivery efficiency in PM tumors. Inspired by the histopathological examinations of PM tumors from colon cancer patients, a C[RGDfK] peptide-modified bioinspired lipoprotein (R-BLP) system was optimized from 8 formulations with profound penetrating ability in PM tumors of colorectal cancers. Then, a chemotherapeutic 7-ethyl-10-hydroxy-camptothecin (SN38)-loaded R-BLP (termed SR-BLP) was designed to promote their penetration in PM tumors and improve the chemotherapeutic efficacy. In CT26-induced PM models, SR-BLP exhibited better penetration profiles in PM tumors over a counterpart liposomal formulation. SR-BLP treatment produced an 80.03% suppression of PM incidence with obvious DNA damage and topoisomerase I (TOP I) downregulation and caused a 1.78-fold prolongation of survival time. Therefore, the histopathological features-inspired R-BLP provides an encouraging tumor-penetrating delivery platform for the effective chemotherapy of colorectal PM.
Collapse
Affiliation(s)
- Yifei Chi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yi Lu
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, P.R. China
| | - Jingbo Wu
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Shanghai 200240, P.R. China
| | - Guodong Li
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, P.R. China
| | - Xianlu Li
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, P.R. China
| | - Yao Wu
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, P.R. China
| | - Xiao Zhao
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, P.R. China
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Shanghai 200240, P.R. China
| | - Jingyuan Wen
- The University of Auckland, Auckland 1142, New Zealand
| | - Xianyi Sha
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, P.R. China
| | - Guo-Liang Lu
- The University of Auckland, Auckland 1142, New Zealand
| | - Kaili Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Zhiwen Zhang
- School of Pharmacy, Key laboratory of smart drug delivery (Ministry of Education) & National key laboratory of complex drug formulations for overcoming delivery barriers, Fudan University, Shanghai 201203, P.R. China
- Department of Pathology, Shanghai Fifth People's Hospital, Fudan University, 801 Heqing Road, Shanghai 200240, P.R. China
| |
Collapse
|
7
|
Amodeo ME, Eyler CE, Johnstone SE. Rewiring cancer: 3D genome determinants of cancer hallmarks. Curr Opin Genet Dev 2025; 91:102307. [PMID: 39862605 PMCID: PMC11867856 DOI: 10.1016/j.gde.2024.102307] [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: 09/09/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025]
Abstract
In modern cancer biology, Hanahan and Weinberg's classic depiction of the Hallmarks of Cancer serves as a heuristic for understanding malignant phenotypes [1]. Genetic determinants of these phenotypes promote cancer induction and progression, and these mutations drive current approaches to understanding and treating cancer. Meanwhile, for over a century, pathologists have noted that profound alterations of nuclear structure accompany transformation, integrating these changes into diagnostic classifications (Figure 1). Nevertheless, the relationship of nuclear organization to malignant phenotypes has lagged. Recent advances yield profound insight into the 3D genome's relationship with cancer phenotypes, suggesting that spatial genome organization influences many, if not all, of these malignant features. Here, we highlight recent discoveries elucidating connections between 3D genome organization and cancer phenotypes.
Collapse
Affiliation(s)
- Maria E Amodeo
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA; Broad Institute, Cambridge, MA, USA
| | - Christine E Eyler
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA.
| | - Sarah E Johnstone
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA; Broad Institute, Cambridge, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
8
|
Smaruj PN, Xiao Y, Fudenberg G. Recipes and ingredients for deep learning models of 3D genome folding. Curr Opin Genet Dev 2025; 91:102308. [PMID: 39862604 PMCID: PMC11867851 DOI: 10.1016/j.gde.2024.102308] [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: 12/19/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025]
Abstract
Three-dimensional genome folding plays roles in gene regulation and disease. In this review, we compare and contrast recent deep learning models for predicting genome contact maps. We survey preprocessing, architecture, training, evaluation, and interpretation methods, highlighting the capabilities and limitations of different models. In each area, we highlight challenges, opportunities, and potential future directions for genome-folding models.
Collapse
Affiliation(s)
- Paulina N Smaruj
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Yao Xiao
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA
| | - Geoffrey Fudenberg
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Murayama Y. Sister chromatid cohesion through the lens of biochemical experiments. Curr Opin Cell Biol 2025; 93:102464. [PMID: 39879903 DOI: 10.1016/j.ceb.2025.102464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 12/29/2024] [Accepted: 01/06/2025] [Indexed: 01/31/2025]
Abstract
Faithful chromosome segregation in eukaryotes relies on physical cohesion between newly duplicated sister chromatids. Cohesin is a ring-shaped ATPase assembly that mediates sister chromatid cohesion through its ability to topologically entrap DNA. Cohesin, assisted by several regulatory proteins, binds to DNA prior to DNA replication and then holds two sister DNAs together when it encounters the replication machinery. Cohesion establishment further requires cohesin acetylation, which confers near eternal stability on chromatin-bound cohesin until the onset of chromosome segregation. In addition to a wealth of experimental evidence from cellular studies, recent advances in reconstitution approaches are now beginning to unravel the biochemical properties of cohesin that underlie its function in sister chromatid cohesion. This review summarizes recent insights into the mechanism of cohesion establishment.
Collapse
Affiliation(s)
- Yasuto Murayama
- Department of Chromosome Science, National Institute of Genetics, Mishima, 411-8540, Japan; Department of Genetics, Graduate University for Advanced Studies (SOKENDAI), Mishima, 411-8540, Japan.
| |
Collapse
|
10
|
Chiolo I, Altmeyer M, Legube G, Mekhail K. Nuclear and genome dynamics underlying DNA double-strand break repair. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00828-1. [PMID: 40097581 DOI: 10.1038/s41580-025-00828-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2025] [Indexed: 03/19/2025]
Abstract
Changes in nuclear shape and in the spatial organization of chromosomes in the nucleus commonly occur in cancer, ageing and other clinical contexts that are characterized by increased DNA damage. However, the relationship between nuclear architecture, genome organization, chromosome stability and health remains poorly defined. Studies exploring the connections between the positioning and mobility of damaged DNA relative to various nuclear structures and genomic loci have revealed nuclear and cytoplasmic processes that affect chromosome stability. In this Review, we discuss the dynamic mechanisms that regulate nuclear and genome organization to promote DNA double-strand break (DSB) repair, genome stability and cell survival. Genome dynamics that support DSB repair rely on chromatin states, repair-protein condensates, nuclear or cytoplasmic microtubules and actin filaments, kinesin or myosin motor proteins, the nuclear envelope, various nuclear compartments, chromosome topology, chromatin loop extrusion and diverse signalling cues. These processes are commonly altered in cancer and during natural or premature ageing. Indeed, the reshaping of the genome in nuclear space during DSB repair points to new avenues for therapeutic interventions that may take advantage of new cancer cell vulnerabilities or aim to reverse age-associated defects.
Collapse
Affiliation(s)
- Irene Chiolo
- Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA.
| | - Matthias Altmeyer
- Department of Molecular Mechanisms of Disease, University of Zurich (UZH), Zurich, Switzerland.
| | - Gaëlle Legube
- MCD, Centre de Biologie Intégrative (CBI), CNRS, Université de Toulouse, UT3, Toulouse, France.
| | - Karim Mekhail
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
11
|
Ambrosio S, Noviello A, Di Fusco G, Gorini F, Piscone A, Amente S, Majello B. Interplay and Dynamics of Chromatin Architecture and DNA Damage Response: An Overview. Cancers (Basel) 2025; 17:949. [PMID: 40149285 PMCID: PMC11940107 DOI: 10.3390/cancers17060949] [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: 02/07/2025] [Revised: 03/06/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
Genome stability is safeguarded by a finely orchestrated cascade of events that collectively represent the DNA damage response (DDR). In eukaryotes, the DDR operates within the dynamic chromatin landscape, where the interplay between DNA repair factors, chromatin remodeling, replication, transcription, spatial genome organization, and cytoskeletal forces is tightly coordinated. High-resolution studies have unveiled chromatin alterations spanning multiple scales, from localized kilobase-level changes to megabase-scale reorganization, which impact chromatin's physical properties and enhance the mobility of damaged regions. Leveraging this knowledge could pave the way for innovative therapeutic strategies, particularly in targeting chromatin dynamics to destabilize cancer cells selectively. This review, focusing on DNA double-strand breaks (DSBs), sheds light on how chromatin undergoes dynamic modifications in response to damage and how these changes influence the DDR at both local and global levels, offering a glimpse into how nuclear architecture contributes to the delicate balance between genome stability and adaptability and highlighting the importance of exploring these interactions in the context of cancer therapy.
Collapse
Affiliation(s)
- Susanna Ambrosio
- Department of Biology, University of Naples “Federico II”, 80126 Naples, Italy; (A.N.); (G.D.F.)
| | - Anna Noviello
- Department of Biology, University of Naples “Federico II”, 80126 Naples, Italy; (A.N.); (G.D.F.)
| | - Giovanni Di Fusco
- Department of Biology, University of Naples “Federico II”, 80126 Naples, Italy; (A.N.); (G.D.F.)
| | - Francesca Gorini
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy; (F.G.); (A.P.); (S.A.)
| | - Anna Piscone
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy; (F.G.); (A.P.); (S.A.)
| | - Stefano Amente
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples “Federico II”, 80131 Naples, Italy; (F.G.); (A.P.); (S.A.)
| | - Barbara Majello
- Department of Biology, University of Naples “Federico II”, 80126 Naples, Italy; (A.N.); (G.D.F.)
| |
Collapse
|
12
|
Machour FE, Barisaac AS, Ayoub N. Why are RNA processing factors recruited to DNA double-strand breaks? Trends Genet 2025; 41:194-200. [PMID: 39567312 DOI: 10.1016/j.tig.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 11/22/2024]
Abstract
DNA double-strand break (DSB) induction leads to local transcriptional silencing at damage sites, raising the question: Why are RNA processing factors (RPFs), including splicing factors, rapidly recruited to these sites? Recent findings show that DSBs cluster in a chromatin compartment termed the 'D compartment', where DNA damage response (DDR) genes relocate and undergo transcriptional activation. Here, we propose two non-mutually exclusive models to elucidate the rationale behind the recruitment of RPFs to DSB sites. First, RPFs circulate through the D compartment to process transcripts of the relocated DDR genes. Second, the D compartment serves as a 'post-translational modifications (PTMs) hub', altering RPF activity and leading to the production of unique DNA damage-induced transcripts, which are essential for orchestrating the DDR.
Collapse
Affiliation(s)
- Feras E Machour
- Department of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Alma Sophia Barisaac
- Department of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel
| | - Nabieh Ayoub
- Department of Biology, Technion - Israel Institute of Technology, Haifa 3200003, Israel.
| |
Collapse
|
13
|
Sebastian R, Sun EG, Fedkenheuer M, Fu H, Jung S, Thakur BL, Redon CE, Pegoraro G, Tran AD, Gross JM, Mosavarpour S, Kusi NA, Ray A, Dhall A, Pongor LS, Casellas R, Aladjem MI. Mechanism for local attenuation of DNA replication at double-strand breaks. Nature 2025; 639:1084-1092. [PMID: 39972127 DOI: 10.1038/s41586-024-08557-9] [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: 12/22/2023] [Accepted: 12/09/2024] [Indexed: 02/21/2025]
Abstract
DNA double-strand breaks (DSBs) disrupt the continuity of the genome, with consequences for malignant transformation. Massive DNA damage can elicit a cellular checkpoint response that prevents cell proliferation1,2. However, how highly aggressive cancer cells, which can tolerate widespread DNA damage, respond to DSBs alongside continuous chromosome duplication is unknown. Here we show that DSBs induce a local genome maintenance mechanism that inhibits replication initiation in DSB-containing topologically associating domains (TADs) without affecting DNA synthesis at other genomic locations. This process is facilitated by mediators of replication and DSBs (MRDs). In normal and cancer cells, MRDs include the TIMELESS-TIPIN complex and the WEE1 kinase, which actively dislodges the TIMELESS-TIPIN complex from replication origins adjacent to DSBs and prevents initiation of DNA synthesis at DSB-containing TADs. Dysregulation of MRDs, or disruption of 3D chromatin architecture by dissolving TADs, results in inadvertent replication in damaged chromatin and increased DNA damage in cancer cells. We propose that the intact MRD cascade precedes DSB repair to prevent genomic instability, which is otherwise observed when replication is forced, or when genome architecture is challenged, in the presence of DSBs3-5. These observations reveal a previously unknown vulnerability in the DNA replication machinery that may be exploited to therapeutically target cancer cells.
Collapse
Affiliation(s)
- Robin Sebastian
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eric G Sun
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Tri-Institutional MD-PhD Program, Weill Cornell Medicine, Rockefeller University, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Fedkenheuer
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Haiqing Fu
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - SeolKyoung Jung
- Biodata Mining and Discovery Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Bhushan L Thakur
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christophe E Redon
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gianluca Pegoraro
- High Throughput Imaging Facility (HiTIF), National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andy D Tran
- CCR Microscopy Core Facility, Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jacob M Gross
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sara Mosavarpour
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nana Afua Kusi
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anagh Ray
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anjali Dhall
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lorinc S Pongor
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Genomics and Epigenetics Core Group, HCEMM, Szeged, Hungary
| | - Rafael Casellas
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
- Department of Hematopoietic Biology and Malignancy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mirit I Aladjem
- Developmental Therapeutics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
14
|
Giles KA, Taberlay PC, Cesare AJ, Jones MJK. Roles for the 3D genome in the cell cycle, DNA replication, and double strand break repair. Front Cell Dev Biol 2025; 13:1548946. [PMID: 40083661 PMCID: PMC11903485 DOI: 10.3389/fcell.2025.1548946] [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: 12/20/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Large eukaryotic genomes are packaged into the restricted area of the nucleus to protect the genetic code and provide a dedicated environment to read, copy and repair DNA. The physical organisation of the genome into chromatin loops and self-interacting domains provides the basic structural units of genome architecture. These structural arrangements are complex, multi-layered, and highly dynamic and influence how different regions of the genome interact. The role of chromatin structures during transcription via enhancer-promoter interactions is well established. Less understood is how nuclear architecture influences the plethora of chromatin transactions during DNA replication and repair. In this review, we discuss how genome architecture is regulated during the cell cycle to influence the positioning of replication origins and the coordination of DNA double strand break repair. The role of genome architecture in these cellular processes highlights its critical involvement in preserving genome integrity and cancer prevention.
Collapse
Affiliation(s)
- Katherine A. Giles
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Phillippa C. Taberlay
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Anthony J. Cesare
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW, Australia
| | - Mathew J. K. Jones
- Faculty of Medicine, Frazer Institute, University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
15
|
Liao Q, Brandão HB, Ren Z, Wang X. Replisomes restrict SMC-mediated DNA-loop extrusion in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.23.639750. [PMID: 40027636 PMCID: PMC11870623 DOI: 10.1101/2025.02.23.639750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Structural maintenance of chromosomes (SMC) complexes organize genomes by extruding DNA loops, while replisomes duplicate entire chromosomes. These essential molecular machines must collide frequently in every cell cycle, yet how such collisions are resolved in vivo remains poorly understood. Taking advantage of the ability to load SMC complexes at defined sites in the Bacillus subtilis genome, we engineered head-on and head-to-tail collisions between SMC complexes and the replisome. Replisome progression was monitored by marker frequency analysis, and SMC translocation was monitored by time-resolved ChIP-seq and Hi-C. We found that SMC complexes do not impede replisome progression. By contrast, replisomes restrict SMC translocation regardless of collision orientations. Combining experimental data with simulations, we determined that SMC complexes are blocked by the replisome and then released from the chromosome. Occasionally, SMC complexes can bypass the replisome and continue translocating. Our findings establish that the replisome is a barrier to SMC-mediated DNA-loop extrusion in vivo , with implications for processes such as chromosome segregation, DNA repair, and gene regulation that require dynamic chromosome organization in all organisms.
Collapse
|
16
|
Selivanovskiy AV, Molodova MN, Khrameeva EE, Ulianov SV, Razin SV. Liquid condensates: a new barrier to loop extrusion? Cell Mol Life Sci 2025; 82:80. [PMID: 39976773 PMCID: PMC11842697 DOI: 10.1007/s00018-024-05559-8] [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: 07/10/2024] [Revised: 12/04/2024] [Accepted: 12/20/2024] [Indexed: 02/23/2025]
Abstract
Liquid-liquid phase separation (LLPS), driven by dynamic, low-affinity multivalent interactions of proteins and RNA, results in the formation of macromolecular condensates on chromatin. These structures are likely to provide high local concentrations of effector factors responsible for various processes including transcriptional regulation and DNA repair. In particular, enhancers, super-enhancers, and promoters serve as platforms for condensate assembly. In the current paradigm, enhancer-promoter (EP) interaction could be interpreted as a result of enhancer- and promoter-based condensate contact/fusion. There is increasing evidence that the spatial juxtaposition of enhancers and promoters could be provided by loop extrusion (LE) by SMC complexes. Here, we propose that condensates may act as barriers to LE, thereby contributing to various nuclear processes including spatial contacts between regulatory genomic elements.
Collapse
Affiliation(s)
- Arseniy V Selivanovskiy
- Institute of Gene Biology, Russian Academy of Sciences, 119334, Moscow, Russia
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia
| | - Maria N Molodova
- Institute of Gene Biology, Russian Academy of Sciences, 119334, Moscow, Russia
- Skolkovo Institute of Science and Technology, 121205, Moscow, Russia
| | | | - Sergey V Ulianov
- Institute of Gene Biology, Russian Academy of Sciences, 119334, Moscow, Russia
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia
| | - Sergey V Razin
- Institute of Gene Biology, Russian Academy of Sciences, 119334, Moscow, Russia.
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119234, Moscow, Russia.
| |
Collapse
|
17
|
Marin-Gonzalez A, Rybczynski AT, Nilavar NM, Nguyen D, Karwacki-Neisius V, Li AG, Zou RS, Avilés-Vázquez FJ, Kanemaki MT, Scully R, Ha T. Cohesin drives chromatin scanning during the RAD51-mediated homology search. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637451. [PMID: 39990468 PMCID: PMC11844420 DOI: 10.1101/2025.02.10.637451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Cohesin folds genomes into chromatin loops, whose roles are under debate. We report that double strand breaks (DSB) induce de novo formation of chromatin loops, with the break positioned at the loop base. These loops form only in S/G2 phases and occur during repair via homologous recombination (HR), concomitant with DNA end resection and RAD51 assembly. RAD51 showed two-tiered accumulation around DSBs, with a broad (~Mb) domain arising from the homology search. This domain is regulated by cohesin unloader, is constrained by TAD boundaries, and it overlaps with chromatin regions reeled through the break-anchored loop, suggesting that loop extrusion regulates the homology search. Indeed, depletion of NIPBL results in reduced HR, and this effect is more pronounced when the HR donor is far (~100 kb) from the break. Our data indicates that loop-extruding cohesin promotes the mammalian homology search by facilitating break-chromatin interactions within the damaged TAD.
Collapse
Affiliation(s)
- Alberto Marin-Gonzalez
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Adam T. Rybczynski
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Namrata M. Nilavar
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Daniel Nguyen
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Violetta Karwacki-Neisius
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Andrew G. Li
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Roger S. Zou
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Franklin J. Avilés-Vázquez
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University, Baltimore, MD, USA
| | - Masato T. Kanemaki
- Department of Chromosome Science, National Institute of Genetics, Mishima, Japan
- Graduate Institute for Advanced Studies, SOKENDAI, Mishima, Japan
- Department of Biological Science, Graduate School of Science, The University of Tokyo, Tokyo, Japan
| | - Ralph Scully
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Taekjip Ha
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
18
|
Marin-Gonzalez A, Rybczynski AT, Zou RS, Ha T. Measuring double-strand break repair events in mammalian cells with multi-target CRISPR. Methods Enzymol 2025; 712:1-22. [PMID: 40121069 DOI: 10.1016/bs.mie.2025.01.011] [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: 03/25/2025]
Abstract
A mechanistic understanding of the different pathways involved in the repair of DSBs is a timely, yet challenging task. CRISPR-Cas9 is a powerful tool to induce DNA double-strand breaks (DSB) at defined genomic locations to study the ensuing repair response, but Cas9 studies are typically limited by i) low-throughput induction of DSB, by targeting only one or a few genomic sites, or ii) the use of genetically integrated reporter systems, which do not always reflect endogenous phenotypes. To address these limitations, we developed multi-target CRISPR, a Cas9-based tool to controllably induce DSBs in high-throughput at endogenous sites, by leveraging repetitive genomic regions. In this Chapter, we describe how to design and execute a multi-target CRISPR experiment. We also detail how to analyze next-generation sequencing data for characterization of DSB repair events at multiple cut sites. We envision that multi-target CRISPR will become a valuable tool for the study of mammalian DSB repair mechanisms.
Collapse
Affiliation(s)
- Alberto Marin-Gonzalez
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States; Howard Hughes Medical Institute, Boston, MA, United States
| | - Adam T Rybczynski
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Roger S Zou
- Department of Medicine, Massachusetts General Hospital, Boston, MA, United States
| | - Taekjip Ha
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, United States; Department of Pediatrics, Harvard Medical School, Boston, MA, United States; Howard Hughes Medical Institute, Boston, MA, United States.
| |
Collapse
|
19
|
Lang F, Kaur K, Fu H, Zaheer J, Ribeiro DL, Aladjem MI, Yang C. D-2-hydroxyglutarate impairs DNA repair through epigenetic reprogramming. Nat Commun 2025; 16:1431. [PMID: 39920158 PMCID: PMC11806014 DOI: 10.1038/s41467-025-56781-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
Cancer-associated mutations in IDH are associated with multiple types of human malignancies, which exhibit distinctive metabolic reprogramming, production of oncometabolite D-2-HG, and shifted epigenetic landscape. IDH mutated malignancies are signatured with "BRCAness", highlighted with the sensitivity to DNA repair inhibitors and genotoxic agents, although the underlying molecular mechanism remains elusive. In the present study, we demonstrate that D-2-HG impacts the chromatin conformation adjustments, which are associated with DNA repair process. Mechanistically, D-2-HG diminishes the chromatin interactions in the DNA damage regions via revoking CTCF binding. The hypermethylation of cytosine, resulting from the suppression of TET1 and TET2 activities by D-2-HG, contributes to the dissociation of CTCF from DNA damage regions. CTCF depletion leads to the disruption of chromatin organization around the DNA damage sites, which abolishes the recruitment of essential DNA damage repair proteins BRCA2 and RAD51, as well as impairs homologous repair in the IDH mutant cancer cells. These findings provide evidence that CTCF-mediated chromatin interactions play a key role in DNA damage repair proceedings. Oncometabolites jeopardize genome stability and DNA repair by affecting high-order chromatin structure.
Collapse
Affiliation(s)
- Fengchao Lang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Karambir Kaur
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Haiqing Fu
- Developmental Therapeutic Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Javeria Zaheer
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Diego Luis Ribeiro
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Mirit I Aladjem
- Developmental Therapeutic Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Chunzhang Yang
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
20
|
Barth R, Davidson IF, van der Torre J, Taschner M, Gruber S, Peters JM, Dekker C. SMC motor proteins extrude DNA asymmetrically and can switch directions. Cell 2025; 188:749-763.e21. [PMID: 39824185 DOI: 10.1016/j.cell.2024.12.020] [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: 12/22/2023] [Revised: 09/13/2024] [Accepted: 12/13/2024] [Indexed: 01/20/2025]
Abstract
Structural maintenance of chromosomes (SMC) complexes organize the genome via DNA loop extrusion. Although some SMCs were reported to do so symmetrically, reeling DNA from both sides into the extruded DNA loop simultaneously, others perform loop extrusion asymmetrically toward one direction only. The mechanism underlying this variability remains unclear. Here, we examine the directionality of DNA loop extrusion by SMCs using in vitro single-molecule experiments. We find that cohesin and SMC5/6 do not reel in DNA from both sides, as reported before, but instead extrude DNA asymmetrically, although the direction can switch over time. Asymmetric DNA loop extrusion thus is the shared mechanism across all eukaryotic SMC complexes. For cohesin, direction switches strongly correlate with the turnover of the subunit NIPBL, during which DNA strand switching may occur. Apart from expanding by extrusion, loops frequently diffuse and shrink. The findings reveal that SMCs, surprisingly, can switch directions.
Collapse
Affiliation(s)
- Roman Barth
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, the Netherlands
| | - Iain F Davidson
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Jaco van der Torre
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, the Netherlands
| | - Michael Taschner
- Department of Fundamental Microbiology (DMF), Faculty of Biology and Medicine (FBM), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Stephan Gruber
- Department of Fundamental Microbiology (DMF), Faculty of Biology and Medicine (FBM), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Jan-Michael Peters
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC), Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Cees Dekker
- Department of Bionanoscience, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, the Netherlands.
| |
Collapse
|
21
|
Kaya VO, Adebali O. UV-induced reorganization of 3D genome mediates DNA damage response. Nat Commun 2025; 16:1376. [PMID: 39910043 PMCID: PMC11799157 DOI: 10.1038/s41467-024-55724-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 12/20/2024] [Indexed: 02/07/2025] Open
Abstract
While it is well-established that UV radiation threatens genomic integrity, the precise mechanisms by which cells orchestrate DNA damage response and repair within the context of 3D genome architecture remain unclear. Here, we address this gap by investigating the UV-induced reorganization of the 3D genome and its critical role in mediating damage response. Employing temporal maps of contact matrices and transcriptional profiles, we illustrate the immediate and holistic changes in genome architecture post-irradiation, emphasizing the significance of this reconfiguration for effective DNA repair processes. We demonstrate that UV radiation triggers a comprehensive restructuring of the 3D genome organization at all levels, including loops, topologically associating domains and compartments. Through the analysis of DNA damage and excision repair maps, we uncover a correlation between genome folding, gene regulation, damage formation probability, and repair efficacy. We show that adaptive reorganization of the 3D genome is a key mediator of the damage response, providing new insights into the complex interplay of genomic structure and cellular defense mechanisms against UV-induced damage, thereby advancing our understanding of cellular resilience.
Collapse
Affiliation(s)
- Veysel Oğulcan Kaya
- Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, Türkiye
| | - Ogün Adebali
- Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul, Türkiye.
| |
Collapse
|
22
|
Wong LH, Tremethick DJ. Multifunctional histone variants in genome function. Nat Rev Genet 2025; 26:82-104. [PMID: 39138293 DOI: 10.1038/s41576-024-00759-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2024] [Indexed: 08/15/2024]
Abstract
Histones are integral components of eukaryotic chromatin that have a pivotal role in the organization and function of the genome. The dynamic regulation of chromatin involves the incorporation of histone variants, which can dramatically alter its structural and functional properties. Contrary to an earlier view that limited individual histone variants to specific genomic functions, new insights have revealed that histone variants exert multifaceted roles involving all aspects of genome function, from governing patterns of gene expression at precise genomic loci to participating in genome replication, repair and maintenance. This conceptual change has led to a new understanding of the intricate interplay between chromatin and DNA-dependent processes and how this connection translates into normal and abnormal cellular functions.
Collapse
Affiliation(s)
- Lee H Wong
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - David J Tremethick
- The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capial Territory, Australia.
| |
Collapse
|
23
|
Tang W, Costantino L, Stocsits R, Wutz G, Ladurner R, Hudecz O, Mechtler K, Peters JM. Cohesin positions the epigenetic reader Phf2 within the genome. EMBO J 2025; 44:736-766. [PMID: 39748119 PMCID: PMC11790891 DOI: 10.1038/s44318-024-00348-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Genomic DNA is assembled into chromatin by histones, and extruded into loops by cohesin. These mechanisms control important genomic functions, but whether histones and cohesin cooperate in genome regulation is poorly understood. Here we identify Phf2, a member of the Jumonji-C family of histone demethylases, as a cohesin-interacting protein. Phf2 binds to H3K4me3 nucleosomes at active transcription start sites (TSSs), but also co-localizes with cohesin. Cohesin depletion reduces Phf2 binding at sites lacking H3K4me3, and depletion of Wapl and CTCF re-positions Phf2 together with cohesin in the genome, resulting in the accumulation of both proteins in chromosomal regions called vermicelli and cohesin islands. Conversely, Phf2 depletion reduces cohesin binding at TSSs lacking CTCF and decreases the number of short cohesin loops, while increasing the length of heterochromatic B compartments. These results suggest that Phf2 is an 'epigenetic reader', which is translocated through the genome by cohesin-mediated DNA loop extrusion, and which recruits cohesin to active TSSs and limits the size of B compartments. These findings reveal an unexpected degree of cooperativity between epigenetic and architectural mechanisms of eukaryotic genome regulation.
Collapse
Affiliation(s)
- Wen Tang
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Lorenzo Costantino
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Roman Stocsits
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Gordana Wutz
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Rene Ladurner
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Otto Hudecz
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Karl Mechtler
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria
| | - Jan-Michael Peters
- Research Institute of Molecular Pathology (IMP), Vienna Biocenter (VBC), Campus-Vienna-Biocenter 1, 1030, Vienna, Austria.
| |
Collapse
|
24
|
Rutkauskas M, Kim E. In vitro dynamics of DNA loop extrusion by structural maintenance of chromosomes complexes. Curr Opin Genet Dev 2025; 90:102284. [PMID: 39591812 DOI: 10.1016/j.gde.2024.102284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
Genomic DNA inside the cell's nucleus is highly organized and tightly controlled by the structural maintenance of chromosomes (SMC) protein complexes. These complexes fold genomes by creating and processively enlarging loops, a process called loop extrusion. After more than a decade of accumulating indirect evidence, recent in vitro single-molecule studies confirmed loop extrusion as an evolutionarily conserved function among eukaryotic and prokaryotic SMCs. These studies further provided important insights into mechanisms and regulations of these universal molecular machines, which will be discussed in this minireview.
Collapse
Affiliation(s)
- Marius Rutkauskas
- Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Eugene Kim
- Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
25
|
He L, Moon J, Cai C, Hao Y, Lee H, Kim W, Zhao F, Lou Z. The interplay between chromatin remodeling and DNA double-strand break repair: Implications for cancer biology and therapeutics. DNA Repair (Amst) 2025; 146:103811. [PMID: 39848026 DOI: 10.1016/j.dnarep.2025.103811] [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: 11/25/2024] [Revised: 01/08/2025] [Accepted: 01/12/2025] [Indexed: 01/25/2025]
Abstract
Proper chromatin remodeling is crucial for many cellular physiological processes, including the repair of DNA double-strand break (DSB). While the mechanism of DSB repair is well understood, the connection between chromatin remodeling and DSB repair remains incompletely elucidated. In this review, we aim to highlight recent studies demonstrating the close relationship between chromatin remodeling and DSB repair. We summarize the impact of DSB repair on chromatin, including nucleosome arrangement, chromatin organization, and dynamics, and conversely, the role of chromatin architecture in regulating DSB repair. Additionally, we also summarize the contribution of chromatin remodeling complexes to cancer biology through DNA repair and discuss their potential as therapeutic targets for cancer.
Collapse
Affiliation(s)
- Liujun He
- College of Biology, Hunan University, Changsha 410082, China
| | - Jaeyoung Moon
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Chungcheongnam-do 31151, Republic of Korea
| | - Chenghui Cai
- College of Biology, Hunan University, Changsha 410082, China
| | - Yalan Hao
- Analytical Instrumentation Center, Hunan University, Changsha 410082, China
| | - Hyorin Lee
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Chungcheongnam-do 31151, Republic of Korea
| | - Wootae Kim
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-bio Science (SIMS), Soonchunhyang University, Cheonan, Chungcheongnam-do 31151, Republic of Korea.
| | - Fei Zhao
- College of Biology, Hunan University, Changsha 410082, China.
| | - Zhenkun Lou
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
26
|
Scott JS, Al Ayadi L, Epeslidou E, van Scheppingen RH, Mukha A, Kaaij LJT, Lutz C, Prekovic S. Emerging roles of cohesin-STAG2 in cancer. Oncogene 2025; 44:277-287. [PMID: 39613934 DOI: 10.1038/s41388-024-03221-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 10/30/2024] [Accepted: 11/04/2024] [Indexed: 12/01/2024]
Abstract
Cohesin, a crucial regulator of genome organisation, plays a fundamental role in maintaining chromatin architecture as well as gene expression. Among its subunits, STAG2 stands out because of its frequent deleterious mutations in various cancer types, such as bladder cancer and melanoma. Loss of STAG2 function leads to significant alterations in chromatin structure, disrupts transcriptional regulation, and impairs DNA repair pathways. In this review, we explore the molecular mechanisms underlying cohesin-STAG2 function, highlighting its roles in healthy cells and its contributions to cancer biology, showing how STAG2 dysfunction promotes tumourigenesis and presents opportunities for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Julia S Scott
- Center for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | - Loubna Al Ayadi
- Center for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | | | | | - Anna Mukha
- Department of Medical BioSciences, RadboudUMC, Nijmegen, The Netherlands
| | - Lucas J T Kaaij
- Center for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | - Catrin Lutz
- Division of Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Stefan Prekovic
- Center for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
27
|
Zhang QS, Wang JN, Yang TL, Li SY, Li JQ, Liu DN, Shang H, Zhang ZN. SHMT2 regulates CD8+ T cell senescence via the reactive oxygen species axis in HIV-1 infected patients on antiretroviral therapy. EBioMedicine 2025; 112:105533. [PMID: 39808948 PMCID: PMC11782833 DOI: 10.1016/j.ebiom.2024.105533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Although antiretroviral therapy (ART) effectively inhibits viral replication, it does not fully mitigate the immunosenescence instigated by HIV infection. Cellular metabolism regulates cellular differentiation, survival, and senescence. Serine hydroxymethyltransferase 2 (SHMT2) is the first key enzyme for the entry of serine into the mitochondria from the de novo synthesis pathway that orchestrates its conversion glutathione (GSH), a key molecule in neutralising ROS and ensuring the stability of the immune system. It remains incompletely understood whether SHMT2 is involved in the senescence of CD8+ T cells, crucial for immune vigilance against HIV. METHODS HIV-infected individuals receiving antiretroviral therapy were enrolled in our study. SHMT2-siRNA was electroporated into T cells to disrupt the gene expression of SHMT2, followed by the quantification of mRNA levels of crucial serine metabolism enzymes using real-time PCR. Immunophenotyping, proliferation, cellular and mitochondrial function, and senescence-associated signalling pathways were examined using flow cytometry in CD8+ T cell subsets. FINDINGS Our findings revealed that CD8+ T cells in HIV-infected subjects are inclined towards senescence, and we identified that SHMT2, a key enzyme in serine metabolism, plays a role in CD8+ T cell senescence. SHMT2 can regulate glutathione (GSH) synthesis and protect mitochondrial function, thus effectively controlling intracellular reactive oxygen species (ROS) levels. Moreover, SHMT2 significantly contributes to averting immunosenescence and sustaining CD8+ T cell competence by modulating downstream DNA damage and phosphorylation cascades in pathways intricately linked to cellular senescence. Additionally, our study identified glycine can ameliorate CD8+ T cell senescence in HIV-infected individuals. INTERPRETATION Decreased SHMT2 levels in HIV-infected CD8+ T cells affect ROS levels by altering mitochondrial function and GSH content. Increased ROS levels activate senescence-related signalling pathways in the nucleus. However, glycine supplementation counteracts these effects and moderates senescence. FUNDING This study was supported by grants from the National Key R&D Program of China (2021YFC2301900-2021YFC2301901), National Natural Science Foundation of China (82372240), and Department of Science and Technology of Liaoning Province Project for the High-Quality Scientific and Technological Development of China Medical University (2022JH2/20200074).
Collapse
Affiliation(s)
- Qi-Sheng Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, 110001, China; The First Affiliated Hospital of Baotou Medical College, Baotou, 014010, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001, China
| | - Jia-Ning Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, 110001, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001, China
| | - Tian-Ling Yang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, 110001, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001, China
| | - Si-Yao Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, 110001, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001, China
| | - Jia-Qi Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, 110001, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001, China
| | - Ding-Ning Liu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, 110001, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001, China
| | - Hong Shang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, 110001, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001, China.
| | - Zi-Ning Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, NHC Key Laboratory of AIDS Prevention and Treatment, National Clinical Research Center for Laboratory Medicine, The First Hospital of China Medical University, China Medical University, Shenyang, 110001, China; Key Laboratory of AIDS Immunology, Chinese Academy of Medical Sciences, Shenyang, 110001, China; Key Laboratory of AIDS Immunology of Liaoning Province, Shenyang, 110001, China.
| |
Collapse
|
28
|
Fedkenheuer M, Shang Y, Jung S, Fedkenheuer K, Park S, Mazza D, Sebastian R, Nagashima H, Zong D, Tan H, Jaiswal SK, Fu H, Cruz A, Vartak SV, Wisniewski J, Sartorelli V, O'Shea JJ, Elnitski L, Nussenzweig A, Aladjem MI, Meng FL, Casellas R. A dual role of Cohesin in DNA DSB repair. Nat Commun 2025; 16:843. [PMID: 39833168 PMCID: PMC11747280 DOI: 10.1038/s41467-025-56086-4] [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: 04/06/2024] [Accepted: 01/08/2025] [Indexed: 01/22/2025] Open
Abstract
Cells undergo tens of thousands of DNA-damaging events each day. Defects in repairing double-stranded breaks (DSBs) can lead to genomic instability, contributing to cancer, genetic disorders, immunological diseases, and developmental defects. Cohesin, a multi-subunit protein complex, plays a crucial role in both chromosome organization and DNA repair by creating architectural loops through chromatin extrusion. However, the mechanisms by which cohesin regulates these distinct processes are not fully understood. In this study, we identify two separate roles for cohesin in DNA repair within mammalian cells. First, cohesin serves as an intrinsic architectural factor that normally prevents interactions between damaged chromatin. Second, cohesin has an architecture-independent role triggered by ATM phosphorylation of SMC1, which enhances the efficiency of repair. Our findings suggest that these two functions work together to reduce the occurrence of translocations and deletions associated with non-homologous end joining, thereby maintaining genomic stability.
Collapse
Affiliation(s)
- Michael Fedkenheuer
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Yafang Shang
- Key Laboratory of RNA Innovation, Science and Engineering, 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, 200031, China
| | - Seolkyoung Jung
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kevin Fedkenheuer
- Translational and Functional Analysis Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Solji Park
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Davide Mazza
- Experimental Imaging Center, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, Milano, Italy
| | - Robin Sebastian
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892-4255, USA
| | - Hiroyuki Nagashima
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dali Zong
- Laboratory of Genome Integrity, National Cancer Institute NIH, Bethesda, MD, USA
| | - Hua Tan
- Translational and Functional Analysis Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sushil Kumar Jaiswal
- Translational and Functional Analysis Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Haiqing Fu
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892-4255, USA
| | - Anthony Cruz
- Translational Genetics and Genomics Section, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Supriya V Vartak
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jan Wisniewski
- EIB Microscopy and Digital Imaging Facility, National Cancer Institute NIH, Bethesda, MD, USA
| | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Laura Elnitski
- Translational and Functional Analysis Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andre Nussenzweig
- Laboratory of Genome Integrity, National Cancer Institute NIH, Bethesda, MD, USA
| | - Mirit I Aladjem
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, 20892-4255, USA
| | - Fei-Long Meng
- Key Laboratory of RNA Innovation, Science and Engineering, 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, 200031, China
| | - Rafael Casellas
- Department of Hematopoietic Biology & Malignancy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
29
|
López-Hernández L, Toolan-Kerr P, Bannister AJ, Millán-Zambrano G. Dynamic histone modification patterns coordinating DNA processes. Mol Cell 2025; 85:225-237. [PMID: 39824165 DOI: 10.1016/j.molcel.2024.10.034] [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: 08/12/2024] [Revised: 10/02/2024] [Accepted: 10/25/2024] [Indexed: 01/20/2025]
Abstract
Significant effort has been spent attempting to unravel the causal relationship between histone post-translational modifications and fundamental DNA processes, including transcription, replication, and repair. However, less attention has been paid to understanding the reciprocal influence-that is, how DNA processes, in turn, shape the distribution and patterns of histone modifications and how these changes convey information, both temporally and spatially, from one process to another. Here, we review how histone modifications underpin the widespread bidirectional crosstalk between different DNA processes, which allow seemingly distinct phenomena to operate as a unified whole.
Collapse
Affiliation(s)
- Laura López-Hernández
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, 41092 Seville, Spain; Departamento de Genética, Universidad de Sevilla, 41012 Seville, Spain
| | - Patrick Toolan-Kerr
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, 41092 Seville, Spain; Departamento de Genética, Universidad de Sevilla, 41012 Seville, Spain
| | - Andrew J Bannister
- Gurdon Institute and Department of Pathology, University of Cambridge, Cambridge CB2 1QN, UK.
| | - Gonzalo Millán-Zambrano
- Centro Andaluz de Biología Molecular y Medicina Regenerativa-CABIMER, Universidad de Sevilla-CSIC-Universidad Pablo de Olavide, 41092 Seville, Spain; Departamento de Genética, Universidad de Sevilla, 41012 Seville, Spain.
| |
Collapse
|
30
|
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
|
31
|
Okumura H, Hayashi R, Unami D, Isono M, Yamauchi M, Otsuka K, Kato Y, Oike T, Uchihara Y, Shibata A. MeCP2 deficiency leads to the γH2AX nano foci expansion after ionizing radiation. DNA Repair (Amst) 2025; 145:103790. [PMID: 39647429 DOI: 10.1016/j.dnarep.2024.103790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/03/2024] [Accepted: 11/21/2024] [Indexed: 12/10/2024]
Abstract
DNA double-strand breaks (DSBs) trigger the recruitment of repair protein and promote signal transduction through posttranslational modifications such as phosphorylation. After DSB induction, ataxia telangiectasia mutated (ATM) phosphorylates H2AX on chromatin surrounds the mega-base pairs proximal to the DSBs. Advanced super-resolution microscopic technology has demonstrated the formation of γH2AX nano foci as a unit of nano domain comprised of multiple nucleosomes. The formation of γH2AX nano foci could be potentially affected by pre-existing chromatin structure prior to DSB induction; however, it remains unclear whether chromatin status around DSBs influences the formation of γH2AX nano foci. In this study, to investigate γH2AX nano foci formation in the context of chromatin relaxation, γH2AX nano foci were examined following the depletion of MeCP2, which is a factor promoting chromatin condensation. Remarkably, by using super-resolution imaging analysis, we found that the volume of γH2AX nano foci cluster in MeCP2-depleted cells was significantly greater than that in control cells, both 5 and 30 min after ionizing radiation (IR). Corresponding to the increased volume size, the number of γH2AX nano foci per cluster was greater than that in control cells, while the distance of each nano focus within foci clusters remained unchanged. These findings suggest that relaxed chromatin condition by MeCP2 depletion facilitates faster and more extensive γH2AX nano foci formation after IR. Collectively, our super-resolution analysis suggests that the chromatin status surrounding DSBs influences the expansion of γH2AX nano foci formation, thus, potentially influencing the DSB repair and signaling.
Collapse
Affiliation(s)
- Hikaru Okumura
- Division of Molecular Oncological Pharmacy, Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Ryota Hayashi
- Division of Molecular Oncological Pharmacy, Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Daiki Unami
- Division of Molecular Oncological Pharmacy, Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Mayu Isono
- Division of Molecular Oncological Pharmacy, Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Motohiro Yamauchi
- Hospital Campus Laboratory, Radioisotope Center, Central Institute of Radioisotope Science and Safety Management, Kyushu University, Fukuoka 812-8582, Japan
| | - Kensuke Otsuka
- Biology and Environmental Chemistry Division, Sustainable System Research Laboratory, Central Research Institute of Electric Power Industry, Chiba 270-1194, Japan
| | - Yu Kato
- Division of Molecular Oncological Pharmacy, Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Takahiro Oike
- Gunma University Heavy Ion Medical Center, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan; Department of Radiation Oncology, Gunma University Graduate School of Medicine, 3-39-22, Showa-machi, Maebashi, Gunma 371-8511, Japan
| | - Yuki Uchihara
- Division of Molecular Oncological Pharmacy, Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8512, Japan
| | - Atsushi Shibata
- Division of Molecular Oncological Pharmacy, Faculty of Pharmacy, Keio University, 1-5-30, Shibakoen, Minato-ku, Tokyo 105-8512, Japan.
| |
Collapse
|
32
|
Di Nardo M, Musio A. Cohesin - bridging the gap among gene transcription, genome stability, and human diseases. FEBS Lett 2025; 599:190-208. [PMID: 38852996 DOI: 10.1002/1873-3468.14949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/15/2024] [Accepted: 05/08/2024] [Indexed: 06/11/2024]
Abstract
The intricate landscape of cellular processes governing gene transcription, chromatin organization, and genome stability is a fascinating field of study. A key player in maintaining this delicate equilibrium is the cohesin complex, a molecular machine with multifaceted roles. This review presents an in-depth exploration of these intricate connections and their significant impact on various human diseases.
Collapse
Affiliation(s)
- Maddalena Di Nardo
- Institute for Biomedical Technologies (ITB), National Research Council (CNR), Pisa, Italy
| | - Antonio Musio
- Institute for Biomedical Technologies (ITB), National Research Council (CNR), Pisa, Italy
| |
Collapse
|
33
|
Phipps J, Toulouze M, Ducrot C, Costa R, Brocas C, Dubrana K. Cohesin complex oligomerization maintains end-tethering at DNA double-strand breaks. Nat Cell Biol 2025; 27:118-129. [PMID: 39482358 PMCID: PMC11735392 DOI: 10.1038/s41556-024-01552-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 10/01/2024] [Indexed: 11/03/2024]
Abstract
DNA double-strand breaks (DSBs) must be repaired to ensure genome stability. Crucially, DSB-ends must be kept together for timely repair. In Saccharomyces cerevisiae, two pathways mediate DSB end-tethering. One employs the Mre11-Rad50-Xrs2 (MRX) complex to physically bridge DSB-ends. Another requires the conversion of DSB-ends into single-strand DNA (ssDNA) by Exo1, but the bridging proteins are unknown. We uncover that cohesin, its loader and Smc5/6 act with Exo1 to tether DSB-ends. Remarkably, cohesin specifically impaired in oligomerization fails to tether DSB-ends, revealing a function for cohesin oligomerization. In addition to the known importance of sister chromatid cohesion, microscopy-based microfluidic experiments unveil a role for cohesin in repair by ensuring DSB end-tethering. Altogether, our findings demonstrate that oligomerization of cohesin prevents DSB end-separation and promotes DSB repair, revealing a previously undescribed mode of action and role for cohesin in safeguarding genome integrity.
Collapse
Affiliation(s)
- Jamie Phipps
- UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris Cité, INSERM, CEA, Fontenay-aux-Roses, France
- UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Mathias Toulouze
- UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris Cité, INSERM, CEA, Fontenay-aux-Roses, France
- UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Cécile Ducrot
- UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris Cité, INSERM, CEA, Fontenay-aux-Roses, France
- UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Rafaël Costa
- UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris Cité, INSERM, CEA, Fontenay-aux-Roses, France
- UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Clémentine Brocas
- UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris Cité, INSERM, CEA, Fontenay-aux-Roses, France
- UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France
| | - Karine Dubrana
- UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris Cité, INSERM, CEA, Fontenay-aux-Roses, France.
- UMR Stabilité Génétique Cellules Souches et Radiations, Université Paris-Saclay, INSERM, CEA, Fontenay-aux-Roses, France.
| |
Collapse
|
34
|
Lu WT, Zalmas LP, Bailey C, Black JRM, Martinez-Ruiz C, Pich O, Gimeno-Valiente F, Usaite I, Magness A, Thol K, Webber TA, Jiang M, Saunders RE, Liu YH, Biswas D, Ige EO, Aerne B, Grönroos E, Venkatesan S, Stavrou G, Karasaki T, Al Bakir M, Renshaw M, Xu H, Schneider-Luftman D, Sharma N, Tovini L, Jamal-Hanjani M, McClelland SE, Litchfield K, Birkbak NJ, Howell M, Tapon N, Fugger K, McGranahan N, Bartek J, Kanu N, Swanton C. TRACERx analysis identifies a role for FAT1 in regulating chromosomal instability and whole-genome doubling via Hippo signalling. Nat Cell Biol 2025; 27:154-168. [PMID: 39738653 PMCID: PMC11735399 DOI: 10.1038/s41556-024-01558-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/16/2024] [Indexed: 01/02/2025]
Abstract
Chromosomal instability (CIN) is common in solid tumours and fuels evolutionary adaptation and poor prognosis by increasing intratumour heterogeneity. Systematic characterization of driver events in the TRACERx non-small-cell lung cancer (NSCLC) cohort identified that genetic alterations in six genes, including FAT1, result in homologous recombination (HR) repair deficiencies and CIN. Using orthogonal genetic and experimental approaches, we demonstrate that FAT1 alterations are positively selected before genome doubling and associated with HR deficiency. FAT1 ablation causes persistent replication stress, an elevated mitotic failure rate, nuclear deformation and elevated structural CIN, including chromosome translocations and radial chromosomes. FAT1 loss contributes to whole-genome doubling (a form of numerical CIN) through the dysregulation of YAP1. Co-depletion of YAP1 partially rescues numerical CIN caused by FAT1 loss but does not relieve HR deficiencies, nor structural CIN. Importantly, overexpression of constitutively active YAP15SA is sufficient to induce numerical CIN. Taken together, we show that FAT1 loss in NSCLC attenuates HR and exacerbates CIN through two distinct downstream mechanisms, leading to increased tumour heterogeneity.
Collapse
Affiliation(s)
| | | | | | - James R M Black
- The Francis Crick Institute, London, UK
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Carlos Martinez-Ruiz
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | | | - Francisco Gimeno-Valiente
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Ieva Usaite
- The Francis Crick Institute, London, UK
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | | | - Kerstin Thol
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | | | | | | | - Yun-Hsin Liu
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Dhruva Biswas
- The Francis Crick Institute, London, UK
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | | | | | | | - Subramanian Venkatesan
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Georgia Stavrou
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Takahiro Karasaki
- The Francis Crick Institute, London, UK
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
- Department of Thoracic Surgery, Respiratory Center, Toranomon Hospital, Tokyo, Japan
| | - Maise Al Bakir
- The Francis Crick Institute, London, UK
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | | | - Hang Xu
- The Francis Crick Institute, London, UK
| | | | - Natasha Sharma
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Laura Tovini
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Mariam Jamal-Hanjani
- The Francis Crick Institute, London, UK
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | | | - Kevin Litchfield
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Nicolai J Birkbak
- Department of Molecular Medicine, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | | | | | - Kasper Fugger
- University College London Cancer Institute, London, UK
| | - Nicholas McGranahan
- CRUK Lung Cancer Centre of Excellence, London, UK
- University College London Cancer Institute, London, UK
| | - Jiri Bartek
- Danish Cancer Society Research Centre, Copenhagen, Denmark.
- Division of Genome Biology, Department of Medical Biochemistry and Biophysics, Science for Laboratory, Karolinska Institutet, Solna, Sweden.
| | - Nnennaya Kanu
- CRUK Lung Cancer Centre of Excellence, London, UK.
- University College London Cancer Institute, London, UK.
| | - Charles Swanton
- The Francis Crick Institute, London, UK.
- CRUK Lung Cancer Centre of Excellence, London, UK.
- University College London Cancer Institute, London, UK.
| |
Collapse
|
35
|
Liu Z, Xia Q, Wang C, Xu J, Tian K, Wang Z, Li L, Li Y, Shang H, Liu Q, Xin T. Biomimetic astrocyte cell membrane-fused nanovesicles for protecting neurovascular units in hypoxic ischemic encephalopathy. J Nanobiotechnology 2024; 22:766. [PMID: 39695691 DOI: 10.1186/s12951-024-03053-8] [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: 09/22/2024] [Accepted: 11/29/2024] [Indexed: 12/20/2024] Open
Abstract
Hypoxic ischemic encephalopathy (HIE) refers to neonatal hypoxic brain injury caused by severe asphyxia during the perinatal period. With a high incidence rate and poor prognosis, HIE accounts for 2.4% of the global disease burden, imposing a heavy burden on families and society. Current clinical treatment for HIE primarily focuses on symptomatic management and supportive care. Therefore, the developments of effective treatment strategies and new drug formulations are critical for improving the prognosis of HIE patients. In order to protect the compromised neurovascular units after HIE, we prepared membrane-fused nanovesicles for delivering rapamycin and si EDN1 (TRCAM@RAPA@si EDN1). Due to the homotypic targeting feature of membrane-fused nanovesicles, we employed astrocyte membranes as synthetic materials to improve the targeting of astrocytes in brain while reducing the clearance of nanovesicles by circulatory system. Additionally, the surface of cell membrane was modified with CXCR3 receptors, enhancing the homing of nanovesicles to infarcted lesions. Lipid vesicles were modified with TK and RVG29 transmembrane peptides, enabling responsive release of internal drugs and blood-brain barrier penetration. Internally loaded rapamycin could promote protective autophagy in astrocytes, improve cellular oxidative stress, while si EDN1 could reduce the expression level of endothelin gene, thereby reducing secondary damage to neurovascular units.
Collapse
Affiliation(s)
- Zihao Liu
- Department of Neurosurgery, Shandong Provincial Hospital, Shandong First Medical University, Jinan, 250021, China
| | - Qian Xia
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Chanyue Wang
- Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Jiacan Xu
- Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Kangqian Tian
- Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Zhihai Wang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Longji Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Yuchen Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Hao Shang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China
| | - Qian Liu
- Department of Histology and Embryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Tao Xin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, 250014, China.
| |
Collapse
|
36
|
Zhao H, Fu X, Ding X, Zhu L. HMGA1 Plays a Role in Counteracting DNA Damage Induced by BoHV-1 Productive Infection. Int J Mol Sci 2024; 25:13265. [PMID: 39769030 PMCID: PMC11675814 DOI: 10.3390/ijms252413265] [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: 11/04/2024] [Revised: 12/02/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Bovine herpesvirus 1 (BoHV-1) productive infection induces the generation of DNA double-strand breaks (DSBs), which may consequently lead to cell apoptosis. In response to DSBs, the DNA damage repair-related protein 53BP1 is recruited to the sites of DSBs, leading to the formation of 53BP1foci, which are crucial for the repair of damaged DNA and maintaining genomic integrity by repairing DSBs. In this study, we discovered that HMGA1 may play a significant role in counteracting virus infection-induced DNA damage, as the siRNA-mediated knockdown of HMGA1 protein expression or inhibition of HMGA1 activity by the chemical inhibitor Netropsin uniformly exacerbates the DNA damage induced by BoHV-1 productive infection. Interestingly, HMGA1 may positively regulate 53BP1 expression, and treatment with Netropsin reduced the accumulation of 53BP1 protein in the nucleus, suggesting that HMGA1 may potentially influence 53BP1's nuclear localization. However, this effect was reversed in the context of virus infection. Furthermore, Netropsin treatment restored the disruption of 53BP1 foci caused by virus infection, which is consistent with our findings that Netropsin enhances the nuclear accumulation of 53BP1. Collectively, these results indicate that HMGA1 is involved in countering DNA damage induced by virus infection. HMGA1 does indeed modulate the nuclear accumulation of 53BP1 protein, but this effect is counteracted by virus infection. Therefore, the biological function of HMGA1 in countering virus infection-induced DNA damage may be independent of its regulation of 53BP1 signaling. This is the first report suggesting that HMGA1 may be implicated in virus infection-induced DNA damage, although the precise mechanism remains to be elucidated. Furthermore, we report for the first time an interaction between HMGA1 and 53BP1, which is disrupted following virus infection.
Collapse
Affiliation(s)
| | | | - Xiuyan Ding
- College of Life Sciences, Hebei University, Baoding 071002, China; (H.Z.); (X.F.)
| | - Liqian Zhu
- College of Life Sciences, Hebei University, Baoding 071002, China; (H.Z.); (X.F.)
| |
Collapse
|
37
|
Xu Y, Li H, Ding Y, Zhang D, Liu W. How nanoscale plastics facilitate the evolution of antibiotic resistance? JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136157. [PMID: 39423639 DOI: 10.1016/j.jhazmat.2024.136157] [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: 06/18/2024] [Revised: 08/29/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
The plastic can enhance the proliferation of antibiotic resistance genes (ARGs), however, the effect of nanoplastics (NPLs) on bacterial antibiotic resistance has not been clearly explained. Herein, we explored the effects and mechanisms of NPLs of different sizes (200 and 600 nm) on the evolution of antibiotic resistance in Serratia marcescens. The results indicated that the evolution of bacterial antibiotic resistance could be promoted under NPLs exposure, which the median of relative abundance of ARGs was 1.11-1.46 times compared to the treatment without NPLs. Transcriptomic analysis showed that the larger size of NPLs mainly increased the permeability of bacterial cell membranes to efflux antibiotics, thus potentiating antibiotic resistance. While, the smaller NPLs is more than that, its enhanced the expression of antibiotic resistance by modulating bacterial metabolic processes. The genome SNP analysis found that the NPLs could cause the genetic mutation occurrence to alter the membrane transport and metabolism processes, and it increased at a size of 200 nm more than at 600 nm NPLs. Importantly, we demonstrated that the horizontal transfer of ARGs was augmented due to the NPLs could dock to bacterial surface proteins and pull their movement to contact with other bacteria (binding energy of membrane proteins: -8.54 kcal/mol), especially the smaller size. It suggests that NPLs will also contribute to the proliferation of ARGs in the environment. This study provides data for understanding the risk of bacterial resistance.
Collapse
Affiliation(s)
- Yan Xu
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin 300191, China
| | - Houyu Li
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin 300191, China
| | - Yinuo Ding
- Jilin Agriculture University, College of Life Science, Jilin 130118, China
| | - Dandan Zhang
- Agro-Environmental Protection Institute, Ministry of Agriculture and Rural Affairs, Tianjin 300191, China
| | - Wei Liu
- Department F.A. Forel for Environmental and Aquatic Sciences, Section of Earth and Environmental Sciences and Institute for Environmental Sciences, University of Geneva, Switzerland.
| |
Collapse
|
38
|
de Luca KL, Rullens PMJ, Karpinska MA, de Vries SS, Gacek-Matthews A, Pongor LS, Legube G, Jachowicz JW, Oudelaar AM, Kind J. Genome-wide profiling of DNA repair proteins in single cells. Nat Commun 2024; 15:9918. [PMID: 39572529 PMCID: PMC11582664 DOI: 10.1038/s41467-024-54159-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/31/2024] [Indexed: 11/24/2024] Open
Abstract
Accurate repair of DNA damage is critical for maintenance of genomic integrity and cellular viability. Because damage occurs non-uniformly across the genome, single-cell resolution is required for proper interrogation, but sensitive detection has remained challenging. Here, we present a comprehensive analysis of repair protein localization in single human cells using DamID and ChIC sequencing techniques. This study reports genome-wide binding profiles in response to DNA double-strand breaks induced by AsiSI, and explores variability in genomic damage locations and associated repair features in the context of spatial genome organization. By unbiasedly detecting repair factor localization, we find that repair proteins often occupy entire topologically associating domains, mimicking variability in chromatin loop anchoring. Moreover, we demonstrate the formation of multi-way chromatin hubs in response to DNA damage. Notably, larger hubs show increased coordination of repair protein binding, suggesting a preference for cooperative repair mechanisms. Together, our work offers insights into the heterogeneous processes underlying genome stability in single cells.
Collapse
Affiliation(s)
- Kim L de Luca
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) & University Medical Center Utrecht, Utrecht, the Netherlands.
- Oncode Institute, Utrecht, the Netherlands.
| | - Pim M J Rullens
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) & University Medical Center Utrecht, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, the Netherlands
| | - Magdalena A Karpinska
- Genome Organization and Regulation, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sandra S de Vries
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) & University Medical Center Utrecht, Utrecht, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Agnieszka Gacek-Matthews
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - Lőrinc S Pongor
- Cancer Genomics and Epigenetics Core Group, Hungarian Center of Excellence for Molecular Medicine (HCEMM), Szeged, Hungary
| | - Gaëlle Legube
- MCD, Centre de Biologie Intégrative (CBI), CNRS, Université de Toulouse, Toulouse, France
| | - Joanna W Jachowicz
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter (VBC), Vienna, Austria
| | - A Marieke Oudelaar
- Genome Organization and Regulation, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Jop Kind
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) & University Medical Center Utrecht, Utrecht, the Netherlands.
- Oncode Institute, Utrecht, the Netherlands.
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, the Netherlands.
| |
Collapse
|
39
|
Sharma N, Coticchio G, Borini A, Tachibana K, Nasmyth KA, Schuh M. Changes in DNA repair compartments and cohesin loss promote DNA damage accumulation in aged oocytes. Curr Biol 2024; 34:5131-5148.e6. [PMID: 39437784 DOI: 10.1016/j.cub.2024.09.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/20/2024] [Accepted: 09/16/2024] [Indexed: 10/25/2024]
Abstract
Oocyte loss, a natural process that accelerates as women approach their mid-30s, poses a significant challenge to female reproduction. Recent studies have identified DNA damage as a primary contributor to oocyte loss, but the mechanisms underlying DNA damage accumulation remain unclear. Here, we show that aged oocytes have a lower DNA repair capacity and reduced mobility of DNA damage sites compared to young oocytes. Incomplete DNA repair in aged oocytes results in defective chromosome integrity and partitioning, thereby compromising oocyte quality. We found that DNA repair proteins are arranged in spatially distinct DNA repair compartments that form during the late stages of oocyte growth, accompanied by changes in the activity of DNA repair pathways. We demonstrate alterations in these compartments with age, including substantial changes in the levels of key DNA repair proteins and a shift toward error-prone DNA repair pathways. In addition, we show that reduced cohesin levels make aged oocytes more vulnerable to persistent DNA damage and cause changes in DNA repair compartments. Our study links DNA damage accumulation in aged oocytes, a leading cause of oocyte loss, to cohesin deterioration and changes in the organization, abundance, and response of DNA repair machinery.
Collapse
Affiliation(s)
- Ninadini Sharma
- Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany
| | | | - Andrea Borini
- IVIRMA Global Research Alliance, 9.baby, Bologna 40125, Italy
| | - Kikuë Tachibana
- Department of Totipotency, Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried, Munich 82152, Germany
| | - Kim A Nasmyth
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Melina Schuh
- Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, Göttingen 37077, Germany.
| |
Collapse
|
40
|
Ryzhkova A, Maltseva E, Battulin N, Kabirova E. Loop Extrusion Machinery Impairments in Models and Disease. Cells 2024; 13:1896. [PMID: 39594644 PMCID: PMC11592926 DOI: 10.3390/cells13221896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
Structural maintenance of chromosomes (SMC) complexes play a crucial role in organizing the three-dimensional structure of chromatin, facilitating key processes such as gene regulation, DNA repair, and chromosome segregation. This review explores the molecular mechanisms and biological significance of SMC-mediated loop extrusion complexes, including cohesin, condensins, and SMC5/6, focusing on their structure, their dynamic function during the cell cycle, and their impact on chromatin architecture. We discuss the implications of impairments in loop extrusion machinery as observed in experimental models and human diseases. Mutations affecting these complexes are linked to various developmental disorders and cancer, highlighting their importance in genome stability and transcriptional regulation. Advances in model systems and genomic techniques have provided deeper insights into the pathological roles of SMC complex dysfunction, offering potential therapeutic avenues for associated diseases.
Collapse
Affiliation(s)
- Anastasiya Ryzhkova
- Institute of Cytology and Genetics, 630090 Novosibirsk, Russia; (A.R.); (N.B.)
| | - Ekaterina Maltseva
- Department of Genetics and Genetic Technologies, Sirius University of Science and Technology, 354340 Sirius, Russia;
| | - Nariman Battulin
- Institute of Cytology and Genetics, 630090 Novosibirsk, Russia; (A.R.); (N.B.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| | - Evelyn Kabirova
- Institute of Cytology and Genetics, 630090 Novosibirsk, Russia; (A.R.); (N.B.)
- Department of Natural Sciences, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
41
|
Dekker J, Mirny LA. The chromosome folding problem and how cells solve it. Cell 2024; 187:6424-6450. [PMID: 39547207 PMCID: PMC11569382 DOI: 10.1016/j.cell.2024.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024]
Abstract
Every cell must solve the problem of how to fold its genome. We describe how the folded state of chromosomes is the result of the combined activity of multiple conserved mechanisms. Homotypic affinity-driven interactions lead to spatial partitioning of active and inactive loci. Molecular motors fold chromosomes through loop extrusion. Topological features such as supercoiling and entanglements contribute to chromosome folding and its dynamics, and tethering loci to sub-nuclear structures adds additional constraints. Dramatically diverse chromosome conformations observed throughout the cell cycle and across the tree of life can be explained through differential regulation and implementation of these basic mechanisms. We propose that the first functions of chromosome folding are to mediate genome replication, compaction, and segregation and that mechanisms of folding have subsequently been co-opted for other roles, including long-range gene regulation, in different conditions, cell types, and species.
Collapse
Affiliation(s)
- Job Dekker
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| | - Leonid A Mirny
- Institute for Medical Engineering and Science and Department of Physics, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
42
|
Wu Y, Adeel M, Xia D, Sancar A, Li W. Nucleotide excision repair of aflatoxin-induced DNA damage within the 3D human genome organization. Nucleic Acids Res 2024; 52:11704-11719. [PMID: 39258558 PMCID: PMC11514448 DOI: 10.1093/nar/gkae755] [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: 02/18/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/12/2024] Open
Abstract
Aflatoxin B1 (AFB1), a potent mycotoxin, is one of the environmental risk factors that cause liver cancer. In the liver, the bioactivated AFB1 intercalates into the DNA double helix to form a bulky DNA adduct which will lead to mutation if left unrepaired. Here, we adapted the tXR-seq method to measure the nucleotide excision repair of AFB1-induced DNA adducts at single-nucleotide resolution on a genome-wide scale, and compared it with repair data obtained from conventional UV-damage XR-seq. Our results showed that transcription-coupled repair plays a major role in the damage removal process. We further analyzed the distribution of nucleotide excision repair sites for AFB1-induced DNA adducts within the 3D human genome organization. Our analysis revealed a heterogeneous AFB1-dG repair across four different organization levels, including chromosome territories, A/B compartments, TADs, and chromatin loops. We found that chromosomes positioned closer to the nuclear center and regions within A compartments have higher levels of nucleotide excision repair. Notably, we observed high repair activity around both TAD boundaries and loop anchors. These findings provide insights into the complex interplay between AFB1-induced DNA damage repair, transcription, and 3D genome organization, shedding light on the mechanisms underlying AFB1-induced mutagenesis.
Collapse
Affiliation(s)
- Yiran Wu
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA 30602, USA
| | - Muhammad Muzammal Adeel
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA 30602, USA
| | - Dian Xia
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA 30602, USA
| | - Aziz Sancar
- Department of Biochemistry and Biophysics, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Wentao Li
- Department of Environmental Health Science, College of Public Health, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
43
|
Cohen S, Cheradame L, Pratt KJB, Collins S, Barillas A, Carlson A, Ramani V, Legube G, Villeda SA, Mullins RD, Schwer B. Endogenous neuronal DNA double-strand breaks are not sufficient to drive brain aging and neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619740. [PMID: 39484383 PMCID: PMC11526996 DOI: 10.1101/2024.10.22.619740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Loss of genomic information due to the accumulation of somatic DNA damage has been implicated in aging and neurodegeneration 1-3 . Somatic mutations in human neurons increase with age 4 , but it is unclear whether this is a cause or a consequence of brain aging. Here, we clarify the role of endogenous, neuronal DNA double-strand breaks (DSBs) in brain aging and neurodegeneration by generating mice with post-developmental inactivation of the classical non-homologous end-joining (C-NHEJ) core factor Xrcc4 in forebrain neurons. Xrcc4 is critical for the ligation step of C-NHEJ and has no known function outside of DSB repair 5,6 . We find that, unlike their wild-type counterparts, C-NHEJ-deficient neurons accumulate high levels of DSB foci with age, indicating that neurons undergo frequent DSBs that are typically efficiently repaired by C-NHEJ across their lifespan. Genome-wide mapping reveals that endogenous neuronal DSBs preferentially occur in promoter regions and other genic features. Analysis of 3-D genome organization shows intra-chromosomal clustering and loop extrusion of neuronal DSB regions. Strikingly, however, DSB accumulation caused by loss of C-NHEJ induces only minor epigenetic alterations and does not significantly affect gene expression, 3-D genome organization, or mutational outcomes at neuronal DSBs. Despite extensive aging-associated accumulation of neuronal DSBs, mice with neuronal Xrcc4 inactivation do not show neurodegeneration, neuroinflammation, reduced lifespan, or impaired memory and learning behavior. We conclude that the formation of spontaneous neuronal DSBs caused by normal cellular processes is insufficient to cause brain aging and neurodegeneration, even in the absence of C-NHEJ, the principal neuronal DSB repair pathway.
Collapse
|
44
|
Qu SJ, Zhu SY, Wang EY, Yan XL, Cao RF, Li HT, Jiang ZL. Environmental high temperature affects pre-implantation embryo development by impairing the DNA repair ability. J Therm Biol 2024; 125:103968. [PMID: 39312816 DOI: 10.1016/j.jtherbio.2024.103968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 07/25/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024]
Abstract
Environmental high temperature poses a significant threat to human health, however, limited information is available for understanding the relationship between the hot weather and infertility. This study aims to assess the adverse effect of the hot weather to early embryonic cells. Our results indicated that environmental high temperature exposure could cause the decline of early embryo quality and implantation ability. In detail, it led to early embryonic development retardation, embryo degeneration rate increased, the rate of blastocyst and hatching decreased, and reduced the number of implants. And the finding also the impairment of environmental high temperature on early embryonic cells may be due to oxidative damage of DNA caused by ROS, while BER repair ability is decreased, failing to repair oxidative damage of DNA in time, resulting in a large number of early embryonic apoptosis. The work underscored that pregnant women should stay away from high-temperature environments.
Collapse
Affiliation(s)
- Si-Jing Qu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China
| | - Shi-Yong Zhu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China
| | - En-Yan Wang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China
| | - Xin-Lei Yan
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China
| | - Rong-Feng Cao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China
| | - Hua-Tao Li
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China
| | - Zhong-Ling Jiang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
45
|
Smirnov AV, Ryzhkova AS, Yunusova AM. Effects of the auxin-dependent degradation of the cohesin and condensin complexes on the repair of distant DNA double-strand breaks in mouse embryonic stem cells. Vavilovskii Zhurnal Genet Selektsii 2024; 28:583-591. [PMID: 39440310 PMCID: PMC11491485 DOI: 10.18699/vjgb-24-65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/23/2024] [Accepted: 07/22/2024] [Indexed: 10/25/2024] Open
Abstract
The SMC protein family, including cohesin and condensin I/II, plays a pivotal role in maintaining the topological structure of chromosomes and influences many cellular processes, notably the repair of double-stranded DNA breaks (DSBs). The cohesin complex impacts DSB repair by spreading γH2AX signal and containing DNA ends in close proximity by loop extrusion. Cohesin supports DNA stability by sister chromatid cohesion during the S/G2 phase, which limits DNA end mobility. Cohesin knockdown was recently shown to stimulate frequencies of genomic deletions produced by distant paired DSBs, but does not affect DNA repair of a single or close DSBs. We examined how auxin-inducible protein degradation of Rad21 (cohesin) or Smc2 (condensins I+II) changes the frequencies of rearrangements between paired distant DSBs in mouse embryonic stem cells (mESCs). We used Cas9 RNP nucleofection to generate deletions and inversions with high efficiency without additional selection. We determined optimal Neon settings and deletion appearance timings. Two strategies for auxin addition were tested (4 independent experiments in total). We examined deletion/inversion frequencies for two regions spanning 3.5 and 3.9 kbp in size. Contrary to expectations, in our setting, Rad21 depletion did not increase deletion/inversion frequencies, not even for the region with an active Ctcf boundary. We actually observed a 12 % decrease in deletions (but not inversions). At the same time, double condensin depletion (Smc2 degron line) demonstrated high biological variability between experiments, complicating the analysis, and requires additional examination in the future. TIDE analysis revealed that editing frequency was consistent (30-50 %) for most experiments with a minor decrease after auxin addition. In the end, we discuss the Neon/ddPCR method for deletion generation and detection in mESCs.
Collapse
Affiliation(s)
- A V Smirnov
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A S Ryzhkova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A M Yunusova
- Institute of Cytology and Genetics of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
46
|
Downs JA, Gasser SM. Chromatin remodeling and spatial concerns in DNA double-strand break repair. Curr Opin Cell Biol 2024; 90:102405. [PMID: 39083951 DOI: 10.1016/j.ceb.2024.102405] [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: 04/08/2024] [Revised: 07/07/2024] [Accepted: 07/11/2024] [Indexed: 08/02/2024]
Abstract
The substrate for the repair of DNA damage in living cells is not DNA but chromatin. Chromatin bears a range of modifications, which in turn bind ligands that compact or open chromatin structure, and determine its spatial organization within the nucleus. In some cases, RNA in the form of RNA:DNA hybrids or R-loops modulates DNA accessibility. Each of these parameters can favor particular pathways of repair. Chromatin or nucleosome remodelers are key regulators of chromatin structure, and a number of remodeling complexes are implicated in DNA repair. We cover novel insights into the impact of chromatin structure, nuclear organization, R-loop formation, nuclear actin, and nucleosome remodelers in DNA double-strand break repair, focusing on factors that alter repair functional upon ablation.
Collapse
Affiliation(s)
- Jessica A Downs
- Epigenetics and Genome Stability Team, The Institute of Cancer Research, 237 Fulham Road, London, SW3 6JB, UK
| | - Susan M Gasser
- ISREC Foundation, and University of Lausanne, Agora Cancer Research Center, Rue du Bugnon 25a, 1005 Lausanne, Switzerland.
| |
Collapse
|
47
|
Chang LH, Noordermeer D. Permeable TAD boundaries and their impact on genome-associated functions. Bioessays 2024; 46:e2400137. [PMID: 39093600 DOI: 10.1002/bies.202400137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/12/2024] [Accepted: 07/17/2024] [Indexed: 08/04/2024]
Abstract
TAD boundaries are genomic elements that separate biological processes in neighboring domains by blocking DNA loops that are formed through Cohesin-mediated loop extrusion. Most TAD boundaries consist of arrays of binding sites for the CTCF protein, whose interaction with the Cohesin complex blocks loop extrusion. TAD boundaries are not fully impermeable though and allow a limited amount of inter-TAD loop formation. Based on the reanalysis of Nano-C data, a multicontact Chromosome Conformation Capture assay, we propose a model whereby clustered CTCF binding sites promote the successive stalling of Cohesin and subsequent dissociation from the chromatin. A fraction of Cohesin nonetheless achieves boundary read-through. Due to a constant rate of Cohesin dissociation elsewhere in the genome, the maximum length of inter-TAD loops is restricted though. We speculate that the DNA-encoded organization of stalling sites regulates TAD boundary permeability and discuss implications for enhancer-promoter loop formation and other genomic processes.
Collapse
Affiliation(s)
- Li-Hsin Chang
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Blood and Transplant Research Unit in Precision Cellular Therapeutics, National Institute of Health Research, Oxford, UK
| | - Daan Noordermeer
- CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
48
|
Kim J, Wang H, Ercan S. Cohesin organizes 3D DNA contacts surrounding active enhancers in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.18.558239. [PMID: 37786717 PMCID: PMC10541618 DOI: 10.1101/2023.09.18.558239] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
In mammals, cohesin and CTCF organize the 3D genome into topologically associated domains (TADs) to regulate communication between cis-regulatory elements. Many organisms, including S. cerevisiae, C. elegans, and A. thaliana contain cohesin but lack CTCF. Here, we used C. elegans to investigate the function of cohesin in 3D genome organization in the absence of CTCF. Using Hi-C data, we observe cohesin-dependent features called "fountains", which are also reported in zebrafish and mice. These are population average reflections of DNA loops originating from distinct genomic regions and are ~20-40 kb in C. elegans. Hi-C analysis upon cohesin and WAPL depletion support the idea that cohesin is preferentially loaded at NIPBL occupied sites and loop extrudes in an effectively two-sided manner. ChIP-seq analyses show that cohesin translocation along the fountain trajectory depends on a fully intact complex and is extended upon WAPL-1 depletion. Hi-C contact patterns at individual fountains suggest that cohesin processivity is unequal on each side, possibly due to collision with cohesin loaded from surrounding sites. The putative cohesin loading sites are closest to active enhancers and fountain strength is associated with transcription. Compared to mammals, average processivity of C. elegans cohesin is ~10-fold shorter and NIPBL binding does not depend on cohesin. We propose that preferential loading and loop extrusion by cohesin is an evolutionarily conserved mechanism that regulates the 3D interactions of enhancers in animal genomes.
Collapse
Affiliation(s)
- Jun Kim
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Haoyu Wang
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Sevinç Ercan
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| |
Collapse
|
49
|
Danovski G, Panova G, Keister B, Georgiev G, Atemin A, Uzunova S, Stamatov R, Kanev PB, Aleksandrov R, Blagoev KB, Stoynov SS. Diffusion of activated ATM explains γH2AX and MDC1 spread beyond the DNA damage site. iScience 2024; 27:110826. [PMID: 39310780 PMCID: PMC11416226 DOI: 10.1016/j.isci.2024.110826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/12/2024] [Accepted: 08/22/2024] [Indexed: 09/25/2024] Open
Abstract
During DNA repair, ATM-induced H2AX histone phosphorylation and MDC1 recruitment spread megabases beyond the damage site. While loop extrusion has been suggested to drive this spread, the underlying mechanism remains unclear. Herein, we provide two lines of evidence that loop extrusion is not the only driver of damage-induced γH2AX spread. First, cohesin loader NIPBL and cohesin subunit RAD21 accumulate considerably later than the phosphorylation of H2AX and MDC1 recruitment at micro-IR-induced damage. Second, auxin-induced RAD21 depletion does not affect γH2AX/MDC1 spread following micro-irradiation or DSB induction by zeocin. To determine if diffusion of activated ATM could account for the observed behavior, we measured the exchange rate and diffusion constants of ATM and MDC1 within damaged and unperturbed chromatin. Using these measurements, we introduced a quantitative model in which the freely diffusing activated ATM phosphorylates H2AX. This model faithfully describes the dynamics of ATM and subsequent γH2AX/MDC1 spread at complex DNA lesions.
Collapse
Affiliation(s)
- Georgi Danovski
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str, 1113 Sofia, Bulgaria
| | | | | | - Georgi Georgiev
- Faculty of Mathematics and Informatics, Sofia University, St. Kliment Ohridski, 5 James Bourchier Boulevard, 1164 Sofia, Bulgaria
| | - Aleksandar Atemin
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str, 1113 Sofia, Bulgaria
| | - Sonya Uzunova
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str, 1113 Sofia, Bulgaria
| | - Rumen Stamatov
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str, 1113 Sofia, Bulgaria
| | - Petar-Bogomil Kanev
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str, 1113 Sofia, Bulgaria
| | - Radoslav Aleksandrov
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str, 1113 Sofia, Bulgaria
| | - Krastan B. Blagoev
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str, 1113 Sofia, Bulgaria
- National Science Foundation, Alexandria, VA 22230, USA
- Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
- Institut Curie, PSL Research University, Sorbonne Université, CNRS UMR3664, Paris, France
| | - Stoyno S. Stoynov
- Institute of Molecular Biology, Bulgarian Academy of Sciences, 21, G. Bontchev Str, 1113 Sofia, Bulgaria
| |
Collapse
|
50
|
Yao W, Hu X, Wang X. Crossing epigenetic frontiers: the intersection of novel histone modifications and diseases. Signal Transduct Target Ther 2024; 9:232. [PMID: 39278916 PMCID: PMC11403012 DOI: 10.1038/s41392-024-01918-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/11/2024] [Accepted: 06/30/2024] [Indexed: 09/18/2024] Open
Abstract
Histone post-translational modifications (HPTMs), as one of the core mechanisms of epigenetic regulation, are garnering increasing attention due to their close association with the onset and progression of diseases and their potential as targeted therapeutic agents. Advances in high-throughput molecular tools and the abundance of bioinformatics data have led to the discovery of novel HPTMs which similarly affect gene expression, metabolism, and chromatin structure. Furthermore, a growing body of research has demonstrated that novel histone modifications also play crucial roles in the development and progression of various diseases, including various cancers, cardiovascular diseases, infectious diseases, psychiatric disorders, and reproductive system diseases. This review defines nine novel histone modifications: lactylation, citrullination, crotonylation, succinylation, SUMOylation, propionylation, butyrylation, 2-hydroxyisobutyrylation, and 2-hydroxybutyrylation. It comprehensively introduces the modification processes of these nine novel HPTMs, their roles in transcription, replication, DNA repair and recombination, metabolism, and chromatin structure, as well as their involvement in promoting the occurrence and development of various diseases and their clinical applications as therapeutic targets and potential biomarkers. Moreover, this review provides a detailed overview of novel HPTM inhibitors targeting various targets and their emerging strategies in the treatment of multiple diseases while offering insights into their future development prospects and challenges. Additionally, we briefly introduce novel epigenetic research techniques and their applications in the field of novel HPTM research.
Collapse
Affiliation(s)
- Weiyi Yao
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Xinting Hu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, 250021, China.
- Taishan Scholars Program of Shandong Province, Jinan, Shandong, 250021, China.
| |
Collapse
|