1
|
Yu HJ, Byun YH, Park CK. Techniques for assessing telomere length: A methodological review. Comput Struct Biotechnol J 2024; 23:1489-1498. [PMID: 38633384 PMCID: PMC11021795 DOI: 10.1016/j.csbj.2024.04.011] [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: 10/23/2023] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 04/19/2024] Open
Abstract
Telomeres are located at the ends of chromosomes and have specific sequences with a distinctive structure that safeguards genes. They possess capping structures that protect chromosome ends from fusion events and ensure chromosome stability. Telomeres shorten in length during each cycle of cell division. When this length reaches a certain threshold, it can lead to genomic instability, thus being implicated in various diseases, including cancer and neurodegenerative disorders. The possibility of telomeres serving as a biomarker for aging and age-related disease is being explored, and their significance is still under study. This is because post-mitotic cells, which are mature cells that do not undergo mitosis, do not experience telomere shortening due to age. Instead, other causes, for example, exposure to oxidative stress, can directly damage the telomeres, causing genomic instability. Nonetheless, a general agreement has been established that measuring telomere length offers valuable insights and forms a crucial foundation for analyzing gene expression and epigenetic data. Numerous approaches have been developed to accurately measure telomere lengths. In this review, we summarize various methods and their advantages and limitations for assessing telomere length.
Collapse
Affiliation(s)
- Hyeon Jong Yu
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Yoon Hwan Byun
- Department of Neurosurgery, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Chul-Kee Park
- Department of Neurosurgery, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
2
|
Feng Y, Guo X, Luo M, Sun Y, Sun L, Zhang H, Zou Y, Liu D, Lu H. GbHSP90 act as a dual functional role regulated in telomere stability in Ginkgo biloba. Int J Biol Macromol 2024; 279:135240. [PMID: 39250995 DOI: 10.1016/j.ijbiomac.2024.135240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 08/12/2024] [Accepted: 08/29/2024] [Indexed: 09/11/2024]
Abstract
The heat shock protein 90 (HSP90) family members are not only widely involved in animal cellular immune response and signal transduction pathway regulation, but also play an important role in plant development and environmental stress response. Here,we identified a HSP90 family member in Ginkgo biloba, designated as GbHSP90, which performs a dual functional role to regulate telomere stability. GbHSP90 was screened by a yeast one-hybrid library using the Ginkgo biloba telomeric DNA (TTTAGGG)5. Fluorescence polarization, surface plasmon resonance(SPR) and EMSA technologyies revealed a specific interaction between GbHSP90 and the double-stranded telomeric DNA via its N-CR region, with no affinity for the single-stranded telomeric DNA or human double-stranded telomeric DNA. Furthermore, yeast two-hybrid system and Split-LUC assay demonstrated that GbHSP90 can interacts with two telomere end-binding proteins:the ginkgo telomerase reverse transcriptase (GbTERT) and the ginkgo Structural Maintenance of Chromosomes protein 1 (GbSMC1). Overexpression of GbHSP90 in human 293 T and HeLa cells increased cell growth rate, the content of telomerase reverse transcriptase (TERT), and promote cell division and inhibit cell apoptosis. Our results indicated GbHSP90 have dually functions: as a telomere-binding protein that binds specifically to double-stranded telomeric DNA and as a molecular chaperone that modulates cell differentiation and apoptosis by binding to telomere protein complexes in Ginkgo biloba. This study contributes to a significantly understanding of the unique telomere complex structure and regulatory mechanisms in Ginkgo biloba, a long-lived tree species.
Collapse
Affiliation(s)
- Yuping Feng
- State Key Laboratory of Tree Genetics and Breeding, National Engineering Research Center of Tree Breeding and Ecological Restoration, Beijing Forestry University, Beijing 100083, China; The Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Xueqin Guo
- State Key Laboratory of Tree Genetics and Breeding, National Engineering Research Center of Tree Breeding and Ecological Restoration, Beijing Forestry University, Beijing 100083, China; The Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Mei Luo
- State Key Laboratory of Tree Genetics and Breeding, National Engineering Research Center of Tree Breeding and Ecological Restoration, Beijing Forestry University, Beijing 100083, China; The Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China; School of Biology and Engineering (School of Modern Industry for Health and Medicine), Guizhou Medical University, Guiyang 561113, China
| | - Yu Sun
- State Key Laboratory of Tree Genetics and Breeding, National Engineering Research Center of Tree Breeding and Ecological Restoration, Beijing Forestry University, Beijing 100083, China; The Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Leiqian Sun
- State Key Laboratory of Tree Genetics and Breeding, National Engineering Research Center of Tree Breeding and Ecological Restoration, Beijing Forestry University, Beijing 100083, China; The Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Huimin Zhang
- State Key Laboratory of Tree Genetics and Breeding, National Engineering Research Center of Tree Breeding and Ecological Restoration, Beijing Forestry University, Beijing 100083, China; The Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Yirong Zou
- State Key Laboratory of Tree Genetics and Breeding, National Engineering Research Center of Tree Breeding and Ecological Restoration, Beijing Forestry University, Beijing 100083, China; The Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Di Liu
- State Key Laboratory of Tree Genetics and Breeding, National Engineering Research Center of Tree Breeding and Ecological Restoration, Beijing Forestry University, Beijing 100083, China; The Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China
| | - Hai Lu
- State Key Laboratory of Tree Genetics and Breeding, National Engineering Research Center of Tree Breeding and Ecological Restoration, Beijing Forestry University, Beijing 100083, China; The Tree and Ornamental Plant Breeding and Biotechnology Laboratory of National Forestry and Grassland Administration, College of Biological Sciences and Technology, Beijing Forestry University, Beijing 100083, China.
| |
Collapse
|
3
|
Zheng YL, Wu X, Williams M, Verhulst S, Lin J, Takahashi Y, Ma JX, Wang Y. High-throughput single telomere analysis using DNA microarray and fluorescent in situ hybridization. Nucleic Acids Res 2024; 52:e96. [PMID: 39291738 PMCID: PMC11514468 DOI: 10.1093/nar/gkae812] [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: 04/06/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024] Open
Abstract
The human telomere system is highly dynamic. Both short and long leucocyte average telomere lengths (aTL) are associated with an increased risk of cancer and early death, illustrating the complex relationship between TL and human health and the importance of assessing TL distributions with single TL analysis. A DNA microarray and telomere fluorescent in situ hybridization (DNA-array-FISH) approach was developed to measure the base-pair (bp) lengths of single telomeres. On average 32000 telomeres were measured per DNA sample with one microarray chip assaying 96 test DNA samples. Various telomere parameters, i.e. aTL and the frequency of short/long telomeres, were computed to delineate TL distribution. The intra-assay and inter-assay coefficient of variations of aTL ranged from 1.37% to 3.98%. The correlation coefficient (r) of aTL in repeated measurements ranged from 0.91 to 1.00, demonstrating high measurement precision. aTLs measured by DNA-array-FISH predicted aTLs measured by terminal restriction fragment (TRF) analysis with r ranging 0.87-0.99. A new accurate and high-throughput method has been developed to measure the bp lengths of single telomeres. The large number of single TL data provides an opportunity for an in-depth analysis of telomere dynamics and the complex relationship between telomere and age-related diseases.
Collapse
Affiliation(s)
- Yun-Ling Zheng
- Cancer Prevention and Control Program, Department of Oncology, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, USA
| | - Xingjia Wu
- Cancer Prevention and Control Program, Department of Oncology, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, USA
| | - Madeline Williams
- Cancer Prevention and Control Program, Department of Oncology, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, USA
| | - Simon Verhulst
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Jue Lin
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Yusuke Takahashi
- Department of Biochemistry, Wake Forest School of Medicine, NC 27157, USA
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest School of Medicine, NC 27157, USA
| | - Ying Wang
- TelohealthDx, LLC, Clarksburg, MD 20871, USA
| |
Collapse
|
4
|
Coulter T, Hill C, McKnight AJ. Insights into the length and breadth of methodologies harnessed to study human telomeres. Biomark Res 2024; 12:127. [PMID: 39438947 PMCID: PMC11515763 DOI: 10.1186/s40364-024-00668-9] [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: 08/15/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024] Open
Abstract
Telomeres are protective structures at the end of eukaryotic chromosomes that are strongly implicated in ageing and ill health. They attrition upon every cellular reproductive cycle. Evidence suggests that short telomeres trigger DNA damage responses that lead to cellular senescence. Accurate methods for measuring telomeres are required to fully investigate the roles that shortening telomeres play in the biology of disease and human ageing. The last two decades have brought forth several techniques that are used for measuring telomeres. This editorial highlights strengths and limitations of traditional and emerging techniques, guiding researchers to choose the most appropriate methodology for their research needs. These methods include Quantitative Polymerase Chain Reaction (qPCR), Omega qPCR (Ω-qPCR), Terminal Restriction Fragment analysis (TRF), Single Telomere Absolute-length Rapid (STAR) assays, Single TElomere Length Analysis (STELA), TElomere Shortest Length Assays (TESLA), Telomere Combing Assays (TCA), and Long-Read Telomere Sequencing. Challenges include replicating telomere measurement within and across cohorts, measuring the length of telomeres on individual chromosomes, and standardised reporting for publications. Areas of current and future focus have been highlighted, with recent methodical advancements, such as long-read sequencing, providing significant scope to study telomeres at an individual chromosome level.
Collapse
Affiliation(s)
- Tiernan Coulter
- Centre for Public Health, Queen's University Belfast, Institute of Clinical Sciences - Block A, Royal Victoria Hospital, Grosvenor Road, Belfast, BT12 6BJ, UK
| | - Claire Hill
- Centre for Public Health, Queen's University Belfast, Institute of Clinical Sciences - Block A, Royal Victoria Hospital, Grosvenor Road, Belfast, BT12 6BJ, UK.
| | - Amy Jayne McKnight
- Centre for Public Health, Queen's University Belfast, Institute of Clinical Sciences - Block A, Royal Victoria Hospital, Grosvenor Road, Belfast, BT12 6BJ, UK.
| |
Collapse
|
5
|
Wu Z, Qu J, Liu GH. Roles of chromatin and genome instability in cellular senescence and their relevance to ageing and related diseases. Nat Rev Mol Cell Biol 2024:10.1038/s41580-024-00775-3. [PMID: 39363000 DOI: 10.1038/s41580-024-00775-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2024] [Indexed: 10/05/2024]
Abstract
Ageing is a complex biological process in which a gradual decline in physiological fitness increases susceptibility to diseases such as neurodegenerative disorders and cancer. Cellular senescence, a state of irreversible cell-growth arrest accompanied by functional deterioration, has emerged as a pivotal driver of ageing. In this Review, we discuss how heterochromatin loss, telomere attrition and DNA damage contribute to cellular senescence, ageing and age-related diseases by eliciting genome instability, innate immunity and inflammation. We also discuss how emerging therapeutic strategies could restore heterochromatin stability, maintain telomere integrity and boost the DNA repair capacity, and thus counteract cellular senescence and ageing-associated pathologies. Finally, we outline current research challenges and future directions aimed at better comprehending and delaying ageing.
Collapse
Affiliation(s)
- Zeming Wu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jing Qu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Guang-Hui Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Verhoeven JE, Wolkowitz OM, Barr Satz I, Conklin Q, Lamers F, Lavebratt C, Lin J, Lindqvist D, Mayer SE, Melas PA, Milaneschi Y, Picard M, Rampersaud R, Rasgon N, Ridout K, Söderberg Veibäck G, Trumpff C, Tyrka AR, Watson K, Wu GWY, Yang R, Zannas AS, Han LKM, Månsson KNT. The researcher's guide to selecting biomarkers in mental health studies. Bioessays 2024; 46:e2300246. [PMID: 39258367 DOI: 10.1002/bies.202300246] [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: 12/25/2023] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 09/12/2024]
Abstract
Clinical mental health researchers may understandably struggle with how to incorporate biological assessments in clinical research. The options are numerous and are described in a vast and complex body of literature. Here we provide guidelines to assist mental health researchers seeking to include biological measures in their studies. Apart from a focus on behavioral outcomes as measured via interviews or questionnaires, we advocate for a focus on biological pathways in clinical trials and epidemiological studies that may help clarify pathophysiology and mechanisms of action, delineate biological subgroups of participants, mediate treatment effects, and inform personalized treatment strategies. With this paper we aim to bridge the gap between clinical and biological mental health research by (1) discussing the clinical relevance, measurement reliability, and feasibility of relevant peripheral biomarkers; (2) addressing five types of biological tissues, namely blood, saliva, urine, stool and hair; and (3) providing information on how to control sources of measurement variability.
Collapse
Affiliation(s)
- Josine E Verhoeven
- Department of Psychiatry, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, The Netherlands
| | - Owen M Wolkowitz
- Department of Psychiatry and Behavioral Sciences, and Weill Institute for Neurosciences, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Isaac Barr Satz
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Quinn Conklin
- Center for Mind and Brain, University of California, Davis, California, USA
- Center for Health and Community, University of California, San Francisco, California, USA
| | - Femke Lamers
- Department of Psychiatry, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, The Netherlands
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, L8:00, Karolinska University Hospital, Stockholm, Sweden
| | - Jue Lin
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, USA
| | - Daniel Lindqvist
- Unit for Biological and Precision Psychiatry, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Office for Psychiatry and Habilitation, Psychiatry Research Skåne, Region Skåne, Lund, Sweden
| | - Stefanie E Mayer
- Department of Psychiatry and Behavioral Sciences, and Weill Institute for Neurosciences, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Philippe A Melas
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Complex Trait Genetics, Amsterdam, The Netherlands
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA
- Department of Neurology, H. Houston Merritt Center, Columbia Translational Neuroscience Initiative, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA
- New York State Psychiatric Institute, New York, USA
- Robert N Butler Columbia Aging Center, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Ryan Rampersaud
- Department of Psychiatry and Behavioral Sciences, and Weill Institute for Neurosciences, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Natalie Rasgon
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Kathryn Ridout
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
- Department of Psychiatry, Kaiser Permanente, Santa Rosa Medical Center, Santa Rosa, California, USA
| | - Gustav Söderberg Veibäck
- Unit for Biological and Precision Psychiatry, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Office for Psychiatry and Habilitation, Psychiatry Research Skåne, Region Skåne, Lund, Sweden
| | - Caroline Trumpff
- Department of Psychiatry, Division of Behavioral Medicine, College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, USA
| | - Audrey R Tyrka
- Department of Psychiatry and Human Behavior, Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Kathleen Watson
- Department of Psychiatry & Behavioral Sciences, Stanford University School of Medicine, Stanford, California, USA
| | - Gwyneth Winnie Y Wu
- Department of Psychiatry and Behavioral Sciences, and Weill Institute for Neurosciences, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Ruoting Yang
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Anthony S Zannas
- Department of Psychiatry, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Laura K M Han
- Department of Psychiatry, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Centre for Youth Mental Health, The University of Melbourne, Parkville, Victoria, Australia
- Orygen, Parkville, Victoria, Australia
| | - Kristoffer N T Månsson
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Psychology and Psychotherapy, Babeș-Bolyai University, Cluj-Napoca, Romania
| |
Collapse
|
7
|
Colominas-Ciuró R, Gray FE, Arikan K, Zahn S, Meier C, Criscuolo F, Bize P. Effects of persistent organic pollutants on telomere dynamics are sex and age-specific in a wild long-lived bird. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 943:173785. [PMID: 38851349 DOI: 10.1016/j.scitotenv.2024.173785] [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: 03/13/2024] [Revised: 06/02/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Chemical pollution is a major man-made environmental threat to ecosystems and natural animal populations. Of concern are persistent organic pollutants (POPs), which can persist in the environment for many years. While bioaccumulating throughout the lives of wild animals, POPs can affect their health, reproduction, and survival. However, measuring long-term effects of POPs in wild populations is challenging, and therefore appropriate biomarkers are required in wildlife ecotoxicology. One potential target is telomere length, since telomere preservation has been associated to survival and longevity, and stressors as chemical pollution can disrupt its maintenance. Here, we investigated the effects of different classes of POPs on relative telomere length (RTL) and its rate of change (TROC) in wild long-lived Alpine swifts (Tachymarptis melba). As both RTL and TROC are often reported to differ between sexes and with chronological age, we tested for sex- and age-specific (pre-senescent vs. senescent, ≥ 9 age of years, individuals) effects of POPs. Our results showed that senescent females presented longer RTL and elongated telomeres over time compared to pre-senescent females and males. These sex- and age-related differences in RTL and TROC were influenced by POPs, but differently depending on whether they were organochlorine pesticides (OCPs) or industrial polychlorinated biphenyls (PCBs). OCPs (particularly drins) were negatively associated with RTL, with the strongest negative effects being found in senescent females. Conversely, PCBs led to slower rates of telomere shortening, especially in females. Our study indicates diametrically opposed effects of OCPs on RTL and PCBs on TROC, and these effects were more pronounced in females and senescent individuals. The mechanisms behind these effects (e.g., increased oxidative stress by OCPs; upregulation of telomerase activity by PCBs) remain unknown. Our results highlight the importance in wildlife ecotoxicology to account for sex- and age-related effects when investigating the health effects of pollutants on biomarkers such as telomeres.
Collapse
Affiliation(s)
| | | | - Kalender Arikan
- Department of Biology Education, Faculty of Education, Hacettepe University, Turkey
| | - Sandrine Zahn
- Université de Strasbourg, CNRS, IPHC UMR 7178, France
| | | | | | - Pierre Bize
- Swiss Ornithological Institute, Switzerland.
| |
Collapse
|
8
|
Kageler L, Aquilanti E. Discovery of telomerase inhibitors: existing strategies and emerging innovations. Biochem Soc Trans 2024; 52:1957-1968. [PMID: 39194999 DOI: 10.1042/bst20230264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024]
Abstract
Telomerase, crucial for maintaining telomere length, is an attractive target for cancer therapy due to its role in cellular immortality. Despite three decades of research efforts, no small-molecule telomerase inhibitors have been clinically approved, highlighting the extensive challenges in developing effective telomerase-based therapeutics. This review examines conventional and emerging methods to measure telomerase activity and discusses existing inhibitors, including oligonucleotides and small molecules. Furthermore, this review highlights recent breakthroughs in structural studies of telomerase using cryo-electron microscopy, which can facilitate improved structure-based drug design. Altogether, advancements in structural methodologies and high-throughput screening offer promising prospects for telomerase-based cancer therapeutic development.
Collapse
Affiliation(s)
- Lauren Kageler
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, U.S.A
| | - Elisa Aquilanti
- Division of Neuro Oncology, Dana Farber Cancer Institute, Boston, MA, U.S.A
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, U.S.A
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, U.S.A
| |
Collapse
|
9
|
Stuart A, de Lange T. Replicative senescence is ATM driven, reversible, and accelerated by hyperactivation of ATM at normoxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600514. [PMID: 38979390 PMCID: PMC11230194 DOI: 10.1101/2024.06.24.600514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Programmed telomere shortening limits tumorigenesis through the induction of replicative senescence. Here we address three long-standing questions concerning senescence. First, we show that the ATM kinase is solely responsible for the induction of replicative senescence. Senescence was delayed by ATM inhibition (ATMi) or overexpression of TRF2, the shelterin subunit dedicated to ATM repression. In contrast, there was no evidence for ATR signaling contributing to replicative senescence even when ATMi was combined with ATR inhibition. Second, we show ATMi can induce apparently normal cell divisions in a subset of senescent cells, indicating that senescence can be reversed. Third, we show that the extended replicative life span at low (physiological) oxygen is due to diminished ATM activity. At low oxygen, cells show a decreased ATM response to dysfunctional telomeres and genome-wide DSBs compared to 20% oxygen. As this effect could be reversed by NAC, the attenuated response of ATM to critically short telomeres and the resulting extended life span at low oxygen is likely due to ROS-induced formation of cysteine disulfide-bridges that crosslink ATM dimers into a form that is not activated by DSBs. These findings show how primary human cells detect shortened telomeres and reveal the molecular mechanism underlying the telomere tumor suppressor pathway.
Collapse
Affiliation(s)
- Alexander Stuart
- Laboratory for Cell Biology and Genetics, Rockefeller University, New York, USA
| | - Titia de Lange
- Laboratory for Cell Biology and Genetics, Rockefeller University, New York, USA
| |
Collapse
|
10
|
Sun H, Li X, Long Q, Wang X, Zhu W, Chen E, Zhou W, Yang H, Huang C, Deng W, Chen M. TERC promotes non-small cell lung cancer progression by facilitating the nuclear localization of TERT. iScience 2024; 27:109869. [PMID: 38799568 PMCID: PMC11126826 DOI: 10.1016/j.isci.2024.109869] [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: 12/12/2023] [Revised: 03/26/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
The core of telomerase consists of the protein subunit telomerase reverse transcriptase (TERT) and the telomerase RNA component (TERC). So far, the role of TERC in cancer development has remained elusive. Here, we found TERC expression elevated in non-small cell lung cancer (NSCLC) tissues, which was associated with disease progression and poor prognosis in patients. Using NSCLC cell lines and xenograft models, we showed that knockdown of TERC caused cell cycle arrest, and inhibition of cell proliferation and migration. Mechanistically, TERC was exported to the cytoplasm by nuclear RNA export factor 1 (NXF1), where it mediated the interaction of TERT with other telomerase subunits. Depletion of TERC hindered the assembly and subsequent nuclear localization of the telomerase complex, preventing TERT from functioning in telomere maintenance and transcription regulation. Our findings suggest that TERC is a potential biomarker for NSCLC diagnosis and prognosis and can be a target for NSCLC treatment.
Collapse
Affiliation(s)
- Haohui Sun
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Xiaodi Li
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Qian Long
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
- Department of General Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Xiaonan Wang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Wancui Zhu
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Enni Chen
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Wenhao Zhou
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Han Yang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Chuyang Huang
- Department of Urology, Shaoyang Central Hospital, University of South China, Shaoyang, Hunan 422000, China
| | - Wuguo Deng
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| | - Miao Chen
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong 510060, China
| |
Collapse
|
11
|
Graham MK, Xu B, Davis C, Meeker AK, Heaphy CM, Yegnasubramanian S, Dyer MA, Zeineldin M. The TERT Promoter is Polycomb-Repressed in Neuroblastoma Cells with Long Telomeres. CANCER RESEARCH COMMUNICATIONS 2024; 4:1533-1547. [PMID: 38837897 PMCID: PMC11188873 DOI: 10.1158/2767-9764.crc-22-0287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 05/04/2023] [Accepted: 05/22/2024] [Indexed: 06/07/2024]
Abstract
Acquiring a telomere maintenance mechanism is a hallmark of high-risk neuroblastoma and commonly occurs by expressing telomerase (TERT). Telomerase-negative neuroblastoma has long telomeres and utilizes the telomerase-independent alternative lengthening of telomeres (ALT) mechanism. Conversely, no discernable telomere maintenance mechanism is detected in a fraction of neuroblastoma with long telomeres. Here, we show, unlike most cancers, DNA of the TERT promoter is broadly hypomethylated in neuroblastoma. In telomerase-positive neuroblastoma cells, the hypomethylated DNA promoter is approximately 1.5 kb. The TERT locus shows active chromatin marks with low enrichment for the repressive mark, H3K27me3. MYCN, a commonly amplified oncogene in neuroblstoma, binds to the promoter and induces TERT expression. Strikingly, in neuroblastoma with long telomeres, the hypomethylated region spans the entire TERT locus, including multiple nearby genes with enrichment for the repressive H3K27me3 chromatin mark. Furthermore, subtelomeric regions showed enrichment of repressive chromatin marks in neuroblastomas with long telomeres relative to those with short telomeres. These repressive marks were even more evident at the genic loci, suggesting a telomere position effect (TPE). Inhibiting H3K27 methylation by three different EZH2 inhibitors induced the expression of TERT in cell lines with long telomeres and H3K27me3 marks in the promoter region. EZH2 inhibition facilitated MYCN binding to the TERT promoter in neuroblastoma cells with long telomeres. Taken together, these data suggest that epigenetic regulation of TERT expression differs in neuroblastoma depending on the telomere maintenance status, and H3K27 methylation is important in repressing TERT expression in neuroblastoma with long telomeres. SIGNIFICANCE The epigenetic landscape of the TERT locus is unique in neuroblastoma. The DNA at the TERT locus, unlike other cancer cells and similar to normal cells, are hypomethylated in telomerase-positive neuroblastoma cells. The TERT locus is repressed by polycomb repressive complex-2 complex in neuroblastoma cells that have long telomeres and do not express TERT. Long telomeres in neuroblastoma cells are also associated with repressive chromatin states at the chromosomal termini, suggesting TPE.
Collapse
Affiliation(s)
- Mindy K. Graham
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Urology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Beisi Xu
- Center for Applied Bioinformatics, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Christine Davis
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alan K. Meeker
- Department of Urology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher M. Heaphy
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Srinivasan Yegnasubramanian
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael A. Dyer
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, Tennessee
- Howard Hughes Medical Institute, Chevy Chase, Maryland
- Department of Ophthalmology, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Maged Zeineldin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
12
|
Harutyunyan T, Sargsyan A, Kalashyan L, Igityan H, Grigoryan B, Davtyan H, Aroutiounian R, Liehr T, Hovhannisyan G. Changes in Telomere Length in Leukocytes and Leukemic Cells after Ultrashort Electron Beam Radiation. Int J Mol Sci 2024; 25:6709. [PMID: 38928414 PMCID: PMC11203595 DOI: 10.3390/ijms25126709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Application of laser-generated electron beams in radiotherapy is a recent development. Accordingly, mechanisms of biological response to radiation damage need to be investigated. In this study, telomere length (TL) as endpoint of genetic damage was analyzed in human blood cells (leukocytes) and K562 leukemic cells irradiated with laser-generated ultrashort electron beam. Metaphases and interphases were analyzed in quantitative fluorescence in situ hybridization (Q-FISH) to assess TL. TLs were shortened compared to non-irradiated controls in both settings (metaphase and interphase) after irradiation with 0.5, 1.5, and 3.0 Gy in blood leukocytes. Radiation also caused a significant TL shortening detectable in the interphase of K562 cells. Overall, a negative correlation between TL and radiation doses was observed in normal and leukemic cells in a dose-dependent manner. K562 cells were more sensitive than normal blood cells to increasing doses of ultrashort electron beam radiation. As telomere shortening leads to genome instability and cell death, the results obtained confirm the suitability of this biomarker for assessing genotoxic effects of accelerated electrons for their further use in radiation therapy. Observed differences in TL shortening between normal and K562 cells provide an opportunity for further development of optimal radiation parameters to reduce side effects in normal cells during radiotherapy.
Collapse
Affiliation(s)
- Tigran Harutyunyan
- Laboratory of General and Molecular Genetics, Research Institute of Biology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (T.H.); (A.S.); (L.K.); (H.I.); (R.A.); (G.H.)
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia
| | - Anzhela Sargsyan
- Laboratory of General and Molecular Genetics, Research Institute of Biology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (T.H.); (A.S.); (L.K.); (H.I.); (R.A.); (G.H.)
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia
| | - Lily Kalashyan
- Laboratory of General and Molecular Genetics, Research Institute of Biology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (T.H.); (A.S.); (L.K.); (H.I.); (R.A.); (G.H.)
| | - Hovhannes Igityan
- Laboratory of General and Molecular Genetics, Research Institute of Biology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (T.H.); (A.S.); (L.K.); (H.I.); (R.A.); (G.H.)
| | - Bagrat Grigoryan
- CANDLE Synchrotron Research Institute, Acharyan 31, Yerevan 0040, Armenia; (B.G.); (H.D.)
| | - Hakob Davtyan
- CANDLE Synchrotron Research Institute, Acharyan 31, Yerevan 0040, Armenia; (B.G.); (H.D.)
| | - Rouben Aroutiounian
- Laboratory of General and Molecular Genetics, Research Institute of Biology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (T.H.); (A.S.); (L.K.); (H.I.); (R.A.); (G.H.)
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia
| | - Thomas Liehr
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller University, Am Klinikum 1, D-07747 Jena, Germany
| | - Galina Hovhannisyan
- Laboratory of General and Molecular Genetics, Research Institute of Biology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia; (T.H.); (A.S.); (L.K.); (H.I.); (R.A.); (G.H.)
- Department of Genetics and Cytology, Yerevan State University, Alex Manoogian 1, Yerevan 0025, Armenia
| |
Collapse
|
13
|
Sanchez SE, Gu Y, Wang Y, Golla A, Martin A, Shomali W, Hockemeyer D, Savage SA, Artandi SE. Digital telomere measurement by long-read sequencing distinguishes healthy aging from disease. Nat Commun 2024; 15:5148. [PMID: 38890274 PMCID: PMC11189511 DOI: 10.1038/s41467-024-49007-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 05/21/2024] [Indexed: 06/20/2024] Open
Abstract
Telomere length is an important biomarker of organismal aging and cellular replicative potential, but existing measurement methods are limited in resolution and accuracy. Here, we deploy digital telomere measurement (DTM) by nanopore sequencing to understand how distributions of human telomere length change with age and disease. We measure telomere attrition and de novo elongation with up to 30 bp resolution in genetically defined populations of human cells, in blood cells from healthy donors and in blood cells from patients with genetic defects in telomere maintenance. We find that human aging is accompanied by a progressive loss of long telomeres and an accumulation of shorter telomeres. In patients with defects in telomere maintenance, the accumulation of short telomeres is more pronounced and correlates with phenotypic severity. We apply machine learning to train a binary classification model that distinguishes healthy individuals from those with telomere biology disorders. This sequencing and bioinformatic pipeline will advance our understanding of telomere maintenance mechanisms and the use of telomere length as a clinical biomarker of aging and disease.
Collapse
Affiliation(s)
- Santiago E Sanchez
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Medical Scientist Training Program, Stanford University, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuchao Gu
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Yan Wang
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Anudeep Golla
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Annika Martin
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - William Shomali
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Dirk Hockemeyer
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Sharon A Savage
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Steven E Artandi
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
14
|
DeBoy EA, Nicosia AM, Liyanarachchi S, Iyer SS, Shah MH, Ringel MD, Brock P, Armanios M. Telomere-lengthening germline variants predispose to a syndromic papillary thyroid cancer subtype. Am J Hum Genet 2024; 111:1114-1124. [PMID: 38688277 PMCID: PMC11179366 DOI: 10.1016/j.ajhg.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 05/02/2024] Open
Abstract
Papillary thyroid cancer (PTC) is the most common endocrine malignancy. 10% to 15% of individuals show familial clustering with three or more affected members, but the factors underlying this risk are unknown. In a group of recently studied individuals with POT1 pathogenic variants and ultra-long telomere length, PTC was the second most common solid tumor. We tested whether variants in POT1 and four other telomere-maintenance genes associated with familial cancer underlie PTC susceptibility. Among 470 individuals, we identified pathogenic or likely pathogenic variants in three genes encoding telomere-binding proteins: POT1, TINF2, and ACD. They were found in 4.5% and 1.5% of familial and unselected cases, respectively. Individuals harboring these variants had ultra-long telomere length, and 15 of 18 (83%) developed other cancers, of which melanoma, lymphoma, and sarcoma were most common. Among individuals with PTC and melanoma, 22% carried a deleterious germline variant, suggesting that a long telomere syndrome might be clinically recognizable. Successive generations had longer telomere length than their parents and, at times, developed more cancers at younger ages. Tumor sequencing identified a single oncogenic driver, BRAF p.Val600Glu, in 10 of 10 tumors studied, but no telomere-maintenance mechanism, including at the TERT promoter. These data identify a syndromic subset of PTCs with locus heterogeneity and telomere lengthening as a convergent mechanism. They suggest these germline variants lower the threshold to cancer by obviating the need for an acquired telomere-maintenance mechanism in addition to sustaining the longevity of oncogenic mutations.
Collapse
Affiliation(s)
- Emily A DeBoy
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Medical Scientist Training Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Telomere Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anna M Nicosia
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Telomere Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Sheila S Iyer
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Telomere Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Manisha H Shah
- Department of Internal Medicine, Columbus, OH, USA; The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Matthew D Ringel
- Department of Molecular Medicine and Therapeutics, Columbus, OH, USA; Department of Internal Medicine, Columbus, OH, USA; The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Pamela Brock
- Department of Internal Medicine, Columbus, OH, USA; The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Mary Armanios
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Telomere Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
15
|
Stevers NO, Costello JF. Telomeres in glioma: Maintenance mechanisms to therapeutic potential. Neuro Oncol 2024; 26:1025-1026. [PMID: 38466631 PMCID: PMC11145438 DOI: 10.1093/neuonc/noae052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Indexed: 03/13/2024] Open
Affiliation(s)
- Nicholas O Stevers
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Joseph F Costello
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California, USA
| |
Collapse
|
16
|
Kumar A, Nagasaka Y, Jayananthan V, Zidan A, Heisler-Taylor T, Ambati J, Tamiya S, Kerur N. Therapeutic targeting of telomerase ameliorates experimental choroidal neovascularization. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167156. [PMID: 38582267 PMCID: PMC11497592 DOI: 10.1016/j.bbadis.2024.167156] [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: 09/19/2023] [Revised: 03/29/2024] [Accepted: 03/30/2024] [Indexed: 04/08/2024]
Abstract
Choroidal neovascularization (CNV) is the principal driver of blindness in neovascular age-related macular degeneration (nvAMD). Increased activity of telomerase, has been associated with endothelial cell proliferation, survival, migration, and invasion in the context of tumor angiogenesis. Expanding on this knowledge, we investigated the role of telomerase in the development of CNV in mouse model. We observed increased gene expression and activity of telomerase in mouse CNV. Genetic deficiency of the telomerase components, telomerase reverse transcriptase (Tert) and telomerase RNA component (Terc) suppressed laser-induced CNV in mice. Similarly, a small molecule inhibitor of TERT (BIBR 1532), and antisense oligonucleotides (ASOs) targeting Tert and Terc reduced CNV growth. Bone marrow chimera studies suggested that telomerase activity in non-bone marrow-derived cells is crucial for the development of CNV. Comparison of BIBR 1532 with VEGF neutralizing therapeutic strategy in mouse revealed a comparable level of angiosuppressive activity. However, when BIBR and anti-VEGF antibodies were administered as a combination at sub-therapeutic doses, a statistically significant suppression of CNV was observed. These findings underscore the potential benefits of combining sub-therapeutic doses of BIBR and anti-VEGF antibodies for developing newer therapeutic strategies for NV-AMD. Telomerase inhibition with BIBR 1532 suppressed induction of multiple cytokines and growth factors critical for neovascularization. In conclusion, our study identifies telomerase as a promising therapeutic target for treating neovascular disease of the eye and thus provides a proof of principle for further exploration of telomerase inhibition as a novel treatment strategy for nvAMD.
Collapse
Affiliation(s)
- Aman Kumar
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Yosuke Nagasaka
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Vinodhini Jayananthan
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Asmaa Zidan
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Tyler Heisler-Taylor
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Jayakrishna Ambati
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Shigeo Tamiya
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Nagaraj Kerur
- Department of Ophthalmology and Visual Sciences, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Microbial Infection and Immunity, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
17
|
Sadler DE, Watts PC, Uusi-Heikkilä S. Directional selection, not the direction of selection, affects telomere length and copy number at ribosomal RNA loci. Sci Rep 2024; 14:12162. [PMID: 38802448 PMCID: PMC11130246 DOI: 10.1038/s41598-024-63030-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: 03/14/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024] Open
Abstract
Many fisheries exert directional selection on traits such as body size and growth rate. Whether directional selection impacts regions of the genome associated with traits related to growth is unknown. To address this issue, we characterised copy number variation in three regions of the genome associated with cell division, (1) telomeric DNA, (2) loci transcribed as ribosomal RNA (rDNA), and (3) mitochondrial DNA (mtDNA), in three selection lines of zebrafish reared at three temperatures (22 °C, 28 °C, and 34 °C). Selection lines differed in (1) the direction of selection (two lines experienced directional selection for large or small body size) and (2) whether they experienced any directional selection itself. Lines that had experienced directional selection were smaller, had lower growth rate, shorter telomeres, and lower rDNA copy number than the line that experiencing no directional selection. Neither telomere length nor rDNA copy number were affected by temperature. In contrast, mtDNA content increased at elevated temperature but did not differ among selection lines. Though directional selection impacts rDNA and telomere length, direction of such selection did not matter, whereas mtDNA acts as a stress marker for temperature. Future work should examine the consequences of these genomic changes in natural fish stocks.
Collapse
Affiliation(s)
- Daniel E Sadler
- Department of Biological and Environmental Science, University of Jyväskylä, 40014, Jyväskylä, Finland.
| | - Phillip C Watts
- Department of Biological and Environmental Science, University of Jyväskylä, 40014, Jyväskylä, Finland
| | - Silva Uusi-Heikkilä
- Department of Biological and Environmental Science, University of Jyväskylä, 40014, Jyväskylä, Finland
| |
Collapse
|
18
|
Li Y, Tian X, Luo J, Bao T, Wang S, Wu X. Molecular mechanisms of aging and anti-aging strategies. Cell Commun Signal 2024; 22:285. [PMID: 38790068 PMCID: PMC11118732 DOI: 10.1186/s12964-024-01663-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Aging is a complex and multifaceted process involving a variety of interrelated molecular mechanisms and cellular systems. Phenotypically, the biological aging process is accompanied by a gradual loss of cellular function and the systemic deterioration of multiple tissues, resulting in susceptibility to aging-related diseases. Emerging evidence suggests that aging is closely associated with telomere attrition, DNA damage, mitochondrial dysfunction, loss of nicotinamide adenine dinucleotide levels, impaired macro-autophagy, stem cell exhaustion, inflammation, loss of protein balance, deregulated nutrient sensing, altered intercellular communication, and dysbiosis. These age-related changes may be alleviated by intervention strategies, such as calorie restriction, improved sleep quality, enhanced physical activity, and targeted longevity genes. In this review, we summarise the key historical progress in the exploration of important causes of aging and anti-aging strategies in recent decades, which provides a basis for further understanding of the reversibility of aging phenotypes, the application prospect of synthetic biotechnology in anti-aging therapy is also prospected.
Collapse
Affiliation(s)
- Yumeng Li
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences; National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| | - Xutong Tian
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences; National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| | - Juyue Luo
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences; National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| | - Tongtong Bao
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences; National Center of Technology Innovation for Synthetic Biology, Tianjin, China
| | - Shujin Wang
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Xin Wu
- Tianjin Institute of Industrial Biotechnology, Chinese Academy of Sciences; National Center of Technology Innovation for Synthetic Biology, Tianjin, China.
| |
Collapse
|
19
|
Stephens Z, Kocher JP. Characterization of telomere variant repeats using long reads enables allele-specific telomere length estimation. BMC Bioinformatics 2024; 25:194. [PMID: 38755561 PMCID: PMC11100205 DOI: 10.1186/s12859-024-05807-5] [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] [Received: 11/27/2023] [Accepted: 05/09/2024] [Indexed: 05/18/2024] Open
Abstract
Telomeres are regions of repetitive DNA at the ends of linear chromosomes which protect chromosome ends from degradation. Telomere lengths have been extensively studied in the context of aging and disease, though most studies use average telomere lengths which are of limited utility. We present a method for identifying all 92 telomere alleles from long read sequencing data. Individual telomeres are identified using variant repeats proximal to telomere regions, which are unique across alleles. This high-throughput and high-resolution characterization of telomeres could be foundational to future studies investigating the roles of specific telomeres in aging and disease.
Collapse
|
20
|
Karimian K, Groot A, Huso V, Kahidi R, Tan KT, Sholes S, Keener R, McDyer JF, Alder JK, Li H, Rechtsteiner A, Greider CW. Human telomere length is chromosome end-specific and conserved across individuals. Science 2024; 384:533-539. [PMID: 38603523 DOI: 10.1126/science.ado0431] [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: 01/14/2024] [Accepted: 03/30/2024] [Indexed: 04/13/2024]
Abstract
Short telomeres cause age-related disease, and long telomeres contribute to cancer; however, the mechanisms regulating telomere length are unclear. We developed a nanopore-based method, which we call Telomere Profiling, to determine telomere length at nearly single-nucleotide resolution. Mapping telomere reads to chromosome ends showed chromosome end-specific length distributions that could differ by more than six kilobases. Examination of telomere lengths in 147 individuals revealed that certain chromosome ends were consistently longer or shorter. The same rank order was found in newborn cord blood, suggesting that telomere length is determined at birth and that chromosome end-specific telomere length differences are maintained as telomeres shorten with age. Telomere Profiling makes precision investigation of telomere length widely accessible for laboratory, clinical, and drug discovery efforts and will allow deeper insights into telomere biology.
Collapse
Affiliation(s)
- Kayarash Karimian
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aljona Groot
- Department of Molecular Cell and Developmental Biology, University of California, Santa Cruz, CA, USA
| | - Vienna Huso
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ramin Kahidi
- Health Sciences Program, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kar-Tong Tan
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Cancer Program, The Broad Institute, Cambridge, MA, USA
| | - Samantha Sholes
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Rebecca Keener
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - John F McDyer
- Pulmonary, Allergy, Critical Care, and Sleep Medicine Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jonathan K Alder
- Pulmonary, Allergy, Critical Care, and Sleep Medicine Division, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Heng Li
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Andreas Rechtsteiner
- Department of Molecular Cell and Developmental Biology, University of California, Santa Cruz, CA, USA
| | - Carol W Greider
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Molecular Cell and Developmental Biology, University of California, Santa Cruz, CA, USA
| |
Collapse
|
21
|
Guo X, Li J, Qi Y, Chen J, Jiang M, Zhu L, Liu Z, Wang H, Wang G, Wang X. Telomere length and micronuclei trajectories in APP/PS1 mouse model of Alzheimer's disease: Correlating with cognitive impairment and brain amyloidosis in a sexually dimorphic manner. Aging Cell 2024; 23:e14121. [PMID: 38450924 PMCID: PMC11113262 DOI: 10.1111/acel.14121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 12/31/2023] [Accepted: 02/08/2024] [Indexed: 03/08/2024] Open
Abstract
Although studies have demonstrated that genome instability is accumulated in patients with Alzheimer's disease (AD), the specific types of genome instability linked to AD pathogenesis remain poorly understood. Here, we report the first characterization of the age- and sex-related trajectories of telomere length (TL) and micronuclei in APP/PS1 mice model and wild-type (WT) controls (C57BL/6). TL was measured in brain (prefrontal cortex, cerebellum, pituitary gland, and hippocampus), colon and skin, and MN was measured in bone marrow in 6- to 14-month-old mice. Variation in TL was attributable to tissue type, age, genotype and, to a lesser extent, sex. Compared to WT, APP/PS1 had a significantly shorter baseline TL across all examined tissues. TL was inversely associated with age in both genotypes and TL shortening was accelerated in brain of APP/PS1. Age-related increase of micronuclei was observed in both genotypes but was accelerated in APP/PS1. We integrated TL and micronuclei data with data on cognition performance and brain amyloidosis. TL and micronuclei were linearly correlated with cognition performance or Aβ40 and Aβ42 levels in both genotypes but to a greater extent in APP/PS1. These associations in APP/PS1 mice were dominantly driven by females. Together, our findings provide foundational knowledge to infer the TL and micronuclei trajectories in APP/PS1 mice during disease progression, and strongly support that TL attrition and micronucleation are tightly associated with AD pathogenesis in a female-biased manner.
Collapse
Affiliation(s)
- Xihan Guo
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Jianfei Li
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Yanmei Qi
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Juanlin Chen
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Minyan Jiang
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Lina Zhu
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Zetong Liu
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Han Wang
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Gongwu Wang
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
| | - Xu Wang
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass EnergyYunnan Normal UniversityKunmingYunnanChina
- Yeda Institute of Gene and Cell TherapyTaizhouZhejiangChina
| |
Collapse
|
22
|
Ju W, Zhao Y, Yu Y, Zhao S, Xiang S, Lian F. Mechanisms of mitochondrial dysfunction in ovarian aging and potential interventions. Front Endocrinol (Lausanne) 2024; 15:1361289. [PMID: 38694941 PMCID: PMC11061492 DOI: 10.3389/fendo.2024.1361289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 03/22/2024] [Indexed: 05/04/2024] Open
Abstract
Mitochondria plays an essential role in regulating cellular metabolic homeostasis, proliferation/differentiation, and cell death. Mitochondrial dysfunction is implicated in many age-related pathologies. Evidence supports that the dysfunction of mitochondria and the decline of mitochondrial DNA copy number negatively affect ovarian aging. However, the mechanism of ovarian aging is still unclear. Treatment methods, including antioxidant applications, mitochondrial transplantation, emerging biomaterials, and advanced technologies, are being used to improve mitochondrial function and restore oocyte quality. This article reviews key evidence and research updates on mitochondrial damage in the pathogenesis of ovarian aging, emphasizing that mitochondrial damage may accelerate and lead to cellular senescence and ovarian aging, as well as exploring potential methods for using mitochondrial mechanisms to slow down aging and improve oocyte quality.
Collapse
Affiliation(s)
- Wenhan Ju
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuewen Zhao
- CReATe Fertility Centre, Toronto, ON, Canada
| | - Yi Yu
- Department of Reproduction and Genetics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuai Zhao
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shan Xiang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fang Lian
- Department of Reproduction and Genetics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
23
|
Blanco MB, Smith DL, Greene LK, Yoder AD, Ehmke EE, Lin J, Klopfer PH. Telomere dynamics during hibernation in a tropical primate. J Comp Physiol B 2024; 194:213-219. [PMID: 38466418 DOI: 10.1007/s00360-024-01541-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/13/2024] [Accepted: 01/23/2024] [Indexed: 03/13/2024]
Abstract
Hibernation is a widespread metabolic strategy among mammals for surviving periods of food scarcity. During hibernation, animals naturally alternate between metabolically depressed torpor bouts and energetically expensive arousals without ill effects. As a result, hibernators are promising models for investigating mechanisms that buffer against cellular stress, including telomere protection and restoration. In non-hibernators, telomeres, the protective structural ends of chromosomes, shorten with age and metabolic stress. In temperate hibernators, however, telomere shortening and elongation can occur in response to changing environmental conditions and associated metabolic state. We investigate telomere dynamics in a tropical hibernating primate, the fat-tailed dwarf lemur (Cheirogaleus medius). In captivity, these lemurs can hibernate when maintained under cold temperatures (11-15 °C) with limited food provisioning. We study telomere dynamics in eight fat-tailed dwarf lemurs at the Duke Lemur Center, USA, from samples collected before, during, and after the hibernation season and assayed via qPCR. Contrary to our predictions, we found that telomeres were maintained or even lengthened during hibernation, but shortened immediately thereafter. During hibernation, telomere lengthening was negatively correlated with time in euthermia. Although preliminary in scope, our findings suggest that there may be a preemptive, compensatory mechanism to maintain telomere integrity in dwarf lemurs during hibernation. Nevertheless, telomere shortening immediately afterward may broadly result in similar outcomes across seasons. Future studies could profitably investigate the mechanisms that offset telomere shortening within and outside of the hibernation season and whether those mechanisms are modulated by energy surplus or crises.
Collapse
Affiliation(s)
- M B Blanco
- Duke Lemur Center, Durham, NC, 27705, USA.
- Department of Biology, Duke University, Durham, NC, 27708, USA.
| | - D L Smith
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, 94143, USA
| | - L K Greene
- Duke Lemur Center, Durham, NC, 27705, USA
- Department of Biology, Duke University, Durham, NC, 27708, USA
| | - A D Yoder
- Department of Biology, Duke University, Durham, NC, 27708, USA
| | - E E Ehmke
- Duke Lemur Center, Durham, NC, 27705, USA
| | - J Lin
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, 94143, USA
| | - P H Klopfer
- Department of Biology, Duke University, Durham, NC, 27708, USA
| |
Collapse
|
24
|
Córdova-Oriz I, Polonio AM, Cuadrado-Torroglosa I, Chico-Sordo L, Medrano M, García-Velasco JA, Varela E. Chromosome ends and the theory of marginotomy: implications for reproduction. Biogerontology 2024; 25:227-248. [PMID: 37943366 DOI: 10.1007/s10522-023-10071-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/21/2023] [Indexed: 11/10/2023]
Abstract
Telomeres are the protective structures located at the ends of linear chromosomes. They were first described in the 1930s, but their biology remained unexplored until the early 70s, when Alexey M. Olovnikov, a theoretical biologist, suggested that telomeres cannot be fully copied during DNA replication. He proposed a theory that linked this phenomenon with the limit of cell proliferation capacity and the "duration of life" (theory of marginotomy), and suggested a potential of telomere lenghthening for the prevention of aging (anti-marginotomy). The impact of proliferative telomere shortening on life expectancy was later confirmed. In humans, telomere shortening is counteracted by telomerase, an enzyme that is undetectable in most adult somatic cells, but present in cancer cells and adult and embryonic stem and germ cells. Although telomere length dynamics are different in male and female gametes during gametogenesis, telomere lengths are reset at the blastocyst stage, setting the initial length of the species. The role of the telomere pathway in reproduction has been explored for years, mainly because of increased infertility resulting from delayed childbearing. Short telomere length in ovarian somatic cells is associated to decreased fertility and higher aneuploidy rates in embryos. Consequently, there is a growing interest in telomere lengthening strategies, aimed at improving fertility. It has also been observed that lifestyle factors can affect telomere length and improve fertility outcomes. In this review, we discuss the implications of telomere theory in fertility, especially in oocytes, spermatozoa, and embryos, as well as therapies to enhance reproductive success.
Collapse
Affiliation(s)
- Isabel Córdova-Oriz
- IVIRMA Global Research Alliance, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Alba M Polonio
- IVIRMA Global Research Alliance, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Isabel Cuadrado-Torroglosa
- IVIRMA Global Research Alliance, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Lucía Chico-Sordo
- IVIRMA Global Research Alliance, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Marta Medrano
- IVIRMA Global Research Alliance, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Juan A García-Velasco
- IVIRMA Global Research Alliance, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
- IVIRMA Global Research Alliance, IVIRMA Madrid, Madrid, Spain
- Department of Medical Specialties and Public Health, Edificio Departamental II, Rey Juan Carlos University, Av. de Atenas, s/n, 28922, Alcorcón, Madrid, Spain
| | - Elisa Varela
- IVIRMA Global Research Alliance, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain.
- Department of Medical Specialties and Public Health, Edificio Departamental II, Rey Juan Carlos University, Av. de Atenas, s/n, 28922, Alcorcón, Madrid, Spain.
| |
Collapse
|
25
|
Savage SA. Telomere length and cancer risk: finding Goldilocks. Biogerontology 2024; 25:265-278. [PMID: 38109000 DOI: 10.1007/s10522-023-10080-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023]
Abstract
Telomeres are the nucleoprotein complex at chromosome ends essential in genomic stability. Baseline telomere length (TL) is determined by rare and common germline genetic variants but shortens with age and is susceptible to certain environmental exposures. Cellular senescence or apoptosis are normally triggered when telomeres reach a critically short length, but cancer cells overcome these protective mechanisms and continue to divide despite chromosomal instability. Rare germline variants in telomere maintenance genes cause exceedingly short telomeres for age (< 1st percentile) and the telomere biology disorders, which are associated with elevated risks of bone marrow failure, myelodysplastic syndrome, acute myeloid leukemia, and squamous cell carcinoma of the head/neck and anogenital regions. Long telomeres due to rare germline variants in the same or different telomere maintenance genes are associated with elevated risks of other cancers, such as chronic lymphocytic leukemia or sarcoma. Early epidemiology studies of TL in the general population lacked reproducibility but new methods, including creation of a TL polygenic score using common variants, have found longer telomeres associated with excess risks of renal cell carcinoma, glioma, lung cancer, and others. It has become clear that when it comes to TL and cancer etiology, not too short, not too long, but "just right" telomeres are important in minimizing cancer risk.
Collapse
Affiliation(s)
- Sharon A Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Drive, 6E456, Bethesda, MD, 20892-6772, USA.
| |
Collapse
|
26
|
Gellert-Kristensen H, Bojesen SE, Tybjærg Hansen A, Stender S. Telomere length and risk of cirrhosis, hepatocellular carcinoma, and cholangiocarcinoma in 63,272 individuals from the general population. Hepatology 2024; 79:857-868. [PMID: 37732945 DOI: 10.1097/hep.0000000000000608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/12/2023] [Indexed: 09/22/2023]
Abstract
BACKGROUND AND AIMS Inherited short telomeres are associated with a risk of liver disease, whereas longer telomeres predispose to cancer. The association between telomere length and risk of HCC and cholangiocarcinoma remains unknown. APPROACH AND RESULTS We measured leukocyte telomere length using multiplex PCR in 63,272 individuals from the Danish general population. Telomere length and plasma ALT concentration were not associated (β = 4 ×10 -6 , p -value = 0.06) in a linear regression model, without any signs of a nonlinear relationship. We tested the association between telomere length and risk of cirrhosis, HCC, and cholangiocarcinoma using Cox regression. During a median follow-up of 11 years, 241, 76, and 112 individuals developed cirrhosis, HCC, and cholangiocarcinoma, respectively. Telomere length and risk of cirrhosis were inversely and linearly associated ( p -value = 0.004, p for nonlinearity = 0.27). Individuals with telomeres in the shortest vs. longest quartile had a 2.25-fold higher risk of cirrhosis. Telomere length and risk of HCC were nonlinearly associated ( p -value = 0.009, p -value for nonlinearity = 0.01). This relationship resembled an inverted J-shape, with the highest risk observed in individuals with short telomeres. Individuals with telomeres in the shortest versus longest quartile had a 2.29-fold higher risk of HCC. Telomere length was inversely and linearly associated with the risk of cholangiocarcinoma. Individuals with telomeres in the shortest versus longest quartile had a 1.86-fold higher risk of cholangiocarcinoma. CONCLUSIONS Shorter telomere length is associated with a higher risk of cirrhosis, HCC, and cholangiocarcinoma.
Collapse
Affiliation(s)
- Helene Gellert-Kristensen
- Department of Clinical Biochemistry, Rigshospitalet, Denmark
- Copenhagen University Hospitals and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Stig E Bojesen
- Copenhagen University Hospitals and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Denmark
| | - Anne Tybjærg Hansen
- Department of Clinical Biochemistry, Rigshospitalet, Denmark
- Copenhagen University Hospitals and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| | - Stefan Stender
- Department of Clinical Biochemistry, Rigshospitalet, Denmark
- Copenhagen University Hospitals and Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
27
|
Ojeda-Rodriguez A, Rangel-Zuñiga OA, Arenas-de Larriva AP, Gutierrez-Mariscal FM, Torres-Peña JD, Romero-Cabrera JL, Podadera-Herreros A, García-Fernandez H, Porras-Pérez E, Luque RM, Kales SN, Perez-Martinez P, Delgado-Lista J, Yubero-Serrano EM, Lopez-Miranda J. Telomere length as biomarker of nutritional therapy for prevention of type 2 diabetes mellitus development in patients with coronary heart disease: CORDIOPREV randomised controlled trial. Cardiovasc Diabetol 2024; 23:98. [PMID: 38493287 PMCID: PMC10944592 DOI: 10.1186/s12933-024-02175-5] [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: 12/19/2023] [Accepted: 02/20/2024] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Telomere Length (TL), a marker of cellular aging, holds promise as a biomarker to elucidate the molecular mechanism of diabetes. This study aimed to investigate whether shorter telomeres are associated with a higher risk of type 2 diabetes mellitus (T2DM) incidence in patients with coronary heart disease; and to determine whether the most suitable dietary patterns, particularly a Mediterranean diet or a low-fat diet, can mitigate the development of diabetes in these patients after a follow-up period of five years. METHODS The CORonary Diet Intervention with Olive oil and cardiovascular PREVention study (CORDIOPREV study) was a single-centre, randomised clinical trial done at the Reina Sofia University Hospital in Córdoba, Spain. Patients with established coronary heart disease (aged 20-75 years) were randomly assigned in a 1:1 ratio by the Andalusian School of Public Health to receive two healthy diets. Clinical investigators were masked to treatment assignment; participants were not. Quantitative-PCR was used to assess TL measurements. FINDINGS 1002 patients (59.5 ± 8.7 years and 82.5% men) were enrolled into Mediterranean diet (n = 502) or a low-fat diet (n = 500) groups. In this analysis, we included all 462 patients who did not have T2DM at baseline. Among them, 107 patients developed T2DM after a median of 60 months. Cox regression analyses showed that patients at risk of short telomeres (TL < percentile 20th) are more likely to experience T2DM than those at no risk of short telomeres (HR 1.65, p-value 0.023). In terms of diet, patients at high risk of short telomeres had a higher risk of T2DM incidence after consuming a low-fat diet compared to patients at no risk of short telomeres (HR 2.43, 95CI% 1.26 to 4.69, p-value 0.008), while no differences were observed in the Mediterranean diet group. CONCLUSION Patients with shorter TL presented a higher risk of developing T2DM. This association could be mitigated with a specific dietary pattern, in our case a Mediterranean diet, to prevent T2DM in patients with coronary heart disease. TRIAL REGISTRATION Clinicaltrials.gov number NCT00924937.
Collapse
Affiliation(s)
- Ana Ojeda-Rodriguez
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, Cordoba, 14004, Spain
- Department of Medical and Surgical Science, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Oriol A Rangel-Zuñiga
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, Cordoba, 14004, Spain
- Department of Medical and Surgical Science, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Antonio P Arenas-de Larriva
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, Cordoba, 14004, Spain
- Department of Medical and Surgical Science, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Francisco M Gutierrez-Mariscal
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, Cordoba, 14004, Spain
- Department of Medical and Surgical Science, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Jose D Torres-Peña
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, Cordoba, 14004, Spain
- Department of Medical and Surgical Science, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Juan L Romero-Cabrera
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, Cordoba, 14004, Spain
- Department of Medical and Surgical Science, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Alicia Podadera-Herreros
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, Cordoba, 14004, Spain
- Department of Medical and Surgical Science, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Helena García-Fernandez
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, Cordoba, 14004, Spain
- Department of Medical and Surgical Science, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Esther Porras-Pérez
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, Cordoba, 14004, Spain
- Department of Medical and Surgical Science, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Raul M Luque
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, 14004, Spain
| | - Stefanos N Kales
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Occupational Medicine, Cambridge Health Alliance, Harvard Medical School, Cambridge, MA, USA
| | - Pablo Perez-Martinez
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, Cordoba, 14004, Spain
- Department of Medical and Surgical Science, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, Cordoba, 14004, Spain
- Department of Medical and Surgical Science, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Elena M Yubero-Serrano
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, Cordoba, 14004, Spain
- Department of Medical and Surgical Science, University of Cordoba, Cordoba, 14004, Spain
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, Cordoba, 14004, Spain.
- Department of Medical and Surgical Science, University of Cordoba, Cordoba, 14004, Spain.
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, Cordoba, 14004, Spain.
- CIBER Fisiopatologia de la Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, Madrid, 28029, Spain.
| |
Collapse
|
28
|
Kraft BD, Verhulst S, Lai TP, Sullenger BA, Wang Y, Rountree W, Chen L, Woods CW, Denny TN, Aviv A. T-cell count and T-cell telomere length in patients with severe COVID-19. Front Immunol 2024; 15:1356638. [PMID: 38550590 PMCID: PMC10972845 DOI: 10.3389/fimmu.2024.1356638] [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: 12/15/2023] [Accepted: 02/19/2024] [Indexed: 04/02/2024] Open
Abstract
Lymphocyte telomere length (TL) is highly variable and shortens with age. Short telomeres may impede TL-dependent T-cell clonal expansion with viral infection. As SARS-CoV-2 infection can induce prolonged and severe T-cell lymphopenia, infected adults, and particularly older adults with short telomeres, may display severe T-cell lymphopenia. To examine the relationship between T-cell TL parameters and T-cell counts, we studied 40 patients hospitalized with severe COVID-19. T-cells were isolated from lymphocytes, counted using flow cytometry, and their TL parameters were measured using the Telomere Shortest Length Assay. The cohort (median age = 62 years, 27% female) was racially and ethnically diverse (33% White, 35% Black, and 33% Other). On intensive care unit study day 1, T-cell count (mean=1.03 x109/L) was inversely related to age (p=0.007) and higher in females than males (p=0.025). Mean TL was 3.88 kilobases (kb), and 45.3% of telomeres were shorter than 3 kb. Using multiple regression analysis and adjusting for age and sex, T-cell count decreased with increased proportion of T-cell telomeres shorter than 3 kb (p=0.033) and increased with mean TL (p=0.052). Our findings suggest an association between the buildup of short telomeres within T-cells and explain in part reduced peripheral blood T-cell counts in patients with severe COVID-19. Shortened T-cell telomeres may be a risk factor for COVID-19-associated T-cell lymphopenia.
Collapse
Affiliation(s)
- Bryan D. Kraft
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Durham, NC, United States
| | - Simon Verhulst
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, Netherlands
| | - Tsung-Po Lai
- Center of Human Development and Aging, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Bruce A. Sullenger
- Department of Surgery, Duke University Medical Center, Durham, NC, United States
| | - Yunfei Wang
- Duke Human Vaccine Institute, Durham, NC, United States
| | - Wes Rountree
- Duke Human Vaccine Institute, Durham, NC, United States
| | - Lingye Chen
- Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University Medical Center, Durham, NC, United States
| | - Christopher W. Woods
- Center for Infectious Disease Diagnostics and Innovation, Duke University Medical Center, Durham, NC, United States
- Department of Medicine, Durham Veterans Affairs Health Care System, Durham, NC, United States
| | | | - Abraham Aviv
- Center of Human Development and Aging, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Department of Pediatrics, New Jersey Medical School, Rutgers, The State University of New Jersey, Newark, NJ, United States
| |
Collapse
|
29
|
Lunghi E, Bilandžija H. Telomere length and dynamics in Astyanax mexicanus cave and surface morphs. PeerJ 2024; 12:e16957. [PMID: 38435987 PMCID: PMC10908260 DOI: 10.7717/peerj.16957] [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: 09/13/2023] [Accepted: 01/25/2024] [Indexed: 03/05/2024] Open
Abstract
Background Telomeres are non-coding DNA repeats at the chromosome ends and their shortening is considered one of the major causes of aging. However, they also serve as a biomarker of environmental exposures and their length and attrition is affected by various stressors. In this study, we examined the average telomere length in Astyanax mexicanus, a species that has both surface-dwelling and cave-adapted populations. The cave morph descended from surface ancestors and adapted to a markedly different environment characterized by specific biotic and abiotic stressors, many of which are known to affect telomere length. Our objective was to explore whether telomere length differs between the two morphs and whether it serves as a biological marker of aging or correlates with the diverse environments the morphs are exposed to. Methods We compared telomere length and shortening between laboratory-reared Pachón cavefish and Rio Choy surface fish of A. mexicanus across different tissues and ages. Results Astyanax mexicanus surface fish exhibited longer average telomere length compared to cavefish. In addition, we did not observe telomere attrition in either cave or surface form as a result of aging in adults up to 9 years old, suggesting that efficient mechanisms prevent telomere-mediated senescence in laboratory stocks of this species, at least within this time frame. Our results suggest that telomere length in Astyanax may be considered a biomarker of environmental exposures. Cavefish may have evolved shorter and energetically less costly telomeres due to the absence of potential stressors known to affect surface species, such as predator pressure and ultra-violet radiation. This study provides the first insights into telomere dynamics in Astyanax morphs and suggests that shorter telomeres may have evolved as an adaptation to caves.
Collapse
Affiliation(s)
- Enrico Lunghi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy
| | - Helena Bilandžija
- Division of Molecular Biology, Ruder Bošković Institute, Zagreb, Croatia
| |
Collapse
|
30
|
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
|
31
|
Karimian K, Groot A, Huso V, Kahidi R, Tan KT, Sholes S, Keener R, McDyer JF, Alder JK, Li H, Rechtsteiner A, Greider CW. Human telomere length is chromosome specific and conserved across individuals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.21.572870. [PMID: 38187739 PMCID: PMC10769321 DOI: 10.1101/2023.12.21.572870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Short telomeres cause age-related disease and long telomeres predispose to cancer; however, the mechanisms regulating telomere length are unclear. To probe these mechanisms, we developed a nanopore sequencing method, Telomere Profiling, that is easy to implement, precise, and cost effective with broad applications in research and the clinic. We sequenced telomeres from individuals with short telomere syndromes and found similar telomere lengths to the clinical FlowFISH assay. We mapped telomere reads to specific chromosome end and identified both chromosome end-specific and haplotype-specific telomere length distributions. In the T2T HG002 genome, where the average telomere length is 5kb, we found a remarkable 6kb difference in lengths between some telomeres. Further, we found that specific chromosome ends were consistently shorter or longer than the average length across 147 individuals. The presence of conserved chromosome end-specific telomere lengths suggests there are new paradigms in telomere biology that are yet to be explored. Understanding the mechanisms regulating length will allow deeper insights into telomere biology that can lead to new approaches to disease.
Collapse
Affiliation(s)
- Kayarash Karimian
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Aljona Groot
- Department of Molecular Cell and Developmental Biology, University of California, Santa Cruz
| | - Vienna Huso
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | | | - Kar-Tong Tan
- Harvard Medical School, Department of Genetics, Boston, MA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute, Cancer Program, Cambridge, MA
| | - Samantha Sholes
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Biochemistry, Cellular and Molecular Biology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Present address Merck & Co., 770 Sumneytown Pike, West Point, PA 19486
| | - Rebecca Keener
- Department of Biomedical Engineering, Johns Hopkins University
| | - John F. McDyer
- Pulmonary, Allergy, Critical Care, and Sleep Medicine Division, Department of Medicine, University of Pittsburgh
| | - Jonathan K. Alder
- Pulmonary, Allergy, Critical Care, and Sleep Medicine Division, Department of Medicine, University of Pittsburgh
| | - Heng Li
- Dana-Farber Cancer Institute, Department of Data Sciences, Boston, MA
- Harvard Medical School, Department of Biomedical Informatics, Boston, MA
| | - Andreas Rechtsteiner
- Department of Molecular Cell and Developmental Biology, University of California, Santa Cruz
| | - Carol W. Greider
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Molecular Cell and Developmental Biology, University of California, Santa Cruz
| |
Collapse
|
32
|
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: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [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
|
33
|
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: 0] [Impact Index Per Article: 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
|
34
|
Schmit MM, Baxley RM, Wang L, Hinderlie P, Kaufman M, Simon E, Raju A, Miller JS, Bielinsky AK. A critical threshold of MCM10 is required to maintain genome stability during differentiation of induced pluripotent stem cells into natural killer cells. Open Biol 2024; 14:230407. [PMID: 38262603 PMCID: PMC10805602 DOI: 10.1098/rsob.230407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/23/2023] [Indexed: 01/25/2024] Open
Abstract
Natural killer (NK) cell deficiency (NKD) is a rare disease in which NK cell function is reduced, leaving affected individuals susceptible to repeated viral infections and cancer. Recently, a patient with NKD was identified carrying compound heterozygous variants of MCM10 (minichromosome maintenance protein 10), an essential gene required for DNA replication, that caused a significant decrease in the amount of functional MCM10. NKD in this patient presented as loss of functionally mature late-stage NK cells. To understand how MCM10 deficiency affects NK cell development, we generated MCM10 heterozygous (MCM10+/-) induced pluripotent stem cell (iPSC) lines. Analyses of these cell lines demonstrated that MCM10 was haploinsufficient, similar to results in other human cell lines. Reduced levels of MCM10 in mutant iPSCs was associated with impaired clonogenic survival and increased genomic instability, including micronuclei formation and telomere erosion. The severity of these phenotypes correlated with the extent of MCM10 depletion. Significantly, MCM10+/- iPSCs displayed defects in NK cell differentiation, exhibiting reduced yields of hematopoietic stem cells (HSCs). Although MCM10+/- HSCs were able to give rise to lymphoid progenitors, these did not generate mature NK cells. The lack of mature NK cells coincided with telomere erosion, suggesting that NKD caused by these MCM10 variants arose from the accumulation of genomic instability including degradation of chromosome ends.
Collapse
Affiliation(s)
- Megan M. Schmit
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Ryan M. Baxley
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Liangjun Wang
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Peter Hinderlie
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Marissa Kaufman
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Emily Simon
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Anjali Raju
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey S. Miller
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis, MN, USA
| | - Anja-Katrin Bielinsky
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
35
|
Sanchez SE, Gu J, Golla A, Martin A, Shomali W, Hockemeyer D, Savage SA, Artandi SE. Digital telomere measurement by long-read sequencing distinguishes healthy aging from disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.29.569263. [PMID: 38077053 PMCID: PMC10705489 DOI: 10.1101/2023.11.29.569263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2023]
Abstract
Telomere length is an important biomarker of organismal aging and cellular replicative potential, but existing measurement methods are limited in resolution and accuracy. Here, we deploy digital telomere measurement by nanopore sequencing to understand how distributions of human telomere length change with age and disease. We measure telomere attrition and de novo elongation with unprecedented resolution in genetically defined populations of human cells, in blood cells from healthy donors and in blood cells from patients with genetic defects in telomere maintenance. We find that human aging is accompanied by a progressive loss of long telomeres and an accumulation of shorter telomeres. In patients with defects in telomere maintenance, the accumulation of short telomeres is more pronounced and correlates with phenotypic severity. We apply machine learning to train a binary classification model that distinguishes healthy individuals from those with telomere biology disorders. This sequencing and bioinformatic pipeline will advance our understanding of telomere maintenance mechanisms and the use of telomere length as a clinical biomarker of aging and disease.
Collapse
Affiliation(s)
- Santiago E. Sanchez
- Stanford Cancer Institute, Stanford University School of Medicine; Stanford, CA, USA
- Cancer Biology Program, Stanford University School of Medicine; Stanford, CA, USA
- Medical Scientist Training Program, Stanford University; Stanford CA, USA
| | - Jessica Gu
- Stanford Cancer Institute, Stanford University School of Medicine; Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine; Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine; Stanford, CA, USA
| | - Anudeep Golla
- Stanford Cancer Institute, Stanford University School of Medicine; Stanford, CA, USA
| | - Annika Martin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA
| | - William Shomali
- Department of Medicine, Stanford University School of Medicine; Stanford, CA, USA
| | - Dirk Hockemeyer
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA
- Chan Zuckerberg Biohub, San Francisco, CA
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA
| | - Sharon A. Savage
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Steven E. Artandi
- Stanford Cancer Institute, Stanford University School of Medicine; Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine; Stanford, CA, USA
- Department of Biochemistry, Stanford University School of Medicine; Stanford, CA, USA
| |
Collapse
|
36
|
Raj HA, Lai TP, Niewisch MR, Giri N, Wang Y, Spellman SR, Aviv A, Gadalla SM, Savage SA. The distribution and accumulation of the shortest telomeres in telomere biology disorders. Br J Haematol 2023; 203:820-828. [PMID: 37354000 PMCID: PMC10748793 DOI: 10.1111/bjh.18945] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/19/2023] [Accepted: 06/02/2023] [Indexed: 06/25/2023]
Abstract
Individuals with telomere biology disorders (TBDs) have very short telomeres, high risk of bone marrow failure (BMF), and reduced survival. Using data from TBD patients, a mean leukocyte Southern blot telomere length (TL) of 5 kilobases (kb) was estimated as the 'telomere brink' at which human survival is markedly reduced. However, the shortest telomere, not the mean TL, signals replicative senescence. We used the Telomere Shortest Length Assay (TeSLA) to tally TL of all 46 chromosomes in blood-derived DNA and examined its relationship with TBDs. Patients (n = 18) had much shorter mean TL (TeSmTL) (2.54 ± 0.41 kb vs. 4.48 ± 0.52 kb, p < 0.0001) and more telomeres <3 kb than controls (n = 22) (70.43 ± 8.76% vs. 33.05 ± 6.93%, p < 0.0001). The proportion of ultrashort telomeres (<1.6 kb) was also higher in patients than controls (39.29 ± 10.69% vs. 10.40 ± 4.09%, p < 0.0001). TeS <1.6 kb was associated with severe (n = 11) compared with non-severe (n = 7) BMF (p = 0.027). Patients with multi-organ manifestations (n = 10) had more telomeres <1.6 kb than those with one affected organ system (n = 8) (p = 0.029). Findings suggest that TBD clinical manifestations are associated with a disproportionately higher number of haematopoietic cell telomeres reaching a telomere brink, whose length at the single telomere level is yet to be determined.
Collapse
Affiliation(s)
- Hannah A. Raj
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | - Tsung-Po Lai
- Center of Human Development and Aging, Rutgers University of New Jersey, New Jersey Medical School, Newark, NJ
| | - Marena R. Niewisch
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
- Department of Pediatric Hematology and Oncology, Hannover Medical School, Hannover, Germany
| | - Neelam Giri
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | - Youjin Wang
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | - Stephen R. Spellman
- Center for International Blood and Marrow Transplant Research, National Marrow Donor Program/Be The Match, Minneapolis, MN
| | - Abraham Aviv
- Center of Human Development and Aging, Rutgers University of New Jersey, New Jersey Medical School, Newark, NJ
| | - Shahinaz M. Gadalla
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | - Sharon A. Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| |
Collapse
|
37
|
Guo M, Songyang Z, Xiong Y. ChArmTelo Enables Large-Scale Chromosome Arm-Level Telomere Analysis across Human Populations and in Cancer Patients. SMALL METHODS 2023; 7:e2300385. [PMID: 37526331 DOI: 10.1002/smtd.202300385] [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: 03/23/2023] [Revised: 06/29/2023] [Indexed: 08/02/2023]
Abstract
Telomeres are structures protecting chromosome ends. However, a scalable and cost-effective method to investigate chromosome arm-level (ChArm) telomeres (Telos) in large-scale projects is still lacking, hindering intensive investigation of high-resolution telomeres across cancers and other diseases. Here, ChArmTelo, the first computational toolbox to analyze telomeres at chromosome arm level in human and other animal species, using 10X linked-read and similar technologies, is presented. ChArmTelo currently consists of two algorithms, TeloEM and TeloKnow, for arm-level telomere length (TL) analysis. The algorithms are demonstrated by comprehensive analysis of chromosome arm-level telomere lengths (chArmTLs) in nearly 400 whole genome sequencing samples (WGS) from human populations and animals, including healthy and cancer samples. Notably, considerable performance improvement contributed by using the latest complete telomere-to-telomere reference genome (CHM13v2), compared to hg38, is shown. ChArmTelo reveals population-specific chArmTL differences and liver cancer signatures of chArmTLs and that DNA replication origin disruption may contribute to cancer by affecting TLs. Importantly, ChArmTelo can be readily applied to tens of thousands of cancer and healthy samples with published WGS data.
Collapse
Affiliation(s)
- Mengbiao Guo
- Key Laboratory of Gene Engineering of the Ministry of Education, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhou Songyang
- Key Laboratory of Gene Engineering of the Ministry of Education, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yuanyan Xiong
- Key Laboratory of Gene Engineering of the Ministry of Education, Institute of Healthy Aging Research, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| |
Collapse
|
38
|
Zhang N, Baker EC, Welsh TH, Riley DG. Telomere Dynamics in Livestock. BIOLOGY 2023; 12:1389. [PMID: 37997988 PMCID: PMC10669808 DOI: 10.3390/biology12111389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/25/2023]
Abstract
Telomeres are repeated sequences of nucleotides at the end of chromosomes. They deteriorate across mitotic divisions of a cell. In Homo sapiens this process of lifetime reduction has been shown to correspond with aspects of organismal aging and exposure to stress or other insults. The early impetus to characterize telomere dynamics in livestock related to the concern that aged donor DNA would result in earlier cell senescence and overall aging in cloned animals. Telomere length investigations in dairy cows included breed effects, estimates of additive genetic control (heritability 0.12 to 0.46), and effects of external stressors on telomere degradation across animal life. Evaluation of telomeres with respect to aging has also been conducted in pigs and horses, and there are fewer reports of telomere biology in beef cattle, sheep, and goats. There were minimal associations of telomere length with animal productivity measures. Most, but not all, work in livestock has documented an inverse relationship between peripheral blood cell telomere length and age; that is, a longer telomere length was associated with younger age. Because livestock longevity affects productivity and profitability, the role of tissue-specific telomere attrition in aging may present alternative improvement strategies for genetic improvement while also providing translational biomedical knowledge.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (N.Z.); (T.H.W.J.)
| | - Emilie C. Baker
- Department of Agricultural Sciences, West Texas A&M University, Canyon, TX 79016, USA;
| | - Thomas H. Welsh
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (N.Z.); (T.H.W.J.)
- Texas A&M AgriLife Research, College Station, TX 77843, USA
| | - David G. Riley
- Department of Animal Science, Texas A&M University, College Station, TX 77843, USA; (N.Z.); (T.H.W.J.)
- Texas A&M AgriLife Research, College Station, TX 77843, USA
| |
Collapse
|
39
|
Smoom R, May CL, Ortiz V, Tigue M, Kolev HM, Rowe M, Reizel Y, Morgan A, Egyes N, Lichtental D, Skordalakes E, Kaestner KH, Tzfati Y. Telomouse-a mouse model with human-length telomeres generated by a single amino acid change in RTEL1. Nat Commun 2023; 14:6708. [PMID: 37872177 PMCID: PMC10593777 DOI: 10.1038/s41467-023-42534-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/14/2023] [Indexed: 10/25/2023] Open
Abstract
Telomeres, the ends of eukaryotic chromosomes, protect genome integrity and enable cell proliferation. Maintaining optimal telomere length in the germline and throughout life limits the risk of cancer and enables healthy aging. Telomeres in the house mouse, Mus musculus, are about five times longer than human telomeres, limiting the use of this common laboratory animal for studying the contribution of telomere biology to aging and cancer. We identified a key amino acid variation in the helicase RTEL1, naturally occurring in the short-telomere mouse species M. spretus. Introducing this variation into M. musculus is sufficient to reduce the telomere length set point in the germline and generate mice with human-length telomeres. While these mice are fertile and appear healthy, the regenerative capacity of their colonic epithelium is compromised. The engineered Telomouse reported here demonstrates a dominant role of RTEL1 in telomere length regulation and provides a unique model for aging and cancer.
Collapse
Affiliation(s)
- Riham Smoom
- Department of Genetics, The Silberman Institute of Life Sciences, Safra Campus, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Catherine Lee May
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Vivian Ortiz
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mark Tigue
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Hannah M Kolev
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Melissa Rowe
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yitzhak Reizel
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Faculty of Biotechnology and Food Engineering, Technion, Haifa, 3200003, Israel
| | - Ashleigh Morgan
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Nachshon Egyes
- Department of Genetics, The Silberman Institute of Life Sciences, Safra Campus, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Dan Lichtental
- Department of Genetics, The Silberman Institute of Life Sciences, Safra Campus, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel
| | - Emmanuel Skordalakes
- Department of Pharmacology and Toxicology, Massey Cancer Center, Virginia Commonwealth University, 401 College St, Richmond, VA, 23298, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Yehuda Tzfati
- Department of Genetics, The Silberman Institute of Life Sciences, Safra Campus, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel.
| |
Collapse
|
40
|
Lyons CE, Razzoli M, Bartolomucci A. The impact of life stress on hallmarks of aging and accelerated senescence: Connections in sickness and in health. Neurosci Biobehav Rev 2023; 153:105359. [PMID: 37586578 PMCID: PMC10592082 DOI: 10.1016/j.neubiorev.2023.105359] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 07/03/2023] [Accepted: 08/10/2023] [Indexed: 08/18/2023]
Abstract
Chronic stress is a risk factor for numerous aging-related diseases and has been shown to shorten lifespan in humans and other social mammals. Yet how life stress causes such a vast range of diseases is still largely unclear. In recent years, the impact of stress on health and aging has been increasingly associated with the dysregulation of the so-called hallmarks of aging. These are basic biological mechanisms that influence intrinsic cellular functions and whose alteration can lead to accelerated aging. Here, we review correlational and experimental literature (primarily focusing on evidence from humans and murine models) on the contribution of life stress - particularly stress derived from adverse social environments - to trigger hallmarks of aging, including cellular senescence, sterile inflammation, telomere shortening, production of reactive oxygen species, DNA damage, and epigenetic changes. We also evaluate the validity of stress-induced senescence and accelerated aging as an etiopathological proposition. Finally, we highlight current gaps of knowledge and future directions for the field, and discuss perspectives for translational geroscience.
Collapse
Affiliation(s)
- Carey E Lyons
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA; Graduate Program in Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Maria Razzoli
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA
| | - Alessandro Bartolomucci
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, MN, USA; Department of Medicine and Surgery, University of Parma, Parma, Italy.
| |
Collapse
|
41
|
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: 1] [Impact Index Per Article: 1.0] [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
|
42
|
Sameni S, Viswanathan R, Ng GYQ, Martinez-Lopez W, Hande MP. Telomerase Inhibition by MST-312 Sensitizes Breast Cancer Cells to the Anti-cancer Properties of Plumbagin. Genome Integr 2023; 14:e20230002. [PMID: 38765717 PMCID: PMC11102071 DOI: 10.14293/genint.14.1.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
Breast cancer is the most common cause of malignancy and the second most common cause of death due to cancer in women. This heterogeneous disease is currently broadly classified as estrogen receptor (ER), progesterone receptor (PR) positive luminal tumors, human epidermal growth factor receptor 2 (HER2) amplified tumors and triple-negative breast cancers (TNBC). Phytochemicals are proven to be promising anti-cancer chemotherapeutics agents with minimal cytotoxic effects on normal cells. Plumbagin (5-hydroxy-2-methyl-1, 4-naphthoquinone) is a phytochemical derived from the roots of Plumbago zeylanica and it is known to possess anti-cancer properties similar to other compounds of naphthoquinones. In about 90% of cancer cells, the telomerase enzyme activity is revived to add telomeric repeats to evade apoptosis. In this study, a combinatorial approach of combining the anti-cancer compound plumbagin to induce genotoxicity and a potent telomerase inhibitor, MST-312 (synthetic derivative of tea catechins), was used to determine the combinational treatment-induced lethality in breast cancer cells such as MDA-MB-231 (TNBC) and MCF-7 (lumina) cells. MDA-MB-231 cells were responsive to combination treatment in both short-term (48 h) and long-term treatment (14 days) in a synergistic manner, whereas in MCF-7, the combination treatment was more effective in the long-term regimen. Furthermore, the cytotoxic effects of the plumbagin and MST-312 combination treatment were not recoverable after the short-term treatment. In conclusion, a combination treatment of MST-312 and plumbagin is proven to be more effective than a single plumbagin compound treatment in inducing DNA damage and telomere dysfunction leading to greater genome instability, cell cycle arrest and eventually cell death in cancer cells.
Collapse
Affiliation(s)
- Safoura Sameni
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ramya Viswanathan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Gavin Yong-Quan Ng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wilner Martinez-Lopez
- Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
- Associate Unit on Genomic Stability, Faculty of Medicine, University of the Republic (UdelaR), Montevideo, Uruguay
| | - M. Prakash Hande
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
43
|
Ojeda-Rodriguez A, Alcala-Diaz JF, Rangel-Zuñiga OA, Arenas-de Larriva AP, Gutierrez-Mariscal FM, Gómez-Luna P, Torres-Peña JD, Garcia-Rios A, Romero-Cabrera JL, Malagon MM, Perez-Martinez P, Ordovas JM, Delgado-Lista J, Yubero-Serrano EM, Lopez-Miranda J. Association between telomere length and intima-media thickness of both common carotid arteries in patients with coronary heart disease: From the CORDIOPREV randomized controlled trial. Atherosclerosis 2023; 380:117193. [PMID: 37549582 DOI: 10.1016/j.atherosclerosis.2023.117193] [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: 03/13/2023] [Revised: 06/23/2023] [Accepted: 07/26/2023] [Indexed: 08/09/2023]
Abstract
BACKGROUND AND AIMS A critical telomere length (TL) is associated with cardiovascular mortality. Dietary habits have been demonstrated to affect cardiovascular risk. However, it remains unclear how exactly TL determines the response to specific dietary approaches in the reduction of arterial injury. We aimed to evaluate whether TL was associated with the progression of arterial injury (assessed by intima-media thickness of both common carotid arteries: IMT-CC), after long-term consumption of two healthy dietary models in patients with coronary heart disease (CHD). METHODS From the 1002 CHD patients of the CORDIOPREV study, 903 completed IMT-CC and TL evaluation at baseline and were randomized to follow a Mediterranean diet or a low-fat diet for 5 years. RESULTS Patients at risk of short TL (TL < 20th percentile) presented an elevated IMT-CC, (0.79 ± 0.17 vs patients at non-risk 0.74 ± 0.17 p < 0.001). TL and IMT-CC showed an inverse association (β = -0.035, p = 0.002). Patients who consumed a Mediterranean diet, regardless of the risk of short TL, showed a significant decrease in IMT-CC, with a higher reduction in those patients with risk of short TL (-0.03 ± 0.11, p = 0.036). TL (β = 0.019, p = 0.024), age (β = -0.001, p = 0.031), energy intake (β = -0.000, p = 0.036), use of statins (β = -0.027, p = 0.028) and allocation into the Mediterranean diet (vs low-fat diet) (β = -0.024, p = 0.003) were significant contributors to changes in IMT-CC. CONCLUSIONS Patients who had a reduced TL exhibited a greater decrease in IMT-CC after consuming a Mediterranean diet.
Collapse
Affiliation(s)
- Ana Ojeda-Rodriguez
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004, Cordoba, Spain; Department of Medical and Surgical Science, University of Cordoba, 14004, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Juan F Alcala-Diaz
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004, Cordoba, Spain; Department of Medical and Surgical Science, University of Cordoba, 14004, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Oriol Alberto Rangel-Zuñiga
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004, Cordoba, Spain; Department of Medical and Surgical Science, University of Cordoba, 14004, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Antonio Pablo Arenas-de Larriva
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004, Cordoba, Spain; Department of Medical and Surgical Science, University of Cordoba, 14004, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Francisco M Gutierrez-Mariscal
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004, Cordoba, Spain; Department of Medical and Surgical Science, University of Cordoba, 14004, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Purificación Gómez-Luna
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004, Cordoba, Spain; Department of Medical and Surgical Science, University of Cordoba, 14004, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Jose D Torres-Peña
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004, Cordoba, Spain; Department of Medical and Surgical Science, University of Cordoba, 14004, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Antonio Garcia-Rios
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004, Cordoba, Spain; Department of Medical and Surgical Science, University of Cordoba, 14004, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Juan L Romero-Cabrera
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004, Cordoba, Spain; Department of Medical and Surgical Science, University of Cordoba, 14004, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Maria M Malagon
- Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, 14004, Cordoba, Spain
| | - Pablo Perez-Martinez
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004, Cordoba, Spain; Department of Medical and Surgical Science, University of Cordoba, 14004, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Jose M Ordovas
- Nutrition and Genomics Laboratory, J.M.-US Department of Agriculture Human Nutrition Research Center on Aging, At Tufts University, Boston, MA, 02111, USA; IMDEA Alimentacion, Madrid, Spain; CNIC, 28049, Madrid, Spain
| | - Javier Delgado-Lista
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004, Cordoba, Spain; Department of Medical and Surgical Science, University of Cordoba, 14004, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Elena M Yubero-Serrano
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004, Cordoba, Spain; Department of Medical and Surgical Science, University of Cordoba, 14004, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Jose Lopez-Miranda
- Lipids and Atherosclerosis Unit, Internal Medicine Unit, Reina Sofia University Hospital, 14004, Cordoba, Spain; Department of Medical and Surgical Science, University of Cordoba, 14004, Córdoba, Spain; Maimonides Biomedical Research Institute of Cordoba (IMIBIC), Av. Menendez Pidal, s/n, 14004, Cordoba, Spain; CIBER Fisiopatologia de La Obesidad y Nutricion (CIBEROBN), Instituto de Salud Carlos III, 28029, Madrid, Spain.
| |
Collapse
|
44
|
Yung Y, Maydan SA, Bart Y, Orvieto R, Aizer A. Human granulosa cells of poor ovarian responder patients display telomeres shortening. J Assist Reprod Genet 2023; 40:1943-1947. [PMID: 37432588 PMCID: PMC10371957 DOI: 10.1007/s10815-023-02860-6] [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: 04/23/2023] [Accepted: 06/12/2023] [Indexed: 07/12/2023] Open
Abstract
OBJECTIVE We aimed to compare the telomere length in granulosa cells of the young normal and poor ovarian responder patients and elderly patients undergoing ovarian stimulation for IVF. METHODS The main outcome measures granulosa cells telomere Length in the 3 study groups of patients undergoing IVF treatment in our center. 1) young normal responder patients (< 35 years); 2) young (< 35 years) poor ovarian responder patients; and 3) Elderly patients (40-45 years). Granulosa cells were obtained at the time of oocyte retrieval. Granulosa cells telomere length was assessed by absolute human telomere length quantification qPCR Assay. RESULTS The telomere length of the young normal responder was significantly longer as compared to young poor ovarian responder (15.5 vs 9.6 KB, p < 0.001) and the elderly patients (15.5 vs 10.66 KB, p < 0.002). No significant difference was observed in the telomere length between the young poor ovarian responder and the elderly patients. CONCLUSIONS Granulosa cells telomere length of the young normal responder was found to be significantly longer than young poor ovarian responder or elderly patients, highlighting the role of telomere length as a predictor, or contributor to poor oocyte yield following IVF treatment.
Collapse
Affiliation(s)
- Yuval Yung
- Department of Obstetrics and Gynecology, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Sharon Avhar Maydan
- Department of Obstetrics and Gynecology, Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Yossi Bart
- Department of Obstetrics and Gynecology, Chaim Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Raoul Orvieto
- Department of Obstetrics and Gynecology, Chaim Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
- The Tarnesby-Tarnowski Chair for Family Planning and Fertility Regulation, Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| | - Adva Aizer
- Department of Obstetrics and Gynecology, Chaim Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel Aviv-Yafo, Israel
| |
Collapse
|
45
|
Stock AJ, Ayyar S, Kashyap A, Wang Y, Yanai H, Starost MF, Tanaka-Yano M, Bodogai M, Sun C, Wang Y, Gong Y, Puligilla C, Fang EF, Bohr VA, Liu Y, Beerman I. Boosting NAD ameliorates hematopoietic impairment linked to short telomeres in vivo. GeroScience 2023; 45:2213-2228. [PMID: 36826621 PMCID: PMC10651621 DOI: 10.1007/s11357-023-00752-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 02/03/2023] [Indexed: 02/25/2023] Open
Abstract
Short telomeres are a defining feature of telomere biology disorders (TBDs), including dyskeratosis congenita (DC), for which there is no effective general cure. Patients with TBDs often experience bone marrow failure. NAD, an essential metabolic coenzyme, is decreased in models of DC. Herein, using telomerase reverse transcriptase null (Tert-/-) mice with critically short telomeres, we investigated the effect of NAD supplementation with the NAD precursor, nicotinamide riboside (NR), on features of health span disrupted by telomere impairment. Our results revealed that NR ameliorated body weight loss in Tert-/- mice and improved telomere integrity and telomere dysfunction-induced systemic inflammation. NR supplementation also mitigated myeloid skewing of Tert-/- hematopoietic stem cells. Furthermore, NR alleviated villous atrophy and inflammation in the small intestine of Tert-/- transplant recipient mice. Altogether, our findings support NAD intervention as a potential therapeutic strategy to enhance aspects of health span compromised by telomere attrition.
Collapse
Affiliation(s)
- Amanda J Stock
- Laboratory of Genetics and Genomics, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA
| | - Saipriya Ayyar
- Translational Gerontology Branch, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA
| | - Amogh Kashyap
- Translational Gerontology Branch, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA
| | - Yunong Wang
- Laboratory of Genetics and Genomics, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA
| | - Hagai Yanai
- Translational Gerontology Branch, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA
| | - Matthew F Starost
- Division of Veterinary Resources, Building 14E, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, USA
| | - Mayuri Tanaka-Yano
- Translational Gerontology Branch, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA
| | - Monica Bodogai
- Laboratory of Molecular Biology and Immunology, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA
| | - Chongkui Sun
- Laboratory of Genetics and Genomics, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA
| | - Yajun Wang
- Laboratory of Genetics and Genomics, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA
| | - Yi Gong
- Laboratory of Genetics and Genomics, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA
| | - Chandrakala Puligilla
- Laboratory of Genetics and Genomics, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA
| | - Evandro F Fang
- DNA Repair Section, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, 1478, Lørenskog, Norway
| | - Vilhelm A Bohr
- DNA Repair Section, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA
| | - Yie Liu
- Laboratory of Genetics and Genomics, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA.
| | - Isabel Beerman
- Translational Gerontology Branch, Biomedical Research Center, National Institute On Aging/National Institutes of Health, 251 Bayview Blvd., Baltimore, MD, USA.
| |
Collapse
|
46
|
Bhala S, Savage SA. What is the future of telomere length testing in telomere biology disorders? Expert Rev Hematol 2023; 16:475-478. [PMID: 37191632 PMCID: PMC10330493 DOI: 10.1080/17474086.2023.2215423] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/15/2023] [Indexed: 05/17/2023]
Affiliation(s)
- Sonia Bhala
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, USA
| | - Sharon A. Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, USA
| |
Collapse
|
47
|
Weng NP. Transcriptome-based measurement of CD8 + T cell age and its applications. Trends Immunol 2023; 44:542-550. [PMID: 37248098 PMCID: PMC10330598 DOI: 10.1016/j.it.2023.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/31/2023]
Abstract
The ability of T cells to undergo robust cell division in response to antigenic stimulation is essential for competent T cell function. However, this ability is reduced with aging and contributes to increased susceptibility to infectious diseases, cancers, and other diseases among older adults. To better understand T cell aging, improved measurements of age-related cellular changes in T cells are necessary. The recent development of machine learning (ML)-assisted transcriptome-based quantification of individual CD8+ T cell age represents a significant step forward in this regard. It reveals both prominent and subtle changes in gene expression and points to potential functional alterations of CD8+ T cells with aging. I argue that single-cell transcriptome-based age prediction in the immune system may have promising future applications.
Collapse
Affiliation(s)
- Nan-Ping Weng
- Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
48
|
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
|
49
|
Kliszczak M, Moralli D, Jankowska JD, Bryjka P, Subha Meem L, Goncalves T, Hester SS, Fischer R, Clynes D, Green CM. Loss of FAM111B protease mutated in hereditary fibrosing poikiloderma negatively regulates telomere length. Front Cell Dev Biol 2023; 11:1175069. [PMID: 37342232 PMCID: PMC10277729 DOI: 10.3389/fcell.2023.1175069] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 05/02/2023] [Indexed: 06/22/2023] Open
Abstract
Hereditary fibrosing poikiloderma (HFP) is a rare human dominant negative disorder caused by mutations in the FAM111B gene that encodes a nuclear trypsin-like serine protease. HFP patients present with symptoms including skin abnormalities, tendon contractures, myopathy and lung fibrosis. We characterized the cellular roles of human FAM111B using U2OS and MCF7 cell lines and report here that the protease interacts with components of the nuclear pore complex. Loss of FAM111B expression resulted in abnormal nuclear shape and reduced telomeric DNA content suggesting that FAM111B protease is required for normal telomere length; we show that this function is independent of telomerase or recombination driven telomere extension. Even though FAM111B-deficient cells were proficient in DNA repair, they showed hallmarks of genomic instability such as increased levels of micronuclei and ultra-fine DNA bridges. When mutated as in HFP, FAM111B was more frequently localized to the nuclear envelope, suggesting that accumulation of the mutated protease at the nuclear periphery may drive the disease pathology.
Collapse
Affiliation(s)
- Maciej Kliszczak
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Daniela Moralli
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Julia D. Jankowska
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Paulina Bryjka
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Lamia Subha Meem
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Tomas Goncalves
- Oncology Department, Weatherall Institute for Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Svenja S. Hester
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom
| | - Roman Fischer
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom
- Chinese Academy of Medical Sciences Oxford Institute, Oxford, United Kingdom
| | - David Clynes
- Oncology Department, Weatherall Institute for Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Catherine M. Green
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
50
|
Rouan A, Pousse M, Djerbi N, Porro B, Bourdin G, Carradec Q, Hume BC, Poulain J, Lê-Hoang J, Armstrong E, Agostini S, Salazar G, Ruscheweyh HJ, Aury JM, Paz-García DA, McMinds R, Giraud-Panis MJ, Deshuraud R, Ottaviani A, Morini LD, Leone C, Wurzer L, Tran J, Zoccola D, Pey A, Moulin C, Boissin E, Iwankow G, Romac S, de Vargas C, Banaigs B, Boss E, Bowler C, Douville E, Flores M, Reynaud S, Thomas OP, Troublé R, Thurber RV, Planes S, Allemand D, Pesant S, Galand PE, Wincker P, Sunagawa S, Röttinger E, Furla P, Voolstra CR, Forcioli D, Lombard F, Gilson E. Telomere DNA length regulation is influenced by seasonal temperature differences in short-lived but not in long-lived reef-building corals. Nat Commun 2023; 14:3038. [PMID: 37263999 DOI: 10.1038/s41467-023-38499-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 04/26/2023] [Indexed: 06/03/2023] Open
Abstract
Telomeres are environment-sensitive regulators of health and aging. Here,we present telomere DNA length analysis of two reef-building coral genera revealing that the long- and short-term water thermal regime is a key driver of between-colony variation across the Pacific Ocean. Notably, there are differences between the two studied genera. The telomere DNA lengths of the short-lived, more stress-sensitive Pocillopora spp. colonies were largely determined by seasonal temperature variation, whereas those of the long-lived, more stress-resistant Porites spp. colonies were insensitive to seasonal patterns, but rather influenced by past thermal anomalies. These results reveal marked differences in telomere DNA length regulation between two evolutionary distant coral genera exhibiting specific life-history traits. We propose that environmentally regulated mechanisms of telomere maintenance are linked to organismal performances, a matter of paramount importance considering the effects of climate change on health.
Collapse
Affiliation(s)
- Alice Rouan
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France.
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France.
| | - Melanie Pousse
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France
- Institut Fédératif de Recherche-Ressources Marines (IFR MARRES), Université Côte d'Azur, Nice, France
| | - Nadir Djerbi
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France
- Institut Fédératif de Recherche-Ressources Marines (IFR MARRES), Université Côte d'Azur, Nice, France
| | - Barbara Porro
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France
- Institut Fédératif de Recherche-Ressources Marines (IFR MARRES), Université Côte d'Azur, Nice, France
| | | | - Quentin Carradec
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, 91057, Evry, France
- Research Federation for the Study of Global Ocean Systems Ecology and Evolution, FR2022/Tara Oceans GO-SEE, 75016, Paris, France
| | - Benjamin Cc Hume
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Julie Poulain
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, 91057, Evry, France
- Research Federation for the Study of Global Ocean Systems Ecology and Evolution, FR2022/Tara Oceans GO-SEE, 75016, Paris, France
| | - Julie Lê-Hoang
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, 91057, Evry, France
- Research Federation for the Study of Global Ocean Systems Ecology and Evolution, FR2022/Tara Oceans GO-SEE, 75016, Paris, France
| | - Eric Armstrong
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, 91057, Evry, France
- Research Federation for the Study of Global Ocean Systems Ecology and Evolution, FR2022/Tara Oceans GO-SEE, 75016, Paris, France
| | - Sylvain Agostini
- Shimoda Marine Research Center, University of Tsukuba, Shimoda, Japan
| | - Guillem Salazar
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zurich, 8092, Zurich, Switzerland
| | - Hans-Joachim Ruscheweyh
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zurich, 8092, Zurich, Switzerland
| | - Jean-Marc Aury
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, 91057, Evry, France
- Research Federation for the Study of Global Ocean Systems Ecology and Evolution, FR2022/Tara Oceans GO-SEE, 75016, Paris, France
| | - David A Paz-García
- Centro de Investigaciones Biológicas del Noroeste (CIBNOR), Av. IPN 195, La Paz, Baja California Sur, 23096, La Paz, México
| | - Ryan McMinds
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
- University of South Florida Center for Global Health and Infectious Diseases Research, Tampa, FL, USA
- Maison de la Modélisation, de la Simulation et des Interactions (MSI),, Université Côte d'Azur, Nice, France
| | - Marie-Josèphe Giraud-Panis
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France
- Institut Fédératif de Recherche-Ressources Marines (IFR MARRES), Université Côte d'Azur, Nice, France
| | - Romane Deshuraud
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France
- Institut Fédératif de Recherche-Ressources Marines (IFR MARRES), Université Côte d'Azur, Nice, France
| | - Alexandre Ottaviani
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France
- Institut Fédératif de Recherche-Ressources Marines (IFR MARRES), Université Côte d'Azur, Nice, France
| | - Lycia Die Morini
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
| | - Camille Leone
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
| | - Lia Wurzer
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
| | - Jessica Tran
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
| | - Didier Zoccola
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France
- Centre Scientifique de Monaco, Principality of Monaco, Monaco, Monaco
| | - Alexis Pey
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France
- Institut Fédératif de Recherche-Ressources Marines (IFR MARRES), Université Côte d'Azur, Nice, France
| | - Clémentine Moulin
- Research Federation for the Study of Global Ocean Systems Ecology and Evolution, FR2022/Tara Oceans GO-SEE, 75016, Paris, France
- Tara Ocean Foundation, 8 rue de Prague, 75012, Paris, France
| | - Emilie Boissin
- Laboratoire d'Excellence "CORAIL," PSL Research University: EPHE-UPVD-CNRS, USR 3278 CRIOBE, Université de Perpignan, Perpignan Cedex, France
| | - Guillaume Iwankow
- Laboratoire d'Excellence "CORAIL," PSL Research University: EPHE-UPVD-CNRS, USR 3278 CRIOBE, Université de Perpignan, Perpignan Cedex, France
| | - Sarah Romac
- Sorbonne Université, CNRS, Station Biologique de Roscoff, AD2M, UMR 7144, ECOMAP, Roscoff, France
| | - Colomban de Vargas
- Research Federation for the Study of Global Ocean Systems Ecology and Evolution, FR2022/Tara Oceans GO-SEE, 75016, Paris, France
- Sorbonne Université, CNRS, Station Biologique de Roscoff, AD2M, UMR 7144, ECOMAP, Roscoff, France
| | - Bernard Banaigs
- Laboratoire d'Excellence "CORAIL," PSL Research University: EPHE-UPVD-CNRS, USR 3278 CRIOBE, Université de Perpignan, Perpignan Cedex, France
| | - Emmanuel Boss
- School of Marine Sciences, University of Maine, Orono, ME, USA
| | - Chris Bowler
- Research Federation for the Study of Global Ocean Systems Ecology and Evolution, FR2022/Tara Oceans GO-SEE, 75016, Paris, France
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole normale supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Eric Douville
- Laboratoire des Sciences du Climat et de l'Environnement, LSCE/IPSL, CEA-CNRS-UVSQ, Université Paris-Saclay, 91191, Gif-sur-Yvette, France
| | - Michel Flores
- Weizmann Institute of Science, Department of Earth, and Planetary Sciences, 76100, Rehovot, Israel
| | - Stéphanie Reynaud
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France
- Centre Scientifique de Monaco, Principality of Monaco, Monaco, Monaco
| | - Olivier P Thomas
- School of Biological and Chemical Sciences, Ryan Institute, University of Galway, University Road, H91TK33, Galway, Ireland
| | - Romain Troublé
- Research Federation for the Study of Global Ocean Systems Ecology and Evolution, FR2022/Tara Oceans GO-SEE, 75016, Paris, France
- Tara Ocean Foundation, 8 rue de Prague, 75012, Paris, France
| | - Rebecca Vega Thurber
- Oregon State University, Department of Microbiology, 220 Nash Hall, Corvallis, OR, 97331, USA
| | - Serge Planes
- Research Federation for the Study of Global Ocean Systems Ecology and Evolution, FR2022/Tara Oceans GO-SEE, 75016, Paris, France
- Laboratoire d'Excellence "CORAIL," PSL Research University: EPHE-UPVD-CNRS, USR 3278 CRIOBE, Université de Perpignan, Perpignan Cedex, France
| | - Denis Allemand
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France
- Centre Scientifique de Monaco, Principality of Monaco, Monaco, Monaco
| | - Stephane Pesant
- European Bioinformatics Institute, Wellcome Genome Campus, European Molecular Biology Laboratory, Wellcome Genome Campus, Cambridge CB10 1SD, UK, UK
| | - Pierre E Galand
- Research Federation for the Study of Global Ocean Systems Ecology and Evolution, FR2022/Tara Oceans GO-SEE, 75016, Paris, France
- Sorbonne Université, CNRS, Laboratoire d'Ecogéochimie des Environnements Benthiques (LECOB), Observatoire Océanologique de Banyuls, Banyuls-sur-Mer, France
| | - Patrick Wincker
- Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, 91057, Evry, France
- Research Federation for the Study of Global Ocean Systems Ecology and Evolution, FR2022/Tara Oceans GO-SEE, 75016, Paris, France
| | - Shinichi Sunagawa
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zurich, 8092, Zurich, Switzerland
| | - Eric Röttinger
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France
- Institut Fédératif de Recherche-Ressources Marines (IFR MARRES), Université Côte d'Azur, Nice, France
| | - Paola Furla
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France
- Institut Fédératif de Recherche-Ressources Marines (IFR MARRES), Université Côte d'Azur, Nice, France
| | | | - Didier Forcioli
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France
- Institut Fédératif de Recherche-Ressources Marines (IFR MARRES), Université Côte d'Azur, Nice, France
| | - Fabien Lombard
- Research Federation for the Study of Global Ocean Systems Ecology and Evolution, FR2022/Tara Oceans GO-SEE, 75016, Paris, France
- Sorbonne Université, Institut de la Mer de Villefranche sur mer, Laboratoire d'Océanographie de Villefranche, Villefranche-sur-Mer, France
- Institut Universitaire de France, Ministère chargé de l'enseignement supérieur, Paris, France
| | - Eric Gilson
- Université Côte d'Azur-CNRS-Inserm-Institute for Research on Cancer and Ageing, Nice (IRCAN), Medical School, Nice, France.
- Laboratoire International Associé Université Côte d'Azur-Centre Scientifique de Monaco (LIA ROPSE), Monaco, Nice, France.
- Institut Fédératif de Recherche-Ressources Marines (IFR MARRES), Université Côte d'Azur, Nice, France.
- Department of Medical Genetics, CHU, Nice, France.
| |
Collapse
|