1
|
Hernández-Reyes Y, Fonseca-Rodríguez C, Freire R, Smits VAJ. DDX37 and DDX50 Maintain Genome Stability by Preventing Transcription-dependent R-loop Formation. J Mol Biol 2025; 437:169061. [PMID: 40043837 DOI: 10.1016/j.jmb.2025.169061] [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: 12/12/2024] [Revised: 02/19/2025] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
R-loops consist of an RNA-DNA hybrid and a displaced single-stranded DNA strand that play a central role in several biological processes. However, as the presence of aberrant R-loops forms a significant threat to genome stability, R-loop formation and resolution is strictly controlled by RNAse H and helicases. In a screening for RNA helicases, previously described as RNA-DNA hybrid interactors, that control genome integrity, we identified for the first time DDX37 and DDX50. Depletion of DDX37 and DDX50 promotes DNA damage, as demonstrated by H2AX phosphorylation and increased comet tail length. In addition, knock down of these RNA helicases decreases the DNA replication track length and leads to RPA focus formation, results that are indicative of replication stress. Downregulation of DDX37 and DDX50 triggers an increase in RNA-DNA hybrids, that can be reverted by the overexpression of RNase H1. Interestingly, inhibition of transcription prevented the increased RNA-DNA hybrid formation and DNA damage upon DDX37 or DDX50 depletion. Together these results demonstrate that DDX37 and DDX50 are important for resolving RNA-DNA hybrids appearing during transcription and thereby preventing DNA damage by replication stress.
Collapse
Affiliation(s)
- Yeray Hernández-Reyes
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Investigación Sanitaria de Canarias (IISC), Santa Cruz de Tenerife, Spain; Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain; Escuela de Doctorado y Estudio de Postgrado, Universidad de la Laguna, Santa Cruz de Tenerife, Spain
| | - Cintia Fonseca-Rodríguez
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Investigación Sanitaria de Canarias (IISC), Santa Cruz de Tenerife, Spain; Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain; Escuela de Doctorado y Estudio de Postgrado, Universidad de la Laguna, Santa Cruz de Tenerife, Spain
| | - Raimundo Freire
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Investigación Sanitaria de Canarias (IISC), Santa Cruz de Tenerife, Spain; Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain; Universidad Fernando Pessoa Canarias, Santa María de Guía, Las Palmas, Spain
| | - Veronique A J Smits
- Unidad de Investigación, Hospital Universitario de Canarias, Instituto de Investigación Sanitaria de Canarias (IISC), Santa Cruz de Tenerife, Spain; Instituto de Tecnologías Biomédicas, Universidad de La Laguna, Santa Cruz de Tenerife, Spain; Universidad Fernando Pessoa Canarias, Santa María de Guía, Las Palmas, Spain.
| |
Collapse
|
2
|
Zhu M, Xu R, Yuan J, Wang J, Ren X, Cong T, You Y, Ju A, Xu L, Wang H, Zheng P, Tao H, Lin C, Yu H, Du J, Lin X, Xie W, Li Y, Lan X. Tracking-seq reveals the heterogeneity of off-target effects in CRISPR-Cas9-mediated genome editing. Nat Biotechnol 2025; 43:799-810. [PMID: 38956324 DOI: 10.1038/s41587-024-02307-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: 08/12/2023] [Accepted: 06/06/2024] [Indexed: 07/04/2024]
Abstract
The continued development of novel genome editors calls for a universal method to analyze their off-target effects. Here we describe a versatile method, called Tracking-seq, for in situ identification of off-target effects that is broadly applicable to common genome-editing tools, including Cas9, base editors and prime editors. Through tracking replication protein A (RPA)-bound single-stranded DNA followed by strand-specific library construction, Tracking-seq requires a low cell input and is suitable for in vitro, ex vivo and in vivo genome editing, providing a sensitive and practical genome-wide approach for off-target detection in various scenarios. We show, using the same guide RNA, that Tracking-seq detects heterogeneity in off-target effects between different editor modalities and between different cell types, underscoring the necessity of direct measurement in the original system.
Collapse
Affiliation(s)
- Ming Zhu
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China.
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China.
- MOE Key Laboratory of Bioinformatics, State Key Laboratory of Molecular Oncology, Tsinghua University, Beijing, China.
| | - Runda Xu
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- MOE Key Laboratory of Bioinformatics, State Key Laboratory of Molecular Oncology, Tsinghua University, Beijing, China
| | - Junsong Yuan
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
- IDG-McGovern Institute for Brain Research, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Jiacheng Wang
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
- MOE Key Laboratory of Bioinformatics, State Key Laboratory of Molecular Oncology, Tsinghua University, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Xiaoyu Ren
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
- IDG-McGovern Institute for Brain Research, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Tingting Cong
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
- IDG-McGovern Institute for Brain Research, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Yaxian You
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- MOE Key Laboratory of Bioinformatics, State Key Laboratory of Molecular Oncology, Tsinghua University, Beijing, China
| | - Anji Ju
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- MOE Key Laboratory of Bioinformatics, State Key Laboratory of Molecular Oncology, Tsinghua University, Beijing, China
| | - Longchen Xu
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Huimin Wang
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Peiyuan Zheng
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
- IDG-McGovern Institute for Brain Research, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Huiying Tao
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Department of Urology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Chunhua Lin
- Department of Urology, Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Honghao Yu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
- Key Laboratory of Medical Biotechnology and Translational Medicine, Guilin Medical University, Guilin, China
| | - Juanjuan Du
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
- IDG-McGovern Institute for Brain Research, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Xin Lin
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China
| | - Wei Xie
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Yinqing Li
- MOE Key Laboratory of Bioinformatics, State Key Laboratory of Molecular Oncology, Tsinghua University, Beijing, China.
- IDG-McGovern Institute for Brain Research, Center for Synthetic and Systems Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
| | - Xun Lan
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, Beijing, China.
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China.
- MOE Key Laboratory of Bioinformatics, State Key Laboratory of Molecular Oncology, Tsinghua University, Beijing, China.
| |
Collapse
|
3
|
Chen Y, Wang L, Zhou Q, Wei W, Wei H, Ma Y, Han T, Ma S, Huang X, Zhang M, Gao F, Liu C, Li W. Dynamic R-loops at centromeres ensure chromosome alignment during oocyte meiotic divisions in mice. Sci Bull (Beijing) 2025; 70:1311-1327. [PMID: 39984387 DOI: 10.1016/j.scib.2025.02.009] [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: 09/04/2024] [Revised: 01/12/2025] [Accepted: 01/24/2025] [Indexed: 02/23/2025]
Abstract
R-loops play various roles in many physiological processes, however, their role in meiotic division remains largely unknown. Here we show that R-loops and their regulator RNase H1 are present at centromeres during oocyte meiotic divisions. Proper centromeric R-loops are essential to ensure chromosome alignment in oocytes during metaphase I (MI). Remarkably, both Rnaseh1 knockout and overexpression in oocytes lead to severe spindle assembly defects and chromosome misalignment due to dysregulation of R-loops at centromeres. Furthermore, we find that replication protein A (RPA) is recruited to centromeric R-loops, facilitating the deposition of ataxia telangiectasia-mutated and Rad3-related (ATR) kinase at centromeres by interacting with the ATR-interaction protein (ATRIP). The ATR kinase deposition triggers the activity of CHK1, stimulating the phosphorylation of Aurora B to finally promote proper spindle assembly and chromosome alignment at the equatorial plate. Most importantly, the application of ATR, CHK1, and Aurora B inhibitors could efficiently rescue the defects in spindle assembly and chromosome alignment due to RNase H1 deficiency in oocytes. Overall, our findings uncover a critical role of R-loops during mouse oocyte meiotic divisions, suggesting that dysregulation of R-loops may be associated with female infertility. Additionally, ATR, CHK1, and Aurora B inhibitors may potentially be used to treat some infertile patients.
Collapse
Affiliation(s)
- Yinghong Chen
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China; State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Liying Wang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Qiuxing Zhou
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Wei Wei
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Huafang Wei
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China; Sino-French Hoffmann Institute, School of Basic Medical Science, Guangzhou Medical University, Guangzhou 511436, China
| | - Yanjie Ma
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China; State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tingting Han
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Shuang Ma
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China; Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Xiaoming Huang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Meijia Zhang
- The Innovation Centre of Ministry of Education for Development and Diseases, the Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou 510006, China.
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Chao Liu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China; State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Wei Li
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China; State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
4
|
Cheng B, Ding Z, Hong Y, Wang Y, Zhou Y, Chen J, Peng X, Zeng C. Research progress in DNA damage response (DDR)-targeting modulators: From hits to clinical candidates. Eur J Med Chem 2025; 287:117347. [PMID: 39908794 DOI: 10.1016/j.ejmech.2025.117347] [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: 12/25/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/07/2025]
Abstract
In recent years, synthetic lethality has been regarded as a sound example of cancer treatment. Identifying a growing number of synthetic lethality targets has led to a substantial broadening of the application of synthetic lethality, well beyond the PAPR inhibitors employed for treating tumors with BRCA1/2 deficiencies. Especially, molecular targets within the DDR have furnished inhibitor sources and have rapidly advanced to clinical trials. In this review, we summarize the DDR-associated synthetic lethality targets such as WRN, USP1, PARP, ATR, DNA-PK, PRMT5, POLQ, and WEE1. These targets allow for the development of targeted modulators like inhibitors and degraders. Additionally, we emphasize the rational design, advantages, and potential limitations. Furthermore, we outline the promising future of DDR-targeted drug development.
Collapse
Affiliation(s)
- Binbin Cheng
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University, Huangshi, Hubei, 435003, China; Department of Cardiology, Central Laboratory of Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang, 323000, China
| | - Zongbao Ding
- Department of Bioengineering, Zhuhai Campus of Zunyi Medical University, Zhuhai, 519041, China
| | - Yimeng Hong
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University, Huangshi, Hubei, 435003, China
| | - Yaping Wang
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University, Huangshi, Hubei, 435003, China
| | - Yingxing Zhou
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University, Huangshi, Hubei, 435003, China; Huangshi Key Laboratory of Molecular Diagnosis and Individualized Treatment, Huangshi Love&health Hospital Affiliated of Hubei Polytechnic University, China.
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China.
| | - Xiaopeng Peng
- College of Pharmacy, Gannan Medical University, Ganzhou, 314000, China.
| | - Chunlai Zeng
- Department of Cardiology, Central Laboratory of Lishui Hospital of Wenzhou Medical University, The First Affiliated Hospital of Lishui University, Lishui People's Hospital, Lishui, Zhejiang, 323000, China.
| |
Collapse
|
5
|
Liu Q, Liu Y, Yi C, Gao Z, Zhang Z, Zhu C, Birchler JA, Han F. Genome assembly of the maize B chromosome provides insight into its epigenetic characteristics and effects on the host genome. Genome Biol 2025; 26:47. [PMID: 40050975 PMCID: PMC11887103 DOI: 10.1186/s13059-025-03517-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/25/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND B chromosomes contribute to the genetic variation in numerous eukaryotes. Yet their genetic and epigenetic characteristics, as well as their effects on the host genome remain poorly understood. RESULTS Here, we present a comprehensive genome assembly of diploid maize B73 with two copies of B chromosomes using long-read sequencing. We annotate a total of 1124 high-confidence protein-coding genes and 119,579,190 bp repeat elements representing 88.55% of the B chromosome assembly. Using CENH3 ChIP-seq data, we accurately determined the position of the B chromosome centromere, which features a unique monomer-composed satellite array distinct from that found on the chromosome arms. Our research provides detailed genetic and epigenetic maps of the B chromosome, shedding light on its molecular landscape, including DNA sequence composition, DNA methylation patterns, histone modifications, and R-loop distributions across various chromatin regions. Consistent with the cytological morphology of the B chromosome, the less condensed euchromatin regions displayed high levels of H3K4me3, H3K9ac, gene expression, and dense R-loop distributions. DNA methylation on the B chromosome was primarily observed at CG sites. The centromeric region is notably enriched with H3K4me3 and H3K9ac histone modifications and has lower CHG methylation compared to the pericentromeric regions. Moreover, our findings reveal that B chromosome accumulation affects R-loop formation on A chromosomes, and exerts tissue-specific influences on A chromosome gene expression. CONCLUSIONS The accurate assembly and detailed epigenetic maps of the maize B chromosome will help understand the drive mechanism, reveal its conflict with the host genome, and accelerate the construction of artificial chromosomes.
Collapse
Affiliation(s)
- Qian Liu
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yang Liu
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Congyang Yi
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhi Gao
- Division of Biological Science, University of Missouri-Columbia, 311 Tucker Hall, Columbia, MO, 65211-7400, USA
| | - Zeyan Zhang
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Congle Zhu
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - James A Birchler
- Division of Biological Science, University of Missouri-Columbia, 311 Tucker Hall, Columbia, MO, 65211-7400, USA.
| | - Fangpu Han
- State Key Laboratory of Seed Innovation, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
6
|
Li D, Shao F, Li X, Yu Q, Wu R, Wang J, Wang Z, Wusiman D, Ye L, Guo Y, Tuo Z, Wei W, Yoo KH, Cho WC, Feng D. Advancements and challenges of R-loops in cancers: Biological insights and future directions. Cancer Lett 2025; 610:217359. [PMID: 39613219 DOI: 10.1016/j.canlet.2024.217359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 12/01/2024]
Abstract
R-loops involve in various biological processes under human normal physiological conditions. Disruption of R-loops can lead to disease onset and affect the progression of illnesses, particularly in cancers. Herein, we summarized and discussed the regulative networks, phenotypes and future directions of R-loops in cancers. In this review, we highlighted the following insights: (1) R-loops significantly influence cancer development, progression and treatment efficiency by regulating key genes, such as PARPs, BRCA1/2, sex hormone receptors, DHX9, and TOP1. (2) Currently, the ATM, ATR, cGAS/STING, and noncanonical pathways are the main pathways that involve in the regulatory network of R-loops in cancer. (3) Cancer biology can be modulated by R-loops-regulated phenotypes, including RNA methylation, DNA and histone methylation, oxidative stress, immune and inflammation regulation, and senescence. (4) Regulation of R-loops induces kinds of drug resistance in various cancers, suggesting that targeting R-loops maybe a promising way to overcome treatment resistance. (5) The role of R-loops in tumorigenesis remains controversial, and senescence may be a crucial research direction to unravel the mechanism of R-loop-induced tumorigenesis. Looking forward, further studies are needed to elucidate the specific mechanisms of R-loops in cancer, laying the groundwork for preclinical and clinical research.
Collapse
Affiliation(s)
- Dengxiong Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Fanglin Shao
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xinrui Li
- Department of Rehabilitation, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qingxin Yu
- Department of Pathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo City, Zhejiang Province, 315211, China
| | - Ruicheng Wu
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jie Wang
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhipeng Wang
- Department of Urology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Dilinaer Wusiman
- Department of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA
| | - Luxia Ye
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Yiqing Guo
- Department of Public Research Platform, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, China
| | - Zhouting Tuo
- Department of Urological Surgery, Daping Hospital, Army Medical Center of PLA, Army Medical University, Chongqing, China
| | - Wuran Wei
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Koo Han Yoo
- Department of Urology, Kyung Hee University, South Korea.
| | - William C Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong Special Administrative Region of China.
| | - Dechao Feng
- Division of Surgery & Interventional Science, University College London, London, W1W 7TS, UK.
| |
Collapse
|
7
|
Grillo G, Boyarchuk E, Mihic S, Ivkovic I, Bertrand M, Jouneau A, Dahlet T, Dumas M, Weber M, Velasco G, Francastel C. ZBTB24 is a conserved multifaceted transcription factor at genes and centromeres that governs the DNA methylation state and expression of satellite repeats. Hum Mol Genet 2025; 34:161-177. [PMID: 39562305 PMCID: PMC11780882 DOI: 10.1093/hmg/ddae163] [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: 06/24/2024] [Revised: 10/07/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Since its discovery as a causative gene of the Immunodeficiency with Centromeric instability and Facial anomalies syndrome, ZBTB24 has emerged as a key player in DNA methylation, immunity and development. By extensively analyzing ZBTB24 genomic functions in ICF-relevant mouse and human cellular models, we document here its multiple facets as a transcription factor, with key roles in immune response-related genes expression and also in early embryonic development. Using a constitutive Zbtb24 ICF-like mutant and an auxin-inducible degron system in mouse embryonic stem cells, we showed that ZBTB24 is recruited to centromeric satellite DNA where it is required to establish and maintain the correct DNA methylation patterns through the recruitment of DNMT3B. The ability of ZBTB24 to occupy centromeric satellite DNA is conserved in human cells. Together, our results unveiled an essential and underappreciated role for ZBTB24 at mouse and human centromeric satellite repeat arrays by controlling their DNA methylation and transcription status.
Collapse
Affiliation(s)
- Giacomo Grillo
- UMR7216 Epigénétique et Destin Cellulaire, CNRS, Université de Paris Cité, Epigenetics and Cell Fate, Lamarck building, 35 rue Hélène Brion, Paris F-75013, France
| | - Ekaterina Boyarchuk
- UMR7216 Epigénétique et Destin Cellulaire, CNRS, Université de Paris Cité, Epigenetics and Cell Fate, Lamarck building, 35 rue Hélène Brion, Paris F-75013, France
- UMR7216, Genome engineering in epigenetics platform (GENIE), Lamarck building, 35 rue Hélène Brion, Paris F-75013, France
| | - Seed Mihic
- UMR7216 Epigénétique et Destin Cellulaire, CNRS, Université de Paris Cité, Epigenetics and Cell Fate, Lamarck building, 35 rue Hélène Brion, Paris F-75013, France
| | - Ivana Ivkovic
- UMR7216 Epigénétique et Destin Cellulaire, CNRS, Université de Paris Cité, Epigenetics and Cell Fate, Lamarck building, 35 rue Hélène Brion, Paris F-75013, France
| | - Mathilde Bertrand
- Bioinformatics and Biostatistics Core Facility, iCONICS, Institut du Cerveau (ICM), Sorbonne Université, INSERM, CNRS, Hôpital Pitié-Salpêtrière, 47 bd de l'hôpital, Paris F-75013, France
| | - Alice Jouneau
- Université Paris-Saclay, UVSQ, INRAE, BREED, Bâtiment 230, Domaine de Vilvert, Jouy-en-Josas 78350, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, 7 av. du Général de Gaulle, Maisons-Alfort 94700, France
| | - Thomas Dahlet
- University of Strasbourg, 4 rue Blaise Pascal, Strasbourg 67081, France
- CNRS UMR7242, Biotechnology and Cell Signaling, 300 bd Sébastien Brant, Illkirch 67412, France
| | - Michael Dumas
- University of Strasbourg, 4 rue Blaise Pascal, Strasbourg 67081, France
- CNRS UMR7242, Biotechnology and Cell Signaling, 300 bd Sébastien Brant, Illkirch 67412, France
| | - Michael Weber
- University of Strasbourg, 4 rue Blaise Pascal, Strasbourg 67081, France
- CNRS UMR7242, Biotechnology and Cell Signaling, 300 bd Sébastien Brant, Illkirch 67412, France
| | - Guillaume Velasco
- UMR7216 Epigénétique et Destin Cellulaire, CNRS, Université de Paris Cité, Epigenetics and Cell Fate, Lamarck building, 35 rue Hélène Brion, Paris F-75013, France
| | - Claire Francastel
- UMR7216 Epigénétique et Destin Cellulaire, CNRS, Université de Paris Cité, Epigenetics and Cell Fate, Lamarck building, 35 rue Hélène Brion, Paris F-75013, France
| |
Collapse
|
8
|
Day CA, Grigore F, Hakkim FL, Paul S, Langfald A, Weberg M, Fadness S, Schwab P, Sepaniac L, Stumpff J, Vaughan KT, Daniels DJ, Robinson JP, Hinchcliffe EH. The histone H3.3 K27M mutation suppresses Ser31phosphorylation and mitotic fidelity, which can directly drive gliomagenesis. Curr Biol 2025; 35:354-362.e7. [PMID: 39729988 DOI: 10.1016/j.cub.2024.11.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 12/29/2024]
Abstract
Serine 31 is a phospho-site unique to the histone H3.3 variant; mitotic phospho-Ser31 is restricted to pericentromeric heterochromatin, and disruption of phospho-Ser31 results in chromosome segregation defects and loss of p53-dependant G1 cell-cycle arrest.1,2,3,4 Ser31 is proximal to the H3.3 lysine 27-to-methionine (K27M) mutation that drives ∼80% of pediatric diffuse midline gliomas.5,6,7,8,9,10,11,12 Here, we show that expression of the H3.3 K27M mutant in normal, diploid cells results in increased chromosome missegregation and failure to arrest in the following G1. Expression of a non-phosphorylatable S31A mutant also drives chromosome missegregation, while the expression of a double K27M + phosphomimetic S31E mutant restores mitotic fidelity and the p53 response to chromosome missegregation. We show that patient-derived H3.3 K27M tumor cells have decreased mitotic Ser31 phosphorylation and increased frequency of chromosome missegregation. CRISPR reversion of the K27M mutation to wild type (WT) restores phospho-Ser31 levels and results in a decrease in chromosome missegregation. However, inserting an S31A mutation by CRISPR into these revertant cells disrupts mitotic fidelity. In vitro and in vivo analyses reveal that Chk1-the mitotic Ser31 kinase-is preferentially retained at pericentromeres in K27M-expressing tumor cells, compared with MLysine27-to-methionine mutation (M27K) isogenic revertants, correlating with both diminished phospho-Ser31 and mitotic defects. Interestingly, whereas M27K revertant cells do not form xenograft tumors in mice, H3.3 S31A cells do, similar to those formed by H3.3 K27M cells. Replication-competent avian leukosis virus splice-acceptor (RCAS)/cellular receptor for subgroup A avian sarcoma and leukosis virus (TVA) mice expressing S31A also form diffuse midline gliomas morphologically indistinguishable from K27M tumors. Together, our results reveal that the H3.3 K27M mutant alters H3.3 Ser31 phosphorylation, which, in turn, has profound impacts on chromosome segregation/cell-cycle regulation.
Collapse
Affiliation(s)
- Charles A Day
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; Neuro-Oncology Training Program, Mayo Clinic, Rochester, MN 55905, USA
| | - Florina Grigore
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; Department of Neurosurgery, University of Minnesota, Minneapolis, MN 55455, USA
| | - Faruck L Hakkim
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Souren Paul
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Alyssa Langfald
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Molly Weberg
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Sela Fadness
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Paiton Schwab
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Leslie Sepaniac
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05405, USA
| | - Jason Stumpff
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT 05405, USA; Vermont Cancer Center, University of Vermont, Burlington, VT 05405, USA
| | - Kevin T Vaughan
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA; Harper Cancer Center, University of Notre Dame, Notre Dame, IN 46556, USA
| | - David J Daniels
- Department of Neurosurgery, Mayo Clinic College of Medicine, Rochester, MN 55905, USA; Mayo Clinic Cancer Center, Mayo Clinic, Rochester, MN 55905, USA
| | - James P Robinson
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| | - Edward H Hinchcliffe
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA; Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
9
|
Zhang Y, Su M, Chen Y, Cui L, Xia W, Xu R, Xue D, Zhang X, Feng X. EHMT2-mediated R-loop formation promotes the malignant progression of prostate cancer via activating Aurora B. Clin Transl Med 2025; 15:e70164. [PMID: 39763034 PMCID: PMC11705492 DOI: 10.1002/ctm2.70164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 12/11/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Chromosomal instability (CIN), a hallmark of cancer, is commonly linked to poor prognosis in high-grade prostate cancer (PCa). Paradoxically, excessively high levels of CIN may impair cancer cell viability. Consequently, understanding how tumours adapt to CIN is critical for identifying novel therapeutic targets. METHODS Bioinformatic analyses were conducted to identify genes overexpressed in PCa tissues using The Cancer Genome Atlas (TCGA) and GEO datasets. Western blotting and immunohistochemistry assays were applied to determine the expression levels of euchromatic histone lysine methyltransferase 2 (EHMT2), pT232-Aurora B and Cullin 3 (CUL3). The proliferation of cells was measured through CCK-8 tests, clonogenesis and subcutaneous xenografts of human PCa cells in BALB/c nude mice. Live cell imaging, immunofluorescence (IF) and flow cytometry were used to confirm the role of EHMT2 in PCa cell mitosis. Co-immunoprecipitation, Western blotting and IF assays further elucidated the underlying molecular mechanisms. RESULTS EHMT2 was highly expressed in metastatic PCa tissues exhibiting elevated CIN and was strongly associated with adverse clinical outcomes in patients with PCa. Silencing EHMT2 impaired cell division, inducing G2/M-phase arrest and mitotic catastrophe in PCa cells. Mechanistically, EHMT2 is indispensable to ensure the full activation of Aurora B through centromeric R-loop-driven ATR-CHK1 pathway, with EHMT2 protein expression peaking during the G2/M-phase. Moreover, CUL3 was identified as a binding partner of EHMT2, mediating its polyubiquitination and destabilising its protein levels. CONCLUSIONS This study reveals a CUL3-EHMT2-Aurora B regulatory axis that safeguards accurate chromosome segregation in PCa cells, supporting the potential therapeutic application of EHMT2 inhibitors. KEY POINTS Euchromatic histone lysine methyltransferase 2 (EHMT2) is overexpressed in advanced prostate cancer, restraining catastrophic chromosomal instability (CIN) and enhancing cell fitness. EHMT2 functions via the centromeric R-loop-driven ATR-CHK1-Aurora B pathway to promote chromosomal stability. EHMT2 confers enzalutamide resistance via activating Aurora B. Cullin 3 (CUL3) promotes EHMT2 destabilisation via deubiquitination.
Collapse
Affiliation(s)
- Yuyang Zhang
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- Institute of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityHefeiAnhuiChina
| | - Mingqin Su
- Department of PathologyThe Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical UniversityHefeiAnhuiChina
| | - Yiming Chen
- Department of UrologyThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
- Department of UrologyThe First People's Hospital of ChangzhouChangzhouJiangsuChina
| | - Li Cui
- Department of UrologyThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
- Department of UrologyThe First People's Hospital of ChangzhouChangzhouJiangsuChina
| | - Wei Xia
- Department of UrologyThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
- Department of UrologyThe First People's Hospital of ChangzhouChangzhouJiangsuChina
| | - Renfang Xu
- Department of UrologyThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
- Department of UrologyThe First People's Hospital of ChangzhouChangzhouJiangsuChina
| | - Dong Xue
- Department of UrologyThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
- Department of UrologyThe First People's Hospital of ChangzhouChangzhouJiangsuChina
| | - Xiansheng Zhang
- Department of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- Institute of UrologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhuiChina
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical TransformationAnhui Medical UniversityHefeiAnhuiChina
| | - Xingliang Feng
- Department of UrologyThe Third Affiliated Hospital of Soochow UniversityChangzhouJiangsuChina
- Department of UrologyThe First People's Hospital of ChangzhouChangzhouJiangsuChina
| |
Collapse
|
10
|
Stratigi K, Siametis A, Garinis GA. Looping forward: exploring R-loop processing and therapeutic potential. FEBS Lett 2025; 599:244-266. [PMID: 38844597 PMCID: PMC11771710 DOI: 10.1002/1873-3468.14947] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 01/28/2025]
Abstract
Recently, there has been increasing interest in the complex relationship between transcription and genome stability, with specific attention directed toward the physiological significance of molecular structures known as R-loops. These structures arise when an RNA strand invades into the DNA duplex, and their formation is involved in a wide range of regulatory functions affecting gene expression, DNA repair processes or cell homeostasis. The persistent presence of R-loops, if not effectively removed, contributes to genome instability, underscoring the significance of the factors responsible for their resolution and modification. In this review, we provide a comprehensive overview of how R-loop processing can drive either a beneficial or a harmful outcome. Additionally, we explore the potential for manipulating such structures to devise rationalized therapeutic strategies targeting the aberrant accumulation of R-loops.
Collapse
Affiliation(s)
- Kalliopi Stratigi
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology‐HellasHeraklionCreteGreece
| | - Athanasios Siametis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology‐HellasHeraklionCreteGreece
- Department of BiologyUniversity of CreteHeraklionCreteGreece
| | - George A. Garinis
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology‐HellasHeraklionCreteGreece
- Department of BiologyUniversity of CreteHeraklionCreteGreece
| |
Collapse
|
11
|
Ouyang J. Transcription as a double-edged sword in genome maintenance. FEBS Lett 2025; 599:147-156. [PMID: 39704019 DOI: 10.1002/1873-3468.15080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 12/21/2024]
Abstract
Genome maintenance is essential for the integrity of the genetic blueprint, of which only a small fraction is transcribed in higher eukaryotes. DNA lesions occurring in the transcribed genome trigger transcription pausing and transcription-coupled DNA repair. There are two major transcription-coupled DNA repair pathways. The transcription-coupled nucleotide excision repair (TC-NER) pathway has been well studied for decades, while the transcription-coupled homologous recombination repair (TC-HR) pathway has recently gained attention. Importantly, recent studies have uncovered crucial roles of RNA transcripts in TC-HR, opening exciting directions for future research. Transcription also plays pivotal roles in regulating the stability of highly specialized genomic structures such as telomeres, centromeres, and fragile sites. Despite their positive function in genome maintenance, transcription and RNA transcripts can also be the sources of genomic instability, especially when colliding with DNA replication and forming unscheduled pathological RNA:DNA hybrids (R-loops), respectively. Pathological R-loops can result from transcriptional stress, which may be induced by transcription dysregulation. Future investigation into the interplay between transcription and DNA repair will reveal novel molecular bases for genome maintenance and transcriptional stress-associated genomic instability, providing therapeutic targets for human disease intervention.
Collapse
Affiliation(s)
- Jian Ouyang
- Department of Biochemistry and Molecular Biology
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
12
|
Black EM, Ramírez Parrado CA, Trier I, Li W, Joo YK, Pichurin J, Liu Y, Kabeche L. Chk2 sustains PLK1 activity in mitosis to ensure proper chromosome segregation. Nat Commun 2024; 15:10782. [PMID: 39737931 PMCID: PMC11685634 DOI: 10.1038/s41467-024-54922-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Polo-like kinase 1 (PLK1) protects against genome instability by ensuring timely and accurate mitotic cell division, and its activity is tightly regulated throughout the cell cycle. Although the pathways that initially activate PLK1 in G2 are well-characterized, the factors that directly regulate mitotic PLK1 remain poorly understood. Here, we identify that human PLK1 activity is sustained by the DNA damage response kinase Checkpoint kinase 2 (Chk2) in mitosis. Chk2 directly phosphorylates PLK1 T210, a residue on its T-loop whose phosphorylation is essential for full PLK1 kinase activity. Loss of Chk2-dependent PLK1 activity causes increased mitotic errors, including chromosome misalignment, chromosome missegregation, and cytokinetic defects. Moreover, Chk2 deficiency increases sensitivity to PLK1 inhibitors, suggesting that Chk2 status may be an informative biomarker for PLK1 inhibitor efficacy. This work demonstrates that Chk2 sustains mitotic PLK1 activity and protects genome stability through discrete functions in interphase DNA damage repair and mitotic chromosome segregation.
Collapse
Affiliation(s)
- Elizabeth M Black
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Carlos Andrés Ramírez Parrado
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Isabelle Trier
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Wenxue Li
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
- Department of Pharmacology, Yale University, New Haven, CT, 06511, USA
| | - Yoon Ki Joo
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Jennifer Pichurin
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
| | - Yansheng Liu
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA
- Department of Pharmacology, Yale University, New Haven, CT, 06511, USA
| | - Lilian Kabeche
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, 06511, USA.
- Yale Cancer Biology Institute, Yale University, West Haven, CT, 06516, USA.
| |
Collapse
|
13
|
Sobczyńska-Konefał A, Jasek M, Karabon L, Jaskuła E. Insights into genetic aberrations and signalling pathway interactions in chronic lymphocytic leukemia: from pathogenesis to treatment strategies. Biomark Res 2024; 12:162. [PMID: 39732734 DOI: 10.1186/s40364-024-00710-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024] Open
Abstract
Chronic lymphocytic leukemia (CLL) is prevalent in adults and is characterized by the accumulation of mature B cells in the blood, bone marrow, lymph nodes, and spleens. Recent progress in therapy and the introduction of targeted treatments [inhibitors of Bruton's tyrosine kinase (BTKi) or inhibitor of anti-apoptotic B-cell lymphoma-2 (Bcl-2i) protein (venetoclax)] in place of chemoimmunotherapy have significantly improved the outcomes of patients with CLL. These advancements have shifted the importance of traditional predictive markers, leading to a greater focus on resistance genes and reducing the significance of mutations, such as TP53 and del(17p). Despite the significant progress in CLL treatment, some patients still experience disease relapse. This is due to the substantial heterogeneity of CLL as well as the interconnected genetic resistance mechanisms and pathway adaptive resistance mechanisms to targeted therapies in CLL. Although the knowledge of the pathomechanism of CLL has expanded significantly in recent years, the precise origins of CLL and the interplay between various genetic factors remain incompletely understood, necessitating further research. This review enhances the molecular understanding of CLL by describing how BCR signalling, NF-κB PI3K/AKT, and ROR1 pathways sustain CLL cell survival, proliferation, and resistance to apoptosis. It also presents genetic and pathway-adaptive resistance mechanisms in CLL. Identifying B-cell receptor (BCR) signalling as a pivotal driver of CLL progression, the findings advocate personalized treatment strategies based on molecular profiling, emphasizing the need for further research to unravel the complex interplay between BCR signalling and its associated pathways to improve patient outcomes.
Collapse
Affiliation(s)
- Anna Sobczyńska-Konefał
- L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolf Weigl 12, 53-114, Wroclaw, Poland
- Lower Silesian Oncology Hematology and Pulmonology Center, Ludwik Hirszfeld square 12, 53-413, Wroclaw, Poland
| | - Monika Jasek
- L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolf Weigl 12, 53-114, Wroclaw, Poland
| | - Lidia Karabon
- L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolf Weigl 12, 53-114, Wroclaw, Poland
| | - Emilia Jaskuła
- L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolf Weigl 12, 53-114, Wroclaw, Poland.
- Lower Silesian Oncology Hematology and Pulmonology Center, Ludwik Hirszfeld square 12, 53-413, Wroclaw, Poland.
| |
Collapse
|
14
|
Iurchenko NP, Glushchenko NM, Nespryadko SV, Buchynska LG. SIGNIFICANCE OF miRNA-185-5P AND miRNA-424-5P AS PROGNOSTIC MARKERS IN PROGRESSION OF EARLY-STAGE ENDOMETRIAL CANCER. Exp Oncol 2024; 46:244-252. [PMID: 39704456 DOI: 10.15407/exp-oncology.2024.03.244] [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: 12/19/2024] [Indexed: 12/21/2024]
Abstract
AIM To compare the expression of miRNA-185-5p and miRNA-424-5p in tumor cells and peripheral blood serum (PBS) of patients with endometrioid carcinoma of the endometrium (ECE) and to evaluate the significance of these biomarkers in cancer progression. MATERIALS AND METHODS The study was conducted on the samples of peripheral blood serum (PBS) and tumor tissue of 58 patients with stage I ECE using clinical and morphological methods and real-time polymerase chain reaction. RESULTS A significant increase in the levels of circulating and tumor-associated miRNA-424-5p was established in ECE patients with a history of recurrences compared to patients without recurrences. To the contrary, the expression level of miRNA-185-5p increased in the PBS and decreased in the tumor tissue of ECE patients with recurrences compared to the patients without recurrence. In addition, we revealed that the expression levels of the studied miRNAs were associated with the differentiation grade and degree of tumor invasion. We established that miRNA-424-5p levels in PBS could serve as the most significant indicator for predicting the occurrence of recurrence in patients with ECE (AUC = 0.991; Sp 94.0%; Se 99.9%). CONCLUSIONS The expression features of miRNA-185-5p and miRNA-424-5p in the PBS and tumor tissue of patients with ECE are associated with the aggressiveness of cancer course and the risk of recurrence.
Collapse
Affiliation(s)
- N P Iurchenko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - N M Glushchenko
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | | | - L G Buchynska
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
15
|
Tang Z, Liu Q, Pan Z, Liu C, Dong J, Han F, Fu S. Stable minichromosome and functional neocentromere derived from rye 7R chromosome arm. BMC PLANT BIOLOGY 2024; 24:1185. [PMID: 39695363 DOI: 10.1186/s12870-024-05918-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND The study of newly formed centromere with stable transmission ability can provide theoretical guidance for the construction of artificial chromosomes. More neocentromeres are needed to study the mechanisms of their formation. RESULTS In this study, a minichromosome 7RLmini was derived from the progeny of wheat-rye 7R monosomic addition line. The minichromosome 7RLmini contained subtelomeric tandem repeats pSc119.2 and rye-specific pSc200, and it came from the distal region of the long arm of 7R chromosome. A neocentromere was formed in this minichromosome, and it did not contain centromeric repetitive sequences CCS1 and pAWRC.1. CENH3 ChIP-seq and ssDRIP-seq data confirmed that a 2.4 Mb segment from the rye 7R chromosome was involved in the neocentromere formation and enrichment of R-loops in this region. Within the 2.4 Mb segment, the GC content was higher that of AT, and a major binding position of CENH3 nucleosomes was identified on a 6 kb unknown LTR retrotransposon TE00002448. This unknown LTR retrotransposon was rye-specific and distributed through all the arms of rye chromosomes. The minichromosome exhibited stable generational transmission. CONCLUSION A minichromosome from rye 7R with neocentromere was obtained in this study and the neocentromere was formed at the position far away from its native equivalent. This minichromosome provides additional material for the research on the mechanism of neocentromere formation. We theorize that R-loops and transposable element might be involved in the positioning of CENH3 nucleosomes in a functional neocentromere.
Collapse
Affiliation(s)
- Zongxiang Tang
- College of Agronomy, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
- Provincial Key Laboratory for Plant Genetics and Breeding, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Qian Liu
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zijin Pan
- College of Agronomy, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
- Provincial Key Laboratory for Plant Genetics and Breeding, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Chang Liu
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jieran Dong
- College of Agronomy, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
- Provincial Key Laboratory for Plant Genetics and Breeding, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China
| | - Fangpu Han
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shulan Fu
- College of Agronomy, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China.
- Provincial Key Laboratory for Plant Genetics and Breeding, Sichuan Agricultural University, Wenjiang, Chengdu, 611130, China.
| |
Collapse
|
16
|
Tseng CC, Obeng EA. RNA splicing as a therapeutic target in myelodysplastic syndromes. Semin Hematol 2024; 61:431-441. [PMID: 39542752 DOI: 10.1053/j.seminhematol.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 10/18/2024] [Indexed: 11/17/2024]
Abstract
Myelodysplastic syndromes (MDS) represent a heterogeneous group of hematological disorders and are more commonly found in people over the age of 60. MDS patients exhibit peripheral blood cytopenias and carry an increased risk of disease progression to acute myeloid leukemia (AML). Splicing factor mutations (including genes SF3B1, SRSF2, U2AF1, and ZRSR2) are early events identified in more than 50% of MDS cases. These mutations cause aberrant pre-mRNA splicing and impact MDS pathophysiology. Emerging evidence shows that splicing factor-mutant cells are more sensitive to perturbations targeting the spliceosome, aberrantly spliced genes and/or their regulated molecular pathways. This review summarizes current therapeutic strategies and ongoing efforts targeting splicing factor mutations for the treatment of MDS.
Collapse
Affiliation(s)
- Chun-Chih Tseng
- Division of Molecular Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN
| | - Esther A Obeng
- Division of Molecular Oncology, Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN.
| |
Collapse
|
17
|
Joo YK, Ramirez C, Kabeche L. A TRilogy of ATR's Non-Canonical Roles Throughout the Cell Cycle and Its Relation to Cancer. Cancers (Basel) 2024; 16:3536. [PMID: 39456630 PMCID: PMC11506335 DOI: 10.3390/cancers16203536] [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: 09/17/2024] [Revised: 10/12/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Ataxia Telangiectasia and Rad3-related protein (ATR) is an apical kinase of the DNA Damage Response (DDR) pathway responsible for detecting and resolving damaged DNA. Because cancer cells depend heavily on the DNA damage checkpoint for their unchecked proliferation and propagation, ATR has gained enormous popularity as a cancer therapy target in recent decades. Yet, ATR inhibitors have not been the silver bullets as anticipated, with clinical trials demonstrating toxicity and mixed efficacy. To investigate whether the toxicity and mixed efficacy of ATR inhibitors arise from their off-target effects related to ATR's multiple roles within and outside the DDR pathway, we have analyzed recently published studies on ATR's non-canonical roles. Recent studies have elucidated that ATR plays a wide role throughout the cell cycle that is separate from its function in the DDR. This includes maintaining nuclear membrane integrity, detecting mechanical forces, and promoting faithful chromosome segregation during mitosis. In this review, we summarize the canonical, DDR-related roles of ATR and also focus on the non-canonical, multifaceted roles of ATR throughout the cell cycle and their clinical relevance. Through this summary, we also address the need for re-assessing clinical strategies targeting ATR as a cancer therapy based on these newly discovered roles for ATR.
Collapse
Affiliation(s)
- Yoon Ki Joo
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Carlos Ramirez
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| | - Lilian Kabeche
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06511, USA
- Yale Cancer Biology Institute, Yale University, West Haven, CT 06516, USA
| |
Collapse
|
18
|
Wang B, Kou H, Wang Y, Zhang Q, Jiang D, Wang J, Zhao Z, Zhou Y, Zhang M, Sui L, Zhao M, Liu Y, Liu Y, Shi L, Wang F. LAP2α orchestrates alternative lengthening of telomeres suppression through telomeric heterochromatin regulation with HDAC1: unveiling a potential therapeutic target. Cell Death Dis 2024; 15:761. [PMID: 39426946 PMCID: PMC11490576 DOI: 10.1038/s41419-024-07116-4] [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: 02/01/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/21/2024]
Abstract
In response to the challenge of telomere attrition during DNA replication, cancer cells predominantly employ telomerase or, in 10-15% of cases, the alternative lengthening of telomeres (ALT). The intricate details of ALT, however, remain elusive. In this study, we unveil that the knockdown of lamina-associated polypeptide 2 alpha (LAP2α) in ALT cells results in telomere dysfunction, triggering a notable increase in ALT-associated hallmarks, including high frequencies of PML bodies (APBs), C-rich extrachromosomal circles (C-circles), and telomere sister chromatid exchange (T-SCE). Furthermore, LAP2α emerges as a crucial player in break-induced telomere replication for telomerase-positive cells following telomeric double-strand breaks. Mechanistically, our investigation suggests that LAP2α may influence the regulation of the heterochromatic state of telomeres, thereby affecting telomeric accessibility. In line with our findings, LAP2α expression is markedly reduced in ALT-positive osteosarcoma. And the use of methotrexate (MTX) can restore the heterochromatin state altered by LAP2α depletion. This is evidenced by a significant inhibition of tumor proliferation in ALT-positive patient-derived xenograft (PDX) mouse models. These results indicate the important role of LAP2α in regulating ALT activity and offer insights into the interplay between lamina-associated proteins and telomeres in maintaining telomere length. Importantly, our findings may help identify a more appropriate target population for the osteosarcoma therapeutic drug, MTX.
Collapse
Grants
- 32170762 National Natural Science Foundation of China (National Science Foundation of China)
- This work was supported by the grant from the National Natural Science Foundation of China (No. 32170762, 3217050514, 31771520, 31471293, 91649102, 92149302, 81772243, 81771135, 81970958, 82303619), Tianjin Health Research Project (No. 19YFZCSY00600), Science and Technology Project of Tianjin Municipal Health Committee (No. TJWJ2022XK018, TJWJ2022QN030) and the Natural Science Foundation of Tianjin City (No. 19JCJQJC63500)
Collapse
Affiliation(s)
- Bing Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Genetics, School of Basic Medical Science, Institute of Prosthodontics School and Hospital of Stomatology, General Hospital, Tianjin Medical University, 300070, Tianjin, P. R. China
- Department of Hematology, Tianjin First Central Hospital, 300192, Tianjin, P. R. China
| | - Haomeng Kou
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Genetics, School of Basic Medical Science, Institute of Prosthodontics School and Hospital of Stomatology, General Hospital, Tianjin Medical University, 300070, Tianjin, P. R. China
| | - Yuwen Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Genetics, School of Basic Medical Science, Institute of Prosthodontics School and Hospital of Stomatology, General Hospital, Tianjin Medical University, 300070, Tianjin, P. R. China
| | - Qi Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, P. R. China
| | - Duo Jiang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Genetics, School of Basic Medical Science, Institute of Prosthodontics School and Hospital of Stomatology, General Hospital, Tianjin Medical University, 300070, Tianjin, P. R. China
| | - Juan Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Genetics, School of Basic Medical Science, Institute of Prosthodontics School and Hospital of Stomatology, General Hospital, Tianjin Medical University, 300070, Tianjin, P. R. China
| | - Ziqin Zhao
- Department of Pathology, Tianjin Hospital, 300221, Tianjin, P. R. China
| | - Yao Zhou
- Department of Bioinformatics, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, P. R. China
| | - Miaomiao Zhang
- Department of Pathology, Jining No.1 People's Hospital, 272000, Jining, Shandong, P. R. China
| | - Lei Sui
- Department of Prosthodontics, School and Hospital of Stomatology, Tianjin Medical University, 300070, Tianjin, P. R. China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, 300192, Tianjin, P. R. China
| | - Yancheng Liu
- Department of Bone and Soft Tissue Oncology, Tianjin Hospital, 300221, Tianjin, P. R. China.
| | - Yang Liu
- Department of Radiobiology, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, 300192, Tianjin, P. R. China.
| | - Lei Shi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, 300070, Tianjin, P. R. China.
| | - Feng Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Genetics, School of Basic Medical Science, Institute of Prosthodontics School and Hospital of Stomatology, General Hospital, Tianjin Medical University, 300070, Tianjin, P. R. China.
| |
Collapse
|
19
|
Kang Z, Li R, Liu C, Dong X, Hu Y, Xu L, Liu X, Xiang Y, Gao L, Si W, Wang L, Li Q, Zhang L, Wang H, Yang X, Liu J. m 6A-modified cenRNA stabilizes CENPA to ensure centromere integrity in cancer cells. Cell 2024; 187:6035-6054.e27. [PMID: 39305902 DOI: 10.1016/j.cell.2024.08.040] [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: 12/12/2023] [Revised: 01/10/2024] [Accepted: 08/20/2024] [Indexed: 10/20/2024]
Abstract
m6A modification is best known for its critical role in controlling multiple post-transcriptional processes of the mRNAs. Here, we discovered elevated levels of m6A modification on centromeric RNA (cenRNA) in cancerous cells compared with non-cancerous cells. We then identified CENPA, an H3 variant, as an m6A reader of cenRNA. CENPA is localized at centromeres and is essential in preserving centromere integrity and function during mitosis. The m6A-modified cenRNA stabilizes centromeric localization of CENPA in cancer cells during the S phase of the cell cycle. Mutations of CENPA at the Leu61 and the Arg63 or removal of cenRNA m6A modification lead to loss of centromere-bound CENPA during S phase. This in turn results in compromised centromere integrity and abnormal chromosome separation and hinders cancer cell proliferation and tumor growth. Our findings unveil an m6A reading mechanism by CENPA that epigenetically governs centromere integrity in cancer cells, providing potential targets for cancer therapy.
Collapse
Affiliation(s)
- Zihong Kang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China
| | - Ruimeng Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Joint Graduate Program of Peking-Tsinghua-National Institute of Biological Science, Tsinghua University, 100084 Beijing, China
| | - Chang Liu
- Department of Genetics, Stanford University, School of Medicine, Stanford, CA, USA
| | - Xiaozhe Dong
- College of Chemistry and Molecular Engineering, Peking University, 100871 Beijing, China
| | - Yuxuan Hu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 211198 Nanjing, China
| | - Lei Xu
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008 Nanjing, China
| | - Xinyu Liu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China
| | - Yunfan Xiang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China
| | - Liming Gao
- School of Science, China Pharmaceutical University, 211198 Nanjing, China
| | - Wenzhe Si
- State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Laboratory Medicine, Peking University Third Hospital, 100191 Beijing, China
| | - Lei Wang
- Department of Gastroenterology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 210008 Nanjing, China
| | - Qing Li
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China
| | - Liang Zhang
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, 310022 Hangzhou, China
| | - Huan Wang
- College of Chemistry and Molecular Engineering, Peking University, 100871 Beijing, China
| | - Xuerui Yang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Joint Graduate Program of Peking-Tsinghua-National Institute of Biological Science, Tsinghua University, 100084 Beijing, China.
| | - Jun Liu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking-Tsinghua Center for Life Sciences, Peking University, 100871 Beijing, China; Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, China.
| |
Collapse
|
20
|
Mendes IC, Dos Reis Bertoldo W, Miranda-Junior AS, Assis AVD, Repolês BM, Ferreira WRR, Chame DF, Souza DDL, Pavani RS, Macedo AM, Franco GR, Serra E, Perdomo V, Menck CFM, da Silva Leandro G, Fragoso SP, Barbosa Elias MCQ, Machado CR. DNA lesions that block transcription induce the death of Trypanosoma cruzi via ATR activation, which is dependent on the presence of R-loops. DNA Repair (Amst) 2024; 141:103726. [PMID: 39096697 DOI: 10.1016/j.dnarep.2024.103726] [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: 11/23/2023] [Revised: 06/25/2024] [Accepted: 07/07/2024] [Indexed: 08/05/2024]
Abstract
Trypanosoma cruzi is the etiological agent of Chagas disease and a peculiar eukaryote with unique biological characteristics. DNA damage can block RNA polymerase, activating transcription-coupled nucleotide excision repair (TC-NER), a DNA repair pathway specialized in lesions that compromise transcription. If transcriptional stress is unresolved, arrested RNA polymerase can activate programmed cell death. Nonetheless, how this parasite modulates these processes is unknown. Here, we demonstrate that T. cruzi cell death after UV irradiation, a genotoxic agent that generates lesions resolved by TC-NER, depends on active transcription and is signaled mainly by an apoptotic-like pathway. Pre-treated parasites with α-amanitin, a selective RNA polymerase II inhibitor, become resistant to such cell death. Similarly, the gamma pre-irradiated cells are more resistant to UV when the transcription processes are absent. The Cockayne Syndrome B protein (CSB) recognizes blocked RNA polymerase and can initiate TC-NER. Curiously, CSB overexpression increases parasites' cell death shortly after UV exposure. On the other hand, at the same time after irradiation, the single-knockout CSB cells show resistance to the same treatment. UV-induced fast death is signalized by the exposition of phosphatidylserine to the outer layer of the membrane, indicating a cell death mainly by an apoptotic-like pathway. Furthermore, such death is suppressed in WT parasites pre-treated with inhibitors of ataxia telangiectasia and Rad3-related (ATR), a key DDR kinase. Signaling for UV radiation death may be related to R-loops since the overexpression of genes associated with the resolution of these structures suppress it. Together, results suggest that transcription blockage triggered by UV radiation activates an ATR-dependent apoptosis-like mechanism in T. cruzi, with the participation of CSB protein in this process.
Collapse
Affiliation(s)
- Isabela Cecilia Mendes
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Willian Dos Reis Bertoldo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Adalberto Sales Miranda-Junior
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Antônio Vinícius de Assis
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Bruno Marçal Repolês
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Wesley Roger Rodrigues Ferreira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Daniela Ferreira Chame
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Daniela De Laet Souza
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Raphael Souza Pavani
- Laboratório Especial de Ciclo Celular, Instituto Butantan, São Paulo, MG, São Paulo, SP 05503-900, Brazil
| | - Andrea Mara Macedo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Glória Regina Franco
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil
| | - Esteban Serra
- Instituto de Biología Molecular y Celular de Rosario, CONICET, 2000 Rosario, Santa Fe, Argentina; Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, 2000 Rosario, Santa Fe, Argentina
| | - Virginia Perdomo
- Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, 2000 Rosario, Santa Fe, Argentina
| | - Carlos Frederico Martins Menck
- Departamento de Microbiologia, Instituto de Ciências Biomédicas (ICB), Universidade de São Paulo (USP), São Paulo, SP 05508-900, Brazil
| | - Giovana da Silva Leandro
- Departamento de Microbiologia, Instituto de Ciências Biomédicas (ICB), Universidade de São Paulo (USP), São Paulo, SP 05508-900, Brazil
| | | | | | - Carlos Renato Machado
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, MG 30161-970, Brazil.
| |
Collapse
|
21
|
Liu C, Fu S, Yi C, Liu Y, Huang Y, Guo X, Zhang K, Liu Q, Birchler JA, Han F. Unveiling the distinctive traits of functional rye centromeres: minisatellites, retrotransposons, and R-loop formation. SCIENCE CHINA. LIFE SCIENCES 2024; 67:1989-2002. [PMID: 38805064 DOI: 10.1007/s11427-023-2524-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 11/14/2023] [Indexed: 05/29/2024]
Abstract
Centromeres play a vital role in cellular division by facilitating kinetochore assembly and spindle attachments. Despite their conserved functionality, centromeric DNA sequences exhibit rapid evolution, presenting diverse sizes and compositions across species. The functional significance of rye centromeric DNA sequences, particularly in centromere identity, remains unclear. In this study, we comprehensively characterized the sequence composition and organization of rye centromeres. Our findings revealed that these centromeres are primarily composed of long terminal repeat retrotransposons (LTR-RTs) and interspersed minisatellites. We systematically classified LTR-RTs into five categories, highlighting the prevalence of younger CRS1, CRS2, and CRS3 of CRSs (centromeric retrotransposons of Secale cereale) were primarily located in the core centromeres and exhibited a higher association with CENH3 nucleosomes. The minisatellites, mainly derived from retrotransposons, along with CRSs, played a pivotal role in establishing functional centromeres in rye. Additionally, we observed the formation of R-loops at specific regions of CRS1, CRS2, and CRS3, with both rye pericentromeres and centromeres exhibiting enrichment in R-loops. Notably, these R-loops selectively formed at binding regions of the CENH3 nucleosome in rye centromeres, suggesting a potential role in mediating the precise loading of CENH3 to centromeres and contributing to centromere specification. Our work provides insights into the DNA sequence composition, distribution, and potential function of R-loops in rye centromeres. This knowledge contributes valuable information to understanding the genetics and epigenetics of rye centromeres, offering implications for the development of synthetic centromeres in future plant modifications and beyond.
Collapse
Affiliation(s)
- Chang Liu
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shulan Fu
- Key Laboratory for Plant Genetics and Breeding, Sichuan Agricultural University, Chengdu, 611130, China
| | - Congyang Yi
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yang Liu
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuhong Huang
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xianrui Guo
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Kaibiao Zhang
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qian Liu
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - James A Birchler
- Division of Biological Science, University of Missouri, Columbia, 65211-7400, USA
| | - Fangpu Han
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
22
|
Salinas-Luypaert C, Fachinetti D. Canonical and noncanonical regulators of centromere assembly and maintenance. Curr Opin Cell Biol 2024; 89:102396. [PMID: 38981198 DOI: 10.1016/j.ceb.2024.102396] [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: 04/09/2024] [Revised: 06/10/2024] [Accepted: 06/15/2024] [Indexed: 07/11/2024]
Abstract
Centromeres are specialized chromosomal domains where the kinetochores assemble during cell division to ensure accurate transmission of the genetic information to the two daughter cells. The centromeric function is evolutionary conserved and, in most organisms, centromeres are epigenetically defined by a unique chromatin containing the histone H3 variant CENP-A. The canonical regulators of CENP-A assembly and maintenance are well-known, yet some of the molecular mechanisms regulating this complex process have only recently been unveiled. We review the most recent advances on the topic, including the emergence of new and unexpected factors that favor and regulate CENP-A assembly and/or maintenance.
Collapse
Affiliation(s)
- Catalina Salinas-Luypaert
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR 144 & UMR3664, 26 rue d'Ulm, 75005, Paris, France.
| | - Daniele Fachinetti
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR 144 & UMR3664, 26 rue d'Ulm, 75005, Paris, France.
| |
Collapse
|
23
|
Wu T, Hou H, Dey A, Bachu M, Chen X, Wisniewski J, Kudoh F, Chen C, Chauhan S, Xiao H, Pan R, Ozato K. Bromodomain protein BRD4 directs mitotic cell division of mouse fibroblasts by inhibiting DNA damage. iScience 2024; 27:109797. [PMID: 38993671 PMCID: PMC11237862 DOI: 10.1016/j.isci.2024.109797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 10/30/2023] [Accepted: 04/18/2024] [Indexed: 07/13/2024] Open
Abstract
Bromodomain protein BRD4 binds to acetylated histones to regulate transcription. BRD4 also drives cancer cell proliferation. However, the role of BRD4 in normal cell growth has remained unclear. Here, we investigated this question by using mouse embryonic fibroblasts with conditional Brd4 knockout (KO). We found that Brd4KO cells grow more slowly than wild type cells; they do not complete replication, fail to achieve mitosis, and exhibit extensive DNA damage throughout all cell cycle stages. BRD4 was required for expression of more than 450 cell cycle genes including genes encoding core histones and centromere/kinetochore proteins that are critical for genome replication and chromosomal segregation. Moreover, we show that many genes controlling R-loop formation and DNA damage response (DDR) require BRD4 for expression. Finally, BRD4 constitutively occupied genes controlling R-loop, DDR and cell cycle progression. In summary, BRD4 epigenetically marks above genes and serves as a master regulator of normal cell growth.
Collapse
Affiliation(s)
- Tiyun Wu
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haitong Hou
- Science Center for Future Foods, Jiangnan University, 1800 Lihu Road, Wuxi, Jiangsu 214122, China
| | - Anup Dey
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mahesh Bachu
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
- Weill Cornell Medicine, Graduate School of Medical Sciences, 1300 York Avenue Box 65, New York, NY 10065, USA
| | - Xiongfong Chen
- CCR-SF Bioinformatics Group, Advanced Biomedical and Computational Sciences, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA
| | - Jan Wisniewski
- Confocal Microscopy and Digital Imaging Facility, Experimental Immunology Branch, CCR, NCI NIH Bldg 10 Rm 4A05, Bethesda, MD 20892, USA
| | - Fuki Kudoh
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chao Chen
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
- Division of Hematology/Oncology, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Sakshi Chauhan
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Hua Xiao
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard Pan
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Keiko Ozato
- Division of Developmental Biology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
24
|
Luna R, Gómez-González B, Aguilera A. RNA biogenesis and RNA metabolism factors as R-loop suppressors: a hidden role in genome integrity. Genes Dev 2024; 38:504-527. [PMID: 38986581 PMCID: PMC11293400 DOI: 10.1101/gad.351853.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Genome integrity relies on the accuracy of DNA metabolism, but as appreciated for more than four decades, transcription enhances mutation and recombination frequencies. More recent research provided evidence for a previously unforeseen link between RNA and DNA metabolism, which is often related to the accumulation of DNA-RNA hybrids and R-loops. In addition to physiological roles, R-loops interfere with DNA replication and repair, providing a molecular scenario for the origin of genome instability. Here, we review current knowledge on the multiple RNA factors that prevent or resolve R-loops and consequent transcription-replication conflicts and thus act as modulators of genome dynamics.
Collapse
Affiliation(s)
- Rosa Luna
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Universidad de Sevilla-Spanish National Research Council (CSIC), 41092 Seville, Spain
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| | - Belén Gómez-González
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Universidad de Sevilla-Spanish National Research Council (CSIC), 41092 Seville, Spain
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| | - Andrés Aguilera
- Andalusian Center of Molecular Biology and Regenerative Medicine (CABIMER), Universidad de Sevilla-Spanish National Research Council (CSIC), 41092 Seville, Spain;
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| |
Collapse
|
25
|
Chen YL, Jones AN, Crawford A, Sattler M, Ettinger A, Torres-Padilla ME. Determinants of minor satellite RNA function in chromosome segregation in mouse embryonic stem cells. J Cell Biol 2024; 223:e202309027. [PMID: 38625077 PMCID: PMC11022885 DOI: 10.1083/jcb.202309027] [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: 09/05/2023] [Revised: 03/06/2024] [Accepted: 03/29/2024] [Indexed: 04/17/2024] Open
Abstract
The centromere is a fundamental higher-order structure in chromosomes ensuring their faithful segregation upon cell division. Centromeric transcripts have been described in several species and suggested to participate in centromere function. However, low sequence conservation of centromeric repeats appears inconsistent with a role in recruiting highly conserved centromeric proteins. Here, we hypothesized that centromeric transcripts may function through a secondary structure rather than sequence conservation. Using mouse embryonic stem cells (ESCs), we show that an imbalance in the levels of forward or reverse minor satellite (MinSat) transcripts leads to severe chromosome segregation defects. We further show that MinSat RNA adopts a stem-loop secondary structure, which is conserved in human α-satellite transcripts. We identify an RNA binding region in CENPC and demonstrate that MinSat transcripts function through the structured region of the RNA. Importantly, mutants that disrupt MinSat secondary structure do not cause segregation defects. We propose that the conserved role of centromeric transcripts relies on their secondary RNA structure.
Collapse
Affiliation(s)
- Yung-Li Chen
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Munich, München, Germany
| | - Alisha N. Jones
- Institute of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
| | - Amy Crawford
- Department of Chemistry, New York University, New York, NY, USA
| | - Michael Sattler
- Institute of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Munich, Neuherberg, Germany
- Department of Bioscience, Bavarian NMR Center, School of Natural Sciences, Technical University of Munich, Garching, Germany
| | - Andreas Ettinger
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Munich, München, Germany
| | - Maria-Elena Torres-Padilla
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Munich, München, Germany
- Faculty of Biology, Ludwig-Maximilians Universität, München, Germany
| |
Collapse
|
26
|
Zhao Y, He S, Zhao M, Huang Q. Surviving the Storm: The Role of Poly- and Depolyploidization in Tissues and Tumors. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306318. [PMID: 38629780 PMCID: PMC11199982 DOI: 10.1002/advs.202306318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 03/18/2024] [Indexed: 06/27/2024]
Abstract
Polyploidization and depolyploidization are critical processes in the normal development and tissue homeostasis of diploid organisms. Recent investigations have revealed that polyaneuploid cancer cells (PACCs) exploit this ploidy variation as a survival strategy against anticancer treatment and for the repopulation of tumors. Unscheduled polyploidization and chromosomal instability in PACCs enhance malignancy and treatment resistance. However, their inability to undergo mitosis causes catastrophic cellular death in most PACCs. Adaptive ploid reversal mechanisms, such as multipolar mitosis, centrosome clustering, meiosis-like division, and amitosis, counteract this lethal outcome and drive cancer relapse. The purpose of this work is to focus on PACCs induced by cytotoxic therapy, highlighting the latest discoveries in ploidy dynamics in physiological and pathological contexts. Specifically, by emphasizing the role of "poly-depolyploidization" in tumor progression, the aim is to identify novel therapeutic targets or paradigms for combating diseases associated with aberrant ploidies.
Collapse
Affiliation(s)
- Yucui Zhao
- Cancer CenterShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620China
- Department of Radiation OncologySecond Affiliated HospitalZhejiang University School of MedicineHangzhou310009China
| | - Sijia He
- Cancer CenterShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620China
| | - Minghui Zhao
- Cancer CenterShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620China
- Department of Radiation OncologyFirst Affiliated Hospital of Nanjing Medical UniversityNanjing210029China
| | - Qian Huang
- Cancer CenterShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620China
- Shanghai Key Laboratory of Pancreatic DiseasesShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai201620China
| |
Collapse
|
27
|
Choudhury R, Venkateswaran Venkatasubramani A, Hua J, Borsò M, Franconi C, Kinkley S, Forné I, Imhof A. The role of RNA in the maintenance of chromatin domains as revealed by antibody-mediated proximity labelling coupled to mass spectrometry. eLife 2024; 13:e95718. [PMID: 38717135 PMCID: PMC11147508 DOI: 10.7554/elife.95718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/26/2024] [Indexed: 06/04/2024] Open
Abstract
Eukaryotic chromatin is organized into functional domains, that are characterized by distinct proteomic compositions and specific nuclear positions. In contrast to cellular organelles surrounded by lipid membranes, the composition of distinct chromatin domains is rather ill described and highly dynamic. To gain molecular insight into these domains and explore their composition, we developed an antibody-based proximity biotinylation method targeting the RNA and proteins constituents. The method that we termed antibody-mediated proximity labelling coupled to mass spectrometry (AMPL-MS) does not require the expression of fusion proteins and therefore constitutes a versatile and very sensitive method to characterize the composition of chromatin domains based on specific signature proteins or histone modifications. To demonstrate the utility of our approach we used AMPL-MS to characterize the molecular features of the chromocenter as well as the chromosome territory containing the hyperactive X chromosome in Drosophila. This analysis identified a number of known RNA-binding proteins in proximity of the hyperactive X and the centromere, supporting the accuracy of our method. In addition, it enabled us to characterize the role of RNA in the formation of these nuclear bodies. Furthermore, our method identified a new set of RNA molecules associated with the Drosophila centromere. Characterization of these novel molecules suggested the formation of R-loops in centromeres, which we validated using a novel probe for R-loops in Drosophila. Taken together, AMPL-MS improves the selectivity and specificity of proximity ligation allowing for novel discoveries of weak protein-RNA interactions in biologically diverse domains.
Collapse
Affiliation(s)
- Rupam Choudhury
- Department of Molecular Biology, Biomedical Center Munich, Ludwig-Maximilians UniversityPlanegg-MartinsriedGermany
| | - Anuroop Venkateswaran Venkatasubramani
- Department of Molecular Biology, Biomedical Center Munich, Ludwig-Maximilians UniversityPlanegg-MartinsriedGermany
- Graduate School of Quantitative Biosciences (QBM), Ludwig-Maximilians-Universität MünchenMunichGermany
| | - Jie Hua
- Department of Molecular Biology, Biomedical Center Munich, Ludwig-Maximilians UniversityPlanegg-MartinsriedGermany
| | - Marco Borsò
- Protein Analysis Unit, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians, University (LMU) MunichPlanegg-MartinsriedGermany
| | - Celeste Franconi
- Chromatin Structure and Function group, Department of Computational Molecular Biology, Max Planck Institute for Molecular GeneticsBerlinGermany
| | - Sarah Kinkley
- Chromatin Structure and Function group, Department of Computational Molecular Biology, Max Planck Institute for Molecular GeneticsBerlinGermany
| | - Ignasi Forné
- Protein Analysis Unit, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians, University (LMU) MunichPlanegg-MartinsriedGermany
| | - Axel Imhof
- Department of Molecular Biology, Biomedical Center Munich, Ludwig-Maximilians UniversityPlanegg-MartinsriedGermany
- Protein Analysis Unit, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians, University (LMU) MunichPlanegg-MartinsriedGermany
| |
Collapse
|
28
|
Ngoi NYL, Pilié PG, McGrail DJ, Zimmermann M, Schlacher K, Yap TA. Targeting ATR in patients with cancer. Nat Rev Clin Oncol 2024; 21:278-293. [PMID: 38378898 DOI: 10.1038/s41571-024-00863-5] [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: 01/22/2024] [Indexed: 02/22/2024]
Abstract
Pharmacological inhibition of the ataxia telangiectasia and Rad3-related protein serine/threonine kinase (ATR; also known as FRAP-related protein (FRP1)) has emerged as a promising strategy for cancer treatment that exploits synthetic lethal interactions with proteins involved in DNA damage repair, overcomes resistance to other therapies and enhances antitumour immunity. Multiple novel, potent ATR inhibitors are being tested in clinical trials using biomarker-directed approaches and involving patients across a broad range of solid cancer types; some of these inhibitors have now entered phase III trials. Further insight into the complex interactions of ATR with other DNA replication stress response pathway components and with the immune system is necessary in order to optimally harness the potential of ATR inhibitors in the clinic and achieve hypomorphic targeting of the various ATR functions. Furthermore, a deeper understanding of the diverse range of predictive biomarkers of response to ATR inhibitors and of the intraclass differences between these agents could help to refine trial design and patient selection strategies. Key challenges that remain in the clinical development of ATR inhibitors include the optimization of their therapeutic index and the development of rational combinations with these agents. In this Review, we detail the molecular mechanisms regulated by ATR and their clinical relevance, and discuss the challenges that must be addressed to extend the benefit of ATR inhibitors to a broad population of patients with cancer.
Collapse
Affiliation(s)
- Natalie Y L Ngoi
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Patrick G Pilié
- Department of Genitourinary Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel J McGrail
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | | | - Katharina Schlacher
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Khalifa Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
29
|
Cook D, Kozmin SG, Yeh E, Petes TD, Bloom K. Dicentric chromosomes are resolved through breakage and repair at their centromeres. Chromosoma 2024; 133:117-134. [PMID: 38165460 PMCID: PMC11180013 DOI: 10.1007/s00412-023-00814-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/11/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Chromosomes with two centromeres provide a unique opportunity to study chromosome breakage and DNA repair using completely endogenous cellular machinery. Using a conditional transcriptional promoter to control the second centromere, we are able to activate the dicentric chromosome and follow the appearance of DNA repair products. We find that the rate of appearance of DNA repair products resulting from homology-based mechanisms exceeds the expected rate based on their limited centromere homology (340 bp) and distance from one another (up to 46.3 kb). In order to identify whether DNA breaks originate in the centromere, we introduced 12 single-nucleotide polymorphisms (SNPs) into one of the centromeres. Analysis of the distribution of SNPs in the recombinant centromeres reveals that recombination was initiated with about equal frequency within the conserved centromere DNA elements CDEII and CDEIII of the two centromeres. The conversion tracts range from about 50 bp to the full length of the homology between the two centromeres (340 bp). Breakage and repair events within and between the centromeres can account for the efficiency and distribution of DNA repair products. We propose that in addition to providing a site for kinetochore assembly, the centromere may be a point of stress relief in the face of genomic perturbations.
Collapse
Affiliation(s)
- Diana Cook
- Department of Biology, University of North Carolina Chapel Hill, Chapel Hill, NC, 27599-3280, USA
| | - Stanislav G Kozmin
- Department of Molecular Genetics & Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Elaine Yeh
- Department of Biology, University of North Carolina Chapel Hill, Chapel Hill, NC, 27599-3280, USA
| | - Thomas D Petes
- Department of Molecular Genetics & Microbiology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Kerry Bloom
- Department of Biology, University of North Carolina Chapel Hill, Chapel Hill, NC, 27599-3280, USA.
| |
Collapse
|
30
|
Piguet B, Houseley J. Transcription as source of genetic heterogeneity in budding yeast. Yeast 2024; 41:171-185. [PMID: 38196235 DOI: 10.1002/yea.3926] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 12/10/2023] [Accepted: 12/20/2023] [Indexed: 01/11/2024] Open
Abstract
Transcription presents challenges to genome stability both directly, by altering genome topology and exposing single-stranded DNA to chemical insults and nucleases, and indirectly by introducing obstacles to the DNA replication machinery. Such obstacles include the RNA polymerase holoenzyme itself, DNA-bound regulatory factors, G-quadruplexes and RNA-DNA hybrid structures known as R-loops. Here, we review the detrimental impacts of transcription on genome stability in budding yeast, as well as the mitigating effects of transcription-coupled nucleotide excision repair and of systems that maintain DNA replication fork processivity and integrity. Interactions between DNA replication and transcription have particular potential to induce mutation and structural variation, but we conclude that such interactions must have only minor effects on DNA replication by the replisome with little if any direct mutagenic outcome. However, transcription can significantly impair the fidelity of replication fork rescue mechanisms, particularly Break Induced Replication, which is used to restart collapsed replication forks when other means fail. This leads to de novo mutations, structural variation and extrachromosomal circular DNA formation that contribute to genetic heterogeneity, but only under particular conditions and in particular genetic contexts, ensuring that the bulk of the genome remains extremely stable despite the seemingly frequent interactions between transcription and DNA replication.
Collapse
|
31
|
Showman S, Talbert PB, Xu Y, Adeyemi RO, Henikoff S. Expansion of human centromeric arrays in cells undergoing break-induced replication. Cell Rep 2024; 43:113851. [PMID: 38427559 PMCID: PMC11034957 DOI: 10.1016/j.celrep.2024.113851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/10/2024] [Accepted: 02/07/2024] [Indexed: 03/03/2024] Open
Abstract
Human centromeres are located within α-satellite arrays and evolve rapidly, which can lead to individual variation in array length. Proposed mechanisms for such alterations in length are unequal crossover between sister chromatids, gene conversion, and break-induced replication. However, the underlying molecular mechanisms responsible for the massive, complex, and homogeneous organization of centromeric arrays have not been experimentally validated. Here, we use droplet digital PCR assays to demonstrate that centromeric arrays can expand and contract within ∼20 somatic cell divisions of an alternative lengthening of telomere (ALT)-positive cell line. We find that the frequency of array variation among single-cell-derived subclones ranges from a minimum of ∼7% to a maximum of ∼100%. Further clonal evolution revealed that centromere expansion is favored over contraction. We find that the homologous recombination protein RAD52 and the helicase PIF1 are required for extensive array change, suggesting that centromere sequence evolution can occur via break-induced replication.
Collapse
Affiliation(s)
- Soyeon Showman
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Paul B Talbert
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Yiling Xu
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Richard O Adeyemi
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA
| | - Steven Henikoff
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| |
Collapse
|
32
|
Yin L, Jiang N, Li T, Zhang Y, Yuan S. Telomeric function and regulation during male meiosis in mice and humans. Andrology 2024. [PMID: 38511802 DOI: 10.1111/andr.13631] [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: 07/06/2023] [Revised: 02/26/2024] [Accepted: 03/03/2024] [Indexed: 03/22/2024]
Abstract
BACKGROUND Telomeres are unique structures situated at the ends of chromosomes. Preserving the structure and function of telomeres is essential for maintaining genomic stability and promoting genetic diversity during male meiosis in mammals. MATERIAL-METHODS This review compiled recent literature on the function and regulation of telomeres during male meiosis in both mice and humans, and also highlighted the critical roles of telomeres in reproductive biology and medicine. RESULTS-DISCUSSION Various structures, consisting of the LINC complex (SUN-KASH), SPDYA-CDK2, TTM trimer (TERB1-TERB2-MAJIN), and shelterin, are critical in controlling telomeric activities, such as nuclear envelope attachment and bouquet formation. Other than telomere-related proteins, cohesins and genes responsible for regulating telomere function are also highlighted, though the exact mechanism remains unclear. The gene-mutant mouse models with meiotic defects directly reveal the essential roles of telomeres in male meiosis. Recently reported mutant genes associated with telomere activity in clinical practice have also been illustrated in detail. CONCLUSIONS Proper regulation of telomere activities is essential for male meiosis progression in mice and humans.
Collapse
Affiliation(s)
- Lisha Yin
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Nan Jiang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tao Li
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Youzhi Zhang
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology; Hubei Engineering Research Center of Traditional Chinese Medicine of South Hubei Province, Xianning, China
| | - Shuiqiao Yuan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Laboratory of Animal Center, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
33
|
Scelfo A, Angrisani A, Grillo M, Barnes BM, Muyas F, Sauer CM, Leung CWB, Dumont M, Grison M, Mazaud D, Garnier M, Guintini L, Nelson L, Esashi F, Cortés-Ciriano I, Taylor SS, Déjardin J, Wilhelm T, Fachinetti D. Specialized replication mechanisms maintain genome stability at human centromeres. Mol Cell 2024; 84:1003-1020.e10. [PMID: 38359824 DOI: 10.1016/j.molcel.2024.01.018] [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: 03/13/2023] [Revised: 12/12/2023] [Accepted: 01/19/2024] [Indexed: 02/17/2024]
Abstract
The high incidence of whole-arm chromosome aneuploidy and translocations in tumors suggests instability of centromeres, unique loci built on repetitive sequences and essential for chromosome separation. The causes behind this fragility and the mechanisms preserving centromere integrity remain elusive. We show that replication stress, hallmark of pre-cancerous lesions, promotes centromeric breakage in mitosis, due to spindle forces and endonuclease activities. Mechanistically, we unveil unique dynamics of the centromeric replisome distinct from the rest of the genome. Locus-specific proteomics identifies specialized DNA replication and repair proteins at centromeres, highlighting them as difficult-to-replicate regions. The translesion synthesis pathway, along with other factors, acts to sustain centromere replication and integrity. Prolonged stress causes centromeric alterations like ruptures and translocations, as observed in ovarian cancer models experiencing replication stress. This study provides unprecedented insights into centromere replication and integrity, proposing mechanistic insights into the origins of centromere alterations leading to abnormal cancerous karyotypes.
Collapse
Affiliation(s)
- Andrea Scelfo
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR144, 26 rue d'Ulm, Paris 75005, France
| | - Annapaola Angrisani
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR144, 26 rue d'Ulm, Paris 75005, France
| | - Marco Grillo
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR144, 26 rue d'Ulm, Paris 75005, France
| | - Bethany M Barnes
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Cancer Research Centre, Wilmslow Road, Manchester M20 4GJ, UK
| | - Francesc Muyas
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge CB10 1SD, UK
| | - Carolin M Sauer
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge CB10 1SD, UK
| | | | - Marie Dumont
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR144, 26 rue d'Ulm, Paris 75005, France
| | - Marine Grison
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR144, 26 rue d'Ulm, Paris 75005, France
| | - David Mazaud
- Plateforme Imagerie PICT-IBiSA, Institut Curie, PSL Research University, Paris 75005, France; Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR3664, 26 rue d'Ulm, Paris 75005, France
| | - Mickaël Garnier
- Plateforme Imagerie PICT-IBiSA, Institut Curie, PSL Research University, Paris 75005, France; Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR3664, 26 rue d'Ulm, Paris 75005, France
| | - Laetitia Guintini
- Institute of Human Genetics, CNRS-Université de Montpellier, Montpellier 34396, France
| | - Louisa Nelson
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Cancer Research Centre, Wilmslow Road, Manchester M20 4GJ, UK
| | - Fumiko Esashi
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, UK
| | - Isidro Cortés-Ciriano
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, Cambridge CB10 1SD, UK
| | - Stephen S Taylor
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Cancer Research Centre, Wilmslow Road, Manchester M20 4GJ, UK
| | - Jérôme Déjardin
- Institute of Human Genetics, CNRS-Université de Montpellier, Montpellier 34396, France
| | - Therese Wilhelm
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR144, 26 rue d'Ulm, Paris 75005, France; Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR3664, 26 rue d'Ulm, Paris 75005, France.
| | - Daniele Fachinetti
- Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR144, 26 rue d'Ulm, Paris 75005, France; Institut Curie, PSL Research University, Sorbonne Université, CNRS, UMR3664, 26 rue d'Ulm, Paris 75005, France.
| |
Collapse
|
34
|
Graham E, Esashi F. DNA strand breaks at centromeres: Friend or foe? Semin Cell Dev Biol 2024; 156:141-151. [PMID: 37872040 DOI: 10.1016/j.semcdb.2023.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 09/22/2023] [Accepted: 10/11/2023] [Indexed: 10/25/2023]
Abstract
Centromeres are large structural regions in the genomic DNA, which are essential for accurately transmitting a complete set of chromosomes to daughter cells during cell division. In humans, centromeres consist of highly repetitive α-satellite DNA sequences and unique epigenetic components, forming large proteinaceous structures required for chromosome segregation. Despite their biological importance, there is a growing body of evidence for centromere breakage across the cell cycle, including periods of quiescence. In this review, we provide an up-to-date examination of the distinct centromere environments at different stages of the cell cycle, highlighting their plausible contribution to centromere breakage. Additionally, we explore the implications of these breaks on centromere function, both in terms of negative consequences and potential positive effects.
Collapse
Affiliation(s)
- Emily Graham
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Fumiko Esashi
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| |
Collapse
|
35
|
Di Tommaso E, Giunta S. Dynamic interplay between human alpha-satellite DNA structure and centromere functions. Semin Cell Dev Biol 2024; 156:130-140. [PMID: 37926668 DOI: 10.1016/j.semcdb.2023.10.002] [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: 08/24/2023] [Revised: 10/04/2023] [Accepted: 10/10/2023] [Indexed: 11/07/2023]
Abstract
Maintenance of genome stability relies on functional centromeres for correct chromosome segregation and faithful inheritance of the genetic information. The human centromere is the primary constriction within mitotic chromosomes made up of repetitive alpha-satellite DNA hierarchically organized in megabase-long arrays of near-identical higher order repeats (HORs). Centromeres are epigenetically specified by the presence of the centromere-specific histone H3 variant, CENP-A, which enables the assembly of the kinetochore for microtubule attachment. Notably, centromeric DNA is faithfully inherited as intact haplotypes from the parents to the offspring without intervening recombination, yet, outside of meiosis, centromeres are akin to common fragile sites (CFSs), manifesting crossing-overs and ongoing sequence instability. Consequences of DNA changes within the centromere are just starting to emerge, with unclear effects on intra- and inter-generational inheritance driven by centromere's essential role in kinetochore assembly. Here, we review evidence of meiotic selection operating to mitigate centromere drive, as well as recent reports on centromere damage, recombination and repair during the mitotic cell division. We propose an antagonistic pleiotropy interpretation to reconcile centromere DNA instability as both driver of aneuploidy that underlies degenerative diseases, while also potentially necessary for the maintenance of homogenized HORs for centromere function. We attempt to provide a framework for this conceptual leap taking into consideration the structural interface of centromere-kinetochore interaction and present case scenarios for its malfunctioning. Finally, we offer an integrated working model to connect DNA instability, chromatin, and structural changes with functional consequences on chromosome integrity.
Collapse
Affiliation(s)
- Elena Di Tommaso
- Laboratory of Genome Evolution, Department of Biology & Biotechnology Charles Darwin, Sapienza University of Rome, Rome 00185, Italy
| | - Simona Giunta
- Laboratory of Genome Evolution, Department of Biology & Biotechnology Charles Darwin, Sapienza University of Rome, Rome 00185, Italy.
| |
Collapse
|
36
|
Marcon F, Giunta S, Bignami M. Emerging roles of DNA repair factors in the stability of centromeres. Semin Cell Dev Biol 2024; 156:121-129. [PMID: 37852903 DOI: 10.1016/j.semcdb.2023.10.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 10/07/2023] [Accepted: 10/10/2023] [Indexed: 10/20/2023]
Abstract
Satellite DNA sequences are an integral part of centromeres, regions critical for faithful segregation of chromosomes during cell division. Because of their complex repetitive structure, satellite DNA may act as a barrier to DNA replication and other DNA based transactions ultimately resulting in chromosome breakage. Over the past two decades, several DNA repair proteins have been shown to bind and function at centromeres. While the importance of these repair factors is highlighted by various structural and numerical chromosome aberrations resulting from their inactivation, their roles in helping to maintain genome stability by solving the intrinsic difficulties of satellite DNA replication or promoting their repair are just starting to emerge. In this review, we summarize the current knowledge on the role of DNA repair and DNA damage response proteins in maintaining the structure and function of centromeres in different contexts. We also report the recent connection between the roles of specific DNA repair factors at these genomic loci with age-related increase of chromosomal instability under physiological and pathological conditions.
Collapse
Affiliation(s)
- Francesca Marcon
- Department of Environment and Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| | - Simona Giunta
- Laboratory of Genome Evolution, Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185, Italy
| | - Margherita Bignami
- Department of Environment and Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
37
|
Black EM, Ramírez Parrado CA, Trier I, Li W, Joo YK, Pichurin J, Liu Y, Kabeche L. Chk2 sustains PLK1 activity in mitosis to ensure proper chromosome segregation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.08.584115. [PMID: 38559033 PMCID: PMC10979866 DOI: 10.1101/2024.03.08.584115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Polo-like kinase 1 (PLK1) protects against genome instability by ensuring timely and accurate mitotic cell division. PLK1 activity is tightly regulated throughout the cell cycle. Although the pathways that initially activate PLK1 in G2 are well-characterized, the factors that directly regulate PLK1 in mitosis remain poorly understood. Here, we identify that human PLK1 activity is sustained by the DNA damage response kinase Checkpoint kinase 2 (Chk2) in mitosis. Chk2 directly phosphorylates PLK1 T210, a residue on its T-loop whose phosphorylation is essential for full PLK1 kinase activity. Loss of Chk2-dependent PLK1 activity causes increased mitotic errors, including chromosome misalignment, chromosome missegregation, and cytokinetic defects. Moreover, Chk2 deficiency increases sensitivity to PLK1 inhibitors, suggesting that Chk2 status may be an informative biomarker for PLK1 inhibitor efficacy. This work demonstrates that Chk2 sustains mitotic PLK1 activity and protects genome stability through discrete functions in interphase DNA damage repair and mitotic chromosome segregation.
Collapse
|
38
|
Contreras A, Perea-Resa C. Transcriptional repression across mitosis: mechanisms and functions. Biochem Soc Trans 2024; 52:455-464. [PMID: 38372373 PMCID: PMC10903446 DOI: 10.1042/bst20231071] [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: 12/07/2023] [Revised: 02/05/2024] [Accepted: 02/06/2024] [Indexed: 02/20/2024]
Abstract
Transcription represents a central aspect of gene expression with RNA polymerase machineries (RNA Pol) driving the synthesis of RNA from DNA template molecules. In eukaryotes, a total of three RNA Pol enzymes generate the plethora of RNA species and RNA Pol II is the one transcribing all protein-coding genes. A high number of cis- and trans-acting factors orchestrates RNA Pol II-mediated transcription by influencing the chromatin recruitment, activation, elongation, and/or termination steps. The levels of DNA accessibility, defining open-euchromatin versus close-heterochromatin, delimits RNA Pol II activity as well as the encounter with other factors acting on chromatin such as the DNA replication or DNA repair machineries. The stage of the cell cycle highly influences RNA Pol II activity with mitosis representing the major challenge. In fact, there is a massive inhibition of transcription during the mitotic entry coupled with chromatin dissociation of most of the components of the transcriptional machinery. Mitosis, as a consequence, highly compromises the transcriptional memory and the perpetuation of cellular identity. Once mitosis ends, transcription levels immediately recover to define the cell fate and to safeguard the proper progression of daughter cells through the cell cycle. In this review, we evaluate our current understanding of the transcriptional repression associated with mitosis with a special focus on the molecular mechanisms involved, on the potential function behind the general repression, and on the transmission of the transcriptional machinery into the daughter cells. We finally discuss the contribution that errors in the inheritance of the transcriptional machinery across mitosis might play in stem cell aging.
Collapse
Affiliation(s)
- A. Contreras
- Centro de Biología Molecular Severo Ochoa (CBMSO-CSIC), C/Nicolas Cabrera 1, 28049 Madrid, Spain
| | - C. Perea-Resa
- Centro de Biología Molecular Severo Ochoa (CBMSO-CSIC), C/Nicolas Cabrera 1, 28049 Madrid, Spain
| |
Collapse
|
39
|
Pinto LM, Pailas A, Bondarchenko M, Sharma AB, Neumann K, Rizzo AJ, Jeanty C, Nicot N, Racca C, Graham MK, Naughton C, Liu Y, Chen CL, Meakin PJ, Gilbert N, Britton S, Meeker AK, Heaphy CM, Larminat F, Van Dyck E. DAXX promotes centromeric stability independently of ATRX by preventing the accumulation of R-loop-induced DNA double-stranded breaks. Nucleic Acids Res 2024; 52:1136-1155. [PMID: 38038252 PMCID: PMC10853780 DOI: 10.1093/nar/gkad1141] [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: 08/02/2022] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023] Open
Abstract
Maintaining chromatin integrity at the repetitive non-coding DNA sequences underlying centromeres is crucial to prevent replicative stress, DNA breaks and genomic instability. The concerted action of transcriptional repressors, chromatin remodelling complexes and epigenetic factors controls transcription and chromatin structure in these regions. The histone chaperone complex ATRX/DAXX is involved in the establishment and maintenance of centromeric chromatin through the deposition of the histone variant H3.3. ATRX and DAXX have also evolved mutually-independent functions in transcription and chromatin dynamics. Here, using paediatric glioma and pancreatic neuroendocrine tumor cell lines, we identify a novel ATRX-independent function for DAXX in promoting genome stability by preventing transcription-associated R-loop accumulation and DNA double-strand break formation at centromeres. This function of DAXX required its interaction with histone H3.3 but was independent of H3.3 deposition and did not reflect a role in the repression of centromeric transcription. DAXX depletion mobilized BRCA1 at centromeres, in line with BRCA1 role in counteracting centromeric R-loop accumulation. Our results provide novel insights into the mechanisms protecting the human genome from chromosomal instability, as well as potential perspectives in the treatment of cancers with DAXX alterations.
Collapse
Affiliation(s)
- Lia M Pinto
- DNA Repair and Chemoresistance Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
- Faculty of Science, Technology and Communication, University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
- Discovery & Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Alexandros Pailas
- DNA Repair and Chemoresistance Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
- Faculty of Science, Technology and Communication, University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
| | - Max Bondarchenko
- DNA Repair and Chemoresistance Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
- Faculty of Science, Technology and Communication, University of Luxembourg, L-4365 Esch-sur-Alzette, Luxembourg
| | - Abhishek Bharadwaj Sharma
- DNA Repair and Chemoresistance Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
| | - Katrin Neumann
- DNA Repair and Chemoresistance Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
| | - Anthony J Rizzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Céline Jeanty
- DNA Repair and Chemoresistance Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
| | - Nathalie Nicot
- Translational Medicine Operations Hub, Luxembourg Institute of Health (LIH), Luxembourg, Luxembourg
| | - Carine Racca
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), 31077 Toulouse Cedex 4, France
| | - Mindy K Graham
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Catherine Naughton
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh EH4 1QY, UK
| | - Yaqun Liu
- Institut Curie, PSL Research University, CNRS UMR3244, Dynamics of Genetic Information, Sorbonne Université, 75248 Paris Cedex 05, France
| | - Chun-Long Chen
- Institut Curie, PSL Research University, CNRS UMR3244, Dynamics of Genetic Information, Sorbonne Université, 75248 Paris Cedex 05, France
| | - Paul J Meakin
- Discovery & Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds LS2 9JT, UK
| | - Nick Gilbert
- Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh EH4 1QY, UK
| | - Sébastien Britton
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), 31077 Toulouse Cedex 4, France
| | - Alan K Meeker
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Christopher M Heaphy
- Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Florence Larminat
- Institut de Pharmacologie et Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), 31077 Toulouse Cedex 4, France
| | - Eric Van Dyck
- DNA Repair and Chemoresistance Group, Department of Cancer Research, Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
| |
Collapse
|
40
|
Abstract
Long non-coding RNAs (lncRNAs) are significant contributors in maintaining genomic integrity through epigenetic regulation. LncRNAs can interact with chromatin-modifying complexes in both cis and trans pathways, drawing them to specific genomic loci and influencing gene expression via DNA methylation, histone modifications, and chromatin remodeling. They can also operate as building blocks to assemble different chromatin-modifying components, facilitating their interactions and gene regulatory functions. Deregulation of these molecules has been associated with various human diseases, including cancer, cardiovascular disease, and neurological disorders. Thus, lncRNAs are implicated as potential diagnostic indicators and therapeutic targets. This review discusses the current understanding of how lncRNAs mediate epigenetic control, genomic integrity, and their putative functions in disease pathogenesis.
Collapse
Affiliation(s)
- Ganesan Arunkumar
- The LncRNA, Epigenetics, and Genome Organization Laboratory, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, NM, USA
| |
Collapse
|
41
|
Miglierina E, Ordanoska D, Le Noir S, Laffleur B. RNA processing mechanisms contribute to genome organization and stability in B cells. Oncogene 2024; 43:615-623. [PMID: 38287115 PMCID: PMC10890934 DOI: 10.1038/s41388-024-02952-2] [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: 08/29/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/31/2024]
Abstract
RNA processing includes post-transcriptional mechanisms controlling RNA quality and quantity to ensure cellular homeostasis. Noncoding (nc) RNAs that are regulated by these dynamic processes may themselves fulfill effector and/or regulatory functions, and recent studies demonstrated the critical role of RNAs in organizing both chromatin and genome architectures. Furthermore, RNAs can threaten genome integrity when accumulating as DNA:RNA hybrids, but could also facilitate DNA repair depending on the molecular context. Therefore, by qualitatively and quantitatively fine-tuning RNAs, RNA processing contributes directly or indirectly to chromatin states, genome organization, and genome stability. B lymphocytes represent a unique model to study these interconnected mechanisms as they express ncRNAs transcribed from key specific sequences before undergoing physiological genetic remodeling processes, including V(D)J recombination, somatic hypermutation, and class switch recombination. RNA processing actors ensure the regulation and degradation of these ncRNAs for efficient DNA repair and immunoglobulin gene remodeling while failure leads to B cell development alterations, aberrant DNA repair, and pathological translocations. This review highlights how RNA processing mechanisms contribute to genome architecture and stability, with emphasis on their critical roles during B cell development, enabling physiological DNA remodeling while preventing lymphomagenesis.
Collapse
Affiliation(s)
- Emma Miglierina
- University of Rennes, Inserm, EFS Bretagne, CHU Rennes, UMR, 1236, Rennes, France
| | - Delfina Ordanoska
- University of Rennes, Inserm, EFS Bretagne, CHU Rennes, UMR, 1236, Rennes, France
| | - Sandrine Le Noir
- UMR CNRS 7276, Inserm 1262, Université de Limoges: Contrôle de la Réponse Immune B et des Lymphoproliférations, Team 2, B-NATION: B cell Nuclear Architecture, Immunoglobulin genes and Oncogenes, Limoges, France
| | - Brice Laffleur
- University of Rennes, Inserm, EFS Bretagne, CHU Rennes, UMR, 1236, Rennes, France.
| |
Collapse
|
42
|
Zhan J, Wang J, Liang Y, Zeng X, Li E, Wang H. P53 together with ferroptosis: a promising strategy leaving cancer cells without escape. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1-14. [PMID: 38105650 PMCID: PMC10875350 DOI: 10.3724/abbs.2023270] [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: 06/08/2023] [Accepted: 08/03/2023] [Indexed: 12/19/2023] Open
Abstract
TP53, functioning as the keeper of the genome, assumes a pivotal function in the inhibition of tumorigenesis. Recent studies have revealed that p53 regulates ferroptosis pathways within tumor cells and is closely related to tumorigenesis. Therefore, we summarize the pathways and mechanisms by which p53 regulates ferroptosis and identify a series of upstream and downstream molecules involved in this process. Furthermore, we construct a p53-ferroptosis network centered on p53. Finally, we present the progress of drugs to prevent wild-type p53 (wtp53) degeneration and restore wtp53, highlighting the deficiencies of drug development and the prospects for p53 in cancer treatment. These findings provide novel strategies and directions for future cancer therapy.
Collapse
Affiliation(s)
- Jianhao Zhan
- Department of General SurgerySecond Affiliated Hospital of Nanchang UniversityNanchang330006China
- HuanKui AcademyNanchang UniversityNanchang330006China
| | - Jisheng Wang
- Department of General SurgerySecond Affiliated Hospital of Nanchang UniversityNanchang330006China
| | - Yuqing Liang
- School of Basic Medical SciencesNanchang UniversityNanchang330006China
| | - Xiaoping Zeng
- School of Basic Medical SciencesNanchang UniversityNanchang330006China
- Medical CollegeJinhua PolytechnicJinhua321017China
| | - Enliang Li
- Department of General SurgerySecond Affiliated Hospital of Nanchang UniversityNanchang330006China
| | - Hongmei Wang
- School of Basic Medical SciencesNanchang UniversityNanchang330006China
- Medical CollegeJinhua PolytechnicJinhua321017China
| |
Collapse
|
43
|
Kim S, Shin WH, Kang Y, Kim H, Lee JY. Direct visualization of replication and R-loop collision using single-molecule imaging. Nucleic Acids Res 2024; 52:259-273. [PMID: 37994723 PMCID: PMC10783495 DOI: 10.1093/nar/gkad1101] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/12/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023] Open
Abstract
R-loops are three-stranded nucleic acid structures that can cause replication stress by blocking replication fork progression. However, the detailed mechanism underlying the collision of DNA replication forks and R-loops remains elusive. To investigate how R-loops induce replication stress, we use single-molecule fluorescence imaging to directly visualize the collision of replicating Phi29 DNA polymerase (Phi29 DNAp), the simplest replication system, and R-loops. We demonstrate that a single R-loop can block replication, and the blockage is more pronounced when an RNA-DNA hybrid is on the non-template strand. We show that this asymmetry results from secondary structure formation on the non-template strand, which impedes the progression of Phi29 DNAp. We also show that G-quadruplex formation on the displaced single-stranded DNA in an R-loop enhances the replication stalling. Moreover, we observe the collision between Phi29 DNAp and RNA transcripts synthesized by T7 RNA polymerase (T7 RNAp). RNA transcripts cause more stalling because of the presence of T7 RNAp. Our work provides insights into how R-loops impede DNA replication at single-molecule resolution.
Collapse
Affiliation(s)
- Subin Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Woo Hee Shin
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Yujin Kang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Hongtae Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
| | - Ja Yil Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology, Ulsan 44919, Republic of Korea
- Institute of Basic Science Center for Genomic Integrity, Ulsan 44919, Republic of Korea
| |
Collapse
|
44
|
Xu R, Pan Z, Nakagawa T. Gross Chromosomal Rearrangement at Centromeres. Biomolecules 2023; 14:28. [PMID: 38254628 PMCID: PMC10813616 DOI: 10.3390/biom14010028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/19/2023] [Accepted: 12/22/2023] [Indexed: 01/24/2024] Open
Abstract
Centromeres play essential roles in the faithful segregation of chromosomes. CENP-A, the centromere-specific histone H3 variant, and heterochromatin characterized by di- or tri-methylation of histone H3 9th lysine (H3K9) are the hallmarks of centromere chromatin. Contrary to the epigenetic marks, DNA sequences underlying the centromere region of chromosomes are not well conserved through evolution. However, centromeres consist of repetitive sequences in many eukaryotes, including animals, plants, and a subset of fungi, including fission yeast. Advances in long-read sequencing techniques have uncovered the complete sequence of human centromeres containing more than thousands of alpha satellite repeats and other types of repetitive sequences. Not only tandem but also inverted repeats are present at a centromere. DNA recombination between centromere repeats can result in gross chromosomal rearrangement (GCR), such as translocation and isochromosome formation. CENP-A chromatin and heterochromatin suppress the centromeric GCR. The key player of homologous recombination, Rad51, safeguards centromere integrity through conservative noncrossover recombination between centromere repeats. In contrast to Rad51-dependent recombination, Rad52-mediated single-strand annealing (SSA) and microhomology-mediated end-joining (MMEJ) lead to centromeric GCR. This review summarizes recent findings on the role of centromere and recombination proteins in maintaining centromere integrity and discusses how GCR occurs at centromeres.
Collapse
Affiliation(s)
- Ran Xu
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Ziyi Pan
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Takuro Nakagawa
- Department of Biological Sciences, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Forefront Research Center, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| |
Collapse
|
45
|
Bothra A, Perry ML, Wei E, Moayeri M, Ma Q, Biamonte MA, Siirin M, Leppla SH. S9.6-based hybrid capture immunoassay for pathogen detection. Sci Rep 2023; 13:22562. [PMID: 38110611 PMCID: PMC10728093 DOI: 10.1038/s41598-023-49881-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: 10/20/2023] [Accepted: 12/13/2023] [Indexed: 12/20/2023] Open
Abstract
The detection of pathogens is critical for clinical diagnosis and public health surveillance. Detection is usually done with nucleic acid-based tests (NATs) and rapid antigen tests (e.g., lateral flow assays [LFAs]). Although NATs are more sensitive and specific, their use is often limited in resource-poor settings due to specialized requirements. To address this limitation, we developed a rapid DNA-RNA Hybrid Capture immunoassay (HC) that specifically detects RNA from pathogens. This assay utilizes a unique monoclonal antibody, S9.6, which binds DNA-RNA hybrids. Biotinylated single-stranded DNA probes are hybridized to target RNAs, followed by hybrid capture on streptavidin and detection with S9.6. The HC-ELISA assay can detect as few as 104 RNA molecules that are 2.2 kb in length. We also adapted this assay into a LFA format, where captured Bacillus anthracis rpoB RNA of 3.5 kb length was detectable from a bacterial load equivalent to 107 CFU per 100 mg of mouse tissue using either HC-ELISA or HC-LFA. Importantly, we also demonstrated the versatility of HC by detecting other pathogens, including SARS-CoV-2 and Toxoplasma gondii, showing its potential for broad pathogen detection. Notably, HC does not require amplification of the target nucleic acid and utilizes economical formats like ELISA and LFA, making it suitable for use in sentinel labs for pathogen detection or as a molecular tool in basic research laboratories. Our study highlights the potential of HC as a sensitive and versatile method for RNA-based pathogen detection.
Collapse
Affiliation(s)
- Ankur Bothra
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA.
| | - Megan L Perry
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Elena Wei
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Mahtab Moayeri
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Qian Ma
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | | | - Marina Siirin
- Drugs and Diagnostics for Tropical Diseases, San Diego, CA, USA
| | - Stephen H Leppla
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| |
Collapse
|
46
|
Girasol MJ, Krasilnikova M, Marques CA, Damasceno JD, Lapsley C, Lemgruber L, Burchmore R, Beraldi D, Carruthers R, Briggs EM, McCulloch R. RAD51-mediated R-loop formation acts to repair transcription-associated DNA breaks driving antigenic variation in Trypanosoma brucei. Proc Natl Acad Sci U S A 2023; 120:e2309306120. [PMID: 37988471 PMCID: PMC10691351 DOI: 10.1073/pnas.2309306120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 09/13/2023] [Indexed: 11/23/2023] Open
Abstract
RNA-DNA hybrids are epigenetic features of all genomes that intersect with many processes, including transcription, telomere homeostasis, and centromere function. Increasing evidence suggests that RNA-DNA hybrids can provide two conflicting roles in the maintenance and transmission of genomes: They can be the triggers of DNA damage, leading to genome change, or can aid the DNA repair processes needed to respond to DNA lesions. Evasion of host immunity by African trypanosomes, such as Trypanosoma brucei, relies on targeted recombination of silent Variant Surface Glycoprotein (VSG) genes into a specialized telomeric locus that directs transcription of just one VSG from thousands. How such VSG recombination is targeted and initiated is unclear. Here, we show that a key enzyme of T. brucei homologous recombination, RAD51, interacts with RNA-DNA hybrids. In addition, we show that RNA-DNA hybrids display a genome-wide colocalization with DNA breaks and that this relationship is impaired by mutation of RAD51. Finally, we show that RAD51 acts to repair highly abundant, localised DNA breaks at the single transcribed VSG and that mutation of RAD51 alters RNA-DNA hybrid abundance at 70 bp repeats both around the transcribed VSG and across the silent VSG archive. This work reveals a widespread, generalised role for RNA-DNA hybrids in directing RAD51 activity during recombination and uncovers a specialised application of this interplay during targeted DNA break repair needed for the critical T. brucei immune evasion reaction of antigenic variation.
Collapse
Affiliation(s)
- Mark John Girasol
- College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, University of Glasgow, GlasgowG12 8TA, United Kingdom
- Faculty of the MD-PhD in Molecular Medicine Program, College of Medicine, University of the Philippines Manila, Manila1000, Philippines
| | - Marija Krasilnikova
- College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, University of Glasgow, GlasgowG12 8TA, United Kingdom
| | - Catarina A. Marques
- College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, University of Glasgow, GlasgowG12 8TA, United Kingdom
| | - Jeziel D. Damasceno
- College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, University of Glasgow, GlasgowG12 8TA, United Kingdom
| | - Craig Lapsley
- College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, University of Glasgow, GlasgowG12 8TA, United Kingdom
| | - Leandro Lemgruber
- College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, University of Glasgow, GlasgowG12 8TA, United Kingdom
| | - Richard Burchmore
- College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, University of Glasgow, GlasgowG12 8TA, United Kingdom
| | - Dario Beraldi
- College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, University of Glasgow, GlasgowG12 8TA, United Kingdom
| | - Ross Carruthers
- College of Medical, Veterinary and Life Sciences, School of Cancer Sciences, University of Glasgow, GlasgowG12 0YN, United Kingdom
| | - Emma M. Briggs
- College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, University of Glasgow, GlasgowG12 8TA, United Kingdom
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, EdinburghEH9 3FL, United Kingdom
| | - Richard McCulloch
- College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, University of Glasgow, GlasgowG12 8TA, United Kingdom
| |
Collapse
|
47
|
Showman S, Talbert PB, Xu Y, Adeyemi RO, Henikoff S. Expansion of human centromeric arrays in cells undergoing break-induced replication. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.11.566714. [PMID: 38014305 PMCID: PMC10680626 DOI: 10.1101/2023.11.11.566714] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Human centromeres are located within α-satellite arrays and evolve rapidly, which can lead to individual variation in array lengths. Proposed mechanisms for such alterations in lengths are unequal cross-over between sister chromatids, gene conversion, and break-induced replication. However, the underlying molecular mechanisms responsible for the massive, complex, and homogeneous organization of centromeric arrays have not been experimentally validated. Here, we use droplet digital PCR assays to demonstrate that centromeric arrays can expand and contract within ~20 somatic cell divisions of a cell line. We find that the frequency of array variation among single-cell-derived subclones ranges from a minimum of ~7% to a maximum of ~100%. Further clonal evolution revealed that centromere expansion is favored over contraction. We find that the homologous recombination protein RAD52 and the helicase PIF1 are required for extensive array change, suggesting that centromere sequence evolution can occur via break-induced replication.
Collapse
Affiliation(s)
- Soyeon Showman
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Paul B. Talbert
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Yiling Xu
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Richard O. Adeyemi
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Steven Henikoff
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| |
Collapse
|
48
|
Girasol MJ, Briggs EM, Marques CA, Batista JM, Beraldi D, Burchmore R, Lemgruber L, McCulloch R. Immunoprecipitation of RNA-DNA hybrid interacting proteins in Trypanosoma brucei reveals conserved and novel activities, including in the control of surface antigen expression needed for immune evasion by antigenic variation. Nucleic Acids Res 2023; 51:11123-11141. [PMID: 37843098 PMCID: PMC10639054 DOI: 10.1093/nar/gkad836] [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: 05/26/2023] [Revised: 09/14/2023] [Accepted: 09/28/2023] [Indexed: 10/17/2023] Open
Abstract
RNA-DNA hybrids are epigenetic features of genomes that provide a diverse and growing range of activities. Understanding of these functions has been informed by characterising the proteins that interact with the hybrids, but all such analyses have so far focused on mammals, meaning it is unclear if a similar spectrum of RNA-DNA hybrid interactors is found in other eukaryotes. The African trypanosome is a single-cell eukaryotic parasite of the Discoba grouping and displays substantial divergence in several aspects of core biology from its mammalian host. Here, we show that DNA-RNA hybrid immunoprecipitation coupled with mass spectrometry recovers 602 putative interactors in T. brucei mammal- and insect-infective cells, some providing activities also found in mammals and some lineage-specific. We demonstrate that loss of three factors, two putative helicases and a RAD51 paralogue, alters T. brucei nuclear RNA-DNA hybrid and DNA damage levels. Moreover, loss of each factor affects the operation of the parasite immune survival mechanism of antigenic variation. Thus, our work reveals the broad range of activities contributed by RNA-DNA hybrids to T. brucei biology, including new functions in host immune evasion as well as activities likely fundamental to eukaryotic genome function.
Collapse
Affiliation(s)
- Mark J Girasol
- University of Glasgow, College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, Glasgow, UK
- University of the Philippines Manila, College of Medicine, Manila, Philippines
| | - Emma M Briggs
- University of Glasgow, College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, Glasgow, UK
- University of Edinburgh, Institute for Immunology and Infection Research, School of Biological Sciences, Edinburgh, UK
| | - Catarina A Marques
- University of Glasgow, College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, Glasgow, UK
| | - José M Batista
- University of Glasgow, College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, Glasgow, UK
| | - Dario Beraldi
- University of Glasgow, College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, Glasgow, UK
| | - Richard Burchmore
- University of Glasgow, College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, Glasgow, UK
| | - Leandro Lemgruber
- University of Glasgow, College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, Glasgow, UK
| | - Richard McCulloch
- University of Glasgow, College of Medical, Veterinary and Life Sciences, School of Infection and Immunity, Wellcome Centre for Integrative Parasitology, Glasgow, UK
| |
Collapse
|
49
|
Sokhi S, Lewis CW, Bukhari AB, Hadfield J, Xiao EJ, Fung J, Yoon YJ, Hsu WH, Gamper AM, Chan GK. Myt1 overexpression mediates resistance to cell cycle and DNA damage checkpoint kinase inhibitors. Front Cell Dev Biol 2023; 11:1270542. [PMID: 38020882 PMCID: PMC10652759 DOI: 10.3389/fcell.2023.1270542] [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: 07/31/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Cell cycle checkpoint kinases serve as important therapeutic targets for various cancers. When they are inhibited by small molecules, checkpoint abrogation can induce cell death or further sensitize cancer cells to other genotoxic therapies. Particularly aberrant Cdk1 activation at the G2/M checkpoint by kinase inhibitors causing unscheduled mitotic entry and mitotic arrest was found to lead to DNA damage and cell death selectively in cancer cells. Promising drugs inhibiting kinases like Wee1 (Adavosertib), Wee1+Myt1 (PD166285), ATR (AZD6738) and Chk1 (UCN-01) have been developed, but clinical data has shown variable efficacy for them with poorly understood mechanisms of resistance. Our lab recently identified Myt1 as a predictive biomarker of acquired resistance to the Wee1 kinase inhibitor, Adavosertib. Here, we investigate the role of Myt1 overexpression in promoting resistance to inhibitors (PD166285, UCN-01 and AZD6738) of other kinases regulating cell cycle progression. We demonstrate that Myt1 confers resistance by compensating Cdk1 inhibition in the presence of these different kinase inhibitors. Myt1 overexpression leads to reduced premature mitotic entry and decreased length of mitosis eventually leading to increased survival rates in Adavosertib treated cells. Elevated Myt1 levels also conferred resistance to inhibitors of ATR or Chk1 inhibitor. Our data supports that Myt1 overexpression is a common mechanism by which cancer cells can acquire resistance to a variety of drugs entering the clinic that aim to induce mitotic catastrophe by abrogating the G2/M checkpoint.
Collapse
Affiliation(s)
- Sargun Sokhi
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Experimental Oncology, Cross Cancer Institute, Edmonton, AB, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
| | - Cody W. Lewis
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Experimental Oncology, Cross Cancer Institute, Edmonton, AB, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
| | - Amirali B. Bukhari
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Experimental Oncology, Cross Cancer Institute, Edmonton, AB, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
| | - Joanne Hadfield
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Experimental Oncology, Cross Cancer Institute, Edmonton, AB, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
| | - Edric J. Xiao
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Experimental Oncology, Cross Cancer Institute, Edmonton, AB, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
| | - Jeremy Fung
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Experimental Oncology, Cross Cancer Institute, Edmonton, AB, Canada
| | - Yea Jin Yoon
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Experimental Oncology, Cross Cancer Institute, Edmonton, AB, Canada
| | - Wen-Hsin Hsu
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Experimental Oncology, Cross Cancer Institute, Edmonton, AB, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
| | - Armin M. Gamper
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Experimental Oncology, Cross Cancer Institute, Edmonton, AB, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
| | - Gordon K. Chan
- Department of Oncology, University of Alberta, Edmonton, AB, Canada
- Experimental Oncology, Cross Cancer Institute, Edmonton, AB, Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
50
|
Gambelli A, Ferrando A, Boncristiani C, Schoeftner S. Regulation and function of R-loops at repetitive elements. Biochimie 2023; 214:141-155. [PMID: 37619810 DOI: 10.1016/j.biochi.2023.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/13/2023] [Accepted: 08/19/2023] [Indexed: 08/26/2023]
Abstract
R-loops are atypical, three-stranded nucleic acid structures that contain a stretch of RNA:DNA hybrids and an unpaired, single stranded DNA loop. R-loops are physiological relevant and can act as regulators of gene expression, chromatin structure, DNA damage repair and DNA replication. However, unscheduled and persistent R-loops are mutagenic and can mediate replication-transcription conflicts, leading to DNA damage and genome instability if left unchecked. Detailed transcriptome analysis unveiled that 85% of the human genome, including repetitive regions, hold transcriptional activity. This anticipates that R-loops management plays a central role for the regulation and integrity of genomes. This function is expected to have a particular relevance for repetitive sequences that make up to 75% of the human genome. Here, we review the impact of R-loops on the function and stability of repetitive regions such as centromeres, telomeres, rDNA arrays, transposable elements and triplet repeat expansions and discuss their relevance for associated pathological conditions.
Collapse
Affiliation(s)
- Alice Gambelli
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Via E. Weiss 2, 34127, Trieste, Italy
| | - Alessandro Ferrando
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Via E. Weiss 2, 34127, Trieste, Italy
| | - Chiara Boncristiani
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Via E. Weiss 2, 34127, Trieste, Italy
| | - Stefan Schoeftner
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Via E. Weiss 2, 34127, Trieste, Italy.
| |
Collapse
|