1
|
Jones-Weinert C, Mainz L, Karlseder J. Telomere function and regulation from mouse models to human ageing and disease. Nat Rev Mol Cell Biol 2025; 26:297-313. [PMID: 39614014 DOI: 10.1038/s41580-024-00800-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 12/01/2024]
Abstract
Telomeres protect the ends of chromosomes but shorten following cell division in the absence of telomerase activity. When telomeres become critically short or damaged, a DNA damage response is activated. Telomeres then become dysfunctional and trigger cellular senescence or death. Telomere shortening occurs with ageing and may contribute to associated maladies such as infertility, neurodegeneration, cancer, lung dysfunction and haematopoiesis disorders. Telomere dysfunction (sometimes without shortening) is associated with various diseases, known as telomere biology disorders (also known as telomeropathies). Telomere biology disorders include dyskeratosis congenita, Høyeraal-Hreidarsson syndrome, Coats plus syndrome and Revesz syndrome. Although mouse models have been invaluable in advancing telomere research, full recapitulation of human telomere-related diseases in mice has been challenging, owing to key differences between the species. In this Review, we discuss telomere protection, maintenance and damage. We highlight the differences between human and mouse telomere biology that may contribute to discrepancies between human diseases and mouse models. Finally, we discuss recent efforts to generate new 'humanized' mouse models to better model human telomere biology. A better understanding of the limitations of mouse telomere models will pave the road for more human-like models and further our understanding of telomere biology disorders, which will contribute towards the development of new therapies.
Collapse
Affiliation(s)
| | - Laura Mainz
- The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jan Karlseder
- The Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
2
|
Lu S, Huang X, Chen Y, Lin Y, Zou J, Chen Y, Wu J. The De Ritis ratio mediates the association between creatinine-to-body weight ratio and normoglycemic conversion in Chinese health examinees. Sci Rep 2025; 15:5994. [PMID: 39966552 PMCID: PMC11836274 DOI: 10.1038/s41598-025-90501-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 02/13/2025] [Indexed: 02/20/2025] Open
Abstract
To assess the associations between the creatinine-to-body weight (Cre/BW) ratio and reversion to normoglycemia mediated by the aspartate aminotransferase-to-alanine aminotransferase (De Ritis) ratio. This retrospective cohort study included 24,884 prediabetic participants from health check-ups in 32 regions across 11 Chinese cities. We employed multivariable Cox regression to assess time-to-event outcomes, smooth curve fitting for trend analysis, and inflection point analysis to determine critical thresholds. Subgroup analyses were performed to explore interactions. Mediation analysis was conducted to clarify the mechanisms linking prediabetes with health outcomes. In the analysis of 24,884 individuals, it was observed that 49.08% of the prediabetic subjects experienced normoglycemic conversion during a follow-up period spanning 73,517.66 person-years. Following full adjustment, the Cre/BW ratio was positively associated with normoglycemic conversion among prediabetic individuals (HR 1.42, 95% CI 1.30-1.55). Multivariate smooth spline analysis revealed a nonlinear relationship between the Cre/BW ratio and glucose status conversion (P for nonlinearity < 0.05), with the curves leveling off when the Cre/BW ratio (×100) reached 1.34. Subgroup analysis and additional sensitivity analyses confirmed that the results between the Cre/BW ratio and glucose status conversion were robust. Additionally, the De Ritis ratio was found to explain 50% of the association between the Cre/BW ratio and the prediabetes-to-normoglycemia transition. A higher Cre/BW ratio was associated with a better chance of controlling blood glucose levels in prediabetic individuals, with the De Ritis ratio playing a key mediating role.
Collapse
Affiliation(s)
- Songyao Lu
- Department of Clinical Laboratory, Jieyang People's Hospital, No.107 Tianfu Road, Rongcheng District, Jieyang, 522000, Guangdong, China
| | - Xudong Huang
- Department of Clinical Laboratory, Jieyang People's Hospital, No.107 Tianfu Road, Rongcheng District, Jieyang, 522000, Guangdong, China
| | - Yiying Chen
- Department of Clinical Laboratory, Jieyang People's Hospital, No.107 Tianfu Road, Rongcheng District, Jieyang, 522000, Guangdong, China
| | - Yuye Lin
- Department of Clinical Laboratory, Jieyang People's Hospital, No.107 Tianfu Road, Rongcheng District, Jieyang, 522000, Guangdong, China
| | - Junzhong Zou
- Department of Clinical Laboratory, Jieyang People's Hospital, No.107 Tianfu Road, Rongcheng District, Jieyang, 522000, Guangdong, China
| | - Yimei Chen
- Physical Examination Center, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, Guangdong, China
| | - Juan Wu
- Department of Clinical Laboratory, Jieyang People's Hospital, No.107 Tianfu Road, Rongcheng District, Jieyang, 522000, Guangdong, China.
| |
Collapse
|
3
|
Moreno-Villanueva M, Jimenez-Chavez LE, Krieger S, Ding LH, Zhang Y, Babiak-Vazquez A, Berres M, Splinter S, Pauken KE, Schaefer BC, Crucian BE, Wu H. Transcriptomics analysis reveals potential mechanisms underlying mitochondrial dysfunction and T cell exhaustion in astronauts' blood cells in space. Front Immunol 2025; 15:1512578. [PMID: 39902046 PMCID: PMC11788081 DOI: 10.3389/fimmu.2024.1512578] [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: 10/16/2024] [Accepted: 12/26/2024] [Indexed: 02/05/2025] Open
Abstract
Introduction The impact of spaceflight on the immune system and mitochondria has been investigated for decades. However, the molecular mechanisms underlying spaceflight-induced immune dysregulations are still unclear. Methods In this study, blood from eleven crewmembers was collected before and during International Space Station (ISS) missions. Transcriptomic analysis was performed in isolated peripheral blood mononuclear cells (PBMCs) using RNA-sequencing. Differentially expresses genes (DEG) in space were determined by comparing of the inflight to the preflight samples. Pathways and statistical analyses of these DEG were performed using the Ingenuity Pathway Analysis (IPA) tool. Results In comparison to pre-flight, a total of 2030 genes were differentially expressed in PBMC collected between 135 and 210 days in orbit, which included a significant number of surface receptors. The dysregulated genes and pathways were mostly involved in energy and oxygen metabolism, immune responses, cell adhesion/migration and cell death/survival. Discussion Based on the DEG and the associated pathways and functions, we propose that mitochondria dysfunction was caused by constant modulation of mechano-sensing receptors in microgravity, which triggered a signaling cascade that led to calcium overloading in mitochondria. The response of PBMC in space shares T-cell exhaustion features, likely initiated by microgravity than by infection. Consequences of mitochondria dysfunction include immune dysregulation and prolonged cell survival which potentially explains the reported findings of inhibition of T cell activation and telomere lengthening in astronauts. Conclusion Our study potentially identifies the upstream cause of mitochondria dysfunction and the downstream consequences in immune cells.
Collapse
Affiliation(s)
- Maria Moreno-Villanueva
- National Aeronautics and Space Administration, Johnson Space Center, Houston, TX, United States
- Department of Sport Science, University of Konstanz, Konstanz, Germany
| | - Luis E. Jimenez-Chavez
- National Aeronautics and Space Administration, Johnson Space Center, Houston, TX, United States
- College of Medicine, University of Central Florida, Orlando, FL, United States
| | | | - Liang-Hao Ding
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Ye Zhang
- National Aeronautics and Space Administration, Kennedy Space Center, Cape Canaveral, FL, United States
| | - Adriana Babiak-Vazquez
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Mark Berres
- Bioinformatics Resource and Gene Expression Center, University of Wisconsin, Madison, WI, United States
| | - Sandra Splinter
- Bioinformatics Resource and Gene Expression Center, University of Wisconsin, Madison, WI, United States
| | - Kristen E. Pauken
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Brian C. Schaefer
- Department of Microbiology and Immunology, Uniformed Services University, Bethesda, MD, United States
| | - Brian E. Crucian
- National Aeronautics and Space Administration, Johnson Space Center, Houston, TX, United States
| | - Honglu Wu
- National Aeronautics and Space Administration, Johnson Space Center, Houston, TX, United States
| |
Collapse
|
4
|
He X, Cao L, Fu X, Wu Y, Wen H, Gao Y, Huo W, Wang M, Liu M, Su Y, Liu G, Zhang M, Hu F, Hu D, Zhao Y. The Association Between Telomere Length and Diabetes Mellitus: Accumulated Evidence From Observational Studies. J Clin Endocrinol Metab 2024; 110:e177-e185. [PMID: 39087945 DOI: 10.1210/clinem/dgae536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/11/2024] [Accepted: 07/31/2024] [Indexed: 08/02/2024]
Abstract
OBJECTIVE In order to assess the associations between telomere length (TL) and diabetes mellitus (DM), especially type 2 diabetes (T2DM), we performed this systematic review and meta-analysis. METHODS PubMed, Embase, and Web of Science were thoroughly searched up to July 11, 2023. The pooled standardized mean difference (SMD) and the 95% confidence interval (CI) were evaluated using the random-effects model. Age, sex, study design, duration of diabetes, region, sample size, and body mass index (BMI) were used to stratify subgroup analyses. RESULTS A total of 37 observational studies involving 18 181 participants from 14 countries were included in the quantitative meta-analysis. In this study, patients with diabetes had shorter TL than the non-diabetic, whether those patients had T1DM (-2.70; 95% CI: -4.47, -0.93; P < .001), T2DM (-3.70; 95% CI: -4.20, -3.20; P < .001), or other types of diabetes (-0.71; 95% CI: -1.10, -0.31; P < .001). Additionally, subgroup analysis of T2DM showed that TL was significantly correlated with age, sex, study design, diabetes duration, sample size, detection method, region, and BMI. CONCLUSION A negative correlation was observed between TL and DM. To validate this association in the interim, more extensive, superior prospective investigations and clinical trials are required.
Collapse
Affiliation(s)
- Xinxin He
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Lu Cao
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Xueru Fu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Yuying Wu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Hongwei Wen
- Department of Public Health, Zhengzhou Shuqing Medical College, Zhengzhou, Henan 450000, People's Republic of China
| | - Yajuan Gao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Weifeng Huo
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Mengdi Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Mengna Liu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Yijia Su
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Ge Liu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Ming Zhang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong 518060, People's Republic of China
| | - Fulan Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong 518060, People's Republic of China
| | - Dongsheng Hu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Yang Zhao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| |
Collapse
|
5
|
Mason CE, Sierra MA, Feng HJ, Bailey SM. Telomeres and aging: on and off the planet! Biogerontology 2024; 25:313-327. [PMID: 38581556 PMCID: PMC10998805 DOI: 10.1007/s10522-024-10098-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 04/08/2024]
Abstract
Improving human healthspan in our rapidly aging population has never been more imperative. Telomeres, protective "caps" at the ends of linear chromosomes, are essential for maintaining genome stability of eukaryotic genomes. Due to their physical location and the "end-replication problem" first envisioned by Dr. Alexey Olovnikov, telomeres shorten with cell division, the implications of which are remarkably profound. Telomeres are hallmarks and molecular drivers of aging, as well as fundamental integrating components of the cumulative effects of genetic, lifestyle, and environmental factors that erode telomere length over time. Ongoing telomere attrition and the resulting limit to replicative potential imposed by cellular senescence serves a powerful tumor suppressor function, and also underlies aging and a spectrum of age-related degenerative pathologies, including reduced fertility, dementias, cardiovascular disease and cancer. However, very little data exists regarding the extraordinary stressors and exposures associated with long-duration space exploration and eventual habitation of other planets, nor how such missions will influence telomeres, reproduction, health, disease risk, and aging. Here, we briefly review our current understanding, which has advanced significantly in recent years as a result of the NASA Twins Study, the most comprehensive evaluation of human health effects associated with spaceflight ever conducted. Thus, the Twins Study is at the forefront of personalized space medicine approaches for astronauts and sets the stage for subsequent missions. We also extrapolate from current understanding to future missions, highlighting potential biological and biochemical strategies that may enable human survival, and consider the prospect of longevity in the extreme environment of space.
Collapse
Affiliation(s)
- Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA
| | - Maria A Sierra
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine and WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional Computational Biology & Medicine Program, Weill Cornell Medicine, New York, NY, USA
| | - Henry J Feng
- Department of Biological Sciences, Columbia University, New York, NY, USA
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Susan M Bailey
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
6
|
Thosar SA, Barnes RP, Detwiler A, Bhargava R, Wondisford A, O'Sullivan RJ, Opresko PL. Oxidative guanine base damage plays a dual role in regulating productive ALT-associated homology-directed repair. Cell Rep 2024; 43:113656. [PMID: 38194346 PMCID: PMC10851105 DOI: 10.1016/j.celrep.2023.113656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 11/06/2023] [Accepted: 12/20/2023] [Indexed: 01/10/2024] Open
Abstract
Cancer cells maintain telomeres by upregulating telomerase or alternative lengthening of telomeres (ALT) via homology-directed repair at telomeric DNA breaks. 8-Oxoguanine (8oxoG) is a highly prevalent endogenous DNA lesion in telomeric sequences, altering telomere structure and telomerase activity, but its impact on ALT is unclear. Here, we demonstrate that targeted 8oxoG formation at telomeres stimulates ALT activity and homologous recombination specifically in ALT cancer cells. Mechanistically, an acute 8oxoG induction increases replication stress, as evidenced by increased telomere fragility and ATR kinase activation at ALT telomeres. Furthermore, ALT cells are more sensitive to chronic telomeric 8oxoG damage than telomerase-positive cancer cells, consistent with increased 8oxoG-induced replication stress. However, telomeric 8oxoG production in G2 phase, when ALT telomere elongation occurs, impairs telomeric DNA synthesis. Our study demonstrates that a common oxidative base lesion has a dual role in regulating ALT depending on when the damage arises in the cell cycle.
Collapse
Affiliation(s)
- Sanjana A Thosar
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Ryan P Barnes
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Ariana Detwiler
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Ragini Bhargava
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Anne Wondisford
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Roderick J O'Sullivan
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Patricia L Opresko
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
7
|
Marinaccio J, Micheli E, Udroiu I, Di Nottia M, Carrozzo R, Baranzini N, Grimaldi A, Leone S, Moreno S, Muzzi M, Sgura A. TERT Extra-Telomeric Roles: Antioxidant Activity and Mitochondrial Protection. Int J Mol Sci 2023; 24:ijms24054450. [PMID: 36901881 PMCID: PMC10002448 DOI: 10.3390/ijms24054450] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/18/2023] [Accepted: 02/20/2023] [Indexed: 03/05/2023] Open
Abstract
Telomerase reverse transcriptase (TERT) is the catalytic subunit of telomerase holoenzyme, which adds telomeric DNA repeats on chromosome ends to counteract telomere shortening. In addition, there is evidence of TERT non-canonical functions, among which is an antioxidant role. In order to better investigate this role, we tested the response to X-rays and H2O2 treatment in hTERT-overexpressing human fibroblasts (HF-TERT). We observed in HF-TERT a reduced induction of reactive oxygen species and an increased expression of the proteins involved in the antioxidant defense. Therefore, we also tested a possible role of TERT inside mitochondria. We confirmed TERT mitochondrial localization, which increases after oxidative stress (OS) induced by H2O2 treatment. We next evaluated some mitochondrial markers. The basal mitochondria quantity appeared reduced in HF-TERT compared to normal fibroblasts and an additional reduction was observed after OS; nevertheless, the mitochondrial membrane potential and morphology were better conserved in HF-TERT. Our results suggest a protective function of TERT against OS, also preserving mitochondrial functionality.
Collapse
Affiliation(s)
| | - Emanuela Micheli
- Department of Science, University “ROMA TRE”, 00146 Rome, Italy
- Correspondence:
| | - Ion Udroiu
- Department of Science, University “ROMA TRE”, 00146 Rome, Italy
| | - Michela Di Nottia
- Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
| | - Rosalba Carrozzo
- Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children’s Hospital IRCCS, 00146 Rome, Italy
| | - Nicolò Baranzini
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Annalisa Grimaldi
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
| | - Stefano Leone
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, 00161 Rome, Italy
| | - Sandra Moreno
- Department of Science, University “ROMA TRE”, 00146 Rome, Italy
- IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Maurizio Muzzi
- Department of Science, University “ROMA TRE”, 00146 Rome, Italy
- IRCCS Santa Lucia Foundation, 00179 Rome, Italy
| | - Antonella Sgura
- Department of Science, University “ROMA TRE”, 00146 Rome, Italy
| |
Collapse
|
8
|
de Bardet JC, Cardentey CR, González BL, Patrone D, Mulet IL, Siniscalco D, Robinson-Agramonte MDLA. Cell Immortalization: In Vivo Molecular Bases and In Vitro Techniques for Obtention. BIOTECH 2023; 12:14. [PMID: 36810441 PMCID: PMC9944833 DOI: 10.3390/biotech12010014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Somatic human cells can divide a finite number of times, a phenomenon known as the Hayflick limit. It is based on the progressive erosion of the telomeric ends each time the cell completes a replicative cycle. Given this problem, researchers need cell lines that do not enter the senescence phase after a certain number of divisions. In this way, more lasting studies can be carried out over time and avoid the tedious work involved in performing cell passes to fresh media. However, some cells have a high replicative potential, such as embryonic stem cells and cancer cells. To accomplish this, these cells express the enzyme telomerase or activate the mechanisms of alternative telomere elongation, which favors the maintenance of the length of their stable telomeres. Researchers have been able to develop cell immortalization technology by studying the cellular and molecular bases of both mechanisms and the genes involved in the control of the cell cycle. Through it, cells with infinite replicative capacity are obtained. To obtain them, viral oncogenes/oncoproteins, myc genes, ectopic expression of telomerase, and the manipulation of genes that regulate the cell cycle, such as p53 and Rb, have been used.
Collapse
Affiliation(s)
- Javier Curi de Bardet
- Department of Neurobiology, International Center for Neurological Restoration, Havana 11300, Cuba
| | | | - Belkis López González
- Department of Allergy, Calixto Garcia General University Hospital, Havana 10400, Cuba
| | - Deanira Patrone
- Department of Experimental Medicine, Division of Molecular Biology, Biotechnology and Histology, University of Campania, 80138 Naples, Italy
| | | | - Dario Siniscalco
- Department of Experimental Medicine, Division of Molecular Biology, Biotechnology and Histology, University of Campania, 80138 Naples, Italy
| | | |
Collapse
|
9
|
Čapková Frydrychová R, Mason JM, Peska V. Editorial: Telomere Flexibility and Versatility: A Role of Telomeres in Adaptive Potential. Front Genet 2021; 12:771938. [PMID: 34671387 PMCID: PMC8520972 DOI: 10.3389/fgene.2021.771938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 09/15/2021] [Indexed: 11/30/2022] Open
Affiliation(s)
- Radmila Čapková Frydrychová
- Institute of Entomology, Biology Centre of the Czech Academy of Sciences, České Budějovice, Czechia.,Faculty of Science, University of South Bohemia, České Budějovice, Czechia
| | | | - Vratislav Peska
- Department of Cell Biology and Radiobiology, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czechia
| |
Collapse
|
10
|
Bailey SM, Luxton JJ, McKenna MJ, Taylor LE, George KA, Jhavar SG, Swanson GP. Ad Astra - telomeres in space! Int J Radiat Biol 2021; 98:395-403. [PMID: 34270368 DOI: 10.1080/09553002.2021.1956010] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE My journey to the stars began as I - along with the whole world - stood still and watched Neil Armstrong take those first small steps on the Moon. Fast forward 50 years and NASA astronauts Scott Kelly and Christina Koch each spend nearly a year in space aboard the International Space Station (ISS), a remarkable multinational collaborative project and floating U.S. National Laboratory that has supported continuous human presence in low Earth orbit for the past 20 years. Marking a new era of human space exploration, the first commercial rocket, SpaceX Falcon 9, recently launched NASA astronauts Doug Hurley and Bob Behnken in the Crew Dragon spacecraft Endeavor to the ISS and returned safely to Earth. NASA and its commercial partners are rapidly advancing innovative space technologies, and with the recently announced Artemis team of astronauts, plans to send the first woman and next man back to the moon and establish sustainable exploration by the end of the decade. Humankind will then be poised to take the next giant leap - pioneering human exploration of Mars. CONCLUSIONS Historically, fewer than 600 individuals have participated in spaceflight, the vast majority of whom have been middle aged males (35-55 years) on short duration missions (less than 20 days). Thus, as the number and diversity of space travelers increase, a better understanding of how long-duration spaceflight affects human health is essential to maintaining individual astronaut performance during, and improving disease and aging trajectories following, future exploration missions. Here, I review findings from our NASA Twins Study and Telomeres investigations, highlighting potential mechanistic roles of chronic space radiation exposure in changes in telomere length and persistent DNA damage responses associated with long-duration spaceflight. Importantly, similar trends were observed in prostate cancer patients undergoing intensity-modulated radiation therapy (IMRT), additional support specifically for the role of radiation exposure. Individual differences in response were also observed in both cohorts, underscoring the importance of developing personalized approaches for evaluating human health effects and long-term outcomes associated with radiation exposures, whether on Earth or living in the extreme environment of space.
Collapse
Affiliation(s)
- Susan M Bailey
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA.,Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
| | - Jared J Luxton
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA.,Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
| | - Miles J McKenna
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA.,Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
| | - Lynn E Taylor
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | | | | | | |
Collapse
|
11
|
Nelson CB, Alturki TM, Luxton JJ, Taylor LE, Maranon DG, Muraki K, Murnane JP, Bailey SM. Telomeric Double Strand Breaks in G1 Human Cells Facilitate Formation of 5' C-Rich Overhangs and Recruitment of TERRA. Front Genet 2021; 12:644803. [PMID: 33841503 PMCID: PMC8027502 DOI: 10.3389/fgene.2021.644803] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/08/2021] [Indexed: 12/12/2022] Open
Abstract
Telomeres, repetitive nucleoprotein complexes that protect chromosomal termini and prevent them from activating inappropriate DNA damage responses (DDRs), shorten with cell division and thus with aging. Here, we characterized the human cellular response to targeted telomeric double-strand breaks (DSBs) in telomerase-positive and telomerase-independent alternative lengthening of telomere (ALT) cells, specifically in G1 phase. Telomeric DSBs in human G1 cells elicited early signatures of a DDR; however, localization of 53BP1, an important regulator of resection at broken ends, was not observed at telomeric break sites. Consistent with this finding and previously reported repression of classical non-homologous end-joining (c-NHEJ) at telomeres, evidence for c-NHEJ was also lacking. Likewise, no evidence of homologous recombination (HR)-dependent repair of telomeric DSBs in G1 was observed. Rather, and supportive of rapid truncation events, telomeric DSBs in G1 human cells facilitated formation of extensive tracks of resected 5′ C-rich telomeric single-stranded (ss)DNA, a previously proposed marker of the recombination-dependent ALT pathway. Indeed, induction of telomeric DSBs in human ALT cells resulted in significant increases in 5′ C-rich (ss)telomeric DNA in G1, which rather than RPA, was bound by the complementary telomeric RNA, TERRA, presumably to protect these exposed ends so that they persist into S/G2 for telomerase-mediated or HR-dependent elongation, while also circumventing conventional repair pathways. Results demonstrate the remarkable adaptability of telomeres, and thus they have important implications for persistent telomeric DNA damage in normal human G1/G0 cells (e.g., lymphocytes), as well as for therapeutically relevant targets to improve treatment of ALT-positive tumors.
Collapse
Affiliation(s)
- Christopher B Nelson
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, United States.,Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, United States
| | - Taghreed M Alturki
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, United States.,Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, United States
| | - Jared J Luxton
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, United States.,Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, United States
| | - Lynn E Taylor
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, United States
| | - David G Maranon
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, United States
| | - Keiko Muraki
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, United States
| | - John P Murnane
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, United States
| | - Susan M Bailey
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, United States.,Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
12
|
Stan MS, Badea S, Hermenean A, Herman H, Trica B, Sbarcea BG, Dinischiotu A. New Insights into the Cell Death Signaling Pathways Triggered by Long-Term Exposure to Silicon-Based Quantum Dots in Human Lung Fibroblasts. NANOMATERIALS 2021; 11:nano11020323. [PMID: 33513804 PMCID: PMC7911990 DOI: 10.3390/nano11020323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/18/2021] [Accepted: 01/22/2021] [Indexed: 12/19/2022]
Abstract
This report is the first research study that aims to explore the molecular mechanisms involved in the in vitro pulmonary cytotoxicity triggered by long-term exposure to silicon-based quantum dots (QDs). Human lung fibroblasts (MRC-5 cell line) were exposed to 5 µg/mL silicon-based QDs for 5 weeks and the concentration was increased up to 40 µg/mL QDs during the next 4 weeks. Cell viability and population doubling level were calculated based on Trypan blue staining. The expression levels of proteins were established by Western blotting and the telomeres’ length was determined through Southern blotting. Prolonged exposure of lung fibroblasts to QDs reduced the cell viability by 10% compared to untreated cells. The level of p53 and apoptosis-inducing factor (AIF) expression increased during the exposure, the peak intensity being registered after the seventh week. The expressions of autophagy-related proteins, Beclin-1 and LC-3, were higher compared to untreated cells. Regarding the protein expression of Nrf-2, a progressive decrease was noticed, suggesting the downregulation of a cytoprotective response to oxidative stress. In contrast, the heat shock proteins’ (HSPs) expression was increased or maintained near the control level during QDs exposure in order to promote cell survival. Furthermore, the telomeres’ length was not reduced during this exposure, indicating that QDs did not induce cellular senescence. In conclusion, our study shows that silicon-based QDs triggered the activation of apoptotic and autophagy pathways and downregulation of survival signaling molecules as an adaptive response to cellular stress which was not associated with telomeres shortening.
Collapse
Affiliation(s)
- Miruna S. Stan
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050095 Bucharest, Romania; (M.S.S.); (S.B.); (A.H.)
- Research Institute of the University of Bucharest–ICUB, University of Bucharest, 91-95 Spl. Independentei, 050095 Bucharest, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 1-7 Gheorghe Polizu Str., 011061 Bucharest, Romania
| | - Smaranda Badea
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050095 Bucharest, Romania; (M.S.S.); (S.B.); (A.H.)
| | - Anca Hermenean
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050095 Bucharest, Romania; (M.S.S.); (S.B.); (A.H.)
- Institute of Life Sciences, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania;
| | - Hildegard Herman
- Institute of Life Sciences, Vasile Goldis Western University of Arad, 86 Rebreanu, 310414 Arad, Romania;
| | - Bogdan Trica
- National Institute for Research & Development in Chemistry and Petrochemistry (INCDCP-ICECHIM), 202 Spl. Independentei, 060021 Bucharest, Romania;
| | - Beatrice G. Sbarcea
- Materials Characterization Department, National Institute for Research & Development in Electrical Engineering (ICPE-CA), 313 Splaiul Unirii, 030138 Bucharest, Romania
- Correspondence: (B.G.S.); (A.D.)
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Spl. Independentei, 050095 Bucharest, Romania; (M.S.S.); (S.B.); (A.H.)
- Correspondence: (B.G.S.); (A.D.)
| |
Collapse
|
13
|
Luxton JJ, McKenna MJ, Taylor LE, George KA, Zwart SR, Crucian BE, Drel VR, Garrett-Bakelman FE, Mackay MJ, Butler D, Foox J, Grigorev K, Bezdan D, Meydan C, Smith SM, Sharma K, Mason CE, Bailey SM. Temporal Telomere and DNA Damage Responses in the Space Radiation Environment. Cell Rep 2020; 33:108435. [PMID: 33242411 DOI: 10.1016/j.celrep.2020.108435] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/15/2020] [Accepted: 11/04/2020] [Indexed: 02/08/2023] Open
Abstract
Telomeres, repetitive terminal features of chromosomes essential for maintaining genome integrity, shorten with cell division, lifestyle factors and stresses, and environmental exposures, and so they provide a robust biomarker of health, aging, and age-related diseases. We assessed telomere length dynamics (changes over time) in three unrelated astronauts before, during, and after 1-year or 6-month missions aboard the International Space Station (ISS). Similar to our results for National Aeronautics and Space Administration's (NASA's) One-Year Mission twin astronaut (Garrett-Bakelman et al., 2019), significantly longer telomeres were observed during spaceflight for two 6-month mission astronauts. Furthermore, telomere length shortened rapidly after return to Earth for all three crewmembers and, overall, telomere length tended to be shorter after spaceflight than before spaceflight. Consistent with chronic exposure to the space radiation environment, signatures of persistent DNA damage responses were also detected, including mitochondrial and oxidative stress, inflammation, and telomeric and chromosomal aberrations, which together provide potential mechanistic insight into spaceflight-specific telomere elongation.
Collapse
Affiliation(s)
- Jared J Luxton
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA; Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
| | - Miles J McKenna
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA; Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
| | - Lynn E Taylor
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | | | - Sara R Zwart
- University of Texas Medical Branch, Galveston, TX, USA
| | | | - Viktor R Drel
- Center for Renal Precision Medicine, UT Health, San Antonio, TX, USA
| | - Francine E Garrett-Bakelman
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA; Department of Medicine, University of Virginia, Charlottesville, VA, USA; Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Matthew J Mackay
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Daniel Butler
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Jonathan Foox
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Kirill Grigorev
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Daniela Bezdan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | | | - Kumar Sharma
- Center for Renal Precision Medicine, UT Health, San Antonio, TX, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA; The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
| | - Susan M Bailey
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA; Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
14
|
Luxton JJ, McKenna MJ, Lewis A, Taylor LE, George KA, Dixit SM, Moniz M, Benegas W, Mackay MJ, Mozsary C, Butler D, Bezdan D, Meydan C, Crucian BE, Zwart SR, Smith SM, Mason CE, Bailey SM. Telomere Length Dynamics and DNA Damage Responses Associated with Long-Duration Spaceflight. Cell Rep 2020; 33:108457. [PMID: 33242406 DOI: 10.1016/j.celrep.2020.108457] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/31/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Telomere length dynamics and DNA damage responses were assessed before, during, and after one-year or shorter duration missions aboard the International Space Station (ISS) in a comparatively large cohort of astronauts (n = 11). Although generally healthy individuals, astronauts tended to have significantly shorter telomeres and lower telomerase activity than age- and sex-matched ground controls before and after spaceflight. Although telomeres were longer during spaceflight irrespective of mission duration, telomere length shortened rapidly upon return to Earth, and overall astronauts had shorter telomeres after spaceflight than they did before; inter-individual differences were identified. During spaceflight, all crewmembers experienced oxidative stress, which positively correlated with telomere length dynamics. Significantly increased frequencies of chromosomal inversions were observed during and after spaceflight; changes in cell populations were also detected. We propose a telomeric adaptive response to chronic oxidative damage in extreme environments, whereby the telomerase-independent Alternative Lengthening of Telomeres (ALT) pathway is transiently activated in normal somatic cells.
Collapse
Affiliation(s)
- Jared J Luxton
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA; Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
| | - Miles J McKenna
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA; Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA
| | - Aidan Lewis
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Lynn E Taylor
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | | | - Sameer M Dixit
- Center for Molecular Dynamics - Nepal (CMDN), Kathmandu, Nepal
| | | | | | - Matthew J Mackay
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Christopher Mozsary
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Daniel Butler
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Daniela Bezdan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA
| | - Cem Meydan
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA; The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
| | - Brian E Crucian
- Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX, USA
| | - Sara R Zwart
- University of Texas Medical Branch, Galveston, TX, USA
| | - Scott M Smith
- Human Health and Performance Directorate, NASA Johnson Space Center, Houston, TX, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, USA; The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA; The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA.
| | - Susan M Bailey
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA; Cell and Molecular Biology Program, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
15
|
Polettini J, da Silva MG. Telomere-Related Disorders in Fetal Membranes Associated With Birth and Adverse Pregnancy Outcomes. Front Physiol 2020; 11:561771. [PMID: 33123024 PMCID: PMC7573552 DOI: 10.3389/fphys.2020.561771] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/14/2020] [Indexed: 12/22/2022] Open
Abstract
Telomere disorders have been associated with aging-related diseases, including diabetes, vascular, and neurodegenerative diseases. The main consequence of altered telomere is the induction of the state of irreversible cell cycle arrest. Though several mechanisms responsible for the activation of senescence have been identified, it is still unclear how a cell is indeed induced to become irreversibly arrested. Most tissues in the body will experience senescence throughout its lifespan, but intrinsic and extrinsic stressors, such as chemicals, pollution, oxidative stress (OS), and inflammation accelerate the process. Pregnancy is a state of OS, as the higher metabolic demand of the growing fetus results in increased reactive oxygen species production. As a temporary organ in the mother, senescence in fetal membranes and placenta is expected and linked to term parturition (>37 weeks of gestation). However, a persistent, overwhelming, or premature OS affects placental antioxidant capacity, with consequent accumulation of OS causing damage to lipids, proteins, and DNA in the placental tissues. Therefore, senescence and its main inducer, telomere length (TL) reduction, have been associated with pregnancy complications, including stillbirth, preeclampsia, intrauterine growth restriction, and prematurity. Fetal membranes have a notable role in preterm births, which continue to be a major health issue associated with increased risk of neo and perinatal adverse outcomes and/or predisposition to disease in later life; however, the ability to mediate a delay in parturition during such cases is limited, because the pathophysiology of preterm births and physiological mechanisms of term births are not yet fully elucidated. Here, we review the current knowledge regarding the regulation of telomere-related senescence mechanisms in fetal membranes, highlighting the role of inflammation, methylation, and telomerase activity. Moreover, we present the evidences of TL reduction and senescence in gestational tissues by the time of term parturition. In conclusion, we verified that telomere regulation in fetal membranes requires a more complete understanding, in order to support the development of successful effective interventions of the molecular mechanisms that triggers parturition, including telomere signals, which may vary throughout placental tissues.
Collapse
Affiliation(s)
- Jossimara Polettini
- Universidade Federal da Fronteira Sul (UFFS), Programa de Pós Graduação em Ciências Biomédicas, Faculdade de Medicina, Campus Passo Fundo, Brazil
| | - Marcia Guimarães da Silva
- Universidade Estadual Paulista (UNESP), Faculdade de Medicina, Departamento de Patologia, Botucatu, Brazil
| |
Collapse
|
16
|
Piekna-Przybylska D, Bambara RA, Maggirwar SB, Dewhurst S. G-quadruplex ligands targeting telomeres do not inhibit HIV promoter activity and cooperate with latency reversing agents in killing latently infected cells. Cell Cycle 2020; 19:2298-2313. [PMID: 32807015 DOI: 10.1080/15384101.2020.1796268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Altered telomere maintenance mechanism (TMM) is linked to increased DNA damage at telomeres and telomere uncapping. We previously showed that HIV-1 latent cells have altered TMM and are susceptible to ligands that target G-quadruplexes (G4) at telomeres. Susceptibility of latent cells to telomere targeting could potentially be used to support approaches to eradicate HIV reservoirs. However, G4 ligands also target G-quadruplexes in promoters blocking gene transcription. Since HIV promoter sequence can form G-quadruplexes, we investigated whether G4 ligands interfere with HIV-1 promoter activity and virus reactivation from latency, and whether telomere targeting could be combined with latency reversing agents (LRAs) to promote elimination of HIV reservoirs. Our results indicate that Sp1 binding region in HIV-1 promoter can adopt G4 structures in duplex DNA, and that in vitro binding of Sp1 to G-quadruplex is blocked by G4 ligand, suggesting that agents targeting telomeres interfere with virus reactivation. However, our studies show that G4 agents do not affect HIV-1 promoter activity in cell culture, and do not interfere with latency reversal. Importantly, primary memory CD4 + T cells infected with latent HIV-1 are more susceptible to combined treatment with LRAs and G4 ligands, indicating that drugs targeting TMM may enhance killing of HIV reservoirs. Using a cell-based DNA repair assay, we also found that HIV-1 infected cells have reduced efficiency of DNA mismatch repair (MMR), and base excision repair (BER), suggesting that altered TMM in latently infected cells could be associated with accumulation of DNA damage at telomeres and changes in telomeric caps.
Collapse
Affiliation(s)
- Dorota Piekna-Przybylska
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester , Rochester, NY, USA
| | - Robert A Bambara
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester , Rochester, NY, USA
| | - Sanjay B Maggirwar
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University , Washington, DC, USA
| | - Stephen Dewhurst
- Department of Microbiology and Immunology, School of Medicine and Dentistry, University of Rochester , Rochester, NY, USA
| |
Collapse
|
17
|
Sommer A, Royle NJ. ALT: A Multi-Faceted Phenomenon. Genes (Basel) 2020; 11:E133. [PMID: 32012790 PMCID: PMC7073516 DOI: 10.3390/genes11020133] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/17/2020] [Accepted: 01/22/2020] [Indexed: 01/13/2023] Open
Abstract
One of the hallmarks of cancer cells is their indefinite replicative potential, made possible by the activation of a telomere maintenance mechanism (TMM). The majority of cancers reactivate the reverse transcriptase, telomerase, to maintain their telomere length but a minority (10% to 15%) utilize an alternative lengthening of telomeres (ALT) pathway. Here, we review the phenotypes and molecular markers specific to ALT, and investigate the significance of telomere mutations and sequence variation in ALT cell lines. We also look at the recent advancements in understanding the different mechanisms behind ALT telomere elongation and finally, the progress made in identifying potential ALT-targeted therapies, including those already in use for the treatment of both hematological and solid tumors.
Collapse
Affiliation(s)
| | - Nicola J. Royle
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK;
| |
Collapse
|
18
|
Kent T, Gracias D, Shepherd S, Clynes D. Alternative Lengthening of Telomeres in Pediatric Cancer: Mechanisms to Therapies. Front Oncol 2020; 9:1518. [PMID: 32039009 PMCID: PMC6985284 DOI: 10.3389/fonc.2019.01518] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 12/17/2019] [Indexed: 12/26/2022] Open
Abstract
Achieving replicative immortality is a crucial step in tumorigenesis and requires both bypassing cell cycle checkpoints and the extension of telomeres, sequences that protect the distal ends of chromosomes during replication. In the majority of cancers this is achieved through the enzyme telomerase, however a subset of cancers instead utilize a telomerase-independent mechanism of telomere elongation-the Alternative Lengthening of Telomeres (ALT) pathway. Recent work has aimed to decipher the exact mechanism that underlies this pathway. To this end, this pathway has now been shown to extend telomeres through exploitation of DNA repair machinery in a unique process that may present a number of druggable targets. The identification of such targets, and the subsequent development or repurposing of therapies to these targets may be crucial to improving the prognosis for many ALT-positive cancers, wherein mean survival is lower than non-ALT counterparts and the cancers themselves are particularly unresponsive to standard of care therapies. In this review we summarize the recent identification of many aspects of the ALT pathway, and the therapies that may be employed to exploit these new targets.
Collapse
Affiliation(s)
- Thomas Kent
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Deanne Gracias
- Department of Oncology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - Samuel Shepherd
- Department of Oncology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| | - David Clynes
- Department of Oncology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
19
|
Louzon M, Coeurdassier M, Gimbert F, Pauget B, de Vaufleury A. Telomere dynamic in humans and animals: Review and perspectives in environmental toxicology. ENVIRONMENT INTERNATIONAL 2019; 131:105025. [PMID: 31352262 DOI: 10.1016/j.envint.2019.105025] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 06/19/2019] [Accepted: 07/15/2019] [Indexed: 06/10/2023]
Abstract
Telomeres (TLs) play major roles in stabilizing the genome and are usually shortened with ageing. The maintenance of TLs is ensured by two mechanisms involving telomerase (TA) enzyme and alternative lengthening telomeres (ALT). TL shortening and/or TA inhibition have been related to health effects on organisms (leading to reduced reproductive lifespan and survival), suggesting that they could be key processes in toxicity mechanisms (at molecular and cellular levels) and relevant as an early warning of exposure and effect of chemicals on human health and animal population dynamics. Consequently, a critical analysis of knowledge about relationships between TL dynamic and environmental pollution is essential to highlight the relevance of TL measurement in environmental toxicology. The first objective of this review is to provide a survey on the basic knowledge about TL structure, roles, maintenance mechanisms and causes of shortening in both vertebrates (including humans) and invertebrates. Overall, TL length decreases with ageing but some unexpected exceptions are reported (e.g., in species with different lifespans, such as the nematode Caenorhabditis elegans or the crustacean Homarus americanus). Inconsistent results reported in various biological groups or even between species of the same genus (e.g., the microcrustacean Daphnia sp.) indicate that the relation usually proposed between TL shortening and a decrease in TA activity cannot be generalized and depends on the species, stage of development or lifespan. Although the scientific literature provides evidence of the effect of ageing on TL shortening, much less information on the relationships between shortening, maintenance of TLs, influence of other endogenous and environmental drivers, including exposure to chemical pollutants, is available, especially in invertebrates. The second objective of this review is to connect knowledge on TL dynamic and exposure to contaminants. Most of the studies published on humans rely on correlative epidemiological approaches and few in vitro experiments. They have shown TL attrition when exposed to contaminants, such as polycyclic aromatic hydrocarbons (PAH), polychlorinated biphenyls (PCB), pesticides and metallic elements (ME). In other vertebrates, the studies we found deals mainly with birds and, overall, report a disturbance of TL dynamic consecutively to exposure to chemicals, including metals and organic compounds. In invertebrates, no data are available and the potential of TL dynamic in environmental risk assessment remains to be explored. On the basis of the main gaps identified some research perspectives (e.g., impact of endogenous and environmental drivers, dose response effects, link between TL length, TA activity, longevity and ageing) are proposed to better understand the potential of TL and TA measurements in humans and animals in environmental toxicology.
Collapse
Affiliation(s)
- Maxime Louzon
- Department Chrono-Environnement, UMR UFC/CNRS 6249 USC INRA University of Bourgogne Franche-Comté, 16 route de Gray, 25000 Besançon, France
| | - Michael Coeurdassier
- Department Chrono-Environnement, UMR UFC/CNRS 6249 USC INRA University of Bourgogne Franche-Comté, 16 route de Gray, 25000 Besançon, France
| | - Frédéric Gimbert
- Department Chrono-Environnement, UMR UFC/CNRS 6249 USC INRA University of Bourgogne Franche-Comté, 16 route de Gray, 25000 Besançon, France
| | - Benjamin Pauget
- TESORA, Le Visium, 22 avenue Aristide Briand, 94110 Arcueil, France
| | - Annette de Vaufleury
- Department Chrono-Environnement, UMR UFC/CNRS 6249 USC INRA University of Bourgogne Franche-Comté, 16 route de Gray, 25000 Besançon, France.
| |
Collapse
|
20
|
Co-Operation between Aneuploidy and Metabolic Changes in Driving Tumorigenesis. Int J Mol Sci 2019; 20:ijms20184611. [PMID: 31540349 PMCID: PMC6770258 DOI: 10.3390/ijms20184611] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/05/2019] [Accepted: 09/17/2019] [Indexed: 12/11/2022] Open
Abstract
Alterations from the normal set of chromosomes are extremely common as cells progress toward tumourigenesis. Similarly, we expect to see disruption of normal cellular metabolism, particularly in the use of glucose. In this review, we discuss the connections between these two processes: how chromosomal aberrations lead to metabolic disruption, and vice versa. Both processes typically result in the production of elevated levels of reactive oxygen species, so we particularly focus on their role in mediating oncogenic changes.
Collapse
|
21
|
X-rays Activate Telomeric Homologous Recombination Mediated Repair in Primary Cells. Cells 2019; 8:cells8070708. [PMID: 31336873 PMCID: PMC6678842 DOI: 10.3390/cells8070708] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/27/2019] [Accepted: 07/06/2019] [Indexed: 12/31/2022] Open
Abstract
Cancer cells need to acquire telomere maintenance mechanisms in order to counteract progressive telomere shortening due to multiple rounds of replication. Most human tumors maintain their telomeres expressing telomerase whereas the remaining 15%–20% utilize the alternative lengthening of telomeres (ALT) pathway. Previous studies have demonstrated that ionizing radiations (IR) are able to modulate telomere lengths and to transiently induce some of the ALT-pathway hallmarks in normal primary fibroblasts. In the present study, we investigated the telomere length modulation kinetics, telomeric DNA damage induction, and the principal hallmarks of ALT over a period of 13 days in X-ray-exposed primary cells. Our results show that X-ray-treated cells primarily display telomere shortening and telomeric damage caused by persistent IR-induced oxidative stress. After initial telomere erosion, we observed a telomere elongation that was associated to the transient activation of a homologous recombination (HR) based mechanism, sharing several features with the ALT pathway observed in cancer cells. Data indicate that telomeric damage activates telomeric HR-mediated repair in primary cells. The characterization of HR-mediated telomere repair in normal cells may contribute to the understanding of the ALT pathway and to the identification of novel strategies in the treatment of ALT-positive cancers.
Collapse
|
22
|
Kotla S, Vu HT, Ko KA, Wang Y, Imanishi M, Heo KS, Fujii Y, Thomas TN, Gi YJ, Mazhar H, Paez-Mayorga J, Shin JH, Tao Y, Giancursio CJ, Medina JL, Taunton J, Lusis AJ, Cooke JP, Fujiwara K, Le NT, Abe JI. Endothelial senescence is induced by phosphorylation and nuclear export of telomeric repeat binding factor 2-interacting protein. JCI Insight 2019; 4:124867. [PMID: 31045573 PMCID: PMC6538340 DOI: 10.1172/jci.insight.124867] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/19/2019] [Indexed: 01/03/2023] Open
Abstract
The interplay among signaling events for endothelial cell (EC) senescence, apoptosis, and activation and how these pathological conditions promote atherosclerosis in the area exposed to disturbed flow (d-flow) in concert remain unclear. The aim of this study was to determine whether telomeric repeat-binding factor 2-interacting protein (TERF2IP), a member of the shelterin complex at the telomere, can regulate EC senescence, apoptosis, and activation simultaneously, and if so, by what molecular mechanisms. We found that d-flow induced p90RSK and TERF2IP interaction in a p90RSK kinase activity-dependent manner. An in vitro kinase assay revealed that p90RSK directly phosphorylated TERF2IP at the serine 205 (S205) residue, and d-flow increased TERF2IP S205 phosphorylation as well as EC senescence, apoptosis, and activation by activating p90RSK. TERF2IP phosphorylation was crucial for nuclear export of the TERF2IP-TRF2 complex, which led to EC activation by cytosolic TERF2IP-mediated NF-κB activation and also to senescence and apoptosis of ECs by depleting TRF2 from the nucleus. Lastly, using EC-specific TERF2IP-knockout (TERF2IP-KO) mice, we found that the depletion of TERF2IP inhibited d-flow-induced EC senescence, apoptosis, and activation, as well as atherosclerotic plaque formation. These findings demonstrate that TERF2IP is an important molecular switch that simultaneously accelerates EC senescence, apoptosis, and activation by S205 phosphorylation.
Collapse
Affiliation(s)
- Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hang Thi Vu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kyung Ae Ko
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yin Wang
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Masaki Imanishi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kyung-Sun Heo
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yuka Fujii
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tamlyn N. Thomas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Young Jin Gi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hira Mazhar
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jesus Paez-Mayorga
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo Leon, Mexico
| | - Ji-Hyun Shin
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yunting Tao
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Carolyn J. Giancursio
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Jan L.M. Medina
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jack Taunton
- Department of Cellular and Molecular Pharmacology, UCSF, San Francisco, California, USA
| | - Aldos J. Lusis
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - John P. Cooke
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Keigi Fujiwara
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
23
|
Matzenbacher CA, Da Silva J, Garcia ALH, Cappetta M, de Freitas TRO. Anthropogenic Effects on Natural Mammalian Populations: Correlation Between Telomere Length and Coal Exposure. Sci Rep 2019; 9:6325. [PMID: 31004106 PMCID: PMC6474877 DOI: 10.1038/s41598-019-42804-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/09/2019] [Indexed: 12/31/2022] Open
Abstract
The Candiota coal mine in Rio Grande do Sul (RS) is one of the largest in Brazil. Coal is a fossil fuel that causes environmental impacts from its extraction to combustion due to the release of different agents, such as polycyclic aromatic hydrocarbons (PAH) and heavy metals. Ctenomys torquatus are herbivorous and subterranean rodents that dig tunnels with their paws and teeth and can be exposed to coal through contaminated food. Exposure to pollutants can cause DNA damage and affect different tissues, inducing alterations in the population structure and genetic diversity. Our study aimed to evaluate the effect of exposure to coal and its derivatives on the C. torquatus population and to examine the relationship of coal exposure with variations in absolute telomere length (aTL), global DNA methylation and genotoxicity. Our study showed an inverse correlation between telomere length and coal exposure in addition to an increase in DNA damage. The results indicate that coal and its byproducts can contribute to the alteration of the C. torquatus population structure, as evidenced by a reduction in the number of adults.
Collapse
Affiliation(s)
- Cristina A Matzenbacher
- Department of Genetics, Federal University of Rio Grande do Sul, C.P. 15053, 91501-970, Porto Alegre, RS, Brazil
| | - Juliana Da Silva
- Laboratory of Genetic Toxicology, Lutheran University of Brazil, ULBRA, Canoas, 92425-900, Rio Grande do Sul, Brazil.
| | - Ana Leticia H Garcia
- Laboratory of Genetic Toxicology, Lutheran University of Brazil, ULBRA, Canoas, 92425-900, Rio Grande do Sul, Brazil.,Laboratory of Ecotoxicology, Postgraduate Program in Environmental Quality, University Feevale, ERS-239, 2755, 93525-075, Novo Hamburgo, RS, Brazil
| | - Mónica Cappetta
- Laboratorio de Epidemiología Genética, Departamento de Genética, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Thales R O de Freitas
- Department of Genetics, Federal University of Rio Grande do Sul, C.P. 15053, 91501-970, Porto Alegre, RS, Brazil
| |
Collapse
|
24
|
Bains SK, Chapman K, Bright S, Senan A, Kadhim M, Slijepcevic P. Effects of ionizing radiation on telomere length and telomerase activity in cultured human lens epithelium cells. Int J Radiat Biol 2018; 95:54-63. [PMID: 29667481 DOI: 10.1080/09553002.2018.1466066] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE To investigate the effects of ionizing radiation on telomere length and telomerase activity in human lens epithelial cells. There are studies suggesting evidence of telomere length in association with opacity of the lens; however, these studies have been conducted on Canine Lens cells. Our study was designed to understand further the effects of different doses of ionizing radiation on telomere length and telomerase activity in cultured human lens epithelium cells from three Donors. MATERIALS AND METHODS For this study, embryonic human lens epithelial (HLE) cells from three donors, obtained commercially were cultured. Telomere length and telomerase activity were measured after each passage until cells stopped growing in culture. This was repeated on irradiated (0.001 Gy, 0.01 Gy, 0.02 Gy, 0.1 Gy, 1 Gy and 2 Gy) cells. DNA damage response using the H2AX and telomere dysfunction foci assays were also examined at 30 mins, 24 hours, 48 hours and 72 hours postirradiation. RESULTS AND CONCLUSION We have demonstrated genetic changes in telomere length and oxidative stress, which may be relevant to cataractogenesis. Our study shows that in control cells telomere length increases as passage increases. We have also demonstrated that telomere length increases at higher doses of 1.0 Gy and 2.0 Gy. However, telomerase activity decreases dose dependently and as passages increase. These results are not conclusive and further studies ex vivo measuring lens opacity and telomere length in the model would be beneficial in a bigger cohort, hence confirming a link between telomere length, cataractogenesis and genetic factors.
Collapse
Affiliation(s)
- Savneet Kaur Bains
- a Department of Life Sciences , Brunel University London , Uxbridge , UK.,b Department of Biological and Medical Sciences , Oxford Brookes University , Oxford , UK
| | - Kim Chapman
- b Department of Biological and Medical Sciences , Oxford Brookes University , Oxford , UK.,c Oxford Institute of Nursing , Oxford Brookes University , Oxford , UK
| | - Scott Bright
- b Department of Biological and Medical Sciences , Oxford Brookes University , Oxford , UK.,d Department of Radiation Physics , University of Texas, MD Anderson Cancer Center , Houston , TX , USA
| | - Anish Senan
- b Department of Biological and Medical Sciences , Oxford Brookes University , Oxford , UK
| | - Munira Kadhim
- b Department of Biological and Medical Sciences , Oxford Brookes University , Oxford , UK
| | | |
Collapse
|
25
|
Xie X, Shippen DE. DDM1 guards against telomere truncation in Arabidopsis. PLANT CELL REPORTS 2018; 37:501-513. [PMID: 29392401 PMCID: PMC5880217 DOI: 10.1007/s00299-017-2245-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/26/2017] [Indexed: 05/20/2023]
Abstract
Prolonged hypomethylation of DNA leads to telomere truncation correlated with increased telomere recombination, transposon mobilization and stem cell death. Epigenetic pathways, including DNA methylation, are crucial for telomere maintenance. Deficient in DNA Methylation 1 (DDM1) encodes a nucleosome remodeling protein, required to maintain DNA methylation in Arabidopsis thaliana. Plants lacking DDM1 can be self-propagated, but in the sixth generation (G6) hypomethylation leads to rampant transposon activation and infertility. Here we examine the role of DDM1 in telomere length homeostasis through a longitudinal study of successive generations of ddm1-2 mutants. We report that bulk telomere length remains within the wild-type range for the first five generations (G1-G5), and then precipitously drops in G6. While telomerase activity becomes more variable in later generation ddm1-2 mutants, there is no correlation between enzyme activity and telomere length. Plants lacking DDM1 also exhibit no dysregulation of several known telomere-associated transcripts, including TERRA. Instead, telomere shortening coincides with increased G-overhangs and extra-chromosomal circles, consistent with deletional recombination. Telomere shortening also correlates with transcriptional activation of retrotransposons, and a hypersensitive DNA damage response in root apical meristems. Since abiotic stresses, including DNA damage, stimulate homologous recombination, we hypothesize that telomere deletion in G6 ddm1-2 mutants is a by-product of elevated genome-wide recombination in response to transposon mobilization. Further, we speculate that telomere truncation may be beneficial in adverse environmental conditions by accelerating the elimination of stem cells with aberrant genomes.
Collapse
Affiliation(s)
- Xiaoyuan Xie
- Department of Biochemistry and Biophysics, Texas A&M University, 2128 TAMU, College Station, TX, 77843-2128, USA
| | - Dorothy E Shippen
- Department of Biochemistry and Biophysics, Texas A&M University, 2128 TAMU, College Station, TX, 77843-2128, USA.
| |
Collapse
|
26
|
Su D, Huang X, Dong C, Ren J. Quantitative Determination of Telomerase Activity by Combining Fluorescence Correlation Spectroscopy with Telomerase Repeat Amplification Protocol. Anal Chem 2017; 90:1006-1013. [PMID: 29211436 DOI: 10.1021/acs.analchem.7b04256] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Telomerase is a key enzyme for maintaining the telomere length and is regarded as a versatile cancer biomarker and a potential drug target due to its important role in cancer and aging. It is necessary to develop a sensitive and reliable method for detection of telomerase activity due to its very low level in cells. In this Article, we propose an ultrasensitive and robust method for quantitative determination of telomerase activity by combining single molecule fluorescence correlation spectroscopy (FCS) with telomerase repeat amplification protocol (TRAP). The principle of this new method (FCS-TRAP) is based on measurement of the change in characteristic diffusion time and molecule number of TRAP products by FCS. The characteristic diffusion time is related to the length of TRAP products, and the molecule number represents the concentration of TRAP products. We optimized the conditions of TRAP procedure and FCS measurements. We observed that the telomerase activities are positively correlated to characteristic diffusion time and molecule number of TRAP products at optimal conditions. This method was successfully used for determination of telomerase activity of different cells, and detection of a single cell was realized. Meanwhile, this method was used to evaluate the inhibition efficiency of inhibitors, and the IC50 values obtained were in good agreement with the references. Compared to current TRAP methods, this method shows reliable quantification, ultrahigh sensitivity, and short detection time and is without separation. We believe that the FCS-TRAP method has a potential application in clinical diagnosis and screening of telomerase inhibitors.
Collapse
Affiliation(s)
- Di Su
- School of Chemistry & Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Xiangyi Huang
- School of Chemistry & Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Chaoqing Dong
- School of Chemistry & Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Jicun Ren
- School of Chemistry & Chemical Engineering, State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University , 800 Dongchuan Road, Shanghai, 200240, P. R. China
| |
Collapse
|
27
|
Kamranvar SA, Masucci MG. Regulation of Telomere Homeostasis during Epstein-Barr virus Infection and Immortalization. Viruses 2017; 9:v9080217. [PMID: 28792435 PMCID: PMC5580474 DOI: 10.3390/v9080217] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/11/2022] Open
Abstract
The acquisition of unlimited proliferative potential is dependent on the activation of mechanisms for telomere maintenance, which counteracts telomere shortening and the consequent triggering of the DNA damage response, cell cycle arrest, and apoptosis. The capacity of Epstein Barr virus (EBV) to infect B-lymphocytes in vitro and transform the infected cells into autonomously proliferating immortal cell lines underlies the association of this human gamma-herpesvirus with a broad variety of lymphoid and epithelial cell malignancies. Current evidence suggests that both telomerase-dependent and -independent pathways of telomere elongation are activated in the infected cells during the early and late phases of virus-induced immortalization. Here we review the interaction of EBV with different components of the telomere maintenance machinery and the mechanisms by which the virus regulates telomere homeostasis in proliferating cells. We also discuss how these viral strategies may contribute to malignant transformation.
Collapse
Affiliation(s)
- Siamak A Kamranvar
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, 751 23 Uppsala, Sweden.
| | - Maria G Masucci
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden.
| |
Collapse
|