1
|
Eickhoff P, Sonmez C, Fisher CEL, Inian O, Roumeliotis TI, Dello Stritto A, Mansfeld J, Choudhary JS, Guettler S, Lottersberger F, Douglas ME. Chromosome end protection by RAP1-mediated inhibition of DNA-PK. Nature 2025:10.1038/s41586-025-08896-1. [PMID: 40240611 DOI: 10.1038/s41586-025-08896-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/13/2025] [Indexed: 04/18/2025]
Abstract
During classical non-homologous end joining (cNHEJ), DNA-dependent protein kinase (DNA-PK) encapsulates free DNA ends, forming a recruitment platform for downstream end-joining factors including ligase 4 (LIG4)1. DNA-PK can also bind telomeres and regulate their resection2-4, but does not initiate cNHEJ at this position. How the end-joining process is regulated in this context-specific manner is currently unclear. Here we show that the shelterin components TRF2 and RAP1 form a complex with DNA-PK that directly represses its end-joining function at telomeres. Biochemical experiments and cryo-electron microscopy reveal that when bound to TRF2, RAP1 establishes a network of interactions with KU and DNA that prevents DNA-PK from recruiting LIG4. In mouse and human cells, RAP1 is redundant with the Apollo nuclease in repressing cNHEJ at chromosome ends, demonstrating that the inhibition of DNA-PK prevents telomere fusions in parallel with overhang-dependent mechanisms. Our experiments show that the end-joining function of DNA-PK is directly and specifically repressed at telomeres, establishing a molecular mechanism for how individual linear chromosomes are maintained in mammalian cells.
Collapse
Affiliation(s)
- Patrik Eickhoff
- Telomere Biology Laboratory, The Institute of Cancer Research, London, UK
| | - Ceylan Sonmez
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | | | - Oviya Inian
- Structural Biology of Cell Signalling, The Institute of Cancer Research, London, UK
| | | | - Angela Dello Stritto
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Jörg Mansfeld
- Post-translational Modifications and Cell Proliferation, The Institute of Cancer Research, London, UK
| | | | - Sebastian Guettler
- Structural Biology of Cell Signalling, The Institute of Cancer Research, London, UK
| | | | - Max E Douglas
- Telomere Biology Laboratory, The Institute of Cancer Research, London, UK.
| |
Collapse
|
2
|
Romero-Zamora D, Rogers S, Low RRJ, Page SG, Lane BJE, Kosaka S, Robinson AB, French L, Lamm N, Ishikawa F, Hayashi MT, Cesare AJ. A CPC-shelterin-BTR axis regulates mitotic telomere deprotection. Nat Commun 2025; 16:2277. [PMID: 40097392 PMCID: PMC11914695 DOI: 10.1038/s41467-025-57456-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Telomeres prevent ATM activation by sequestering chromosome termini within telomere loops (t-loops). Mitotic arrest promotes telomere linearity and a localized ATM-dependent telomere DNA damage response (DDR) through an unknown mechanism. Using unbiased interactomics, biochemical screening, molecular biology, and super-resolution imaging, we found that mitotic arrest-dependent (MAD) telomere deprotection requires the combined activities of the Chromosome passenger complex (CPC) on shelterin, and the BLM-TOP3A-RMI1/2 (BTR) complex on t-loops. During mitotic arrest, the CPC component Aurora Kinase B (AURKB) phosphorylated both the TRF1 hinge and TRF2 basic domains. Phosphorylation of the TRF1 hinge domain enhances CPC and TRF1 interaction through the CPC Survivin subunit. Meanwhile, phosphorylation of the TRF2 basic domain promotes telomere linearity, activates a telomere DDR dependent on BTR-mediated double Holliday junction dissolution, and leads to mitotic death. We identify that the TRF2 basic domain functions in mitosis-specific telomere protection and reveal a regulatory role for TRF1 in controlling a physiological ATM-dependent telomere DDR. The data demonstrate that MAD telomere deprotection is a sophisticated active mechanism that exposes telomere ends to signal mitotic stress.
Collapse
Affiliation(s)
- Diana Romero-Zamora
- Graduate School of Biostudies, Kyoto University, Sakyo, Kyoto, Japan
- IFOM-KU Joint Research Laboratory, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto, Japan
| | - Samuel Rogers
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Ronnie Ren Jie Low
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Scott G Page
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Blake J E Lane
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Shunya Kosaka
- Graduate School of Biostudies, Kyoto University, Sakyo, Kyoto, Japan
- IFOM-KU Joint Research Laboratory, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto, Japan
| | - Andrew B Robinson
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Lucy French
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Noa Lamm
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia
| | - Fuyuki Ishikawa
- Graduate School of Biostudies, Kyoto University, Sakyo, Kyoto, Japan
| | - Makoto T Hayashi
- Graduate School of Biostudies, Kyoto University, Sakyo, Kyoto, Japan.
- IFOM-KU Joint Research Laboratory, Graduate School of Medicine, Kyoto University, Sakyo, Kyoto, Japan.
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy.
| | - Anthony J Cesare
- Children's Medical Research Institute, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
3
|
Odintsov I, Papke DJ, George S, Padera RF, Hornick JL, Siegmund SE. Genomic Profiling of Cardiac Angiosarcoma Reveals Novel Targetable KDR Variants, Recurrent MED12 Mutations, and a High Burden of Germline POT1 Alterations. Clin Cancer Res 2025; 31:1091-1102. [PMID: 39820259 DOI: 10.1158/1078-0432.ccr-24-3277] [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: 10/02/2024] [Revised: 12/04/2024] [Accepted: 01/14/2025] [Indexed: 01/19/2025]
Abstract
PURPOSE Cardiac angiosarcoma is a rare, aggressive malignancy with limited treatment options. Both sporadic and familial cases occur, with recent links to germline POT1 mutations. The genomic landscape of this disease is poorly understood. EXPERIMENTAL DESIGN We conducted comprehensive genomic profiling of cardiac angiosarcoma to assess the burden of germline predisposition and identify other recurrent genomic alterations of clinical significance. RESULTS Six patients were female, and four were male. The median age at presentation was 40 years (range, 21-69 years). All cases with available follow-up exhibited an aggressive clinical course (6/8 patients died of disease). KDR alterations, including novel structural variants, were found in 9/11 cases at a rate significantly higher than that in noncardiac angiosarcomas. POT1 mutations were present in 45.5% of cardiac angiosarcoma cases. In three of five POT1-mutant cases, the germline status was confirmed through testing of normal tissue, and in one additional case, the germline status was inferred with high probability through allele frequency analysis. Additionally, we identified novel recurrent MED12 exon 2 mutations in POT1 wild-type cardiac angiosarcoma, suggesting an alternative path to cardiac angiosarcoma oncogenesis. CONCLUSIONS Cardiac angiosarcoma demonstrates a unique genetic profile, distinct from noncardiac angiosarcoma. This study highlights the role of germline POT1 burden on cardiac angiosarcoma development and demonstrates recurrent MED12 alterations for the first time. The reported KDR variants provide a potential avenue for the treatment of this aggressive disease. Given the prevalence of germline POT1 mutations reported in this study, germline genetic testing should be considered in patients diagnosed with cardiac angiosarcoma.
Collapse
Affiliation(s)
- Igor Odintsov
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - David J Papke
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Suzanne George
- Sarcoma Division, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Robert F Padera
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jason L Hornick
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Stephanie E Siegmund
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
4
|
Giles KA, Taberlay PC, Cesare AJ, Jones MJK. Roles for the 3D genome in the cell cycle, DNA replication, and double strand break repair. Front Cell Dev Biol 2025; 13:1548946. [PMID: 40083661 PMCID: PMC11903485 DOI: 10.3389/fcell.2025.1548946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Large eukaryotic genomes are packaged into the restricted area of the nucleus to protect the genetic code and provide a dedicated environment to read, copy and repair DNA. The physical organisation of the genome into chromatin loops and self-interacting domains provides the basic structural units of genome architecture. These structural arrangements are complex, multi-layered, and highly dynamic and influence how different regions of the genome interact. The role of chromatin structures during transcription via enhancer-promoter interactions is well established. Less understood is how nuclear architecture influences the plethora of chromatin transactions during DNA replication and repair. In this review, we discuss how genome architecture is regulated during the cell cycle to influence the positioning of replication origins and the coordination of DNA double strand break repair. The role of genome architecture in these cellular processes highlights its critical involvement in preserving genome integrity and cancer prevention.
Collapse
Affiliation(s)
- Katherine A. Giles
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Phillippa C. Taberlay
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Anthony J. Cesare
- Children’s Medical Research Institute, University of Sydney, Westmead, NSW, Australia
| | - Mathew J. K. Jones
- Faculty of Medicine, Frazer Institute, University of Queensland, Brisbane, QLD, Australia
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
5
|
Xu X, Wang T. Autoregulation of TRF2 through G-Quadruplex-Specific Interaction between the Gene and N-Terminal Domain of the Protein. Biochemistry 2025; 64:57-66. [PMID: 39705116 DOI: 10.1021/acs.biochem.4c00287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2024]
Abstract
Telomere repeat-binding factor 2 (TRF2) is a key component of the shelterin complex which guards the integrity of the telomere. Most of the TRF2 discussed previously was focused on the telomere, and relatively less is discussed on aspects other than that. It is proved that TRF2 also localizes to other potential G-quadruplex-forming sequences among the whole genome besides the telomere. Therefore, it may participate in regulating genes generally except for the well-known function of protecting telomeres. Here, we demonstrate that the N-terminal basic domain of TRF2 (TRF2B) can interact with the G-quadruplex formed by the 5'-UTR sequence of its gene. Subsequently, this interaction was identified as G-quadruplex-specific. Using a reporter gene system, we proved that the translation of the reporter gene was dramatically reduced, triggered by the interaction between TRF2B and the G-quadruplex. Altogether, we propose that TRF2 can be "auto-regulated" through the G-quadruplex formed by its own gene sequence. This finding indicates a potential feedback mechanism in the regulation of the TRF2 gene. Additionally, it suggests a common mode in gene regulation involving the cooperation of TRF2 and the G-quadruplex.
Collapse
Affiliation(s)
- Xiaojuan Xu
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
- School of Biology and Food Engineering, Hefei Normal University, Hefei 230031, China
| | - Tao Wang
- High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China
| |
Collapse
|
6
|
Szmyd R, Casolin S, French L, Manjón AG, Walter M, Cavalli L, Nelson CB, Page SG, Dhawan A, Hau E, Pickett HA, Gee HE, Cesare AJ. Homologous recombination promotes non-immunogenic mitotic cell death upon DNA damage. Nat Cell Biol 2025; 27:59-72. [PMID: 39805921 PMCID: PMC11735404 DOI: 10.1038/s41556-024-01557-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/14/2024] [Indexed: 01/16/2025]
Abstract
Double-strand breaks (DSBs) can initiate mitotic catastrophe, a complex oncosuppressive phenomenon characterized by cell death during or after cell division. Here we unveil how cell cycle-regulated DSB repair guides disparate cell death outcomes through single-cell analysis of extended live imaging. Following DSB induction in S or G2, passage of unresolved homologous recombination intermediates into mitosis promotes non-immunogenic intrinsic apoptosis in the immediate attempt at cell division. Conversely, non-homologous end joining, microhomology-mediated end joining and single-strand annealing cooperate to enable damaged G1 cells to complete the first cell cycle with an aberrant cell division at the cost of delayed extrinsic lethality and interferon production. Targeting non-homologous end joining, microhomology-mediated end joining or single-strand annealing promotes mitotic death, while suppressing mitotic death enhances interferon production. Together the data indicate that a temporal repair hierarchy, coupled with cumulative DSB load, serves as a reliable predictor of mitotic catastrophe outcomes following genome damage. In this pathway, homologous recombination suppresses interferon production by promoting mitotic lethality.
Collapse
Affiliation(s)
- Radoslaw Szmyd
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Sienna Casolin
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Lucy French
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Anna G Manjón
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Melanie Walter
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - Léa Cavalli
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Christopher B Nelson
- Telomere Length Regulation Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Scott G Page
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Andrew Dhawan
- Neurological Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Eric Hau
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia
- Westmead Clinical School, University of Sydney, Westmead, New South Wales, Australia
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Hilda A Pickett
- Telomere Length Regulation Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia
| | - Harriet E Gee
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia.
- Radiation Oncology Network, Western Sydney Local Health District, Sydney, New South Wales, Australia.
- Westmead Clinical School, University of Sydney, Westmead, New South Wales, Australia.
- Translational Radiation Biology and Oncology Laboratory, Centre for Cancer Research, The Westmead Institute for Medical Research, Westmead, New South Wales, Australia.
| | - Anthony J Cesare
- Genome Integrity Unit, Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, Australia.
| |
Collapse
|
7
|
Janovič T, Perez GI, Schmidt JC. TRF1 and TRF2 form distinct shelterin subcomplexes at telomeres. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.23.630076. [PMID: 39763972 PMCID: PMC11703185 DOI: 10.1101/2024.12.23.630076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
The shelterin complex protects chromosome ends from the DNA damage repair machinery and regulates telomerase access to telomeres. Shelterin is composed of six proteins (TRF1, TRF2, TIN2, TPP1, POT1 and RAP1) that can assemble into various subcomplexes in vitro. However, the stoichiometry of the shelterin complex and its dynamic association with telomeres in cells is poorly defined. To quantitatively analyze the shelterin function in living cells we generated a panel of cancer cell lines expressing HaloTagged shelterin proteins from their endogenous loci. We systematically determined the total cellular abundance and telomeric copy number of each shelterin subunit, demonstrating that the shelterin proteins are present at telomeres in equal numbers. In addition, we used single-molecule live-cell imaging to analyze the dynamics of shelterin protein association with telomeres. Our results demonstrate that TRF1-TIN2-TPP1-POT1 and TRF2-RAP1 form distinct subcomplexes that occupy non-overlapping binding sites on telomeric chromatin. TRF1-TIN2-TPP1-POT1 tightly associates with chromatin, while TRF2-RAP1 binding to telomeres is more dynamic, allowing it to recruit a variety of co-factors to chromatin to protect chromosome ends from DNA repair factors. In total, our work provides critical mechanistic insight into how the shelterin proteins carry out multiple essential functions in telomere maintenance and significantly advances our understanding of macromolecular structure of telomeric chromatin.
Collapse
Affiliation(s)
- Tomáš Janovič
- Institute for Quantitative Health Science and Engineering, Gynecology and Reproductive Biology, Michigan State University, East Lansing
| | - Gloria I. Perez
- Institute for Quantitative Health Science and Engineering, Gynecology and Reproductive Biology, Michigan State University, East Lansing
| | - Jens C. Schmidt
- Institute for Quantitative Health Science and Engineering, Gynecology and Reproductive Biology, Michigan State University, East Lansing
- Department of Obstetrics, Gynecology and Reproductive Biology, Michigan State University, East Lansing
| |
Collapse
|
8
|
Zhang B, Zhao Y. Association Analysis of Telomere Length and Vision in a Large Community-Based Survey. Ophthalmic Epidemiol 2024:1-7. [PMID: 39531590 DOI: 10.1080/09286586.2024.2422349] [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: 04/08/2024] [Revised: 10/12/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
PURPOSE To investigate whether there is a direct, age-independent association between telomere length and visual acuity decline in a large community-based cohort study. METHODS Participants older than 40 with linked leukocyte telomere length (LTL) were enrolled in NHANES. LTL was assayed using qPCR from the participants' blood samples. Best corrected visual acuity (BCVA) of the better-seeing eye was analyzed, with visual impairment (VI) defined as the BCVA of 20/40 or worse. LTL was grouped into quartiles, and its association with BCVA and VI was evaluated after adjusting for covariates. RESULTS Among the 4,480 enrolled participants, the weighted means of age, BCVA, and telomere length were 56.1 ± 11.9 years, 0.05 ± 0.08 logMAR, and 5,662 ± 36 base pairs, respectively. The proportion of VI was 2.6%. After adjusting for covariates including sex, ethnicity, education, family poverty income ratio, general health status, hypertension, diabetes, smoking, and body mass index, BCVA was significantly worse in participants with shorter LTL, with a significant trend (p = 0.002). However, after further adjusting for age, the association between LTL and BCVA was no longer significant, without a trend (p = 0.640). No significant association or trend between LTL and VI was found in the stepwise logistic model. CONCLUSIONS No age-independent association between LTL and BCVA was found. Our study indicates LTL may not serve as a biomarker for age-related visual acuity decline.
Collapse
Affiliation(s)
- Bing Zhang
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, China
| | - Yune Zhao
- Eye Hospital and School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Clinical Research Center for Ocular Diseases, Wenzhou, Zhejiang, China
| |
Collapse
|
9
|
Harman A, Bryan TM. Telomere maintenance and the DNA damage response: a paradoxical alliance. Front Cell Dev Biol 2024; 12:1472906. [PMID: 39483338 PMCID: PMC11524846 DOI: 10.3389/fcell.2024.1472906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/07/2024] [Indexed: 11/03/2024] Open
Abstract
Telomeres are the protective caps at the ends of linear chromosomes of eukaryotic organisms. Telomere binding proteins, including the six components of the complex known as shelterin, mediate the protective function of telomeres. They do this by suppressing many arms of the canonical DNA damage response, thereby preventing inappropriate fusion, resection and recombination of telomeres. One way this is achieved is by facilitation of DNA replication through telomeres, thus protecting against a "replication stress" response and activation of the master kinase ATR. On the other hand, DNA damage responses, including replication stress and ATR, serve a positive role at telomeres, acting as a trigger for recruitment of the telomere-elongating enzyme telomerase to counteract telomere loss. We postulate that repression of telomeric replication stress is a shared mechanism of control of telomerase recruitment and telomere length, common to several core telomere binding proteins including TRF1, POT1 and CTC1. The mechanisms by which replication stress and ATR cause recruitment of telomerase are not fully elucidated, but involve formation of nuclear actin filaments that serve as anchors for stressed telomeres. Perturbed control of telomeric replication stress by mutations in core telomere binding proteins can therefore cause the deregulation of telomere length control characteristic of diseases such as cancer and telomere biology disorders.
Collapse
Affiliation(s)
| | - Tracy M. Bryan
- Cell Biology Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| |
Collapse
|
10
|
Schmidt TT, Tyer C, Rughani P, Haggblom C, Jones JR, Dai X, Frazer KA, Gage FH, Juul S, Hickey S, Karlseder J. High resolution long-read telomere sequencing reveals dynamic mechanisms in aging and cancer. Nat Commun 2024; 15:5149. [PMID: 38890299 PMCID: PMC11189484 DOI: 10.1038/s41467-024-48917-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 05/15/2024] [Indexed: 06/20/2024] Open
Abstract
Telomeres are the protective nucleoprotein structures at the end of linear eukaryotic chromosomes. Telomeres' repetitive nature and length have traditionally challenged the precise assessment of the composition and length of individual human telomeres. Here, we present Telo-seq to resolve bulk, chromosome arm-specific and allele-specific human telomere lengths using Oxford Nanopore Technologies' native long-read sequencing. Telo-seq resolves telomere shortening in five population doubling increments and reveals intrasample, chromosome arm-specific, allele-specific telomere length heterogeneity. Telo-seq can reliably discriminate between telomerase- and ALT-positive cancer cell lines. Thus, Telo-seq is a tool to study telomere biology during development, aging, and cancer at unprecedented resolution.
Collapse
Affiliation(s)
| | - Carly Tyer
- Oxford Nanopore Technologies, Inc., New York, NY, USA
| | | | - Candy Haggblom
- Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Jeffrey R Jones
- Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Xiaoguang Dai
- Oxford Nanopore Technologies, Inc., New York, NY, USA
| | - Kelly A Frazer
- Institute of Genomic Medicine, University of California, San Diego, La Jolla, CA, 92093-0761, USA
| | - Fred H Gage
- Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Sissel Juul
- Oxford Nanopore Technologies, Inc., New York, NY, USA
| | - Scott Hickey
- Oxford Nanopore Technologies, Inc., New York, NY, USA.
| | - Jan Karlseder
- Salk Institute for Biological Studies, La Jolla, CA, 92037, USA.
| |
Collapse
|
11
|
Qiu YD, Yan Q, Wang Y, Ye YF, Wang Y, Wang MY, Wang PP, Zhang SY, Wang DL, Yan H, Ruan J, Zhao YJ, Huang LH, Cho N, Wang K, Zheng XH, Liu ZG. Discovery of a selective TRF2 inhibitor FKB04 induced telomere shortening and senescence in liver cancer cells. Acta Pharmacol Sin 2024; 45:1276-1286. [PMID: 38438580 PMCID: PMC11130216 DOI: 10.1038/s41401-024-01243-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 02/11/2024] [Indexed: 03/06/2024]
Abstract
Telomere repeat binding factor 2 (TRF2), a critical element of the shelterin complex, plays a vital role in the maintenance of genome integrity. TRF2 overexpression is found in a wide range of malignant cancers, whereas its down-regulation could cause cell death. Despite its potential role, the selectively small-molecule inhibitors of TRF2 and its therapeutic effects on liver cancer remain largely unknown. Our clinical data combined with bioinformatic analysis demonstrated that TRF2 is overexpressed in liver cancer and that high expression is associated with poor prognosis. Flavokavain B derivative FKB04 potently inhibited TRF2 expression in liver cancer cells while having limited effects on the other five shelterin subunits. Moreover, FKB04 treatment induced telomere shortening and increased the amounts of telomere-free ends, leading to the destruction of T-loop structure. Consequently, FKB04 promoted liver cancer cell senescence without modulating apoptosis levels. In corroboration with these findings, FKB04 inhibited tumor cell growth by promoting telomeric TRF2 deficiency-induced telomere shortening in a mouse xenograft tumor model, with no obvious side effects. These results demonstrate that TRF2 is a potential therapeutic target for liver cancer and suggest that FKB04 may be a selective small-molecule inhibitor of TRF2, showing promise in the treatment of liver cancer.
Collapse
Affiliation(s)
- Yin-da Qiu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
- College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Qi Yan
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yi Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yan-Fei Ye
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yan Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Meng-Ying Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Pei-Pei Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Shu-Yuan Zhang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Da-Long Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hao Yan
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Jing Ruan
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China.
| | - Yun-Jie Zhao
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Le-Hao Huang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Namki Cho
- College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Kun Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiao-Hui Zheng
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China.
| | - Zhi-Guo Liu
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China.
- Oujiang Laboratory, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
12
|
Rai R, Sodeinde T, Boston A, Chang S. Telomeres cooperate with the nuclear envelope to maintain genome stability. Bioessays 2024; 46:e2300184. [PMID: 38047499 DOI: 10.1002/bies.202300184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/12/2023] [Accepted: 11/14/2023] [Indexed: 12/05/2023]
Abstract
Mammalian telomeres have evolved safeguards to prevent their recognition as DNA double-stranded breaks by suppressing the activation of various DNA sensing and repair proteins. We have shown that the telomere-binding proteins TRF2 and RAP1 cooperate to prevent telomeres from undergoing aberrant homology-directed recombination by mediating t-loop protection. Our recent findings also suggest that mammalian telomere-binding proteins interact with the nuclear envelope to maintain chromosome stability. RAP1 interacts with nuclear lamins through KU70/KU80, and disruption of RAP1 and TRF2 function result in nuclear envelope rupture, promoting telomere-telomere recombination to form structures termed ultrabright telomeres. In this review, we discuss the importance of the interactions between shelterin components and the nuclear envelope to maintain telomere homeostasis and genome stability.
Collapse
Affiliation(s)
- Rekha Rai
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Tori Sodeinde
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Molecular, Cellular and Developmental Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ava Boston
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Molecular, Cellular and Developmental Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sandy Chang
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
13
|
Li B. Unwrap RAP1's Mystery at Kinetoplastid Telomeres. Biomolecules 2024; 14:67. [PMID: 38254667 PMCID: PMC10813129 DOI: 10.3390/biom14010067] [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: 12/06/2023] [Revised: 12/27/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
Although located at the chromosome end, telomeres are an essential chromosome component that helps maintain genome integrity and chromosome stability from protozoa to mammals. The role of telomere proteins in chromosome end protection is conserved, where they suppress various DNA damage response machineries and block nucleolytic degradation of the natural chromosome ends, although the detailed underlying mechanisms are not identical. In addition, the specialized telomere structure exerts a repressive epigenetic effect on expression of genes located at subtelomeres in a number of eukaryotic organisms. This so-called telomeric silencing also affects virulence of a number of microbial pathogens that undergo antigenic variation/phenotypic switching. Telomere proteins, particularly the RAP1 homologs, have been shown to be a key player for telomeric silencing. RAP1 homologs also suppress the expression of Telomere Repeat-containing RNA (TERRA), which is linked to their roles in telomere stability maintenance. The functions of RAP1s in suppressing telomere recombination are largely conserved from kinetoplastids to mammals. However, the underlying mechanisms of RAP1-mediated telomeric silencing have many species-specific features. In this review, I will focus on Trypanosoma brucei RAP1's functions in suppressing telomeric/subtelomeric DNA recombination and in the regulation of monoallelic expression of subtelomere-located major surface antigen genes. Common and unique mechanisms will be compared among RAP1 homologs, and their implications will be discussed.
Collapse
Affiliation(s)
- Bibo Li
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, College of Arts and Sciences, Cleveland State University, 2121 Euclid Avenue, Cleveland, OH 44115, USA;
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
- Center for RNA Science and Therapeutics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| |
Collapse
|
14
|
Jeon HJ, Levine MT, Lampson MA. Telomere Elongation During Pre-Implantation Embryo Development. ADVANCES IN ANATOMY, EMBRYOLOGY, AND CELL BIOLOGY 2024; 238:121-129. [PMID: 39030357 DOI: 10.1007/978-3-031-55163-5_6] [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] [Indexed: 07/21/2024]
Abstract
The primary mechanism of telomere elongation in mammals is reverse transcription by telomerase. An alternative (ALT) pathway elongates telomeres by homologous recombination in some cancer cells and during pre-implantation embryo development, when telomere length increases rapidly within a few cell cycles. The maternal and paternal telomeres in the zygote are genetically and epigenetically distinct, with differences in telomere length and in chromatin packaging. We discuss models for how these asymmetries may contribute to telomere regulation during the earliest embryonic cell cycles and suggest directions for future research.
Collapse
Affiliation(s)
- Hyuk-Joon Jeon
- Department of Biology and Penn Center for Genome Integrity, University of Pennsylvania, Philadelphia, PA, USA
| | - Mia T Levine
- Department of Biology and Penn Center for Genome Integrity, University of Pennsylvania, Philadelphia, PA, USA.
| | - Michael A Lampson
- Department of Biology and Penn Center for Genome Integrity, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
15
|
Sobinoff AP, Di Maro S, Low RRJ, Benedetti R, Tomassi S, D'Aniello A, Russo R, Baglivo I, Chianese U, Pedone PV, Chambery A, Cesare AJ, Altucci L, Pickett HA, Cosconati S. Irreversible inhibition of TRF2 TRFH recruiting functions by a covalent cyclic peptide induces telomeric replication stress in cancer cells. Cell Chem Biol 2023; 30:1652-1665.e6. [PMID: 38065101 DOI: 10.1016/j.chembiol.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/28/2023] [Accepted: 11/13/2023] [Indexed: 12/24/2023]
Abstract
The TRF2 shelterin component is an essential regulator of telomere homeostasis and genomic stability. Mutations in the TRF2TRFH domain physically impair t-loop formation and prevent the recruitment of several factors that promote efficient telomere replication, causing telomeric DNA damage. Here, we design, synthesize, and biologically test covalent cyclic peptides that irreversibly target the TRF2TRFH domain. We identify APOD53 as our most promising compound, as it consistently induces a telomeric DNA damage response in cancer cell lines. APOD53 forms a covalent adduct with a reactive cysteine residue present in the TRF2TRFH domain and induces phenotypes consistent with TRF2TRFH domain mutants. These include induction of a telomeric DNA damage response, increased telomeric replication stress, and impaired recruitment of RTEL1 and SLX4 to telomeres. We demonstrate that APOD53 impairs cancer cell growth and find that co-treatment with APOD53 can exacerbate telomere replication stress caused by the G4 stabilizer RHPS4 and low dose aphidicolin (APH).
Collapse
Affiliation(s)
- Alexander P Sobinoff
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Salvatore Di Maro
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Ronnie R J Low
- Genome Integrity Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Rosaria Benedetti
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138 Naples, Italy; Azienda Ospedaliera Universitaria "Luigi Vanvitelli", Medical Epigenetics Program
| | - Stefano Tomassi
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, 80131 Naples, Italy
| | - Antonia D'Aniello
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Rosita Russo
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Ilaria Baglivo
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Ugo Chianese
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138 Naples, Italy
| | - Paolo V Pedone
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Angela Chambery
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy
| | - Anthony J Cesare
- Genome Integrity Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Vico L. De Crecchio 7, 80138 Naples, Italy; BIOGEM, 83031 Ariano Irpino, Italy; Azienda Ospedaliera Universitaria "Luigi Vanvitelli", Medical Epigenetics Program; IEOS CNR, Napoli, Italy
| | - Hilda A Pickett
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia.
| | - Sandro Cosconati
- DiSTABiF, University of Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100 Caserta, Italy.
| |
Collapse
|
16
|
Rivosecchi J, Cusanelli E. TERRA beyond cancer: the biology of telomeric repeat-containing RNAs in somatic and germ cells. FRONTIERS IN AGING 2023; 4:1224225. [PMID: 37636218 PMCID: PMC10448526 DOI: 10.3389/fragi.2023.1224225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023]
Abstract
The telomeric noncoding RNA TERRA is a key component of telomeres and it is widely expressed in normal as well as cancer cells. In the last 15 years, several publications have shed light on the role of TERRA in telomere homeostasis and cell survival in cancer cells. However, only few studies have investigated the regulation or the functions of TERRA in normal tissues. A better understanding of the biology of TERRA in non-cancer cells may provide unexpected insights into how these lncRNAs are transcribed and operate in cells, and their potential role in physiological processes, such as aging, age-related pathologies, inflammatory processes and human genetic diseases. In this review we aim to discuss the findings that have advanced our understanding of the biology of TERRA using non-cancer mammalian cells as a model system.
Collapse
Affiliation(s)
- Julieta Rivosecchi
- Laboratory of Cell Biology and Molecular Genetics, Department of Cellular, Computational and Integrative Biology—CIBIO, University of Trento, Trento, Italy
| | | |
Collapse
|
17
|
Irvin EM, Wang H. Single-molecule imaging of genome maintenance proteins encountering specific DNA sequences and structures. DNA Repair (Amst) 2023; 128:103528. [PMID: 37392578 PMCID: PMC10989508 DOI: 10.1016/j.dnarep.2023.103528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 06/08/2023] [Accepted: 06/21/2023] [Indexed: 07/03/2023]
Abstract
DNA repair pathways are tightly regulated processes that recognize specific hallmarks of DNA damage and coordinate lesion repair through discrete mechanisms, all within the context of a three-dimensional chromatin landscape. Dysregulation or malfunction of any one of the protein constituents in these pathways can contribute to aging and a variety of diseases. While the collective action of these many proteins is what drives DNA repair on the organismal scale, it is the interactions between individual proteins and DNA that facilitate each step of these pathways. In much the same way that ensemble biochemical techniques have characterized the various steps of DNA repair pathways, single-molecule imaging (SMI) approaches zoom in further, characterizing the individual protein-DNA interactions that compose each pathway step. SMI techniques offer the high resolving power needed to characterize the molecular structure and functional dynamics of individual biological interactions on the nanoscale. In this review, we highlight how our lab has used SMI techniques - traditional atomic force microscopy (AFM) imaging in air, high-speed AFM (HS-AFM) in liquids, and the DNA tightrope assay - over the past decade to study protein-nucleic acid interactions involved in DNA repair, mitochondrial DNA replication, and telomere maintenance. We discuss how DNA substrates containing specific DNA sequences or structures that emulate DNA repair intermediates or telomeres were generated and validated. For each highlighted project, we discuss novel findings made possible by the spatial and temporal resolution offered by these SMI techniques and unique DNA substrates.
Collapse
Affiliation(s)
| | - Hong Wang
- Toxicology Program, North Carolina State University, Raleigh, NC, USA; Physics Department, North Carolina State University, Raleigh, NC, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
18
|
Bloom SI, Liu Y, Tucker JR, Islam MT, Machin DR, Abdeahad H, Thomas TG, Bramwell RC, Lesniewski LA, Donato AJ. Endothelial cell telomere dysfunction induces senescence and results in vascular and metabolic impairments. Aging Cell 2023; 22:e13875. [PMID: 37259606 PMCID: PMC10410008 DOI: 10.1111/acel.13875] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/03/2023] [Accepted: 05/04/2023] [Indexed: 06/02/2023] Open
Abstract
In advanced age, increases in oxidative stress and inflammation impair endothelial function, which contributes to the development of cardiovascular disease (CVD). One plausible source of this oxidative stress and inflammation is an increase in the abundance of senescent endothelial cells. Cellular senescence is a cell cycle arrest that occurs in response to various damaging stimuli. In the present study, we tested the hypothesis that advanced age results in endothelial cell telomere dysfunction that induces senescence. In both human and mouse endothelial cells, advanced age resulted in an increased abundance of dysfunctional telomeres, characterized by activation of DNA damage signaling at telomeric DNA. To test whether this results in senescence, we selectively reduced the telomere shelterin protein telomere repeat binding factor 2 (Trf2) from endothelial cells of young mice. Trf2 reduction increased endothelial cell telomere dysfunction and resulted in cellular senescence. Furthermore, induction of endothelial cell telomere dysfunction increased inflammatory signaling and oxidative stress, resulting in impairments in endothelial function. Finally, we demonstrate that endothelial cell telomere dysfunction-induced senescence impairs glucose tolerance. This likely occurs through increases in inflammatory signaling in the liver and adipose tissue, as well as reductions in microvascular density and vasodilation to metabolic stimuli. Cumulatively, the findings of the present study identify age-related telomere dysfunction as a mechanism that leads to endothelial cell senescence. Furthermore, these data provide compelling evidence that senescent endothelial cells contribute to age-related increases in oxidative stress and inflammation that impair arterial and metabolic function.
Collapse
Affiliation(s)
- Samuel I. Bloom
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
| | - Yu Liu
- Department of GeriatricsTongji HospitalWuhanChina
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Jordan R. Tucker
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Md Torikul Islam
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
| | - Daniel R. Machin
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyFlorida State UniversityTallahasseeFloridaUSA
| | - Hossein Abdeahad
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
| | - Tyler G. Thomas
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - R. Colton Bramwell
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
| | - Lisa A. Lesniewski
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
- Geriatric Research, Education and Clinical CenterVeteran's Affairs Medical Center‐Salt Lake CitySalt Lake CityUtahUSA
- Nora Eccles Harrison Cardiovascular Research and Training InstituteThe University of UtahSalt Lake CityUtahUSA
| | - Anthony J. Donato
- Department of Nutrition and Integrative PhysiologyThe University of UtahSalt Lake CityUtahUSA
- Division of Geriatrics, Department of Internal MedicineUniversity of Utah School of MedicineSalt Lake CityUtahUSA
- Geriatric Research, Education and Clinical CenterVeteran's Affairs Medical Center‐Salt Lake CitySalt Lake CityUtahUSA
- Nora Eccles Harrison Cardiovascular Research and Training InstituteThe University of UtahSalt Lake CityUtahUSA
- Department of BiochemistryThe University of UtahSalt Lake CityUtahUSA
| |
Collapse
|
19
|
Carson LM, Flynn RL. Highlighting vulnerabilities in the alternative lengthening of telomeres pathway. Curr Opin Pharmacol 2023; 70:102380. [PMID: 37149932 PMCID: PMC10247456 DOI: 10.1016/j.coph.2023.102380] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/03/2023] [Accepted: 03/28/2023] [Indexed: 05/09/2023]
Abstract
The alternative lengthening of telomeres (ALT) pathway is a telomere elongation mechanism found in a small but often aggressive subset of cancers. Dependent on break-induced replication, telomere extension in ALT-positive cells relies on a baseline level of DNA replication stress to initiate elongation events. This results in an elevated level of DNA damage and presents a possible vulnerability to be exploited in the development of ALT-targeted cancer therapies. Currently, there are no treatment options that target the ALT mechanism or that are specific for ALT-positive tumors. Here, we review recent developments and promising directions in the development of ALT-targeted therapeutics, many of which involve tipping the balance towards inhibition or exacerbation of ALT activity to selectively target these cells.
Collapse
Affiliation(s)
- Lisa M Carson
- Departments of Pharmacology and Experimental Therapeutics, and Medicine, Cancer Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Rachel L Flynn
- Departments of Pharmacology and Experimental Therapeutics, and Medicine, Cancer Center, Boston University School of Medicine, Boston, MA, 02118, USA.
| |
Collapse
|
20
|
Storchova R, Palek M, Palkova N, Veverka P, Brom T, Hofr C, Macurek L. Phosphorylation of TRF2 promotes its interaction with TIN2 and regulates DNA damage response at telomeres. Nucleic Acids Res 2023; 51:1154-1172. [PMID: 36651296 PMCID: PMC9943673 DOI: 10.1093/nar/gkac1269] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 11/25/2022] [Accepted: 12/23/2022] [Indexed: 01/19/2023] Open
Abstract
Protein phosphatase magnesium-dependent 1 delta (PPM1D) terminates the cell cycle checkpoint by dephosphorylating the tumour suppressor protein p53. By targeting additional substrates at chromatin, PPM1D contributes to the control of DNA damage response and DNA repair. Using proximity biotinylation followed by proteomic analysis, we identified a novel interaction between PPM1D and the shelterin complex that protects telomeric DNA. In addition, confocal microscopy revealed that endogenous PPM1D localises at telomeres. Further, we found that ATR phosphorylated TRF2 at S410 after induction of DNA double strand breaks at telomeres and this modification increased after inhibition or loss of PPM1D. TRF2 phosphorylation stimulated its interaction with TIN2 both in vitro and at telomeres. Conversely, induced expression of PPM1D impaired localisation of TIN2 and TPP1 at telomeres. Finally, recruitment of the DNA repair factor 53BP1 to the telomeric breaks was strongly reduced after inhibition of PPM1D and was rescued by the expression of TRF2-S410A mutant. Our results suggest that TRF2 phosphorylation promotes the association of TIN2 within the shelterin complex and regulates DNA repair at telomeres.
Collapse
Affiliation(s)
- Radka Storchova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| | - Matous Palek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| | - Natalie Palkova
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| | - Pavel Veverka
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno CZ-62500, Czech Republic
| | - Tomas Brom
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno CZ-62500, Czech Republic
| | - Ctirad Hofr
- LifeB, Functional Genomics and Proteomics, National Centre for Biomolecular Research, Faculty of Science, Masaryk University, Brno CZ-62500, Czech Republic
| | - Libor Macurek
- Cancer Cell Biology, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague CZ-14220, Czech Republic
| |
Collapse
|
21
|
Romero-Zamora D, Hayashi MT. A non-catalytic N-terminus domain of WRN prevents mitotic telomere deprotection. Sci Rep 2023; 13:645. [PMID: 36635307 PMCID: PMC9837040 DOI: 10.1038/s41598-023-27598-0] [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: 08/19/2022] [Accepted: 01/04/2023] [Indexed: 01/14/2023] Open
Abstract
Telomeric ends form a loop structure (T-loop) necessary for the repression of ATM kinase activation throughout the normal cell cycle. However, cells undergoing a prolonged mitotic arrest are prone to lose the T-loop, resulting in Aurora B kinase-dependent mitotic telomere deprotection, which was proposed as an anti-tumor mechanism that eliminates precancerous cells from the population. The mechanism of mitotic telomere deprotection has not been elucidated. Here, we show that WRN, a RECQ helicase family member, can suppress mitotic telomere deprotection independently of its exonuclease and helicase activities. Truncation of WRN revealed that N-terminus amino acids 168-333, a region that contains a coiled-coil motif, is sufficient to suppress mitotic telomere deprotection without affecting both mitotic Aurora B-dependent spindle checkpoint and ATM kinase activity. The suppressive activity of the WRN168-333 fragment is diminished in cells partially depleted of TRF2, while WRN is required for complete suppression of mitotic telomere deprotection by TRF2 overexpression. Finally, we found that phosphomimetic but not alanine mutations of putative Aurora B target sites in the WRN168-333 fragment abolished its suppressive effect. Our findings reveal a non-enzymatic function of WRN, which may be regulated by phosphorylation in cells undergoing mitotic arrest. We propose that WRN enhances the protective function of TRF2 to counteract the hypothetical pathway that resolves the mitotic T-loop.
Collapse
Affiliation(s)
- Diana Romero-Zamora
- grid.258799.80000 0004 0372 2033Graduate School of Biostudies, Kyoto University, Yoshida-Konoe, Sakyo, Kyoto, 606-8501 Japan ,grid.258799.80000 0004 0372 2033IFOM-KU Joint Research Laboratory, Graduate School of Medicine, Kyoto University, Yoshida-Konoe, Sakyo, Kyoto, 606-8501 Japan
| | - Makoto T. Hayashi
- grid.258799.80000 0004 0372 2033IFOM-KU Joint Research Laboratory, Graduate School of Medicine, Kyoto University, Yoshida-Konoe, Sakyo, Kyoto, 606-8501 Japan ,IFOM ETS, The AIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| |
Collapse
|
22
|
Zhang Y, Liu J, Li X, Zhou G, Sang Y, Zhang M, Gao L, Xue J, Zhao M, Yu H, Zhou X. Dietary selenium excess affected spermatogenesis via DNA damage and telomere-related cell senescence and apoptosis in mice. Food Chem Toxicol 2023; 171:113556. [DOI: 10.1016/j.fct.2022.113556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 11/21/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
|
23
|
Soman A, Korolev N, Nordenskiöld L. Telomeric chromatin structure. Curr Opin Struct Biol 2022; 77:102492. [PMID: 36335846 DOI: 10.1016/j.sbi.2022.102492] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/06/2022]
Abstract
Eukaryotic DNA is packaged into nucleosomes, which further condenses into chromosomes. The telomeres, which form the protective end-capping of chromosomes, play a pivotal role in ageing and cancer. Recently, significant advances have been made in understanding the nucleosomal and telomeric chromatin structure at the molecular level. In addition, recent studies shed light on the nucleosomal organisation at telomeres revealing its ultrastructural organisation, the atomic structure at the nucleosome level, its dynamic properties, and higher-order packaging of telomeric chromatin. Considerable advances have furthermore been made in understanding the structure, function and organisation of shelterin, telomerase and CST complexes. Here we discuss these recent advances in the organisation of telomeric nucleosomes and chromatin and highlight progress in the structural understanding of shelterin, telomerase and CST complexes.
Collapse
Affiliation(s)
- Aghil Soman
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore.
| | - Nikolay Korolev
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Lars Nordenskiöld
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore; NTU Institute of Structural Biology, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
24
|
Gao J, Pickett HA. Targeting telomeres: advances in telomere maintenance mechanism-specific cancer therapies. Nat Rev Cancer 2022; 22:515-532. [PMID: 35790854 DOI: 10.1038/s41568-022-00490-1] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2022] [Indexed: 12/31/2022]
Abstract
Cancer cells establish replicative immortality by activating a telomere-maintenance mechanism (TMM), be it telomerase or the alternative lengthening of telomeres (ALT) pathway. Targeting telomere maintenance represents an intriguing opportunity to treat the vast majority of all cancer types. Whilst telomerase inhibitors have historically been heralded as promising anticancer agents, the reality has been more challenging, and there are currently no therapeutic options for cancer types that use ALT despite their aggressive nature and poor prognosis. In this Review, we discuss the mechanistic differences between telomere maintenance by telomerase and ALT, the current methods used to detect each mechanism, the utility of these tests for clinical diagnosis, and recent developments in the therapeutic strategies being employed to target both telomerase and ALT. We present notable developments in repurposing established therapeutic agents and new avenues that are emerging to target cancer types according to which TMM they employ. These opportunities extend beyond inhibition of telomere maintenance, by finding and exploiting inherent weaknesses in the telomeres themselves to trigger rapid cellular effects that lead to cell death.
Collapse
Affiliation(s)
- Jixuan Gao
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia
| | - Hilda A Pickett
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia.
| |
Collapse
|
25
|
Yu EY, Cheung NKV, Lue NF. Connecting telomere maintenance and regulation to the developmental origin and differentiation states of neuroblastoma tumor cells. J Hematol Oncol 2022; 15:117. [PMID: 36030273 PMCID: PMC9420296 DOI: 10.1186/s13045-022-01337-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/28/2022] [Indexed: 11/18/2022] Open
Abstract
A cardinal feature that distinguishes clinically high-risk neuroblastoma from low-risk tumors is telomere maintenance. Specifically, neuroblastoma tumors with either active telomerase or alternative lengthening of telomeres exhibit aggressive growth characteristics that lead to poor outcomes, whereas tumors without telomere maintenance can be managed with observation or minimal treatment. Even though the need for cancer cells to maintain telomere DNA-in order to sustain cell proliferation-is well established, recent studies suggest that the neural crest origin of neuroblastoma may enforce unique relationships between telomeres and tumor malignancy. Specifically in neuroblastoma, telomere structure and telomerase activity are correlated with the adrenergic/mesenchymal differentiation states, and manipulating telomerase activity can trigger tumor cell differentiation. Both findings may reflect features of normal neural crest development. This review summarizes recent advances in the characterization of telomere structure and telomere maintenance mechanisms in neuroblastoma and discusses the findings in the context of relevant literature on telomeres during embryonic and neural development. Understanding the canonical and non-canonical roles of telomere maintenance in neuroblastoma could reveal vulnerabilities for telomere-directed therapies with potential applications to other pediatric malignancies.
Collapse
Affiliation(s)
- Eun Young Yu
- Department of Microbiology & Immunology, W. R. Hearst Microbiology Research Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Nai-Kong V Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Neal F Lue
- Department of Microbiology & Immunology, W. R. Hearst Microbiology Research Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA.
| |
Collapse
|
26
|
Barnes RP, de Rosa M, Thosar SA, Detwiler AC, Roginskaya V, Van Houten B, Bruchez MP, Stewart-Ornstein J, Opresko PL. Telomeric 8-oxo-guanine drives rapid premature senescence in the absence of telomere shortening. Nat Struct Mol Biol 2022; 29:639-652. [PMID: 35773409 PMCID: PMC9287163 DOI: 10.1038/s41594-022-00790-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 05/16/2022] [Indexed: 01/10/2023]
Abstract
Oxidative stress is a primary cause of cellular senescence and contributes to the etiology of numerous human diseases. Oxidative damage to telomeric DNA has been proposed to cause premature senescence by accelerating telomere shortening. Here, we tested this model directly using a precision chemoptogenetic tool to produce the common lesion 8-oxo-guanine (8oxoG) exclusively at telomeres in human fibroblasts and epithelial cells. A single induction of telomeric 8oxoG is sufficient to trigger multiple hallmarks of p53-dependent senescence. Telomeric 8oxoG activates ATM and ATR signaling, and enriches for markers of telomere dysfunction in replicating, but not quiescent cells. Acute 8oxoG production fails to shorten telomeres, but rather generates fragile sites and mitotic DNA synthesis at telomeres, indicative of impaired replication. Based on our results, we propose that oxidative stress promotes rapid senescence by producing oxidative base lesions that drive replication-dependent telomere fragility and dysfunction in the absence of shortening and shelterin loss. This study uncovers a new mechanism linking oxidative stress to telomere-driven senescence. A common oxidative lesion at telomeres causes rapid premature cellular aging by inducing telomere fragility, rather than telomere shortening.
Collapse
Affiliation(s)
- Ryan P Barnes
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA.,UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Mariarosaria de Rosa
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA.,UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Sanjana A Thosar
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA.,UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Ariana C Detwiler
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA.,UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Vera Roginskaya
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Bennett Van Houten
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Marcel P Bruchez
- Departments of Biological Sciences and Chemistry and the Molecular Biosensors and Imaging Center, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Jacob Stewart-Ornstein
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patricia L Opresko
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA. .,UPMC Hillman Cancer Center, Pittsburgh, PA, USA. .,Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
27
|
Fernandez RJ, Gardner ZJG, Slovik KJ, Liberti DC, Estep KN, Yang W, Chen Q, Santini GT, Perez JV, Root S, Bhatia R, Tobias JW, Babu A, Morley MP, Frank DB, Morrisey EE, Lengner CJ, Johnson FB. GSK3 inhibition rescues growth and telomere dysfunction in dyskeratosis congenita iPSC-derived type II alveolar epithelial cells. eLife 2022; 11:64430. [PMID: 35559731 PMCID: PMC9200405 DOI: 10.7554/elife.64430] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/11/2022] [Indexed: 11/27/2022] Open
Abstract
Dyskeratosis congenita (DC) is a rare genetic disorder characterized by deficiencies in telomere maintenance leading to very short telomeres and the premature onset of certain age-related diseases, including pulmonary fibrosis (PF). PF is thought to derive from epithelial failure, particularly that of type II alveolar epithelial (AT2) cells, which are highly dependent on Wnt signaling during development and adult regeneration. We use human induced pluripotent stem cell-derived AT2 (iAT2) cells to model how short telomeres affect AT2 cells. Cultured DC mutant iAT2 cells accumulate shortened, uncapped telomeres and manifest defects in the growth of alveolospheres, hallmarks of senescence, and apparent defects in Wnt signaling. The GSK3 inhibitor, CHIR99021, which mimics the output of canonical Wnt signaling, enhances telomerase activity and rescues the defects. These findings support further investigation of Wnt agonists as potential therapies for DC-related pathologies.
Collapse
Affiliation(s)
- Rafael Jesus Fernandez
- Medical Scientist Training Program, University of Pennsylvania, Philadelphia, United States
| | - Zachary J G Gardner
- Medical Scientist Training Program, University of Pennsylvania, Philadelphia, United States
| | - Katherine J Slovik
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Derek C Liberti
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, United States
| | - Katrina N Estep
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, United States
| | - Wenli Yang
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Qijun Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | - Garrett T Santini
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Javier V Perez
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | - Sarah Root
- College of Arts and Sciences and Vagelos Scholars Program, University of Pennsylvania, Philadelphia, United States
| | - Ranvir Bhatia
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - John W Tobias
- Penn Genomic Analysis Core, University of Pennsylvania, Philadelphia, United States
| | - Apoorva Babu
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, United States
| | - Michael P Morley
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, United States
| | - David B Frank
- Penn-CHOP Lung Biology Institute, Children's Hospital of Philadelphia, Philadelphia, United States
| | - Edward E Morrisey
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Christopher J Lengner
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, United States
| | - F Brad Johnson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
28
|
Lister-Shimauchi EH, McCarthy B, Lippincott M, Ahmed S. Genetic and Epigenetic Inheritance at Telomeres. EPIGENOMES 2022; 6:9. [PMID: 35323213 PMCID: PMC8947350 DOI: 10.3390/epigenomes6010009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/21/2022] [Accepted: 03/08/2022] [Indexed: 12/17/2022] Open
Abstract
Transgenerational inheritance can occur at telomeres in distinct contexts. Deficiency for telomerase or telomere-binding proteins in germ cells can result in shortened or lengthened chromosome termini that are transmitted to progeny. In human families, altered telomere lengths can result in stem cell dysfunction or tumor development. Genetic inheritance of altered telomeres as well as mutations that alter telomeres can result in progressive telomere length changes over multiple generations. Telomeres of yeast can modulate the epigenetic state of subtelomeric genes in a manner that is mitotically heritable, and the effects of telomeres on subtelomeric gene expression may be relevant to senescence or other human adult-onset disorders. Recently, two novel epigenetic states were shown to occur at C. elegans telomeres, where very low or high levels of telomeric protein foci can be inherited for multiple generations through a process that is regulated by histone methylation.Together, these observations illustrate that information relevant to telomere biology can be inherited via genetic and epigenetic mechanisms, although the broad impact of epigenetic inheritance to human biology remains unclear.
Collapse
Affiliation(s)
- Evan H. Lister-Shimauchi
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599-3280, USA; (E.H.L.-S.); (B.M.); (M.L.)
- Department of Biology, University of North Carolina, Chapel Hill, NC 27599-3280, USA
| | - Benjamin McCarthy
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599-3280, USA; (E.H.L.-S.); (B.M.); (M.L.)
- Department of Biology, University of North Carolina, Chapel Hill, NC 27599-3280, USA
| | - Michael Lippincott
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599-3280, USA; (E.H.L.-S.); (B.M.); (M.L.)
- Department of Biology, University of North Carolina, Chapel Hill, NC 27599-3280, USA
| | - Shawn Ahmed
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599-3280, USA; (E.H.L.-S.); (B.M.); (M.L.)
- Department of Biology, University of North Carolina, Chapel Hill, NC 27599-3280, USA
| |
Collapse
|
29
|
Lu R, Pickett HA. Telomeric replication stress: the beginning and the end for alternative lengthening of telomeres cancers. Open Biol 2022; 12:220011. [PMID: 35259951 PMCID: PMC8905155 DOI: 10.1098/rsob.220011] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Telomeres are nucleoprotein structures that cap the ends of linear chromosomes. Telomeric DNA comprises terminal tracts of G-rich tandem repeats, which are inherently difficult for the replication machinery to navigate. Structural aberrations that promote activation of the alternative lengthening of telomeres (ALT) pathway of telomere maintenance exacerbate replication stress at ALT telomeres, driving fork stalling and fork collapse. This form of telomeric DNA damage perpetuates recombination-mediated repair pathways and break-induced telomere synthesis. The relationship between replication stress and DNA repair is tightly coordinated for the purpose of regulating telomere length in ALT cells, but has been shown to be experimentally manipulatable. This raises the intriguing possibility that induction of replication stress can be used as a means to cause toxic levels of DNA damage at ALT telomeres, thereby selectively disrupting the viability of ALT cancers.
Collapse
Affiliation(s)
- Robert Lu
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Hilda A. Pickett
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| |
Collapse
|
30
|
Rafat A, Dizaji Asl K, Mazloumi Z, Movassaghpour AA, Farahzadi R, Nejati B, Nozad Charoudeh H. Telomerase-based therapies in haematological malignancies. Cell Biochem Funct 2022; 40:199-212. [PMID: 35103334 DOI: 10.1002/cbf.3687] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/10/2022] [Indexed: 02/02/2023]
Abstract
Telomeres are specialized genetic structures present at the end of all eukaryotic linear chromosomes. They progressively get shortened after each cell division due to end replication problems. Telomere shortening (TS) and chromosomal instability cause apoptosis and massive cell death. Following oncogene activation and inactivation of tumour suppressor genes, cells acquire mechanisms such as telomerase expression and alternative lengthening of telomeres to maintain telomere length (TL) and prevent initiation of cellular senescence or apoptosis. Significant TS, telomerase activation and alteration in expression of telomere-associated proteins are frequent features of different haematological malignancies that reflect on the progression, response to therapy and recurrence of these diseases. Telomerase is a ribonucleoprotein enzyme that has a pivotal role in maintaining the TL. However, telomerase activity in most somatic cells is insufficient to prevent TS. In 85-90% of tumour cells, the critically short telomeric length is maintained by telomerase activation. Thus, overexpression of telomerase in most tumour cells is a potential target for cancer therapy. In this review, alteration of telomeres, telomerase and telomere-associated proteins in different haematological malignancies and related telomerase-based therapies are discussed.
Collapse
Affiliation(s)
- Ali Rafat
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khadijeh Dizaji Asl
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Mazloumi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Raheleh Farahzadi
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Babak Nejati
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
31
|
Stem cells at odds with telomere maintenance and protection. Trends Cell Biol 2022; 32:527-536. [DOI: 10.1016/j.tcb.2021.12.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/20/2021] [Accepted: 12/22/2021] [Indexed: 11/23/2022]
|
32
|
Kaur P, Barnes R, Pan H, Detwiler AC, Liu M, Mahn C, Hall J, Messenger Z, You C, Piehler J, Smart R, Riehn R, Opresko PL, Wang H. TIN2 is an architectural protein that facilitates TRF2-mediated trans- and cis-interactions on telomeric DNA. Nucleic Acids Res 2021; 49:13000-13018. [PMID: 34883513 PMCID: PMC8682769 DOI: 10.1093/nar/gkab1142] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/08/2021] [Indexed: 12/23/2022] Open
Abstract
The telomere specific shelterin complex, which includes TRF1, TRF2, RAP1, TIN2, TPP1 and POT1, prevents spurious recognition of telomeres as double-strand DNA breaks and regulates telomerase and DNA repair activities at telomeres. TIN2 is a key component of the shelterin complex that directly interacts with TRF1, TRF2 and TPP1. In vivo, the large majority of TRF1 and TRF2 are in complex with TIN2 but without TPP1 and POT1. Since knockdown of TIN2 also removes TRF1 and TRF2 from telomeres, previous cell-based assays only provide information on downstream effects after the loss of TRF1/TRF2 and TIN2. Here, we investigated DNA structures promoted by TRF2-TIN2 using single-molecule imaging platforms, including tracking of compaction of long mouse telomeric DNA using fluorescence imaging, atomic force microscopy (AFM) imaging of protein-DNA structures, and monitoring of DNA-DNA and DNA-RNA bridging using the DNA tightrope assay. These techniques enabled us to uncover previously unknown unique activities of TIN2. TIN2S and TIN2L isoforms facilitate TRF2-mediated telomeric DNA compaction (cis-interactions), dsDNA-dsDNA, dsDNA-ssDNA and dsDNA-ssRNA bridging (trans-interactions). Furthermore, TIN2 facilitates TRF2-mediated T-loop formation. We propose a molecular model in which TIN2 functions as an architectural protein to promote TRF2-mediated trans and cis higher-order nucleic acid structures at telomeres.
Collapse
Affiliation(s)
- Parminder Kaur
- Physics Department, North Carolina State University, Raleigh, NC27695, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
| | - Ryan Barnes
- Department of Environmental and Occupational Health, University of Pittsburgh, UPMC Hillman Cancer Center, PA 15213, USA
| | - Hai Pan
- Physics Department, North Carolina State University, Raleigh, NC27695, USA
| | - Ariana C Detwiler
- Department of Environmental and Occupational Health, University of Pittsburgh, UPMC Hillman Cancer Center, PA 15213, USA
| | - Ming Liu
- Physics Department, North Carolina State University, Raleigh, NC27695, USA
| | - Chelsea Mahn
- Physics Department, North Carolina State University, Raleigh, NC27695, USA
| | - Jonathan Hall
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
- Toxicology Program, North Carolina State University, Raleigh, NC27695, USA
| | - Zach Messenger
- Toxicology Program, North Carolina State University, Raleigh, NC27695, USA
| | - Changjiang You
- Department of Biology/Chemistry, Universität Osnabrück, Osnabrück 49076, Germany
| | - Jacob Piehler
- Department of Biology/Chemistry, Universität Osnabrück, Osnabrück 49076, Germany
| | - Robert C Smart
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
- Toxicology Program, North Carolina State University, Raleigh, NC27695, USA
| | - Robert Riehn
- Physics Department, North Carolina State University, Raleigh, NC27695, USA
| | - Patricia L Opresko
- Department of Environmental and Occupational Health, University of Pittsburgh, UPMC Hillman Cancer Center, PA 15213, USA
| | - Hong Wang
- Physics Department, North Carolina State University, Raleigh, NC27695, USA
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
- Toxicology Program, North Carolina State University, Raleigh, NC27695, USA
| |
Collapse
|
33
|
Roger L, Tomas F, Gire V. Mechanisms and Regulation of Cellular Senescence. Int J Mol Sci 2021; 22:ijms222313173. [PMID: 34884978 PMCID: PMC8658264 DOI: 10.3390/ijms222313173] [Citation(s) in RCA: 197] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/25/2021] [Accepted: 12/02/2021] [Indexed: 12/23/2022] Open
Abstract
Cellular senescence entails a state of an essentially irreversible proliferative arrest in which cells remain metabolically active and secrete a range of pro-inflammatory and proteolytic factors as part of the senescence-associated secretory phenotype. There are different types of senescent cells, and senescence can be induced in response to many DNA damage signals. Senescent cells accumulate in different tissues and organs where they have distinct physiological and pathological functions. Despite this diversity, all senescent cells must be able to survive in a nondividing state while protecting themselves from positive feedback loops linked to the constant activation of the DNA damage response. This capacity requires changes in core cellular programs. Understanding how different cell types can undergo extensive changes in their transcriptional programs, metabolism, heterochromatin patterns, and cellular structures to induce a common cellular state is crucial to preventing cancer development/progression and to improving health during aging. In this review, we discuss how senescent cells continuously evolve after their initial proliferative arrest and highlight the unifying features that define the senescent state.
Collapse
Affiliation(s)
- Lauréline Roger
- Structure and Instability of Genomes Laboratory, Muséum National d’Histoire Naturelle (MNHN), CNRS-UMR 7196/INSERM U1154, 43 Rue Cuvier, 75005 Paris, France;
| | - Fanny Tomas
- Centre de Recherche en Biologie cellulaire de Montpellier (CRBM), Université de Montpellier, CNRS UMR 5237, 1919 Route de Mende, 34293 Montpellier, France;
| | - Véronique Gire
- Centre de Recherche en Biologie cellulaire de Montpellier (CRBM), Université de Montpellier, CNRS UMR 5237, 1919 Route de Mende, 34293 Montpellier, France;
- Correspondence: ; Tel.: +33-(0)-434359513; Fax: +33-(0)-434359410
| |
Collapse
|
34
|
Paul T, Liou W, Cai X, Opresko PL, Myong S. TRF2 promotes dynamic and stepwise looping of POT1 bound telomeric overhang. Nucleic Acids Res 2021; 49:12377-12393. [PMID: 34850123 PMCID: PMC8643667 DOI: 10.1093/nar/gkab1123] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/20/2021] [Accepted: 11/18/2021] [Indexed: 11/12/2022] Open
Abstract
Human telomeres are protected by shelterin proteins, but how telomeres maintain a dynamic structure remains elusive. Here, we report an unexpected activity of POT1 in imparting conformational dynamics of the telomere overhang, even at a monomer level. Strikingly, such POT1-induced overhang dynamics is greatly enhanced when TRF2 engages with the telomere duplex. Interestingly, TRF2, but not TRF2ΔB, recruits POT1-bound overhangs to the telomere ds/ss junction and induces a discrete stepwise movement up and down the axis of telomere duplex. The same steps are observed regardless of the length of the POT1-bound overhang, suggesting a tightly regulated conformational dynamic coordinated by TRF2 and POT1. TPP1 and TIN2 which physically connect POT1 and TRF2 act to generate a smooth movement along the axis of the telomere duplex. Our results suggest a plausible mechanism wherein telomeres maintain a dynamic structure orchestrated by shelterin.
Collapse
Affiliation(s)
- Tapas Paul
- Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Wilson Liou
- Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Xinyi Cai
- Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Patricia L Opresko
- Department of Environmental and Occupational Health, University of Pittsburgh, Hillman Cancer Center, 5117 Centre Avenue, Suite 2.6a, Pittsburgh, PA 15213, USA
| | - Sua Myong
- Department of Biophysics, Johns Hopkins University, Baltimore, MD 21218, USA.,Physics Frontier Center (Center for Physics of Living Cells), University of Illinois, 1110 W. Green St., Urbana, IL 61801, USA
| |
Collapse
|
35
|
Higa M, Matsuda Y, Fujii J, Sugimoto N, Yoshida K, Fujita M. TRF2-mediated ORC recruitment underlies telomere stability upon DNA replication stress. Nucleic Acids Res 2021; 49:12234-12251. [PMID: 34761263 PMCID: PMC8643664 DOI: 10.1093/nar/gkab1004] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 10/07/2021] [Accepted: 10/12/2021] [Indexed: 12/12/2022] Open
Abstract
Telomeres are intrinsically difficult-to-replicate region of eukaryotic chromosomes. Telomeric repeat binding factor 2 (TRF2) binds to origin recognition complex (ORC) to facilitate the loading of ORC and the replicative helicase MCM complex onto DNA at telomeres. However, the biological significance of the TRF2–ORC interaction for telomere maintenance remains largely elusive. Here, we employed a TRF2 mutant with mutations in two acidic acid residues (E111A and E112A) that inhibited the TRF2–ORC interaction in human cells. The TRF2 mutant was impaired in ORC recruitment to telomeres and showed increased replication stress-associated telomeric DNA damage and telomere instability. Furthermore, overexpression of an ORC1 fragment (amino acids 244–511), which competitively inhibited the TRF2–ORC interaction, increased telomeric DNA damage under replication stress conditions. Taken together, these findings suggest that TRF2-mediated ORC recruitment contributes to the suppression of telomere instability.
Collapse
Affiliation(s)
- Mitsunori Higa
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yukihiro Matsuda
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Jumpei Fujii
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Nozomi Sugimoto
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kazumasa Yoshida
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masatoshi Fujita
- Department of Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
36
|
Dhillon VS, Deo P, Chua A, Thomas P, Fenech M. Sleep duration, Health Promotion Index (HPI), sRAGE and ApoE-ε4 genotype are associated with telomere length (TL) in healthy elderly Australians. J Gerontol A Biol Sci Med Sci 2021; 77:243-249. [PMID: 34508574 DOI: 10.1093/gerona/glab264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Indexed: 12/18/2022] Open
Abstract
Significant alterations in sleep duration and/or quality of sleep become more pronounced as people get older. Poor sleep in elderly people is associated with adverse health outcomes and cellular ageing. We examined the relationship between TL and sleep duration, Health Promotion Index (HPI), and tested whether the presence of ApoE-ε4 allele impacts both sleep and TL. The present study was carried out in 174 healthy elderly subjects (21% male; mean age 53.79 years) from South Australia. Lymphocyte telomere length (TL) was measured by real-time qPCR and ApoE genotype was determined by TaqMan assay. HPI was calculated from a questionnaire regarding 8 lifestyle habits, including sleeping hours. Multivariate regression analysis was used to establish these associations adjusted for specified confounders. TL was found to be inversely associated with age (r = - 0.199; p = 0.008) and BMI (r = - 0.121; p = 0.11), and was significantly shorter in participants who slept for <7 hours (p = 0.001) relative to those sleeping ≥7 hours. TL was positively correlated with HPI (r = 0.195; p = 0.009). ApoE-ε4 allele carriers who slept for less than 7 hours had shortest TL (p = 0.01) compared to non-carriers. Plasma sRAGE level was significantly (p = 0.001) lower in individuals who sleep <7 hours and ApoE-ϵ4 carriers. Our results suggest that inadequate sleep duration or poor HPI is associated with shorter TL in cognitively normal elderly people and that carriage of APOE-ε4 genotype may influence the extent of these effects.
Collapse
Affiliation(s)
- Varinderpal S Dhillon
- Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Permal Deo
- Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Ann Chua
- Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Phil Thomas
- Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia
| | - Michael Fenech
- Health and Biomedical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, 5000, Australia.,Centre of Healthy Ageing and Wellness, Faculty of Health Sciences Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
37
|
Pan H, Kaur P, Barnes R, Detwiler AC, Sanford SL, Liu M, Xu P, Mahn C, Tang Q, Hao P, Bhattaram D, You C, Gu X, Lu W, Piehler J, Xu G, Weninger K, Riehn R, Opresko PL, Wang H. Structure, dynamics, and regulation of TRF1-TIN2-mediated trans- and cis-interactions on telomeric DNA. J Biol Chem 2021; 297:101080. [PMID: 34403696 PMCID: PMC8437784 DOI: 10.1016/j.jbc.2021.101080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 07/30/2021] [Accepted: 08/12/2021] [Indexed: 01/17/2023] Open
Abstract
TIN2 is a core component of the shelterin complex linking double-stranded telomeric DNA-binding proteins (TRF1 and TRF2) and single-strand overhang-binding proteins (TPP1-POT1). In vivo, the large majority of TRF1 and TRF2 exist in complexes containing TIN2 but lacking TPP1/POT1; however, the role of TRF1-TIN2 interactions in mediating interactions with telomeric DNA is unclear. Here, we investigated DNA molecular structures promoted by TRF1-TIN2 interaction using atomic force microscopy (AFM), total internal reflection fluorescence microscopy (TIRFM), and the DNA tightrope assay. We demonstrate that the short (TIN2S) and long (TIN2L) isoforms of TIN2 facilitate TRF1-mediated DNA compaction (cis-interactions) and DNA-DNA bridging (trans-interactions) in a telomeric sequence- and length-dependent manner. On the short telomeric DNA substrate (six TTAGGG repeats), the majority of TRF1-mediated telomeric DNA-DNA bridging events are transient with a lifetime of ~1.95 s. On longer DNA substrates (270 TTAGGG repeats), TIN2 forms multiprotein complexes with TRF1 and stabilizes TRF1-mediated DNA-DNA bridging events that last on the order of minutes. Preincubation of TRF1 with its regulator protein Tankyrase 1 and the cofactor NAD+ significantly reduced TRF1-TIN2 mediated DNA-DNA bridging, whereas TIN2 protected the disassembly of TRF1-TIN2 mediated DNA-DNA bridging upon Tankyrase 1 addition. Furthermore, we showed that TPP1 inhibits TRF1-TIN2L-mediated DNA-DNA bridging. Our study, together with previous findings, supports a molecular model in which protein assemblies at telomeres are heterogeneous with distinct subcomplexes and full shelterin complexes playing distinct roles in telomere protection and elongation.
Collapse
Affiliation(s)
- Hai Pan
- Physics Department, North Carolina State University, Raleigh, North Carolina, USA
| | - Parminder Kaur
- Physics Department, North Carolina State University, Raleigh, North Carolina, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina, USA
| | - Ryan Barnes
- Department of Environmental and Occupational Health, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ariana C Detwiler
- Department of Environmental and Occupational Health, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Samantha Lynn Sanford
- Department of Environmental and Occupational Health, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ming Liu
- Physics Department, North Carolina State University, Raleigh, North Carolina, USA
| | - Pengning Xu
- Physics Department, North Carolina State University, Raleigh, North Carolina, USA
| | - Chelsea Mahn
- Physics Department, North Carolina State University, Raleigh, North Carolina, USA
| | - Qingyu Tang
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina, USA
| | - Pengyu Hao
- Physics Department, North Carolina State University, Raleigh, North Carolina, USA
| | - Dhruv Bhattaram
- Department of Biomedical Engineering, Georgia Institute of Technology & Emory University of Medicine, Atlanta, Georgia, USA
| | - Changjiang You
- Department of Biology/Chemistry, Universität Osnabrück, Osnabrück, Germany
| | - Xinyun Gu
- College of Art and Sciences, New York University, New York City, New York, USA
| | - Warren Lu
- Department of Pathology at NYU Grossman School of Medicine, New York University, New York City, New York, USA
| | - Jacob Piehler
- Department of Biology/Chemistry, Universität Osnabrück, Osnabrück, Germany
| | - Guozhou Xu
- Department of Molecular and Structural Biochemistry, North Carolina State University, Raleigh, North Carolina, USA
| | - Keith Weninger
- Physics Department, North Carolina State University, Raleigh, North Carolina, USA
| | - Robert Riehn
- Physics Department, North Carolina State University, Raleigh, North Carolina, USA
| | - Patricia L Opresko
- Department of Environmental and Occupational Health, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Molecular Biophysics and Structural Biology Graduate Program, Carnegie Mellon University and the University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hong Wang
- Physics Department, North Carolina State University, Raleigh, North Carolina, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, North Carolina, USA; Toxicology Program, North Carolina State University, Raleigh, North Carolina, USA.
| |
Collapse
|
38
|
Li B, Zhao Y. Regulation of Antigenic Variation by Trypanosoma brucei Telomere Proteins Depends on Their Unique DNA Binding Activities. Pathogens 2021; 10:pathogens10080967. [PMID: 34451431 PMCID: PMC8402208 DOI: 10.3390/pathogens10080967] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 01/17/2023] Open
Abstract
Trypanosoma brucei causes human African trypanosomiasis and regularly switches its major surface antigen, Variant Surface Glycoprotein (VSG), to evade the host immune response. Such antigenic variation is a key pathogenesis mechanism that enables T. brucei to establish long-term infections. VSG is expressed exclusively from subtelomere loci in a strictly monoallelic manner, and DNA recombination is an important VSG switching pathway. The integrity of telomere and subtelomere structure, maintained by multiple telomere proteins, is essential for T. brucei viability and for regulating the monoallelic VSG expression and VSG switching. Here we will focus on T. brucei TRF and RAP1, two telomere proteins with unique nucleic acid binding activities, and summarize their functions in telomere integrity and stability, VSG switching, and monoallelic VSG expression. Targeting the unique features of TbTRF and TbRAP1′s nucleic acid binding activities to perturb the integrity of telomere structure and disrupt VSG monoallelic expression may serve as potential therapeutic strategy against T. brucei.
Collapse
Affiliation(s)
- Bibo Li
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, College of Sciences and Health Professions, Cleveland State University, 2121 Euclid Avenue, Cleveland, OH 44115, USA
- Case Comprehensive Cancer Center, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
- Center for RNA Science and Therapeutics, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
- Correspondence: (B.L.); (Y.Z.)
| | - Yanxiang Zhao
- Shenzhen Research Institute, The Hong Kong Polytechnic University, Shenzhen, China
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
- Correspondence: (B.L.); (Y.Z.)
| |
Collapse
|
39
|
Molinaro C, Martoriati A, Cailliau K. Proteins from the DNA Damage Response: Regulation, Dysfunction, and Anticancer Strategies. Cancers (Basel) 2021; 13:3819. [PMID: 34359720 PMCID: PMC8345162 DOI: 10.3390/cancers13153819] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 12/21/2022] Open
Abstract
Cells respond to genotoxic stress through a series of complex protein pathways called DNA damage response (DDR). These monitoring mechanisms ensure the maintenance and the transfer of a correct genome to daughter cells through a selection of DNA repair, cell cycle regulation, and programmed cell death processes. Canonical or non-canonical DDRs are highly organized and controlled to play crucial roles in genome stability and diversity. When altered or mutated, the proteins in these complex networks lead to many diseases that share common features, and to tumor formation. In recent years, technological advances have made it possible to benefit from the principles and mechanisms of DDR to target and eliminate cancer cells. These new types of treatments are adapted to the different types of tumor sensitivity and could benefit from a combination of therapies to ensure maximal efficiency.
Collapse
Affiliation(s)
| | | | - Katia Cailliau
- Univ. Lille, CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, F-59000 Lille, France; (C.M.); (A.M.)
| |
Collapse
|
40
|
Pobiega S, Alibert O, Marcand S. A new assay capturing chromosome fusions shows a protection trade-off at telomeres and NHEJ vulnerability to low-density ionizing radiation. Nucleic Acids Res 2021; 49:6817-6831. [PMID: 34125900 PMCID: PMC8266670 DOI: 10.1093/nar/gkab502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 03/31/2021] [Accepted: 05/27/2021] [Indexed: 11/14/2022] Open
Abstract
Chromosome fusions threaten genome integrity and promote cancer by engaging catastrophic mutational processes, namely chromosome breakage-fusion-bridge cycles and chromothripsis. Chromosome fusions are frequent in cells incurring telomere dysfunctions or those exposed to DNA breakage. Their occurrence and therefore their contribution to genome instability in unchallenged cells is unknown. To address this issue, we constructed a genetic assay able to capture and quantify rare chromosome fusions in budding yeast. This chromosome fusion capture (CFC) assay relies on the controlled inactivation of one centromere to rescue unstable dicentric chromosome fusions. It is sensitive enough to quantify the basal rate of end-to-end chromosome fusions occurring in wild-type cells. These fusions depend on canonical nonhomologous end joining (NHEJ). Our results show that chromosome end protection results from a trade-off at telomeres between positive effectors (Rif2, Sir4, telomerase) and a negative effector partially antagonizing them (Rif1). The CFC assay also captures NHEJ-dependent chromosome fusions induced by ionizing radiation. It provides evidence for chromosomal rearrangements stemming from a single photon-matter interaction.
Collapse
Affiliation(s)
- Sabrina Pobiega
- Université de Paris and Université Paris-Saclay, Inserm, CEA IBFJ/iRCM, UMR Stabilité Génétique Cellules Souches et Radiations, 92265 Fontenay-au-Roses, France
| | | | - Stéphane Marcand
- To whom correspondence should be addressed. Tel: +33 1 46 54 82 33;
| |
Collapse
|
41
|
Ackerson SM, Romney C, Schuck PL, Stewart JA. To Join or Not to Join: Decision Points Along the Pathway to Double-Strand Break Repair vs. Chromosome End Protection. Front Cell Dev Biol 2021; 9:708763. [PMID: 34322492 PMCID: PMC8311741 DOI: 10.3389/fcell.2021.708763] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/17/2021] [Indexed: 01/01/2023] Open
Abstract
The regulation of DNA double-strand breaks (DSBs) and telomeres are diametrically opposed in the cell. DSBs are considered one of the most deleterious forms of DNA damage and must be quickly recognized and repaired. Telomeres, on the other hand, are specialized, stable DNA ends that must be protected from recognition as DSBs to inhibit unwanted chromosome fusions. Decisions to join DNA ends, or not, are therefore critical to genome stability. Yet, the processing of telomeres and DSBs share many commonalities. Accordingly, key decision points are used to shift DNA ends toward DSB repair vs. end protection. Additionally, DSBs can be repaired by two major pathways, namely homologous recombination (HR) and non-homologous end joining (NHEJ). The choice of which repair pathway is employed is also dictated by a series of decision points that shift the break toward HR or NHEJ. In this review, we will focus on these decision points and the mechanisms that dictate end protection vs. DSB repair and DSB repair choice.
Collapse
Affiliation(s)
- Stephanie M Ackerson
- Department of Biological Sciences, University of South Carolina, Columbia, SC, United States
| | - Carlan Romney
- Department of Biological Sciences, University of South Carolina, Columbia, SC, United States
| | - P Logan Schuck
- Department of Biological Sciences, University of South Carolina, Columbia, SC, United States
| | - Jason A Stewart
- Department of Biological Sciences, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
42
|
Multifunctionality of the Telomere-Capping Shelterin Complex Explained by Variations in Its Protein Composition. Cells 2021; 10:cells10071753. [PMID: 34359923 PMCID: PMC8305809 DOI: 10.3390/cells10071753] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/03/2021] [Accepted: 07/09/2021] [Indexed: 12/31/2022] Open
Abstract
Protecting telomere from the DNA damage response is essential to avoid the entry into cellular senescence and organismal aging. The progressive telomere DNA shortening in dividing somatic cells, programmed during development, leads to critically short telomeres that trigger replicative senescence and thereby contribute to aging. In several organisms, including mammals, telomeres are protected by a protein complex named Shelterin that counteract at various levels the DNA damage response at chromosome ends through the specific function of each of its subunits. The changes in Shelterin structure and function during development and aging is thus an intense area of research. Here, we review our knowledge on the existence of several Shelterin subcomplexes and the functional independence between them. This leads us to discuss the possibility that the multifunctionality of the Shelterin complex is determined by the formation of different subcomplexes whose composition may change during aging.
Collapse
|
43
|
Li B. Keeping Balance Between Genetic Stability and Plasticity at the Telomere and Subtelomere of Trypanosoma brucei. Front Cell Dev Biol 2021; 9:699639. [PMID: 34291053 PMCID: PMC8287324 DOI: 10.3389/fcell.2021.699639] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/08/2021] [Indexed: 12/13/2022] Open
Abstract
Telomeres, the nucleoprotein complexes at chromosome ends, are well-known for their essential roles in genome integrity and chromosome stability. Yet, telomeres and subtelomeres are frequently less stable than chromosome internal regions. Many subtelomeric genes are important for responding to environmental cues, and subtelomeric instability can facilitate organismal adaptation to extracellular changes, which is a common theme in a number of microbial pathogens. In this review, I will focus on the delicate and important balance between stability and plasticity at telomeres and subtelomeres of a kinetoplastid parasite, Trypanosoma brucei, which causes human African trypanosomiasis and undergoes antigenic variation to evade the host immune response. I will summarize the current understanding about T. brucei telomere protein complex, the telomeric transcript, and telomeric R-loops, focusing on their roles in maintaining telomere and subtelomere stability and integrity. The similarities and differences in functions and underlying mechanisms of T. brucei telomere factors will be compared with those in human and yeast cells.
Collapse
Affiliation(s)
- Bibo Li
- Center for Gene Regulation in Health and Disease, Department of Biological, Geological, and Environmental Sciences, College of Sciences and Health Professions, Cleveland State University, Cleveland, OH, United States.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, United States.,Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States.,Center for RNA Science and Therapeutics, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
44
|
Telomere Shortening and Fusions: A Link to Aneuploidy in Early Human Embryo Development. Obstet Gynecol Surv 2021; 76:429-436. [PMID: 34324695 DOI: 10.1097/ogx.0000000000000907] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Importance It is known that oocytes undergo aging that is caused by exposure to an aged ovarian microenvironment. Telomere length in mouse and bovine oocytes declines with age, and age-associated telomere shortening in oocytes is considered a sign of poor development competency. Women with advanced age undergoing assisted reproductive technologies have poor outcomes because of increasing aneuploidy rates with age. Research has shown that aneuploidy is associated with DNA damage, reactive oxygen species, and telomere dysfunction. Objective In this review, we focus on the possible relationship between telomere dysfunction and aneuploidy in human early embryo development and several reproductive and perinatal outcomes, discussing the mechanism of aneuploidy caused by telomere shortening and fusion in human embryos. Evidence Acquisition We reviewed the current literature evidence concerning telomere dysfunction and aneuploidy in early human embryo development. Results Shorter telomeres in oocytes, leukocytes, and granulosa cells, related to aging in women, were associated with recurrent miscarriage, trisomy 21, ovarian insufficiency, and decreasing chance of in vitro fertilization success. Telomere length and telomerase activity in embryos have been related to the common genomic instability at the cleavage stage of human development. Complications of assisted reproductive technology pregnancies, such as miscarriage, birth defects, preterm births, and intrauterine growth restriction, also might result from telomere shortening as observed in oocytes, polar body, granulosa cells, and embryos. Conclusions and Relevance Telomere length clearly plays an important role in the development of the embryo and fetus, and the abnormal shortening of telomeres is likely involved in embryo loss during early human development. However, telomere fusion studies have yet to be performed in early human development.
Collapse
|
45
|
Adam N, Beattie TL, Riabowol K. Fluorescence microscopy methods for examining telomeres during cell aging. Ageing Res Rev 2021; 68:101320. [PMID: 33744488 DOI: 10.1016/j.arr.2021.101320] [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: 12/01/2020] [Revised: 02/24/2021] [Accepted: 03/08/2021] [Indexed: 10/21/2022]
Abstract
Telomeres are protective structures, composed of nucleic acids and a complex protein mixture, located at the end of the chromosomes. They play an important role in preventing genomic instability and ensuring cell health. Defects in telomere integrity result in cell dysfunction and the development of diseases, including neurodegenerative disorders, cancer and premature aging syndromes, among others. Loss of telomere integrity during normal cell aging also initiates DNA damage signals that culminate in the senescence phenotype. Fluorescence microscopy has allowed researchers to study the dynamics, shape, localization, and co-distribution of telomeres with proteins of interest. The microscopy tools to investigate these structures have evolved, making it possible to understand in greater detail the molecular mechanisms affecting telomeres that contribute to cell aging and the development of age-related diseases. Using human fibroblasts as an example, we will highlight several characteristics of telomeres that can be investigated using three different microscopy systems, including wide-field microscopy, and the two super-resolution techniques called 3D Structured Illumination Microscopy (3D-SIM) and direct Stochastic Optical Reconstruction Microscopy (dSTORM). In this review, we will also discuss their limitations and highlight their importance in answering telomere-related scientific questions.
Collapse
|
46
|
Lamm N, Rogers S, Cesare AJ. Chromatin mobility and relocation in DNA repair. Trends Cell Biol 2021; 31:843-855. [PMID: 34183232 DOI: 10.1016/j.tcb.2021.06.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/02/2021] [Accepted: 06/03/2021] [Indexed: 01/17/2023]
Abstract
The nucleus is a dynamic environment containing chromatin, membraneless organelles, and specialized molecular structures at the nuclear membrane. Within the spectrum of DNA repair activities are observations of increased mobility of damaged chromatin and the displacement of DNA lesions to specific nuclear environments. Here, we focus on the role that nuclear-specific filamentous actin plays in mobilizing damaged chromatin in response to DNA double-strand breaks and replication stress. We also examine nuclear pore complexes and promyelocytic leukemia-nuclear bodies as specialized platforms for homology-directed repair. The literature suggests an emerging model where specific types of DNA lesions are subjected to nuclear-derived forces that mobilize damaged chromatin and promote interaction with repair hubs to facilitate specialized repair reactions.
Collapse
Affiliation(s)
- Noa Lamm
- Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, 2145, Australia
| | - Samuel Rogers
- Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, 2145, Australia
| | - Anthony J Cesare
- Children's Medical Research Institute, University of Sydney, Westmead, New South Wales, 2145, Australia.
| |
Collapse
|
47
|
Roisné-Hamelin F, Pobiega S, Jézéquel K, Miron S, Dépagne J, Veaute X, Busso D, Du MHL, Callebaut I, Charbonnier JB, Cuniasse P, Zinn-Justin S, Marcand S. Mechanism of MRX inhibition by Rif2 at telomeres. Nat Commun 2021; 12:2763. [PMID: 33980827 PMCID: PMC8115599 DOI: 10.1038/s41467-021-23035-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 04/13/2021] [Indexed: 02/06/2023] Open
Abstract
Specific proteins present at telomeres ensure chromosome end stability, in large part through unknown mechanisms. In this work, we address how the Saccharomyces cerevisiae ORC-related Rif2 protein protects telomere. We show that the small N-terminal Rif2 BAT motif (Blocks Addition of Telomeres) previously known to limit telomere elongation and Tel1 activity is also sufficient to block NHEJ and 5' end resection. The BAT motif inhibits the ability of the Mre11-Rad50-Xrs2 complex (MRX) to capture DNA ends. It acts through a direct contact with Rad50 ATP-binding Head domains. Through genetic approaches guided by structural predictions, we identify residues at the surface of Rad50 that are essential for the interaction with Rif2 and its inhibition. Finally, a docking model predicts how BAT binding could specifically destabilise the DNA-bound state of the MRX complex. From these results, we propose that when an MRX complex approaches a telomere, the Rif2 BAT motif binds MRX Head in its ATP-bound resting state. This antagonises MRX transition to its DNA-bound state, and favours a rapid return to the ATP-bound state. Unable to stably capture the telomere end, the MRX complex cannot proceed with the subsequent steps of NHEJ, Tel1-activation and 5' resection.
Collapse
Affiliation(s)
- Florian Roisné-Hamelin
- Université de Paris, Université Paris-Saclay, Inserm, CEA, Institut de Biologie François Jacob, iRCM, UMR Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, France
| | - Sabrina Pobiega
- Université de Paris, Université Paris-Saclay, Inserm, CEA, Institut de Biologie François Jacob, iRCM, UMR Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, France
| | - Kévin Jézéquel
- Université de Paris, Université Paris-Saclay, Inserm, CEA, Institut de Biologie François Jacob, iRCM, UMR Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, France
| | - Simona Miron
- Université Paris-Saclay, CNRS, CEA, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Jordane Dépagne
- CIGEx, Université de Paris, Université Paris-Saclay, Inserm, CEA, Institut de Biologie François Jacob, iRCM, UMR Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, France
| | - Xavier Veaute
- CIGEx, Université de Paris, Université Paris-Saclay, Inserm, CEA, Institut de Biologie François Jacob, iRCM, UMR Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, France
| | - Didier Busso
- CIGEx, Université de Paris, Université Paris-Saclay, Inserm, CEA, Institut de Biologie François Jacob, iRCM, UMR Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, France
| | - Marie-Hélène Le Du
- Université Paris-Saclay, CNRS, CEA, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Isabelle Callebaut
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, Institut de Minéralogie de Physique des Matériaux et de Cosmochimie (IMPMC), Paris, France
| | - Jean-Baptiste Charbonnier
- Université Paris-Saclay, CNRS, CEA, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Philippe Cuniasse
- Université Paris-Saclay, CNRS, CEA, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Sophie Zinn-Justin
- Université Paris-Saclay, CNRS, CEA, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Stéphane Marcand
- Université de Paris, Université Paris-Saclay, Inserm, CEA, Institut de Biologie François Jacob, iRCM, UMR Stabilité Génétique Cellules Souches et Radiations, Fontenay-aux-Roses, France.
| |
Collapse
|
48
|
Shoeb M, Meier HCS, Antonini JM. Telomeres in toxicology: Occupational health. Pharmacol Ther 2021; 220:107742. [PMID: 33176178 PMCID: PMC7969441 DOI: 10.1016/j.pharmthera.2020.107742] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/03/2020] [Indexed: 12/24/2022]
Abstract
The ends of chromosomes shorten at each round of cell division, and this process is thought to be affected by occupational exposures. Occupational hazards may alter telomere length homeostasis resulting in DNA damage, chromosome aberration, mutations, epigenetic alterations and inflammation. Therefore, for the protection of genetic material, nature has provided a unique nucleoprotein structure known as a telomere. Telomeres provide protection by averting an inappropriate activation of the DNA damage response (DDR) at chromosomal ends and preventing recognition of single and double strand DNA (ssDNA and dsDNA) breaks or chromosomal end-to-end fusion. Telomeres and their interacting six shelterin complex proteins in coordination act as inhibitors of DNA damage machinery by blocking DDR activation at chromosomes, thereby preventing the occurrence of genome instability, perturbed cell cycle, cellular senescence and apoptosis. However, inappropriate DNA repair may result in the inadequate distribution of genetic material during cell division, resulting in the eventual development of tumorigenesis and other pathologies. This article reviews the current literature on the association of changes in telomere length and its interacting proteins with different occupational exposures and the potential application of telomere length or changes in the regulatory proteins as potential biomarkers for exposure and health response, including recent findings and future perspectives.
Collapse
Affiliation(s)
- Mohammad Shoeb
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, United States of America.
| | - Helen C S Meier
- Joseph J. Zilber School of Public Health, University of Wisconsin, Milwaukee, WI, United States of America
| | - James M Antonini
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, United States of America
| |
Collapse
|
49
|
Abstract
Decades of study on cell cycle regulation have provided great insight into human cellular life span barriers, as well as their dysregulation during tumorigenesis. Telomeres, the extremities of linear chromosomes, perform an essential role in implementing these proliferative boundaries and preventing the propagation of potentially cancerous cells. The tumor-suppressive function of telomeres relies on their ability to initiate DNA damage signaling pathways and downstream cellular events, ranging from cell cycle perturbation to inflammation and cell death. While the tumor-suppressor role of telomeres is undoubtable, recent advances have pointed to telomeres as a major source of many of the genomic aberrations found in both early- and late-stage cancers, including the most recently discovered mutational phenomenon of chromothripsis. Telomere shortening appears as a double-edged sword that can function in opposing directions in carcinogenesis. This review focuses on the current knowledge of the dual role of telomeres in cancer and suggests a new perspective to reconcile the paradox of telomeres and their implications in cancer etiology.
Collapse
Affiliation(s)
- Joe Nassour
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Tobias T Schmidt
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | - Jan Karlseder
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, La Jolla, California 92037, USA
| |
Collapse
|
50
|
Liu Y, Wang C, Su H, Birchler JA, Han F. Phosphorylation of histone H3 by Haspin regulates chromosome alignment and segregation during mitosis in maize. JOURNAL OF EXPERIMENTAL BOTANY 2021; 72:1046-1058. [PMID: 33130883 DOI: 10.1093/jxb/eraa506] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/26/2020] [Indexed: 06/11/2023]
Abstract
In human cells, Haspin-mediated histone H3 threonine 3 (H3T3) phosphorylation promotes centromeric localization of the chromosomal passenger complex, thereby ensuring proper kinetochore-microtubule attachment. Haspin also binds to PDS5 cohesin-associated factor B (Pds5B), antagonizing the Wings apart-like protein homolog (Wapl)-Pds5B interaction and thus preventing Wapl from releasing centromeric cohesion during mitosis. However, the role of Haspin in plant chromosome segregation is not well understood. Here, we show that in maize (Zea mays) mitotic cells, ZmHaspin localized to the centromere during metaphase and anaphase, whereas it localized to the telomeres during meiosis. These results suggest that ZmHaspin plays different roles during mitosis and meiosis. Knockout of ZmHaspin led to decreased H3T3 phosphorylation and histone H3 serine 10 phosphorylation, and defects in chromosome alignment and segregation in mitosis. These lines of evidence suggest that Haspin regulates chromosome segregation in plants via the mechanism described for humans, namely, H3T3 phosphorylation. Plant Haspin proteins lack the RTYGA and PxVxL motifs needed to bind Pds5B and heterochromatin protein 1, and no obvious cohesion defects were detected in ZmHaspin knockout plants. Taken together, these results highlight the conserved but slightly different roles of Haspin proteins in cell division in plants and in animals.
Collapse
Affiliation(s)
- Yang Liu
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Chunhui Wang
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Handong Su
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
| | - James A Birchler
- Division of Biological Sciences, University of Missouri, Columbia, MO, USA
| | - Fangpu Han
- State Key Laboratory of Plant Cell and Chromosome Engineering, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|