1
|
Wang J, Zhao Y, Wei Y, Li T, Huang T, Pan T, Wu J, Bai L, Zhu D, Zhao Q, Wang Z, Feng F, Zhou X. Mai-wei-yang-fei decoction protects against pulmonary fibrosis by reducing telomere shortening and inhibiting AECII senescence via FBW7/TPP1 regulation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156682. [PMID: 40215816 DOI: 10.1016/j.phymed.2025.156682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 11/17/2024] [Accepted: 03/21/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Pulmonary fibrosis (PF) is a fatal disease associated with ageing. The senescence of alveolar epithelial type II cells (AECIIs) can drive PF. Therefore, reducing AECII senescence is a promising treatment to prevent PF. Mai-wei-yang-fei decoction (MWYF) has shown significant clinical efficacy in the treatment of patients with PF. However, its mechanism of action remains unclear. PURPOSE To investigate the role and underlying mechanism of MWYF in protecting against PF. METHODS The main chemical components of MWYF were identified using UPLC-MS. The mouse and in vitro cell models of PF were established using BLM. Micro-CT, H&E, and Masson staining were used to observe the protective effect of MWYF on mice with PF. Immunohistochemistry, β-galactosidase staining, and IF-FISH were used to observe the inhibitory effect of MWYF on senescence and telomere shortening in mouse lung tissue or A549 cells. The Transwell assay and cell co-culture method were used to observe the effect of MWYF on the migration and activation of lung fibroblasts by inhibiting AECII senescence. Finally, lentiviral vector was used to overexpress FBW7 gene in A549 cells in vitro to observe the mechanism pathway of MWYF inhibiting AECII senescence and telomere shortening. RESULTS MWYF was effective in protecting against bleomycin (BLM)-induced PF. Furthermore, MWYF alleviated cellular senescence by reducing the DNA damage response (DDR) and shortening of the telomere in AECⅡs in mouse lung tissues. Mechanistically, genes related to telomere disorders were detected in BLM-induced PF mouse models using q-PCR. MWYF mainly inhibited telomere shortening by regulating FBW7 and reducing the degradation of TPP1. In vitro, MWYF reduced BLM-induced senescence in A549 cells, as well as proliferation and migration of MRC5 cells, by inhibiting DDR and telomere shortening via regulation of the FBW7/TPP1 axis. CONCLUSION MWYF is a potential therapeutic agent against PF, as it inhibits telomere shortening and reduces AECII senescence by regulating FBW7/TPP1.
Collapse
Affiliation(s)
- Jing Wang
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Zhao
- First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun Wei
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tingyuan Li
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tongxing Huang
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Tingyu Pan
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jieyu Wu
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Le Bai
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Dongwei Zhu
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qi Zhao
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China; First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhichao Wang
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Fanchao Feng
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Xianmei Zhou
- Department of Respiratory and Critical Care Medicine, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
2
|
Scortecci JF, de Freitas Fernandes A, Serrão VHB, Cornélio ML, de Oliveira Neto M, Thiemann OH. Biophysical analysis of SECIS binding protein 2 (SBP2) from Naegleria gruberi. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2025; 1873:141075. [PMID: 40294688 DOI: 10.1016/j.bbapap.2025.141075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 04/19/2025] [Accepted: 04/25/2025] [Indexed: 04/30/2025]
Abstract
The Selenocysteine (Sec - U) biosynthesis pathway is present in eukaryotes and prokaryotes, in which its incorporation is directed by the stop codon UGA and the structural mRNA element named Sec Insertion Sequence (SECIS) that contain an essential kink-turn motif recognized by specific RNA-binding proteins. SBP2 is the key player in the interaction with the SECIS element in eukaryotes, and it is essential for the biosynthesis pathway. Free-living amoebas are part of the Heterolobosea phylum, and several species, including Naegleria fowleri, are known human pathogens. In 2013 it was reported that Naegleria gruberi (NgSBP2), which is non-pathogenic, had a divergent SBP2 sequence and all the genes essential for Sec synthesis. The identity of NgSBP2 is confirmed experimentally and its binding affinity to the SECIS element is demonstrated. The N-terminal and the C-terminal domains (NgSBP2-NT and NgSBP2-CT, respectively) of NgSBP2 contain disordered regions, particularly in the N-terminal domain. The SECIS element is bonded to NgSBP2-CT, which results in a decrease in the disordered sequence of the domain, and the NgSBP2-NT domain interacts with NgSBP2-CT.SECIS complex, as we present here. The findings reveal the molecular interaction patterns underlying the selenocysteine incorporation pathway in an early-branching eukaryote, which is influenced by multiple protein-RNA interactions.
Collapse
Affiliation(s)
- Jéssica Fernandes Scortecci
- Physics Institute of Sao Carlos, University of Sao Paulo, Trabalhador Sao Carlense Av. 400. Sao Carlos, SP, CEP, 13566-590, Brazil
| | - Adriano de Freitas Fernandes
- Physics Institute of Sao Carlos, University of Sao Paulo, Trabalhador Sao Carlense Av. 400. Sao Carlos, SP, CEP, 13566-590, Brazil
| | - Vitor Hugo Balasco Serrão
- Physics Institute of Sao Carlos, University of Sao Paulo, Trabalhador Sao Carlense Av. 400. Sao Carlos, SP, CEP, 13566-590, Brazil
| | - Marinônio Lopes Cornélio
- Departamento de Física, Instituto de Biociências, Letras e Ciências Exatas (IBILCE), UNESP, Rua Cristovão Colombo 2265, São José do Rio Preto CEP 15054-000, SP, Brazil
| | - Mario de Oliveira Neto
- Instituto de Biociências, Universidade Estadual Paulista, Distrito de Rubião Júnior, Botucatu, SP CEP: 18.618-970, Brazil
| | - Otavio Henrique Thiemann
- Physics Institute of Sao Carlos, University of Sao Paulo, Trabalhador Sao Carlense Av. 400. Sao Carlos, SP, CEP, 13566-590, Brazil; Department of Genetics and Evolution, Federal University of São Carlos - UFSCar, 13565-905 São Carlos, 16 SP, Brazil.
| |
Collapse
|
3
|
Billing D, Sfeir A. The Role of Microhomology-Mediated End Joining (MMEJ) at Dysfunctional Telomeres. Cold Spring Harb Perspect Biol 2025; 17:a041687. [PMID: 39500624 PMCID: PMC11864110 DOI: 10.1101/cshperspect.a041687] [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] [Indexed: 02/05/2025]
Abstract
DNA double-strand break (DSB) repair pathways are crucial for maintaining genome stability and cell viability. However, these pathways can mistakenly recognize chromosome ends as DNA breaks, leading to adverse outcomes such as telomere fusions and malignant transformation. The shelterin complex protects telomeres from activation of DNA repair pathways by inhibiting nonhomologous end joining (NHEJ), homologous recombination (HR), and microhomology-mediated end joining (MMEJ). The focus of this paper is on MMEJ, an error-prone DSB repair pathway characterized by short insertions and deletions flanked by sequence homology. MMEJ is critical in mediating telomere fusions in cells lacking the shelterin complex and at critically short telomeres. Furthermore, studies suggest that MMEJ is the preferred pathway for repairing intratelomeric DSBs and facilitates escape from telomere crisis. Targeting MMEJ to prevent telomere fusions in hematologic malignancies is of potential therapeutic value.
Collapse
Affiliation(s)
- David Billing
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Agnel Sfeir
- Molecular Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| |
Collapse
|
4
|
Shou S, Maolan A, Zhang D, Jiang X, Liu F, Li Y, Zhang X, Geer E, Pu Z, Hua B, Guo Q, Zhang X, Pang B. Telomeres, telomerase, and cancer: mechanisms, biomarkers, and therapeutics. Exp Hematol Oncol 2025; 14:8. [PMID: 39871386 PMCID: PMC11771031 DOI: 10.1186/s40164-025-00597-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
Telomeres and telomerase play crucial roles in the initiation and progression of cancer. As biomarkers, they aid in distinguishing benign from malignant tissues. Despite the promising therapeutic potential of targeting telomeres and telomerase for therapy, translating this concept from the laboratory to the clinic remains challenging. Many candidate drugs remain in the experimental stage, with only a few advancing to clinical trials. This review explores the relationship between telomeres, telomerase, and cancer, synthesizing their roles as biomarkers and reviewing the outcomes of completed trials. We propose that changes in telomere length and telomerase activity can be used to stratify cancer stages. Furthermore, we suggest that differential expression of telomere and telomerase components at the subcellular level holds promise as a biomarker. From a therapeutic standpoint, combining telomerase-targeted therapies with drugs that mitigate the adverse effects of telomerase inhibition may offer a viable strategy.
Collapse
Affiliation(s)
- Songting Shou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ayidana Maolan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Di Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaochen Jiang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fudong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiyuan Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - En Geer
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenqing Pu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baojin Hua
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Qiujun Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xing Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Bo Pang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
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
|
Czaja AJ. Cellular senescence and its pathogenic and therapeutic implications in autoimmune hepatitis. Expert Rev Gastroenterol Hepatol 2024; 18:725-743. [PMID: 39575891 DOI: 10.1080/17474124.2024.2432480] [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: 09/25/2024] [Accepted: 11/18/2024] [Indexed: 12/28/2024]
Abstract
INTRODUCTION Senescent cells are characterized by replicative arrest and phenotypes that produce diverse pro-inflammatory and pro-oxidant mediators. The senescence of diverse hepatic cell types could constitute an unrecognized pathogenic mechanism and prognostic determinant in autoimmune hepatitis. The impact of cellular senescence in autoimmune hepatitis is unknown, and it may suggest adjunctive management strategies. AREAS COVERED This review describes the molecular mechanisms of cellular senescence, indicates its diagnostic features, suggests its consequences, presents possible therapeutic interventions, and encourages investigations of its pathogenic role and management in autoimmune hepatitis. Treatment prospects include elimination or reversal of senescent cells, generation of ectopic telomerase, reactivation of dormant telomerase, neutralization of specific pro-inflammatory secretory products, and mitigation of the effects of mitochondrial dysfunction. EXPERT OPINION The occurrence, nature, and consequences of cellular senescence in autoimmune hepatitis must be determined. The senescence of diverse hepatic cell types could affect the outcome of autoimmune hepatitis by impairing hepatic regeneration, intensifying liver inflammation, and worsening hepatic fibrosis. Cellular senescence could contribute to suboptimal responses during conventional glucocorticoid-based therapy. Interventions that target specific pro-inflammatory products of the senescent phenotype or selectively promote apoptosis of senescent cells may be preferred adjunctive treatments for autoimmune hepatitis depending on the cancer risk.
Collapse
Affiliation(s)
- Albert J Czaja
- Mayo Clinic, Department of Medicine, Division of Gastroenterology and Hepatology, Rochester, MN, USA
| |
Collapse
|
7
|
Nai S, Wang M, Yang J, Ling B, Dong Q, Yang X, Du X, Lu M, Liu L, Yu Z, Chen L. Novel role for Ddx39 in differentiation and telomere length regulation of embryonic stem cells. Cell Death Differ 2024; 31:1534-1544. [PMID: 39107495 PMCID: PMC11519497 DOI: 10.1038/s41418-024-01354-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: 01/19/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 10/30/2024] Open
Abstract
Erk signaling is indispensable for the self-renewal and differentiation of mouse embryonic stem cells (ESCs), as well as telomere homeostasis. But how Erk regulates these biological processes remains unclear. We identified 132 Erk2 interacting proteins by co-immunoprecipitation and mass spectrometric analysis, and focused on Ddx39 as a potential Erk2 substrate. We demonstrated that Erk2 phosphorylates Ddx39 on Y132 and Y138. Ddx39 knockout (KO) ESCs are defective in differentiation, due to reduced H3K27ac level upon differentiation. Phosphorylation of Ddx39 promotes the recruitment of Hat1 to acetylate H3K27 and activate differentiation genes. In addition, Ddx39 KO leads to telomere elongation in ESCs. Ddx39 is recruited to telomeres by the telomere-binding protein Trf1, consequently disrupting the DNA loop formed by Trf1 and suppressing the alternative lengthening of telomeres (ALT). Phosphorylation of Ddx39 weakens its interaction with Trf1, releasing it from telomeres. Thus, ALT activity is enhanced, and telomeres are elongated. Altogether, our studies reveal an essential role of Ddx39 in the differentiation and telomere homeostasis of ESCs.
Collapse
Affiliation(s)
- Shanshan Nai
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University, Tianjin, China
| | - Meijie Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
| | - Jiao Yang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University, Tianjin, China
| | - Bin Ling
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University, Tianjin, China
| | - Qiman Dong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoqiong Yang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University, Tianjin, China
| | - Xiaoling Du
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University, Tianjin, China
| | - Man Lu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
| | - Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University, Tianjin, China
| | - Zhongbo Yu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, Nankai University, Tianjin, China
| | - Lingyi Chen
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Frontiers Science Center for Cell Responses, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University, Tianjin, China.
| |
Collapse
|
8
|
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
|
9
|
Douglas ME. How to write an ending: Telomere replication as a multistep process. DNA Repair (Amst) 2024; 144:103774. [PMID: 39426311 DOI: 10.1016/j.dnarep.2024.103774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
Telomeres are protective nucleoprotein caps found at the natural ends of eukaryotic chromosomes and are crucial for the preservation of stable chromosomal structure. In cycling cells, telomeres are maintained by a multi-step process called telomere replication, which involves the eukaryotic replisome navigating a complex repetitive template tightly bound by specific proteins, before terminating at the chromosome end prior to a 5' resection step that generates a protective 3' overhang. In this review, we examine mechanistic aspects of the telomere replication process and consider how individual parts of this multistep event are integrated and coordinated with one-another.
Collapse
Affiliation(s)
- Max E Douglas
- Telomere Biology Laboratory, The Institute of Cancer Research, 237 Fulham Road, London SW3 6JB, UK.
| |
Collapse
|
10
|
Rubtsova MP, Nikishin DA, Vyssokikh MY, Koriagina MS, Vasiliev AV, Dontsova OA. Telomere Reprogramming and Cellular Metabolism: Is There a Link? Int J Mol Sci 2024; 25:10500. [PMID: 39408829 PMCID: PMC11476947 DOI: 10.3390/ijms251910500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
Telomeres-special DNA-protein structures at the ends of linear eukaryotic chromosomes-define the proliferation potential of cells. Extremely short telomeres promote a DNA damage response and cell death to eliminate cells that may have accumulated mutations after multiple divisions. However, telomere elongation is associated with the increased proliferative potential of specific cell types, such as stem and germ cells. This elongation can be permanent in these cells and is activated temporally during immune response activation and regeneration processes. The activation of telomere lengthening mechanisms is coupled with increased proliferation and the cells' need for energy and building resources. To obtain the necessary nutrients, cells are capable of finely regulating energy production and consumption, switching between catabolic and anabolic processes. In this review, we focused on the interconnection between metabolism programs and telomere lengthening mechanisms during programmed activation of proliferation, such as in germ cell maturation, early embryonic development, neoplastic lesion growth, and immune response activation. It is generally accepted that telomere disturbance influences biological processes and promotes dysfunctionality. Here, we propose that metabolic conditions within proliferating cells should be involved in regulating telomere lengthening mechanisms, and telomere length may serve as a marker of defects in cellular functionality. We propose that it is possible to reprogram metabolism in order to regulate the telomere length and proliferative activity of cells, which may be important for the development of approaches to regeneration, immune response modulation, and cancer therapy. However, further investigations in this area are necessary to improve the understanding and manipulation of the molecular mechanisms involved in the regulation of proliferation, metabolism, and aging.
Collapse
Affiliation(s)
- Maria P. Rubtsova
- Chemistry Department, Lomonosov Moscow State University, Moscow 119234, Russia; (M.S.K.); (O.A.D.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117437, Russia
| | - Denis A. Nikishin
- Department of Embryology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (D.A.N.); (A.V.V.)
| | - Mikhail Y. Vyssokikh
- A.N.Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia;
| | - Maria S. Koriagina
- Chemistry Department, Lomonosov Moscow State University, Moscow 119234, Russia; (M.S.K.); (O.A.D.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117437, Russia
| | - Andrey V. Vasiliev
- Department of Embryology, Faculty of Biology, Lomonosov Moscow State University, Moscow 119234, Russia; (D.A.N.); (A.V.V.)
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia
| | - Olga A. Dontsova
- Chemistry Department, Lomonosov Moscow State University, Moscow 119234, Russia; (M.S.K.); (O.A.D.)
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow 117437, Russia
- A.N.Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia;
- Skolkovo Institute of Science and Technology, Center for Molecular and Cellular Biology, Moscow 121205, Russia
| |
Collapse
|
11
|
Hu H, Yan HL, Nguyen THD. Structural biology of shelterin and telomeric chromatin: the pieces and an unfinished puzzle. Biochem Soc Trans 2024; 52:1551-1564. [PMID: 39109533 PMCID: PMC7617103 DOI: 10.1042/bst20230300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024]
Abstract
The six-subunit shelterin complex binds to mammalian telomeres and protects them from triggering multiple DNA damage response pathways. The loss of this protective function by shelterin can have detrimental effects on cells. In this review, we first discuss structural studies of shelterin, detailing the contributions of each subunit and inter-subunit interactions in protecting chromosome ends. We then examine the influence of telomeric chromatin dynamics on the function of shelterin at telomeres. These studies provide valuable insights and underscore the challenges that future research must tackle to attain high-resolution structures of shelterin.
Collapse
Affiliation(s)
- Hongmiao Hu
- MRC Laboratory of Molecular Biology, Cambridge, U.K
| | | | | |
Collapse
|
12
|
Martin A, Schabort J, Bartke-Croughan R, Tran S, Preetham A, Lu R, Ho R, Gao J, Jenkins S, Boyle J, Ghanim GE, Jagota M, Song YS, Li H, Hockemeyer D. Dissecting the oncogenic mechanisms of POT1 cancer mutations through deep scanning mutagenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608636. [PMID: 39229243 PMCID: PMC11370387 DOI: 10.1101/2024.08.19.608636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Mutations in the shelterin protein POT1 are associated with diverse cancers, but their role in cancer progression remains unclear. To resolve this, we performed deep scanning mutagenesis in POT1 locally haploid human stem cells to assess the impact of POT1 variants on cellular viability and cancer-associated telomeric phenotypes. Though POT1 is essential, frame-shift mutants are rescued by chemical ATR inhibition, indicating that POT1 is not required for telomere replication or lagging strand synthesis. In contrast, a substantial fraction of clinically-validated pathogenic mutations support normal cellular proliferation, but still drive ATR-dependent telomeric DNA damage signaling and ATR-independent telomere elongation. Moreover, this class of cancer-associated POT1 variants elongates telomeres more rapidly than POT1 frame-shifts, indicating they actively drive oncogenesis and are not simple loss-of-function mutations.
Collapse
|
13
|
Pańczyszyn A, Boniewska-Bernacka E, Wertel I, Sadakierska-Chudy A, Goc A. Telomeres and SIRT1 as Biomarkers of Gamete Oxidative Stress, Fertility, and Potential IVF Outcome. Int J Mol Sci 2024; 25:8652. [PMID: 39201341 PMCID: PMC11354255 DOI: 10.3390/ijms25168652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
The number of infertile couples undergoing in vitro fertilisation (IVF) has increased significantly. The efficacy of this procedure is contingent upon a multitude of factors, including gamete quality. One factor influencing gamete quality is oxidative stress, which leads to telomere damage and accelerates cellular ageing. Identifying new biomarkers that can predict the success of assisted reproduction techniques is a current relevant area of research. In this review, we discuss the potential role of SIRT1, a protein known to protect against oxidative stress and telomeres, which are responsible for genome stability, as biomarkers of gamete quality and assisted reproduction technique outcomes.
Collapse
Affiliation(s)
- Anna Pańczyszyn
- Institute of Medical Sciences, Department of Biology and Genetics, Faculty of Medicine, University of Opole, Oleska 48, 45-052 Opole, Poland; (E.B.-B.); (A.G.)
| | - Ewa Boniewska-Bernacka
- Institute of Medical Sciences, Department of Biology and Genetics, Faculty of Medicine, University of Opole, Oleska 48, 45-052 Opole, Poland; (E.B.-B.); (A.G.)
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland;
| | - Anna Sadakierska-Chudy
- Department of Genetics, Faculty of Medicine and Health Sciences, Collegium Medicum, Andrzej Frycz Modrzewski Krakow University, Gustawa Herlinga-Grudzinskiego 1, 30-705 Krakow, Poland;
| | - Anna Goc
- Institute of Medical Sciences, Department of Biology and Genetics, Faculty of Medicine, University of Opole, Oleska 48, 45-052 Opole, Poland; (E.B.-B.); (A.G.)
| |
Collapse
|
14
|
Bailey SM, Kunkel SR, Bedford JS, Cornforth MN. The Central Role of Cytogenetics in Radiation Biology. Radiat Res 2024; 202:227-259. [PMID: 38981612 DOI: 10.1667/rade-24-00038.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/23/2024] [Indexed: 07/11/2024]
Abstract
Radiation cytogenetics has a rich history seldom appreciated by those outside the field. Early radiobiology was dominated by physics and biophysical concepts that borrowed heavily from the study of radiation-induced chromosome aberrations. From such studies, quantitative relationships between biological effect and changes in absorbed dose, dose rate and ionization density were codified into key concepts of radiobiological theory that have persisted for nearly a century. This review aims to provide a historical perspective of some of these concepts, including evidence supporting the contention that chromosome aberrations underlie development of many, if not most, of the biological effects of concern for humans exposed to ionizing radiations including cancer induction, on the one hand, and tumor eradication on the other. The significance of discoveries originating from these studies has widened and extended far beyond their original scope. Chromosome structural rearrangements viewed in mitotic cells were first attributed to the production of breaks by the radiations during interphase, followed by the rejoining or mis-rejoining among ends of other nearby breaks. These relatively modest beginnings eventually led to the discovery and characterization of DNA repair of double-strand breaks by non-homologous end joining, whose importance to various biological processes is now widely appreciated. Two examples, among many, are V(D)J recombination and speciation. Rapid technological advancements in cytogenetics, the burgeoning fields of molecular radiobiology and third-generation sequencing served as a point of confluence between the old and new. As a result, the emergent field of "cytogenomics" now becomes uniquely positioned for the purpose of more fully understanding mechanisms underlying the biological effects of ionizing radiation exposure.
Collapse
Affiliation(s)
- Susan M Bailey
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Stephen R Kunkel
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, Texas
| | - Joel S Bedford
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Michael N Cornforth
- Department of Radiation Oncology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
15
|
Wentworth K, Nandakumar J. At the end, it is POT1 again: Phosphorylation allows human telomeric protein POT1 to recruit the C-rich strand end replication machinery. Mol Cell 2024; 84:2598-2600. [PMID: 39059369 PMCID: PMC11613731 DOI: 10.1016/j.molcel.2024.06.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024]
Abstract
Recently in Cell, Cai et al.1 reported how phosphorylation of human shelterin protein POT1 allows it to recruit the telomeric C-rich strand replication machinery, providing mechanistic insights into an understudied area of telomere biology with implications for telomere biology disorders.
Collapse
Affiliation(s)
- Katherine Wentworth
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jayakrishnan Nandakumar
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
16
|
Bettin N, Querido E, Gialdini I, Grupelli GP, Goretti E, Cantarelli M, Andolfato M, Soror E, Sontacchi A, Jurikova K, Chartrand P, Cusanelli E. TERRA transcripts localize at long telomeres to regulate telomerase access to chromosome ends. SCIENCE ADVANCES 2024; 10:eadk4387. [PMID: 38865460 PMCID: PMC11168465 DOI: 10.1126/sciadv.adk4387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 05/08/2024] [Indexed: 06/14/2024]
Abstract
The function of TERRA in the regulation of telomerase in human cells is still debated. While TERRA interacts with telomerase, how it regulates telomerase function remains unknown. Here, we show that TERRA colocalizes with the telomerase RNA subunit hTR in the nucleoplasm and at telomeres during different phases of the cell cycle. We report that TERRA transcripts relocate away from chromosome ends during telomere lengthening, leading to a reduced number of telomeric TERRA-hTR molecules and consequent increase in "TERRA-free" telomerase molecules at telomeres. Using live-cell imaging and super-resolution microscopy, we show that upon transcription, TERRA relocates from its telomere of origin to long chromosome ends. Furthermore, TERRA depletion by antisense oligonucleotides promoted hTR localization to telomeres, leading to increased residence time and extended half-life of hTR molecules at telomeres. Overall, our findings indicate that telomeric TERRA transcripts inhibit telomere elongation by telomerase acting in trans, impairing telomerase access to telomeres that are different from their chromosome end of origin.
Collapse
Affiliation(s)
- Nicole Bettin
- Laboratory of Cell Biology and Molecular Genetics, Department CIBIO, University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Emmanuelle Querido
- Department of Biochemistry and Molecular Medicine, University of Montreal, 2900 boul. Edouard Montpetit, H3T1J4 Montreal, Canada
| | - Irene Gialdini
- Laboratory of Cell Biology and Molecular Genetics, Department CIBIO, University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Glenda Paola Grupelli
- Laboratory of Cell Biology and Molecular Genetics, Department CIBIO, University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Elena Goretti
- Laboratory of Cell Biology and Molecular Genetics, Department CIBIO, University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Marta Cantarelli
- Laboratory of Cell Biology and Molecular Genetics, Department CIBIO, University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Marta Andolfato
- Laboratory of Cell Biology and Molecular Genetics, Department CIBIO, University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Eslam Soror
- Laboratory of Cell Biology and Molecular Genetics, Department CIBIO, University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Alessandra Sontacchi
- Laboratory of Cell Biology and Molecular Genetics, Department CIBIO, University of Trento, via Sommarive 9, 38123 Trento, Italy
| | - Katarina Jurikova
- Laboratory of Cell Biology and Molecular Genetics, Department CIBIO, University of Trento, via Sommarive 9, 38123 Trento, Italy
- Department of Genetics, Faculty of Natural Sciences, Comenius University in Bratislava, Ilkovičova 6, Mlynská dolina, 84215 Bratislava, Slovakia
| | - Pascal Chartrand
- Department of Biochemistry and Molecular Medicine, University of Montreal, 2900 boul. Edouard Montpetit, H3T1J4 Montreal, Canada
| | - Emilio Cusanelli
- Laboratory of Cell Biology and Molecular Genetics, Department CIBIO, University of Trento, via Sommarive 9, 38123 Trento, Italy
| |
Collapse
|
17
|
Shoeb M, Meighan T, Kodali VK, Abadin H, Faroon O, Zarus GM, Erdely A, Antonini JM. TERT-independent telomere elongation and shelterin dysregulation after pulmonary exposure to stainless-steel welding fume in-vivo. ENVIRONMENTAL RESEARCH 2024; 250:118515. [PMID: 38373547 PMCID: PMC11375608 DOI: 10.1016/j.envres.2024.118515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/16/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
Telomeres are inert DNA sequences (TTAGGG) at the end of chromosomes that protect genetic information and maintain DNA integrity. Emerging evidence has demonstrated that telomere alteration can be closely related to occupational exposure and the development of various disease conditions, including cancer. However, the functions and underlying molecular mechanisms of telomere alteration and shelterin dysregulation after welding fume exposures have not been broadly defined. In this study, we analyzed telomere length and shelterin complex proteins in peripheral blood mononuclear cells (PBMCs) and in lung tissue recovered from male Sprague-Dawley rats following exposure by intratracheal instillation (ITI) to 2 mg/rat of manual metal arc-stainless steel (MMA-SS) welding fume particulate or saline (vehicle control). PBMCs and lung tissue were harvested at 30 d after instillation. Our study identified telomere elongation and shelterin dysregulation in PBMCs and lung tissue after welding fume exposure. Mechanistically, telomere elongation was independent of telomerase reverse transcriptase (TERT) activation. Collectively, our findings demonstrated that welding fume-induced telomere elongation was (a) TERT-independent and (b) associated with shelterin complex dysregulation. It is possible that an alteration of telomere length and its regulatory proteins may be utilized as predictive biomarkers for various disease conditions after welding fume exposure. This needs further investigation.
Collapse
Affiliation(s)
- Mohammad Shoeb
- Office of Innovation and Analytics, Agency for Toxic Substances and Disease Registry (ATSDR), Centers for Disease Control and Prevention (CDC), 4770 Buford Highway, Mailstop S106-5, Chamblee, GA, 30341, USA.
| | - Terence Meighan
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Vamsi K Kodali
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - Henry Abadin
- Office of Innovation and Analytics, Agency for Toxic Substances and Disease Registry (ATSDR), Centers for Disease Control and Prevention (CDC), 4770 Buford Highway, Mailstop S106-5, Chamblee, GA, 30341, USA
| | - Obaid Faroon
- Office of Innovation and Analytics, Agency for Toxic Substances and Disease Registry (ATSDR), Centers for Disease Control and Prevention (CDC), 4770 Buford Highway, Mailstop S106-5, Chamblee, GA, 30341, USA
| | - Gregory M Zarus
- Office of Innovation and Analytics, Agency for Toxic Substances and Disease Registry (ATSDR), Centers for Disease Control and Prevention (CDC), 4770 Buford Highway, Mailstop S106-5, Chamblee, GA, 30341, USA
| | - Aaron Erdely
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| | - James M Antonini
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV, 26505, USA
| |
Collapse
|
18
|
Audry J, Zhang H, Kerr C, Berkner KL, Runge K. Ccq1 restrains Mre11-mediated degradation to distinguish short telomeres from double-strand breaks. Nucleic Acids Res 2024; 52:3722-3739. [PMID: 38321948 PMCID: PMC11040153 DOI: 10.1093/nar/gkae044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 12/21/2023] [Accepted: 01/12/2024] [Indexed: 02/08/2024] Open
Abstract
Telomeres protect chromosome ends and are distinguished from DNA double-strand breaks (DSBs) by means of a specialized chromatin composed of DNA repeats bound by a multiprotein complex called shelterin. We investigated the role of telomere-associated proteins in establishing end-protection by studying viable mutants lacking these proteins. Mutants were studied using a Schizosaccharomyces pombe model system that induces cutting of a 'proto-telomere' bearing telomere repeats to rapidly form a new stable chromosomal end, in contrast to the rapid degradation of a control DSB. Cells lacking the telomere-associated proteins Taz1, Rap1, Poz1 or Rif1 formed a chromosome end that was stable. Surprisingly, cells lacking Ccq1, or impaired for recruiting Ccq1 to the telomere, converted the cleaved proto-telomere to a rapidly degraded DSB. Ccq1 recruits telomerase, establishes heterochromatin and affects DNA damage checkpoint activation; however, these functions were separable from protection of the new telomere by Ccq1. In cells lacking Ccq1, telomere degradation was greatly reduced by eliminating the nuclease activity of Mre11 (part of the Mre11-Rad50-Nbs1/Xrs2 DSB processing complex), and higher amounts of nuclease-deficient Mre11 associated with the new telomere. These results demonstrate a novel function for S. pombe Ccq1 to effect end-protection by restraining Mre11-dependent degradation of the DNA end.
Collapse
Affiliation(s)
- Julien Audry
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Haitao Zhang
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Carly Kerr
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Kathleen L Berkner
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | - Kurt W Runge
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
19
|
Padmanaban S, Lambacher NJ, Tesmer VM, Zhang J, Shibuya H, Nandakumar J. Caenorhabditis elegans telomere-binding proteins TEBP-1 and TEBP-2 adapt the Myb module to dimerize and bind telomeric DNA. Proc Natl Acad Sci U S A 2024; 121:e2316651121. [PMID: 38588418 PMCID: PMC11032478 DOI: 10.1073/pnas.2316651121] [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] [Accepted: 02/17/2024] [Indexed: 04/10/2024] Open
Abstract
Protecting chromosome ends from misrecognition as double-stranded (ds) DNA breaks is fundamental to eukaryotic viability. The protein complex shelterin prevents a DNA damage response at mammalian telomeres. Mammalian shelterin proteins TRF1 and TRF2 and their homologs in yeast and protozoa protect telomeric dsDNA. N-terminal homodimerization and C-terminal Myb-domain-mediated dsDNA binding are two structural hallmarks of end protection by TRF homologs. Yet our understanding of how Caenorhabditis elegans protects its telomeric dsDNA is limited. Recently identified C. elegans proteins TEBP-1 (also called DTN-1) and TEBP-2 (also called DTN-2) are functional homologs of TRF proteins, but how they bind DNA and whether or how they dimerize is not known. TEBP-1 and TEBP-2 harbor three Myb-containing domains (MCDs) and no obvious dimerization domain. We demonstrate biochemically that only the third MCD binds DNA. We solve the X-ray crystal structure of TEBP-2 MCD3 with telomeric dsDNA to reveal the structural mechanism of telomeric dsDNA protection in C. elegans. Mutagenesis of the DNA-binding site of TEBP-1 and TEBP-2 compromises DNA binding in vitro, and increases DNA damage signaling, lengthens telomeres, and decreases brood size in vivo. Via an X-ray crystal structure, biochemical validation of the dimerization interface, and SEC-MALS analysis, we demonstrate that MCD1 and MCD2 form a composite dimerization module that facilitates not only TEBP-1 and TEBP-2 homodimerization but also heterodimerization. These findings provide fundamental insights into C. elegans telomeric dsDNA protection and highlight how different eukaryotes have evolved distinct strategies to solve the chromosome end protection problem.
Collapse
Affiliation(s)
- Shilpa Padmanaban
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI48109
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO63110
| | - Nils J. Lambacher
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden41390
| | - Valerie M. Tesmer
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI48109
| | - Jingjing Zhang
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden41390
| | - Hiroki Shibuya
- Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden41390
- Laboratory for Gametogenesis, RIKEN Center for Biosystems Dynamics Research, Kobe650-0047, Japan
| | - Jayakrishnan Nandakumar
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI48109
| |
Collapse
|
20
|
Olson CL, Wuttke DS. Guardians of the Genome: How the Single-Stranded DNA-Binding Proteins RPA and CST Facilitate Telomere Replication. Biomolecules 2024; 14:263. [PMID: 38540683 PMCID: PMC10968030 DOI: 10.3390/biom14030263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/02/2024] [Accepted: 02/20/2024] [Indexed: 04/26/2024] Open
Abstract
Telomeres act as the protective caps of eukaryotic linear chromosomes; thus, proper telomere maintenance is crucial for genome stability. Successful telomere replication is a cornerstone of telomere length regulation, but this process can be fraught due to the many intrinsic challenges telomeres pose to the replication machinery. In addition to the famous "end replication" problem due to the discontinuous nature of lagging strand synthesis, telomeres require various telomere-specific steps for maintaining the proper 3' overhang length. Bulk telomere replication also encounters its own difficulties as telomeres are prone to various forms of replication roadblocks. These roadblocks can result in an increase in replication stress that can cause replication forks to slow, stall, or become reversed. Ultimately, this leads to excess single-stranded DNA (ssDNA) that needs to be managed and protected for replication to continue and to prevent DNA damage and genome instability. RPA and CST are single-stranded DNA-binding protein complexes that play key roles in performing this task and help stabilize stalled forks for continued replication. The interplay between RPA and CST, their functions at telomeres during replication, and their specialized features for helping overcome replication stress at telomeres are the focus of this review.
Collapse
Affiliation(s)
- Conner L. Olson
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Deborah S. Wuttke
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
21
|
Lu X, Liu L. Genome stability from the perspective of telomere length. Trends Genet 2024; 40:175-186. [PMID: 37957036 DOI: 10.1016/j.tig.2023.10.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/20/2023] [Accepted: 10/23/2023] [Indexed: 11/15/2023]
Abstract
Telomeres and their associated proteins protect the ends of chromosomes to maintain genome stability. Telomeres undergo progressive shortening with each cell division in mammalian somatic cells without telomerase, resulting in genome instability. When telomeres reach a critically short length or are recognized as a damage signal, cells enter a state of senescence, followed by cell cycle arrest, programmed cell death, or immortalization. This review provides an overview of recent advances in the intricate relationship between telomeres and genome instability. Alongside well-established mechanisms such as chromosomal fusion and telomere fusion, we will delve into the perspective on genome stability by examining the role of retrotransposons. Retrotransposons represent an emerging pathway to regulate genome stability through their interactions with telomeres.
Collapse
Affiliation(s)
- Xinyi Lu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, Tianjin 300350, China.
| | - Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, Tianjin 300350, China; Frontiers Science Center for Cell Responses, College of Life Science, Nankai University, Tianjin, Tianjin 300071, China; Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China; Institute of Translational Medicine, Tianjin Union Medical Center, Nankai University, Tianjin 300000, China.
| |
Collapse
|
22
|
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
|
23
|
Lim CJ. Telomere C-Strand Fill-In Machinery: New Insights into the Human CST-DNA Polymerase Alpha-Primase Structures and Functions. Subcell Biochem 2024; 104:73-100. [PMID: 38963484 DOI: 10.1007/978-3-031-58843-3_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Telomeres at the end of eukaryotic chromosomes are extended by a specialized set of enzymes and telomere-associated proteins, collectively termed here the telomere "replisome." The telomere replisome acts on a unique replicon at each chromosomal end of the telomeres, the 3' DNA overhang. This telomere replication process is distinct from the replisome mechanism deployed to duplicate the human genome. The G-rich overhang is first extended before the complementary C-strand is filled in. This overhang is extended by telomerase, a specialized ribonucleoprotein and reverse transcriptase. The overhang extension process is terminated when telomerase is displaced by CTC1-STN1-TEN1 (CST), a single-stranded DNA-binding protein complex. CST then recruits DNA polymerase α-primase to complete the telomere replication process by filling in the complementary C-strand. In this chapter, the recent structure-function insights into the human telomere C-strand fill-in machinery (DNA polymerase α-primase and CST) will be discussed.
Collapse
Affiliation(s)
- Ci Ji Lim
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
24
|
Nassour J, Przetocka S, Karlseder J. Telomeres as hotspots for innate immunity and inflammation. DNA Repair (Amst) 2024; 133:103591. [PMID: 37951043 PMCID: PMC10842095 DOI: 10.1016/j.dnarep.2023.103591] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/05/2023] [Accepted: 10/24/2023] [Indexed: 11/13/2023]
Abstract
Aging is marked by the gradual accumulation of deleterious changes that disrupt organ function, creating an altered physiological state that is permissive for the onset of prevalent human diseases. While the exact mechanisms governing aging remain a subject of ongoing research, there are several cellular and molecular hallmarks that contribute to this biological process. This review focuses on two factors, namely telomere dysfunction and inflammation, which have emerged as crucial contributors to the aging process. We aim to discuss the mechanistic connections between these two distinct hallmarks and provide compelling evidence highlighting the loss of telomere protection as a driver of pro-inflammatory states associated with aging. By reevaluating the interplay between telomeres, innate immunity, and inflammation, we present novel perspectives on the etiology of aging and its associated diseases.
Collapse
Affiliation(s)
- Joe Nassour
- Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, 12801 E. 17th Ave, Aurora, CO 80045, USA; The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Sara Przetocka
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Jan Karlseder
- The Salk Institute for Biological Studies, 10010 North Torrey Pines Rd, La Jolla, CA 92037, USA.
| |
Collapse
|
25
|
Robinson LG, Kalmbach K, Sumerfield O, Nomani W, Wang F, Liu L, Keefe DL. Telomere dynamics and reproduction. Fertil Steril 2024; 121:4-11. [PMID: 37993053 DOI: 10.1016/j.fertnstert.2023.11.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023]
Abstract
The oocyte, a long-lived, postmitotic cell, is the locus of reproductive aging in women. Female germ cells replicate only during fetal life and age throughout reproductive life. Mechanisms of oocyte aging include the accumulation of oxidative damage, mitochondrial dysfunction, and disruption of proteins, including cohesion. Nobel Laureate Bob Edwards also discovered a "production line" during oogonial replication in the mouse, wherein the last oocytes to ovulate in the adult-derived from the last oogonia to exit mitotic replication in the fetus. On the basis of this, we proposed a two-hit "telomere theory of reproductive aging" to integrate the myriad features of oocyte aging. The first hit was that oocytes remaining in older women traversed more cell cycles during fetal oogenesis. The second hit was that oocytes accumulated more environmental and endogenous oxidative damage throughout the life of the woman. Telomeres (Ts) could mediate both of these aspects of oocyte aging. Telomeres provide a "mitotic clock," with T attrition an inevitable consequence of cell division because of the end replication problem. Telomere's guanine-rich sequence renders them especially sensitive to oxidative damage, even in postmitotic cells. Telomerase, the reverse transcriptase that restores Ts, is better at maintaining than elongating T. Moreover, telomerase remains inactive during much of oogenesis and early development. Oocytes are left with short Ts, on the brink of viability. In support of this theory, mice with induced T attrition and women with naturally occurring telomeropathy suffer diminished ovarian reserve, abnormal embryo development, and infertility. In contrast, sperm are produced throughout the life of the male by a telomerase-active progenitor, spermatogonia, resulting in the longest Ts in the body. In mice, cleavage-stage embryos elongate Ts via "alternative lengthening of telomeres," a recombination-based mechanism rarely encountered outside of telomerase-deficient cancers. Many questions about Ts and reproduction are raised by these findings: does the "normal" T attrition observed in human oocytes contribute to their extraordinarily high rate of meiotic nondisjunction? Does recombination-based T elongation render embryos susceptible to mitotic nondisjunction (and mosaicism)? Can some features of Ts serve as markers of oocyte quality?
Collapse
Affiliation(s)
- LeRoy G Robinson
- Department of Obstetrics and Gynecology, New York University Langone Fertility Center, New York University School of Medicine, NYU Langone Health, New York, New York; Department of Biology, San Francisco State University, San Francisco, California
| | - Keri Kalmbach
- Department of Obstetrics and Gynecology, New York University Langone Fertility Center, New York University School of Medicine, NYU Langone Health, New York, New York
| | - Olivia Sumerfield
- Department of Obstetrics and Gynecology, New York University Langone Fertility Center, New York University School of Medicine, NYU Langone Health, New York, New York
| | - Wafa Nomani
- Department of Obstetrics and Gynecology, New York University Langone Fertility Center, New York University School of Medicine, NYU Langone Health, New York, New York
| | - Fang Wang
- Department of Obstetrics and Gynecology, New York University Langone Fertility Center, New York University School of Medicine, NYU Langone Health, New York, New York
| | - Lin Liu
- College of Life Sciences, Nankai University, Tianjin, People's Republic of China
| | - David L Keefe
- Department of Obstetrics and Gynecology, New York University Langone Fertility Center, New York University School of Medicine, NYU Langone Health, New York, New York.
| |
Collapse
|
26
|
Carvalho Borges PC, Bouabboune C, Escandell JM, Matmati S, Coulon S, Ferreira MG. Pot1 promotes telomere DNA replication via the Stn1-Ten1 complex in fission yeast. Nucleic Acids Res 2023; 51:12325-12336. [PMID: 37953281 PMCID: PMC10711446 DOI: 10.1093/nar/gkad1036] [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: 01/20/2023] [Revised: 10/19/2023] [Accepted: 10/31/2023] [Indexed: 11/14/2023] Open
Abstract
Telomeres are nucleoprotein complexes that protect the chromosome-ends from eliciting DNA repair while ensuring their complete duplication. Pot1 is a subunit of telomere capping complex that binds to the G-rich overhang and inhibits the activation of DNA damage checkpoints. In this study, we explore new functions of fission yeast Pot1 by using a pot1-1 temperature sensitive mutant. We show that pot1 inactivation impairs telomere DNA replication resulting in the accumulation of ssDNA leading to the complete loss of telomeric DNA. Recruitment of Stn1 to telomeres, an auxiliary factor of DNA lagging strand synthesis, is reduced in pot1-1 mutants and overexpression of Stn1 rescues loss of telomeres and cell viability at restrictive temperature. We propose that Pot1 plays a crucial function in telomere DNA replication by recruiting Stn1-Ten1 and Polα-primase complex to telomeres via Tpz1, thus promoting lagging-strand DNA synthesis at stalled replication forks.
Collapse
Affiliation(s)
| | - Chaïnez Bouabboune
- CNRS, INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, CRCM, Equipe labellisée par la Ligue Nationale contre le Cancer, Marseille, F-13009, France
| | | | - Samah Matmati
- CNRS, INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, CRCM, Equipe labellisée par la Ligue Nationale contre le Cancer, Marseille, F-13009, France
| | - Stéphane Coulon
- CNRS, INSERM, Aix Marseille Univ, Institut Paoli-Calmettes, CRCM, Equipe labellisée par la Ligue Nationale contre le Cancer, Marseille, F-13009, France
| | - Miguel Godinho Ferreira
- Instituto Gulbenkian de Ciência, Oeiras, 2781-901, Portugal
- Institute for Research on Cancer and Aging of Nice (IRCAN), INSERM U1081 UMR7284 CNRS, 06107 Nice, France
| |
Collapse
|
27
|
Li B. Telomere maintenance in African trypanosomes. Front Mol Biosci 2023; 10:1302557. [PMID: 38074093 PMCID: PMC10704157 DOI: 10.3389/fmolb.2023.1302557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/15/2023] [Indexed: 02/12/2024] Open
Abstract
Telomere maintenance is essential for genome integrity and chromosome stability in eukaryotic cells harboring linear chromosomes, as telomere forms a specialized structure to mask the natural chromosome ends from DNA damage repair machineries and to prevent nucleolytic degradation of the telomeric DNA. In Trypanosoma brucei and several other microbial pathogens, virulence genes involved in antigenic variation, a key pathogenesis mechanism essential for host immune evasion and long-term infections, are located at subtelomeres, and expression and switching of these major surface antigens are regulated by telomere proteins and the telomere structure. Therefore, understanding telomere maintenance mechanisms and how these pathogens achieve a balance between stability and plasticity at telomere/subtelomere will help develop better means to eradicate human diseases caused by these pathogens. Telomere replication faces several challenges, and the "end replication problem" is a key obstacle that can cause progressive telomere shortening in proliferating cells. To overcome this challenge, most eukaryotes use telomerase to extend the G-rich telomere strand. In addition, a number of telomere proteins use sophisticated mechanisms to coordinate the telomerase-mediated de novo telomere G-strand synthesis and the telomere C-strand fill-in, which has been extensively studied in mammalian cells. However, we recently discovered that trypanosomes lack many telomere proteins identified in its mammalian host that are critical for telomere end processing. Rather, T. brucei uses a unique DNA polymerase, PolIE that belongs to the DNA polymerase A family (E. coli DNA PolI family), to coordinate the telomere G- and C-strand syntheses. In this review, I will first briefly summarize current understanding of telomere end processing in mammals. Subsequently, I will describe PolIE-mediated coordination of telomere G- and C-strand synthesis in T. brucei and implication of this recent discovery.
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, 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
|
28
|
Yan M, Cheng S, Wang S, Duan X, Mensah AR, Li L, Zhang Y, Li G, Zhao J, Feng F, Zhou X, Wu Y, Yang Y, Wang W. Association of Genetic Polymorphisms of TERT with Telomere Length in Coke Oven Emissions-Exposed Workers. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2023; 33:1059-1069. [PMID: 35469505 DOI: 10.1080/09603123.2022.2069687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 04/20/2022] [Indexed: 06/14/2023]
Abstract
We explored the association between variations in the telomere maintenance genes and change in telomere length (TL) in workers. The TL of peripheral blood leukocytes from 544 coke oven workers and 238 controls were detected using the Real-time PCR method. Variations in four genes were then detected using the PCR based restriction fragment length polymorphism. The effects of environmental and genetic factors on TL were subsequently analyzed through covariance analysis and a generalized linear model .The TL of subjects with GG genotypes were longer than those with AG genotype in the TERT rs2736098 locus amongst the controls (P = .032). The combined effect of COEs exposure and AG+AA genotypes had a significant effect on TL (P < .001). The interaction between the COEs exposure factor and the rs2736098AG+AA genotypes had a significant effect on the TL (P < .05). The TL in coke oven workers is associated with the interactions between TERT rs2736098 AG+AA and COEs exposure.
Collapse
Affiliation(s)
- Mengqing Yan
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
- The Key Laboratory of Nanomedicine and Health Inspection of Zhengzhou, Zhengzhou, China
| | - Shuai Cheng
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
- The Key Laboratory of Nanomedicine and Health Inspection of Zhengzhou, Zhengzhou, China
| | - Sihua Wang
- Department of Occupational Health, Henan Institute for Occupational Medicine, Zhengzhou, China
| | - Xiaoran Duan
- National Engineering Laboratory for Internet Medical Systems and Applications, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Acquaye Reuben Mensah
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Lei Li
- Department of Occupational Health, Zhengzhou Institute for Occupational Medicine, Zhengzhou, China
| | - Yuhong Zhang
- Department of Occupational Health, Zhengzhou Institute for Occupational Medicine, Zhengzhou, China
| | - Guoyu Li
- Department of Occupational Health, Zhengzhou Institute for Occupational Medicine, Zhengzhou, China
| | - Junfeng Zhao
- Department of Occupational Health, Zhengzhou Institute for Occupational Medicine, Zhengzhou, China
| | - Feifei Feng
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Xiaoshan Zhou
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yongjun Wu
- The Key Laboratory of Nanomedicine and Health Inspection of Zhengzhou, Zhengzhou, China
- Department of Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yongli Yang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Wei Wang
- Department of Occupational and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
- The Key Laboratory of Nanomedicine and Health Inspection of Zhengzhou, Zhengzhou, China
| |
Collapse
|
29
|
He Q, Lim CJ. Models for human telomere C-strand fill-in by CST-Polα-primase. Trends Biochem Sci 2023; 48:860-872. [PMID: 37586999 PMCID: PMC10528720 DOI: 10.1016/j.tibs.2023.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023]
Abstract
Telomere maintenance is essential for the genome integrity of eukaryotes, and this function is underpinned by the two-step telomeric DNA synthesis process: telomere G-overhang extension by telomerase and complementary strand fill-in by DNA polymerase alpha-primase (polα-primase). Compared to the telomerase step, the telomere C-strand fill-in mechanism is less understood. Recent studies have provided new insights into how telomeric single-stranded DNA-binding protein CTC1-STN1-TEN1 (CST) and polα-primase coordinate to synthesize the telomeric C-strand for telomere overhang fill-in. Cryogenic electron microscopy (cryo-EM) structures of CST-polα-primase complexes have provided additional insights into how they assemble at telomeric templates and de novo synthesize the telomere C-strand. In this review, we discuss how these latest findings coalesce with existing understanding to develop a human telomere C-strand fill-in mechanism model.
Collapse
Affiliation(s)
- Qixiang He
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Ci Ji Lim
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
30
|
Sivasangari K, Sivamaruthi BS, Chaiyasut C, Rajan KE. Maternal stress-induced changes in adolescent and adult offspring: Neurobehavioural improvement and telomere maintenance. Heliyon 2023; 9:e20385. [PMID: 37767490 PMCID: PMC10520813 DOI: 10.1016/j.heliyon.2023.e20385] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 07/22/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
Maternal stress (MS) during gestation is known to increase the risk for the development of behavioural and physiological disorders and advances cellular aging. In this study, we investigated whether the supplementation of standardized Bacopa monnieri extract (CDRI-08/BME) or l-Carnosine (L-C) to the mother exposed to social stress during gestation modify the effect on their offspring's neurobehaviour, antioxidant defence gene expression, telomere length, and telomere biology. To test this, timed pregnant rats were subjected to social stress during the gestational day (GD) 16-18. A subset of stressed pregnant rats received either BME [80 mg/kg in 0.5% gum acacia (per-orally; p.o)] or L-C [1 mg/kg (p.o)] every day from GD-10 to until their pup's attained postnatal day (PND)-23. We observed that MS induced anxiety-like behaviour, altered inter-limb coordination, antioxidant defence genes [Superoxide dismutase (SOD1,2), Catalase (CAT), Glutathione peroxidase-3 (GPX3)], telomerase reverse transcriptase (TERT), shelterin complex subunits (TRF1, RAP1B, POT1) protein level and shorten telomere length. Notably, supplementation of BME/L-C dampens the MS, thus the effect on neurobehaviour, antioxidant defence gene expression, and telomere biology is minimized in their offspring. Together, our results suggest that supplementation of BME/L-C during gestation dampens the MS and reduced oxidative stress-mediated changes in telomere shortening/biology and associated neurobehaviour in offspring born following MS.
Collapse
Affiliation(s)
- Karunanithi Sivasangari
- Behavioural Neuroscience Laboratory, Department of Animal Science, Bharathidasan University, Tiruchirappalli, India
| | - Bhagavathi Sundaram Sivamaruthi
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Koilmani Emmanuvel Rajan
- Behavioural Neuroscience Laboratory, Department of Animal Science, Bharathidasan University, Tiruchirappalli, India
| |
Collapse
|
31
|
Takasugi T, Gu P, Liang F, Staco I, Chang S. Pot1b -/- tumors activate G-quadruplex-induced DNA damage to promote telomere hyper-elongation. Nucleic Acids Res 2023; 51:9227-9247. [PMID: 37560909 PMCID: PMC10516629 DOI: 10.1093/nar/gkad648] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/14/2023] [Accepted: 07/22/2023] [Indexed: 08/11/2023] Open
Abstract
Malignant cancers must activate telomere maintenance mechanisms to achieve replicative immortality. Mutations in the human Protection of Telomeres 1 (POT1) gene are frequently detected in cancers with abnormally long telomeres, suggesting that the loss of POT1 function disrupts the regulation of telomere length homeostasis to promote telomere elongation. However, our understanding of the mechanisms leading to elongated telomeres remains incomplete. The mouse genome encodes two POT1 proteins, POT1a and POT1b possessing separation of hPOT1 functions. We performed serial transplantation of Pot1b-/- sarcomas to better understand the role of POT1b in regulating telomere length maintenance. While early-generation Pot1b-/- sarcomas initially possessed shortened telomeres, late-generation Pot1b-/- cells display markedly hyper-elongated telomeres that were recognized as damaged DNA by the Replication Protein A (RPA) complex. The RPA-ATR-dependent DNA damage response at telomeres promotes telomerase recruitment to facilitate telomere hyper-elongation. POT1b, but not POT1a, was able to unfold G-quadruplex present in hyper-elongated telomeres to repress the DNA damage response. Our findings demonstrate that the repression of the RPA-ATR DDR is conserved between POT1b and human POT1, suggesting that similar mechanisms may underly the phenotypes observed in human cancers harboring human POT1 mutations.
Collapse
Affiliation(s)
- Taylor Takasugi
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Peili Gu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Fengshan Liang
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Isabelle Staco
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Sandy Chang
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine, New Haven, CT 06520, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
32
|
Sharaf R, Jin DX, Grady J, Napier C, Ebot E, Frampton GM, Albacker LA, Thomas DM, Montesion M. A pan-sarcoma landscape of telomeric content shows that alterations in RAD51B and GID4 are associated with higher telomeric content. NPJ Genom Med 2023; 8:26. [PMID: 37709802 PMCID: PMC10502097 DOI: 10.1038/s41525-023-00369-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/18/2023] [Indexed: 09/16/2023] Open
Abstract
Tumor cells need to activate a telomere maintenance mechanism, enabling limitless replication. The bulk of evidence supports that sarcomas predominantly use alternative lengthening of telomeres (ALT) mechanism, commonly associated with alterations in ATRX and DAXX. In our dataset, only 12.3% of sarcomas harbored alterations in these genes. Thus, we checked for the presence of other genomic determinants of high telomeric content in sarcomas. Our dataset consisted of 13555 sarcoma samples, sequenced as a part of routine clinical care on the FoundationOne®Heme platform. We observed a median telomeric content of 622.3 telomeric reads per GC-matched million reads (TRPM) across all samples. In agreement with previous studies, telomeric content was significantly higher in ATRX altered and POT1 altered sarcomas. We further observed that sarcomas with alterations in RAD51B or GID4 were enriched in samples with high telomeric content, specifically within uterus leiomyosarcoma for RAD51B and soft tissue sarcoma (not otherwise specified, nos) for GID4, Furthermore, RAD51B and POT1 alterations were mutually exclusive with ATRX and DAXX alterations, suggestive of functional redundancy. Our results propose a role played by RAD51B and GID4 in telomere elongation in sarcomas and open research opportunities for agents aimed at targeting this critical pathway in tumorigenesis.
Collapse
Affiliation(s)
| | | | - John Grady
- Omico Australian Genomic Cancer Medicine, Sydney, Australia
- Garvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - Christine Napier
- Omico Australian Genomic Cancer Medicine, Sydney, Australia
- Garvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - Ericka Ebot
- Foundation Medicine Inc., Cambridge, MA, USA
| | | | | | - David M Thomas
- Omico Australian Genomic Cancer Medicine, Sydney, Australia
- Garvan Institute of Medical Research, Sydney, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | | |
Collapse
|
33
|
Chaux F, Agier N, Garrido C, Fischer G, Eberhard S, Xu Z. Telomerase-independent survival leads to a mosaic of complex subtelomere rearrangements in Chlamydomonas reinhardtii. Genome Res 2023; 33:1582-1598. [PMID: 37580131 PMCID: PMC10620057 DOI: 10.1101/gr.278043.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/09/2023] [Indexed: 08/16/2023]
Abstract
Telomeres and subtelomeres, the genomic regions located at chromosome extremities, are essential for genome stability in eukaryotes. In the absence of the canonical maintenance mechanism provided by telomerase, telomere shortening induces genome instability. The landscape of the ensuing genome rearrangements is not accessible by short-read sequencing. Here, we leverage Oxford Nanopore Technologies long-read sequencing to survey the extensive repertoire of genome rearrangements in telomerase mutants of the model green microalga Chlamydomonas reinhardtii In telomerase-mutant strains grown for hundreds of generations, most chromosome extremities were capped by short telomere sequences that were either recruited de novo from other loci or maintained in a telomerase-independent manner. Other extremities did not end with telomeres but only with repeated subtelomeric sequences. The subtelomeric elements, including rDNA, were massively rearranged and involved in breakage-fusion-bridge cycles, translocations, recombinations, and chromosome circularization. These events were established progressively over time and displayed heterogeneity at the subpopulation level. New telomere-capped extremities composed of sequences originating from more internal genomic regions were associated with high DNA methylation, suggesting that de novo heterochromatin formation contributes to the restoration of chromosome end stability in C. reinhardtii The diversity of alternative strategies present in the same organism to maintain chromosome integrity and the variety of rearrangements found in telomerase mutants are remarkable, and illustrate genome plasticity at short timescales.
Collapse
Affiliation(s)
- Frédéric Chaux
- Sorbonne Université, CNRS, UMR7238, Institut de Biologie Paris-Seine, Laboratory of Computational and Quantitative Biology, 75005 Paris, France
| | - Nicolas Agier
- Sorbonne Université, CNRS, UMR7238, Institut de Biologie Paris-Seine, Laboratory of Computational and Quantitative Biology, 75005 Paris, France
| | - Clotilde Garrido
- Sorbonne Université, CNRS, UMR7238, Institut de Biologie Paris-Seine, Laboratory of Computational and Quantitative Biology, 75005 Paris, France
| | - Gilles Fischer
- Sorbonne Université, CNRS, UMR7238, Institut de Biologie Paris-Seine, Laboratory of Computational and Quantitative Biology, 75005 Paris, France
| | - Stephan Eberhard
- Sorbonne Université, CNRS, UMR7141, Institut de Biologie Physico-Chimique, Laboratory of Chloroplast Biology and Light-Sensing in Microalgae, 75005 Paris, France
| | - Zhou Xu
- Sorbonne Université, CNRS, UMR7238, Institut de Biologie Paris-Seine, Laboratory of Computational and Quantitative Biology, 75005 Paris, France;
| |
Collapse
|
34
|
Tesmer VM, Brenner KA, Nandakumar J. Human POT1 protects the telomeric ds-ss DNA junction by capping the 5' end of the chromosome. Science 2023; 381:771-778. [PMID: 37590346 PMCID: PMC10666826 DOI: 10.1126/science.adi2436] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 07/19/2023] [Indexed: 08/19/2023]
Abstract
Protection of telomeres 1 (POT1) is the 3' single-stranded overhang-binding telomeric protein that prevents an ataxia telangiectasia and Rad3-related (ATR) DNA damage response (DDR) at chromosome ends. What precludes the DDR machinery from accessing the telomeric double-stranded-single-stranded junction is unknown. We demonstrate that human POT1 binds this junction by recognizing the phosphorylated 5' end of the chromosome. High-resolution crystallographic structures reveal that the junction is capped by POT1 through a "POT-hole" surface, the mutation of which compromises junction protection in vitro and telomeric 5'-end definition and DDR suppression in human cells. Whereas both mouse POT1 paralogs bind the single-stranded overhang, POT1a, not POT1b, contains a POT-hole and binds the junction, which explains POT1a's sufficiency for end protection. Our study shifts the paradigm for DDR suppression at telomeres by highlighting the importance of protecting the double-stranded-single-stranded junction.
Collapse
Affiliation(s)
- Valerie M. Tesmer
- Department of Molecular, Cellular and Developmental Biology, University of Michigan; Ann Arbor, 48109, USA
| | - Kirsten A. Brenner
- Department of Molecular, Cellular and Developmental Biology, University of Michigan; Ann Arbor, 48109, USA
| | - Jayakrishnan Nandakumar
- Department of Molecular, Cellular and Developmental Biology, University of Michigan; Ann Arbor, 48109, USA
| |
Collapse
|
35
|
DeBoy EA, Tassia MG, Schratz KE, Yan SM, Cosner ZL, McNally EJ, Gable DL, Xiang Z, Lombard DB, Antonarakis ES, Gocke CD, McCoy RC, Armanios M. Familial Clonal Hematopoiesis in a Long Telomere Syndrome. N Engl J Med 2023; 388:2422-2433. [PMID: 37140166 PMCID: PMC10501156 DOI: 10.1056/nejmoa2300503] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
BACKGROUND Telomere shortening is a well-characterized cellular aging mechanism, and short telomere syndromes cause age-related disease. However, whether long telomere length is advantageous is poorly understood. METHODS We examined the clinical and molecular features of aging and cancer in persons carrying heterozygous loss-of-function mutations in the telomere-related gene POT1 and noncarrier relatives. RESULTS A total of 17 POT1 mutation carriers and 21 noncarrier relatives were initially included in the study, and a validation cohort of 6 additional mutation carriers was subsequently recruited. A majority of the POT1 mutation carriers with telomere length evaluated (9 of 13) had long telomeres (>99th percentile). POT1 mutation carriers had a range of benign and malignant neoplasms involving epithelial, mesenchymal, and neuronal tissues in addition to B- and T-cell lymphoma and myeloid cancers. Five of 18 POT1 mutation carriers (28%) had T-cell clonality, and 8 of 12 (67%) had clonal hematopoiesis of indeterminate potential. A predisposition to clonal hematopoiesis had an autosomal dominant pattern of inheritance, as well as penetrance that increased with age; somatic DNMT3A and JAK2 hotspot mutations were common. These and other somatic driver mutations probably arose in the first decades of life, and their lineages secondarily accumulated a higher mutation burden characterized by a clocklike signature. Successive generations showed genetic anticipation (i.e., an increasingly early onset of disease). In contrast to noncarrier relatives, who had the typical telomere shortening with age, POT1 mutation carriers maintained telomere length over the course of 2 years. CONCLUSIONS POT1 mutations associated with long telomere length conferred a predisposition to a familial clonal hematopoiesis syndrome that was associated with a range of benign and malignant solid neoplasms. The risk of these phenotypes was mediated by extended cellular longevity and by the capacity to maintain telomeres over time. (Funded by the National Institutes of Health and others.).
Collapse
Affiliation(s)
- Emily A DeBoy
- From the Departments of Oncology (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., E.S.A., C.D.G., M.A.), Pathology (C.D.G., M.A.), and Genetic Medicine (M.A.), the Medical Scientist Training Program (E.A.D.), the Telomere Center (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., M.A.), and Sidney Kimmel Comprehensive Cancer Center (K.E.S., E.S.A., C.D.G., M.A.), Johns Hopkins University School of Medicine, and the Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University (M.G.T., S.M.Y., R.C.M.) - both in Baltimore; the Child Neurology Residency Program, Boston Children's Hospital, Boston (D.L.G.); the Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami (D.B.L.); and the Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis (E.S.A.)
| | - Michael G Tassia
- From the Departments of Oncology (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., E.S.A., C.D.G., M.A.), Pathology (C.D.G., M.A.), and Genetic Medicine (M.A.), the Medical Scientist Training Program (E.A.D.), the Telomere Center (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., M.A.), and Sidney Kimmel Comprehensive Cancer Center (K.E.S., E.S.A., C.D.G., M.A.), Johns Hopkins University School of Medicine, and the Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University (M.G.T., S.M.Y., R.C.M.) - both in Baltimore; the Child Neurology Residency Program, Boston Children's Hospital, Boston (D.L.G.); the Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami (D.B.L.); and the Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis (E.S.A.)
| | - Kristen E Schratz
- From the Departments of Oncology (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., E.S.A., C.D.G., M.A.), Pathology (C.D.G., M.A.), and Genetic Medicine (M.A.), the Medical Scientist Training Program (E.A.D.), the Telomere Center (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., M.A.), and Sidney Kimmel Comprehensive Cancer Center (K.E.S., E.S.A., C.D.G., M.A.), Johns Hopkins University School of Medicine, and the Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University (M.G.T., S.M.Y., R.C.M.) - both in Baltimore; the Child Neurology Residency Program, Boston Children's Hospital, Boston (D.L.G.); the Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami (D.B.L.); and the Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis (E.S.A.)
| | - Stephanie M Yan
- From the Departments of Oncology (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., E.S.A., C.D.G., M.A.), Pathology (C.D.G., M.A.), and Genetic Medicine (M.A.), the Medical Scientist Training Program (E.A.D.), the Telomere Center (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., M.A.), and Sidney Kimmel Comprehensive Cancer Center (K.E.S., E.S.A., C.D.G., M.A.), Johns Hopkins University School of Medicine, and the Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University (M.G.T., S.M.Y., R.C.M.) - both in Baltimore; the Child Neurology Residency Program, Boston Children's Hospital, Boston (D.L.G.); the Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami (D.B.L.); and the Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis (E.S.A.)
| | - Zoe L Cosner
- From the Departments of Oncology (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., E.S.A., C.D.G., M.A.), Pathology (C.D.G., M.A.), and Genetic Medicine (M.A.), the Medical Scientist Training Program (E.A.D.), the Telomere Center (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., M.A.), and Sidney Kimmel Comprehensive Cancer Center (K.E.S., E.S.A., C.D.G., M.A.), Johns Hopkins University School of Medicine, and the Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University (M.G.T., S.M.Y., R.C.M.) - both in Baltimore; the Child Neurology Residency Program, Boston Children's Hospital, Boston (D.L.G.); the Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami (D.B.L.); and the Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis (E.S.A.)
| | - Emily J McNally
- From the Departments of Oncology (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., E.S.A., C.D.G., M.A.), Pathology (C.D.G., M.A.), and Genetic Medicine (M.A.), the Medical Scientist Training Program (E.A.D.), the Telomere Center (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., M.A.), and Sidney Kimmel Comprehensive Cancer Center (K.E.S., E.S.A., C.D.G., M.A.), Johns Hopkins University School of Medicine, and the Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University (M.G.T., S.M.Y., R.C.M.) - both in Baltimore; the Child Neurology Residency Program, Boston Children's Hospital, Boston (D.L.G.); the Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami (D.B.L.); and the Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis (E.S.A.)
| | - Dustin L Gable
- From the Departments of Oncology (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., E.S.A., C.D.G., M.A.), Pathology (C.D.G., M.A.), and Genetic Medicine (M.A.), the Medical Scientist Training Program (E.A.D.), the Telomere Center (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., M.A.), and Sidney Kimmel Comprehensive Cancer Center (K.E.S., E.S.A., C.D.G., M.A.), Johns Hopkins University School of Medicine, and the Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University (M.G.T., S.M.Y., R.C.M.) - both in Baltimore; the Child Neurology Residency Program, Boston Children's Hospital, Boston (D.L.G.); the Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami (D.B.L.); and the Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis (E.S.A.)
| | - Zhimin Xiang
- From the Departments of Oncology (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., E.S.A., C.D.G., M.A.), Pathology (C.D.G., M.A.), and Genetic Medicine (M.A.), the Medical Scientist Training Program (E.A.D.), the Telomere Center (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., M.A.), and Sidney Kimmel Comprehensive Cancer Center (K.E.S., E.S.A., C.D.G., M.A.), Johns Hopkins University School of Medicine, and the Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University (M.G.T., S.M.Y., R.C.M.) - both in Baltimore; the Child Neurology Residency Program, Boston Children's Hospital, Boston (D.L.G.); the Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami (D.B.L.); and the Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis (E.S.A.)
| | - David B Lombard
- From the Departments of Oncology (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., E.S.A., C.D.G., M.A.), Pathology (C.D.G., M.A.), and Genetic Medicine (M.A.), the Medical Scientist Training Program (E.A.D.), the Telomere Center (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., M.A.), and Sidney Kimmel Comprehensive Cancer Center (K.E.S., E.S.A., C.D.G., M.A.), Johns Hopkins University School of Medicine, and the Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University (M.G.T., S.M.Y., R.C.M.) - both in Baltimore; the Child Neurology Residency Program, Boston Children's Hospital, Boston (D.L.G.); the Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami (D.B.L.); and the Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis (E.S.A.)
| | - Emmanuel S Antonarakis
- From the Departments of Oncology (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., E.S.A., C.D.G., M.A.), Pathology (C.D.G., M.A.), and Genetic Medicine (M.A.), the Medical Scientist Training Program (E.A.D.), the Telomere Center (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., M.A.), and Sidney Kimmel Comprehensive Cancer Center (K.E.S., E.S.A., C.D.G., M.A.), Johns Hopkins University School of Medicine, and the Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University (M.G.T., S.M.Y., R.C.M.) - both in Baltimore; the Child Neurology Residency Program, Boston Children's Hospital, Boston (D.L.G.); the Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami (D.B.L.); and the Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis (E.S.A.)
| | - Christopher D Gocke
- From the Departments of Oncology (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., E.S.A., C.D.G., M.A.), Pathology (C.D.G., M.A.), and Genetic Medicine (M.A.), the Medical Scientist Training Program (E.A.D.), the Telomere Center (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., M.A.), and Sidney Kimmel Comprehensive Cancer Center (K.E.S., E.S.A., C.D.G., M.A.), Johns Hopkins University School of Medicine, and the Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University (M.G.T., S.M.Y., R.C.M.) - both in Baltimore; the Child Neurology Residency Program, Boston Children's Hospital, Boston (D.L.G.); the Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami (D.B.L.); and the Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis (E.S.A.)
| | - Rajiv C McCoy
- From the Departments of Oncology (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., E.S.A., C.D.G., M.A.), Pathology (C.D.G., M.A.), and Genetic Medicine (M.A.), the Medical Scientist Training Program (E.A.D.), the Telomere Center (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., M.A.), and Sidney Kimmel Comprehensive Cancer Center (K.E.S., E.S.A., C.D.G., M.A.), Johns Hopkins University School of Medicine, and the Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University (M.G.T., S.M.Y., R.C.M.) - both in Baltimore; the Child Neurology Residency Program, Boston Children's Hospital, Boston (D.L.G.); the Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami (D.B.L.); and the Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis (E.S.A.)
| | - Mary Armanios
- From the Departments of Oncology (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., E.S.A., C.D.G., M.A.), Pathology (C.D.G., M.A.), and Genetic Medicine (M.A.), the Medical Scientist Training Program (E.A.D.), the Telomere Center (E.A.D., K.E.S., Z.L.C., E.J.M., Z.X., M.A.), and Sidney Kimmel Comprehensive Cancer Center (K.E.S., E.S.A., C.D.G., M.A.), Johns Hopkins University School of Medicine, and the Department of Biology, Krieger School of Arts and Sciences, Johns Hopkins University (M.G.T., S.M.Y., R.C.M.) - both in Baltimore; the Child Neurology Residency Program, Boston Children's Hospital, Boston (D.L.G.); the Department of Pathology and Laboratory Medicine, Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami (D.B.L.); and the Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis (E.S.A.)
| |
Collapse
|
36
|
Kuse R, Ishii K. Flexible Attachment and Detachment of Centromeres and Telomeres to and from Chromosomes. Biomolecules 2023; 13:1016. [PMID: 37371596 DOI: 10.3390/biom13061016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/15/2023] [Accepted: 06/18/2023] [Indexed: 06/29/2023] Open
Abstract
Accurate transmission of genomic information across multiple cell divisions and generations, without any losses or errors, is fundamental to all living organisms. To achieve this goal, eukaryotes devised chromosomes. Eukaryotic genomes are represented by multiple linear chromosomes in the nucleus, each carrying a centromere in the middle, a telomere at both ends, and multiple origins of replication along the chromosome arms. Although all three of these DNA elements are indispensable for chromosome function, centromeres and telomeres possess the potential to detach from the original chromosome and attach to new chromosomal positions, as evident from the events of telomere fusion, centromere inactivation, telomere healing, and neocentromere formation. These events seem to occur spontaneously in nature but have not yet been elucidated clearly, because they are relatively infrequent and sometimes detrimental. To address this issue, experimental setups have been developed using model organisms such as yeast. In this article, we review some of the key experiments that provide clues as to the extent to which these paradoxical and elusive features of chromosomally indispensable elements may become valuable in the natural context.
Collapse
Affiliation(s)
- Riku Kuse
- Laboratory of Chromosome Function and Regulation, Graduate School of Engineering, Kochi University of Technology, Kochi 782-8502, Japan
| | - Kojiro Ishii
- Laboratory of Chromosome Function and Regulation, Graduate School of Engineering, Kochi University of Technology, Kochi 782-8502, Japan
| |
Collapse
|
37
|
Padmanaban S, Tesmer VM, Nandakumar J. Interaction hub critical for telomerase recruitment and primer-template handling for catalysis. Life Sci Alliance 2023; 6:e202201727. [PMID: 36963832 PMCID: PMC10055720 DOI: 10.26508/lsa.202201727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/26/2023] Open
Abstract
Telomerase processively adds telomeric DNA repeats to chromosome ends using catalytic protein subunit TERT and a template on its RNA subunit TR. Mammalian telomerase is recruited to telomeres by the TEL patch and NOB regions of shelterin component TPP1. Recent cryo-EM structures of human telomerase reveal that a composite TERT TEN-(IFD-TRAP) domain interacts with TPP1. Here, we generate TERT mutants to demonstrate that a three-way TEN-(IFD-TRAP)-TPP1 interaction is critical for telomerase recruitment to telomeres and processive telomere repeat addition. Single mutations of IFD-TRAP at its interface with TR or the DNA primer impair telomerase catalysis. We further reveal the importance of TERT motif 3N and TEN domain loop 99FGF101 in telomerase action. Finally, we demonstrate that TPP1 TEL patch loop residue F172, which undergoes a structural rearrangement to bind telomerase, contributes to the human-mouse species specificity of the telomerase-TPP1 interaction. Our study provides insights into the multiple functions of TERT IFD-TRAP, reveals novel TERT and TPP1 elements critical for function, and helps explain how TPP1 binding licenses robust telomerase action at natural chromosome ends.
Collapse
Affiliation(s)
- Shilpa Padmanaban
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Valerie M Tesmer
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Jayakrishnan Nandakumar
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
38
|
Zade NH, Khattar E. POT1 mutations cause differential effects on telomere length leading to opposing disease phenotypes. J Cell Physiol 2023; 238:1237-1255. [PMID: 37183325 DOI: 10.1002/jcp.31034] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/28/2023] [Accepted: 04/17/2023] [Indexed: 05/16/2023]
Abstract
The protection of telomere protein (POT1) is a telomere-binding protein and is an essential component of the six-membered shelterin complex, which is associated with the telomeres. POT1 directly binds to the 3' single-stranded telomeric overhang and prevents the activation of DNA damage response at telomeres thus preventing the telomere-telomere fusions and genomic instability. POT1 also plays a pivotal role in maintaining telomere length by regulating telomerase-mediated telomere elongation. Mutations in POT1 proteins result in three different telomere phenotypes, which include long, short, or aberrant telomere length. Long telomeres predispose individuals to cancer, while short or aberrant telomere phenotypes result in pro-aging diseases referred to as telomeropathies. Here, we review the function of POT1 proteins in telomere length hemostasis and how the spectrum of mutations reported in POT1 can be segregated toward developing very distinct disease phenotypes of cancer and telomeropathies.
Collapse
Affiliation(s)
- Nikita Harish Zade
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be) University, Mumbai, India
| | - Ekta Khattar
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be) University, Mumbai, India
| |
Collapse
|
39
|
Vijay Kumar MJ, Morales R, Tsvetkov AS. G-quadruplexes and associated proteins in aging and Alzheimer's disease. FRONTIERS IN AGING 2023; 4:1164057. [PMID: 37323535 PMCID: PMC10267416 DOI: 10.3389/fragi.2023.1164057] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 05/17/2023] [Indexed: 06/17/2023]
Abstract
Aging is a prominent risk factor for many neurodegenerative disorders, such as Alzheimer's disease (AD). Alzheimer's disease is characterized by progressive cognitive decline, memory loss, and neuropsychiatric and behavioral symptoms, accounting for most of the reported dementia cases. This disease is now becoming a major challenge and burden on modern society, especially with the aging population. Over the last few decades, a significant understanding of the pathophysiology of AD has been gained by studying amyloid deposition, hyperphosphorylated tau, synaptic dysfunction, oxidative stress, calcium dysregulation, and neuroinflammation. This review focuses on the role of non-canonical secondary structures of DNA/RNA G-quadruplexes (G4s, G4-DNA, and G4-RNA), G4-binding proteins (G4BPs), and helicases, and their roles in aging and AD. Being critically important for cellular function, G4s are involved in the regulation of DNA and RNA processes, such as replication, transcription, translation, RNA localization, and degradation. Recent studies have also highlighted G4-DNA's roles in inducing DNA double-strand breaks that cause genomic instability and G4-RNA's participation in regulating stress granule formation. This review emphasizes the significance of G4s in aging processes and how their homeostatic imbalance may contribute to the pathophysiology of AD.
Collapse
Affiliation(s)
- M. J. Vijay Kumar
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
| | - Rodrigo Morales
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
- Centro Integrativo de Biologia y Quimica Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago, Chile
| | - Andrey S. Tsvetkov
- The Department of Neurology, The University of Texas McGovern Medical School at Houston, Houston, TX, United States
- The University of Texas Graduate School of Biomedical Sciences, Houston, TX, United States
- UTHealth Consortium on Aging, The University of Texas McGovern Medical School, Houston, TX, United States
| |
Collapse
|
40
|
Torres DE, Reckard AT, Klocko AD, Seidl MF. Nuclear genome organization in fungi: from gene folding to Rabl chromosomes. FEMS Microbiol Rev 2023; 47:fuad021. [PMID: 37197899 PMCID: PMC10246852 DOI: 10.1093/femsre/fuad021] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/28/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023] Open
Abstract
Comparative genomics has recently provided unprecedented insights into the biology and evolution of the fungal lineage. In the postgenomics era, a major research interest focuses now on detailing the functions of fungal genomes, i.e. how genomic information manifests into complex phenotypes. Emerging evidence across diverse eukaryotes has revealed that the organization of DNA within the nucleus is critically important. Here, we discuss the current knowledge on the fungal genome organization, from the association of chromosomes within the nucleus to topological structures at individual genes and the genetic factors required for this hierarchical organization. Chromosome conformation capture followed by high-throughput sequencing (Hi-C) has elucidated how fungal genomes are globally organized in Rabl configuration, in which centromere or telomere bundles are associated with opposite faces of the nuclear envelope. Further, fungal genomes are regionally organized into topologically associated domain-like (TAD-like) chromatin structures. We discuss how chromatin organization impacts the proper function of DNA-templated processes across the fungal genome. Nevertheless, this view is limited to a few fungal taxa given the paucity of fungal Hi-C experiments. We advocate for exploring genome organization across diverse fungal lineages to ensure the future understanding of the impact of nuclear organization on fungal genome function.
Collapse
Affiliation(s)
- David E Torres
- Theoretical Biology and Bioinformatics, Department of Biology, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
- Laboratory of Phytopathology, Wageningen University and Research,Droevendaalsesteeg 4, 6708 PB Wageningen, The Netherlands
| | - Andrew T Reckard
- Department of Chemistry and Biochemistry, University of Colorado Colorado Springs, 234 Centennial Hall, 1420 Austin Bluffs Pkwy, Colorado Springs, CO 80918 USA
| | - Andrew D Klocko
- Department of Chemistry and Biochemistry, University of Colorado Colorado Springs, 234 Centennial Hall, 1420 Austin Bluffs Pkwy, Colorado Springs, CO 80918 USA
| | - Michael F Seidl
- Theoretical Biology and Bioinformatics, Department of Biology, Utrecht University, Padualaan 8, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
41
|
Kanoh J. Roles of Specialized Chromatin and DNA Structures at Subtelomeres in Schizosaccharomyces pombe. Biomolecules 2023; 13:biom13050810. [PMID: 37238680 DOI: 10.3390/biom13050810] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
Eukaryotes have linear chromosomes with domains called telomeres at both ends. The telomere DNA consists of a simple tandem repeat sequence, and multiple telomere-binding proteins including the shelterin complex maintain chromosome-end structures and regulate various biological reactions, such as protection of chromosome ends and control of telomere DNA length. On the other hand, subtelomeres, which are located adjacent to telomeres, contain a complex mosaic of multiple common segmental sequences and a variety of gene sequences. This review focused on roles of the subtelomeric chromatin and DNA structures in the fission yeast Schizosaccharomyces pombe. The fission yeast subtelomeres form three distinct chromatin structures; one is the shelterin complex, which is localized not only at the telomeres but also at the telomere-proximal regions of subtelomeres to form transcriptionally repressive chromatin structures. The others are heterochromatin and knob, which have repressive effects in gene expression, but the subtelomeres are equipped with a mechanism that prevents these condensed chromatin structures from invading adjacent euchromatin regions. On the other hand, recombination reactions within or near subtelomeric sequences allow chromosomes to be circularized, enabling cells to survive in telomere shortening. Furthermore, DNA structures of the subtelomeres are more variable than other chromosomal regions, which may have contributed to biological diversity and evolution while changing gene expression and chromatin structures.
Collapse
Affiliation(s)
- Junko Kanoh
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro-ku, Tokyo 153-8902, Japan
| |
Collapse
|
42
|
Xiong Y, Zhang H, Zhou S, Ma L, Xiao W, Wu Y, Yuan YJ. Structural Variations and Adaptations of Synthetic Chromosome Ends Driven by SCRaMbLE in Haploid and Diploid Yeasts. ACS Synth Biol 2023; 12:689-699. [PMID: 36821394 DOI: 10.1021/acssynbio.2c00424] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Variations and adaptations of chromosome ends play an important role in eukaryotic karyotype evolution. Traditional experimental studies of the adaptations of chromosome ends mainly rely on the strategy of introducing defects; thus, the adaptation methods of survivors may vary depending on the initial defects. Here, using the SCRaMbLE strategy, we obtained a library of haploid and diploid synthetic strains with variations in chromosome ends. Analysis of the SCRaMbLEd survivors revealed four routes of adaptation: homologous recombination between nonhomologous chromosome arms (haploids) or homologous chromosome arms (diploids), site-specific recombination between intra- or interchromosomal ends, circularization of chromosomes, and loss of whole chromosomes (diploids). We also found that circularization of synthetic chromosomes can be generated by SCRaMbLE. Our study of various adaptation routes of chromosome ends provides insight into eukaryotic karyotype evolution from the viewpoint of synthetic genomics.
Collapse
Affiliation(s)
- Yao Xiong
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China.,Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Hui Zhang
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China.,Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Sijie Zhou
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China.,Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Lu Ma
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China.,Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Wenhai Xiao
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China.,Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Yi Wu
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China.,Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Ying-Jin Yuan
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300072, China.,Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
43
|
Burroughs A, Aravind L. New biochemistry in the Rhodanese-phosphatase superfamily: emerging roles in diverse metabolic processes, nucleic acid modifications, and biological conflicts. NAR Genom Bioinform 2023; 5:lqad029. [PMID: 36968430 PMCID: PMC10034599 DOI: 10.1093/nargab/lqad029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/10/2023] [Accepted: 03/09/2023] [Indexed: 03/25/2023] Open
Abstract
The protein-tyrosine/dual-specificity phosphatases and rhodanese domains constitute a sprawling superfamily of Rossmannoid domains that use a conserved active site with a cysteine to catalyze a range of phosphate-transfer, thiotransfer, selenotransfer and redox activities. While these enzymes have been extensively studied in the context of protein/lipid head group dephosphorylation and various thiotransfer reactions, their overall diversity and catalytic potential remain poorly understood. Using comparative genomics and sequence/structure analysis, we comprehensively investigate and develop a natural classification for this superfamily. As a result, we identified several novel clades, both those which retain the catalytic cysteine and those where a distinct active site has emerged in the same location (e.g. diphthine synthase-like methylases and RNA 2' OH ribosyl phosphate transferases). We also present evidence that the superfamily has a wider range of catalytic capabilities than previously known, including a set of parallel activities operating on various sugar/sugar alcohol groups in the context of NAD+-derivatives and RNA termini, and potential phosphate transfer activities involving sugars and nucleotides. We show that such activities are particularly expanded in the RapZ-C-DUF488-DUF4326 clade, defined here for the first time. Some enzymes from this clade are predicted to catalyze novel DNA-end processing activities as part of nucleic-acid-modifying systems that are likely to function in biological conflicts between viruses and their hosts.
Collapse
Affiliation(s)
- A Maxwell Burroughs
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| | - L Aravind
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA
| |
Collapse
|
44
|
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
|
45
|
Ueno M. Exploring Genetic Interactions with Telomere Protection Gene pot1 in Fission Yeast. Biomolecules 2023; 13:biom13020370. [PMID: 36830739 PMCID: PMC9953254 DOI: 10.3390/biom13020370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
The regulation of telomere length has a significant impact on cancer risk and aging in humans. Circular chromosomes are found in humans and are often unstable during mitosis, resulting in genome instability. Some types of cancer have a high frequency of a circular chromosome. Fission yeast is a good model for studying the formation and stability of circular chromosomes as deletion of pot1 (encoding a telomere protection protein) results in rapid telomere degradation and chromosome fusion. Pot1 binds to single-stranded telomere DNA and is conserved from fission yeast to humans. Loss of pot1 leads to viable strains in which all three fission yeast chromosomes become circular. In this review, I will introduce pot1 genetic interactions as these inform on processes such as the degradation of uncapped telomeres, chromosome fusion, and maintenance of circular chromosomes. Therefore, exploring genes that genetically interact with pot1 contributes to finding new genes and/or new functions of genes related to the maintenance of telomeres and/or circular chromosomes.
Collapse
Affiliation(s)
- Masaru Ueno
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima 739-8530, Japan; ; Tel.: +81-82-424-7768
- Hiroshima Research Center for Healthy Aging (HiHA), Hiroshima University, Higashi-Hiroshima 739-8530, Japan
| |
Collapse
|
46
|
Yu X, Gray S, Ferreira H. POT-3 preferentially binds the terminal DNA-repeat on the telomeric G-overhang. Nucleic Acids Res 2023; 51:610-618. [PMID: 36583365 PMCID: PMC9881156 DOI: 10.1093/nar/gkac1203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/01/2022] [Accepted: 12/06/2022] [Indexed: 12/31/2022] Open
Abstract
Eukaryotic chromosomes typically end in 3' telomeric overhangs. The safeguarding of telomeric single-stranded DNA overhangs is carried out by factors related to the protection of telomeres 1 (POT1) protein in humans. Of the three POT1-like proteins in Caenorhabditis elegans, POT-3 was the only member thought to not play a role at telomeres. Here, we provide evidence that POT-3 is a bona fide telomere-binding protein. Using a new loss-of-function mutant, we show that the absence of POT-3 causes telomere lengthening and increased levels of telomeric C-circles. We find that POT-3 directly binds the telomeric G-strand in vitro and map its minimal DNA binding site to the six-nucleotide motif, GCTTAG. We further show that the closely related POT-2 protein binds the same motif, but that POT-3 shows higher sequence selectivity. Crucially, in contrast to POT-2, POT-3 prefers binding sites immediately adjacent to the 3' end of DNA. These differences are significant as genetic analyses reveal that pot-2 and pot-3 do not function redundantly with each other in vivo. Our work highlights the rapid evolution and specialisation of telomere binding proteins and places POT-3 in a unique position to influence activities that control telomere length.
Collapse
Affiliation(s)
- Xupeng Yu
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, North Haugh, St Andrews, UK
| | - Sean Gray
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, North Haugh, St Andrews, UK
| | - Helder C Ferreira
- Biomedical Sciences Research Complex, School of Biology, University of St Andrews, North Haugh, St Andrews, UK
| |
Collapse
|
47
|
Kato TA. Nontraditional Method for Telomere Staining by PNA Probes. Methods Mol Biol 2023; 2519:111-116. [PMID: 36066716 DOI: 10.1007/978-1-0716-2433-3_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The standard FISH uses DNA probes to hybridize to the designated complementary strands. This is DNA-DNA interaction, and it usually takes much longer time to obtain detectable signals compared to other reactions such as immunochemical reactions and simple chemical reactions. Certain proteins bind to specific DNA sequences and regulate the biological function of DNA. These DNA-binding proteins have specific domains to interact with single- or double-stranded DNA. Some of telomere proteins apparently bind to telomere sequence and form nucleoprotein complex to protect chromosome ends. Using telomere PNA probes, probes can be accumulated at the telomere sites in a non-hybridization manner. This chapter introduces nontraditional PNA telomere staining protocol without DNA-DNA hybridization to visualize telomere locations on metaphase chromosomes.
Collapse
Affiliation(s)
- Takamitsu A Kato
- Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
48
|
Takeuchi C, Yokoshi M, Kondo S, Shibuya A, Saito K, Fukaya T, Siomi H, Iwasaki Y. Mod(mdg4) variants repress telomeric retrotransposon HeT-A by blocking subtelomeric enhancers. Nucleic Acids Res 2022; 50:11580-11599. [PMID: 36373634 PMCID: PMC9723646 DOI: 10.1093/nar/gkac1034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/17/2022] [Accepted: 10/22/2022] [Indexed: 11/16/2022] Open
Abstract
Telomeres in Drosophila are composed of sequential non-LTR retrotransposons HeT-A, TART and TAHRE. Although they are repressed by the PIWI-piRNA pathway or heterochromatin in the germline, the regulation of these retrotransposons in somatic cells is poorly understood. In this study, we demonstrated that specific splice variants of Mod(mdg4) repress HeT-A by blocking subtelomeric enhancers in ovarian somatic cells. Among the variants, we found that the Mod(mdg4)-N variant represses HeT-A expression the most efficiently. Subtelomeric sequences bound by Mod(mdg4)-N block enhancer activity within subtelomeric TAS-R repeats. This enhancer-blocking activity is increased by the tandem association of Mod(mdg4)-N to repetitive subtelomeric sequences. In addition, the association of Mod(mdg4)-N couples with the recruitment of RNA polymerase II to the subtelomeres, which reinforces its enhancer-blocking function. Our findings provide novel insights into how telomeric retrotransposons are regulated by the specific variants of insulator proteins associated with subtelomeric sequences.
Collapse
Affiliation(s)
- Chikara Takeuchi
- Department of Molecular Biology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Moe Yokoshi
- Laboratory of Transcription Dynamics, Research Center for Biological Visualization, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0032, Japan
| | - Shu Kondo
- Department of Chromosome Science, National Institute of Genetics, Research Organization of Information and Systems (ROIS), Shizuoka 411-8540, Japan
| | - Aoi Shibuya
- Department of Molecular Biology, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kuniaki Saito
- Department of Chromosome Science, National Institute of Genetics, Research Organization of Information and Systems (ROIS), Shizuoka 411-8540, Japan
| | - Takashi Fukaya
- Laboratory of Transcription Dynamics, Research Center for Biological Visualization, Institute for Quantitative Biosciences, The University of Tokyo, Tokyo 113-0032, Japan,Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Tokyo 113-0032, Japan
| | | | - Yuka W Iwasaki
- To whom correspondence should be addressed. Tel: +81 3 5363 3529; Fax: +81 3 5363 3266;
| |
Collapse
|
49
|
Li Y, Xie Y, Wang D, Xu H, Ye J, Yin JC, Chen J, Yan J, Ye B, Chen C. Whole exome sequencing identified a novel POT1 variant as a candidate pathogenic allele underlying a Li-Fraumeni-like family. Front Oncol 2022; 12:963364. [PMID: 36387164 PMCID: PMC9664187 DOI: 10.3389/fonc.2022.963364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/17/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Li-Fraumeni syndrome (LFS) and Li-Fraumeni-like (LFL) syndrome are rare hereditary diseases characterized by predisposition to a diverse spectrum of cancer types, primarily sarcoma. The pathogenic variants underlying the majority of LFL cases remain to be explored. METHODS We performed whole-exome sequencing (WES) on 13 core members of a large LFL family with highly aggregated incidences of cancers, including cases with sarcoma, non-small cell lung cancer and cardiac angiosarcoma, and conducted a comprehensive literature review of candidate gene associations in LFS/LFL syndromes or sarcoma to identify potential pathogenic germline variants. RESULTS No germline variants in the best-known LFL/LFS-associated gene TP53 were detected. Of all the genes associated with LFS/LFL or sarcoma that we have surveyed, we identified a novel p.P35L germline variant in POT1 (protection of telomeres 1). Germline and somatic alterations in POT1 have been implicated in a series of familial cancers, including angiosarcoma, glioma, melanoma and colorectal cancer. This particular variant is located in the telomere-binding OB1 domain, which is important in maintaining the proper telomere length, and showed high conservation across different POT1 orthologues. No record of the variant was found in any of the 1000 genomes, ExAC, gnomAD, dpSNP and COSMIC databases. Prediction algorithms and in silico structural analysis suggested completely disrupted protein structure and function of POT1 in the presence of this mutation. CONCLUSIONS Leveraging WES, we identified a novel germline risk allele, p.P35L in POT1, that likely predisposes to LFL syndrome. Our results support the routine testing of POT1 and other LFL/LFS-associated genes in the risk populations to enable early cancer diagnosis, prevention and intervention.
Collapse
Affiliation(s)
- Yuping Li
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yupeng Xie
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Di Wang
- Medical Department, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Hanyan Xu
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junru Ye
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jiani C. Yin
- Medical Department, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Junjie Chen
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junrong Yan
- Medical Department, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Bin Ye
- Medical Department, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Chengshui Chen
- Key Laboratory of Interventional Pulmonology of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China,*Correspondence: Chengshui Chen,
| |
Collapse
|
50
|
Pan L, Tormey D, Bobon N, Baumann P. Rap1 prevents fusions between long telomeres in fission yeast. EMBO J 2022; 41:e110458. [PMID: 36059259 PMCID: PMC9574727 DOI: 10.15252/embj.2021110458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 11/15/2022] Open
Abstract
The conserved Rap1 protein is part of the shelterin complex that plays critical roles in chromosome end protection and telomere length regulation. Previous studies have addressed how fission yeast Rap1 contributes to telomere length maintenance, but the mechanism by which the protein inhibits end fusions has remained elusive. Here, we use a mutagenesis screen in combination with high‐throughput sequencing to identify several amino acid positions in Rap1 that have key roles in end protection. Interestingly, mutations at these sites render cells susceptible to genome instability in a conditional manner, whereby longer telomeres are prone to undergoing end fusions, while telomeres within the normal length range are sufficiently protected. The protection of long telomeres is in part dependent on their nuclear envelope attachment mediated by the Rap1–Bqt4 interaction. Our data demonstrate that long telomeres represent a challenge for the maintenance of genome integrity, thereby providing an explanation for species‐specific upper limits on telomere length.
Collapse
Affiliation(s)
- Lili Pan
- Department of Biology, Johannes Gutenberg University, Mainz, Germany
| | - Duncan Tormey
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Nadine Bobon
- Department of Biology, Johannes Gutenberg University, Mainz, Germany
| | - Peter Baumann
- Department of Biology, Johannes Gutenberg University, Mainz, Germany.,Institute of Molecular Biology, Mainz, Germany
| |
Collapse
|