1
|
Martinez-Fernandez V, Barascu A, Teixeira MT. Life and Death without Telomerase: The Saccharomyces cerevisiae Model. Cold Spring Harb Perspect Biol 2025; 17:a041699. [PMID: 39694811 PMCID: PMC12047662 DOI: 10.1101/cshperspect.a041699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024]
Abstract
Saccharomyces cerevisiae, a model organism in telomere biology, has been instrumental in pioneering a comprehensive understanding of the molecular processes that occur in the absence of telomerase across eukaryotes. This exploration spans investigations into telomere dynamics, intracellular signaling cascades, and organelle-mediated responses, elucidating their impact on proliferative capacity, genome stability, and cellular variability. Through the lens of budding yeast, numerous sources of cellular heterogeneity have been identified, dissected, and modeled, shedding light on the risks associated with telomeric state transitions, including the evasion of senescence. Moreover, the unraveling of the intricate interplay between the nucleus and other organelles upon telomerase inactivation has provided insights into eukaryotic evolution and cellular communication networks. These contributions, akin to milestones achieved using budding yeast, such as the discovery of the cell cycle, DNA damage checkpoint mechanisms, and DNA replication and repair processes, have been of paramount significance for the telomere field. Particularly, these insights extend to understanding replicative senescence as an anticancer mechanism in humans and enhancing our understanding of eukaryotes' evolution.
Collapse
Affiliation(s)
- Veronica Martinez-Fernandez
- Sorbonne Université, CNRS, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, LBMCE, F-75005 Paris, France
| | - Aurélia Barascu
- Sorbonne Université, CNRS, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, LBMCE, F-75005 Paris, France
| | - Maria Teresa Teixeira
- Sorbonne Université, CNRS, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, LBMCE, F-75005 Paris, France
| |
Collapse
|
2
|
Zeinoun B, Teixeira MT, Barascu A. Hog1 acts in a Mec1-independent manner to counteract oxidative stress following telomerase inactivation in Saccharomyces cerevisiae. Commun Biol 2024; 7:761. [PMID: 38909140 PMCID: PMC11193714 DOI: 10.1038/s42003-024-06464-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 06/18/2024] [Indexed: 06/24/2024] Open
Abstract
Replicative senescence is triggered when telomeres reach critically short length and activate permanent DNA damage checkpoint-dependent cell cycle arrest. Mitochondrial dysfunction and increase in oxidative stress are both features of replicative senescence in mammalian cells. However, how reactive oxygen species levels are controlled during senescence is elusive. Here, we show that reactive oxygen species levels increase in the telomerase-negative cells of Saccharomyces cerevisiae during replicative senescence, and that this coincides with the activation of Hog1, a mammalian p38 MAPK ortholog. Hog1 counteracts increased ROS levels during replicative senescence. While Hog1 deletion accelerates replicative senescence, we found this could stem from a reduced cell viability prior to telomerase inactivation. ROS levels also increase upon telomerase inactivation when Mec1, the yeast ortholog of ATR, is mutated, suggesting that oxidative stress is not simply a consequence of DNA damage checkpoint activation in budding yeast. We speculate that oxidative stress is a conserved hallmark of telomerase-negative eukaryote cells, and that its sources and consequences can be dissected in S. cerevisiae.
Collapse
Affiliation(s)
- Bechara Zeinoun
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005, Paris, France
| | - Maria Teresa Teixeira
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005, Paris, France.
| | - Aurélia Barascu
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005, Paris, France.
| |
Collapse
|
3
|
Jayaprasad AG, Chandrasekharan A, Arun Jyothi SP, John Sam SM, Santhoshkumar TR, Pillai MR. Telomerase inhibitors induce mitochondrial oxidation and DNA damage-dependent cell death rescued by Bcl-2/Bcl-xL. Int J Biol Macromol 2024; 264:130151. [PMID: 38403227 DOI: 10.1016/j.ijbiomac.2024.130151] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/05/2024] [Accepted: 02/11/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND Reactivation of telomerase is a hallmark of cancer and the majority of cancers over-express telomerase. Telomerase-dependent telomere length maintenance confers immortality to cancer cells. However, telomere length-independent cell survival functions of telomerase also play a critical role in tumorigenesis. Multiple telomerase inhibitors have been developed as therapeutics and include anti-sense oligonucleotides, telomerase RNA component targeting agents, chemical inhibitors of telomerase, small molecule inhibitors of hTERT, and telomerase vaccine. In general, telomerase inhibitors affect cell proliferation and survival of cells depending on the telomere length reduction, culminating in replicative senescence or cell death by crisis. However, most telomerase inhibitors kill cancer cells prior to significant reduction in telomere length, suggesting telomere length independent role of telomerase in early telomere dysfunction-dependent cell death. METHODS In this study, we explored the mechanism of cell death induced by three prominent telomerase inhibitors utilizing a series of genetically encoded sensor cells including redox and DNA damage sensor cells. RESULTS We report that telomerase inhibitors induce early cell cycle inhibition, followed by redox alterations at cytosol and mitochondria. Massive mitochondrial oxidation and DNA damage induce classical cell death involving mitochondrial transmembrane potential loss and mitochondrial permeabilization. Real-time imaging of the progression of mitochondrial oxidation revealed that treated cells undergo a biphasic mitochondrial redox alteration during telomerase inhibition, emphasizing the potential role of telomerase in the redox regulation at mitochondria. Additionally, silencing of hTERT confirmed its predominant role in maintaining mitochondrial redox homeostasis. Interestingly, the study also demonstrated that anti-apoptotic Bcl-2 family proteins still confer protection against cell death induced by telomerase inhibitors. CONCLUSION The study demonstrates that redox alterations and DNA damage contribute to early cell death by telomerase inhibitors and anti-apoptotic Bcl-2 family proteins confer protection from cell death by their ability to safeguard mitochondria from oxidation damage.
Collapse
Affiliation(s)
- Aparna Geetha Jayaprasad
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud, Thiruvananthapuram 695014, Kerala, India; PhD Program, Manipal Academy of Higher Education (MAHE), Madhav Nagar, Manipal, Karnataka 576104, India
| | - Aneesh Chandrasekharan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud, Thiruvananthapuram 695014, Kerala, India
| | - S P Arun Jyothi
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud, Thiruvananthapuram 695014, Kerala, India
| | - S M John Sam
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud, Thiruvananthapuram 695014, Kerala, India
| | - T R Santhoshkumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud, Thiruvananthapuram 695014, Kerala, India.
| | - M Radhakrishna Pillai
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Poojappura, Thycaud, Thiruvananthapuram 695014, Kerala, India.
| |
Collapse
|
4
|
Sanpedro-Luna JA, Vega-Alvarado L, Vázquez-Cruz C, Sánchez-Alonso P. Global Gene Expression of Post-Senescent Telomerase-Negative ter1Δ Strain of Ustilago maydis. J Fungi (Basel) 2023; 9:896. [PMID: 37755003 PMCID: PMC10532341 DOI: 10.3390/jof9090896] [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: 05/14/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 09/28/2023] Open
Abstract
We analyzed the global expression patterns of telomerase-negative mutants from haploid cells of Ustilago maydis to identify the gene network required for cell survival in the absence of telomerase. Mutations in either of the telomerase core subunits (trt1 and ter1) of the dimorphic fungus U. maydis cause deficiencies in teliospore formation. We report the global transcriptome analysis of two ter1Δ survivor strains of U. maydis, revealing the deregulation of telomerase-deleted responses (TDR) genes, such as DNA-damage response, stress response, cell cycle, subtelomeric, and proximal telomere genes. Other differentially expressed genes (DEGs) found in the ter1Δ survivor strains were related to pathogenic lifestyle factors, plant-pathogen crosstalk, iron uptake, meiosis, and melanin synthesis. The two ter1Δ survivors were phenotypically comparable, yet DEGs were identified when comparing these strains. Our findings suggest that teliospore formation in U. maydis is controlled by key pathogenic lifestyle and meiosis genes.
Collapse
Affiliation(s)
- Juan Antonio Sanpedro-Luna
- Posgrado en Microbiología, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico;
| | - Leticia Vega-Alvarado
- Instituto de Ciencias Aplicadas y Tecnología, Universidad Nacional Autónoma de México, Ciudad de Mexico 04510, Mexico;
| | - Candelario Vázquez-Cruz
- Centro de Investigaciones en Ciencias Microbiológicas, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico;
| | - Patricia Sánchez-Alonso
- Centro de Investigaciones en Ciencias Microbiológicas, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla, Puebla 72570, Mexico;
| |
Collapse
|
5
|
D’Angiolo M, Yue JX, De Chiara M, Barré BP, Giraud Panis MJ, Gilson E, Liti G. Telomeres are shorter in wild Saccharomyces cerevisiae isolates than in domesticated ones. Genetics 2023; 223:iyac186. [PMID: 36563016 PMCID: PMC9991508 DOI: 10.1093/genetics/iyac186] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/02/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022] Open
Abstract
Telomeres are ribonucleoproteins that cap chromosome-ends and their DNA length is controlled by counteracting elongation and shortening processes. The budding yeast Saccharomyces cerevisiae has been a leading model to study telomere DNA length control and dynamics. Its telomeric DNA is maintained at a length that slightly varies between laboratory strains, but little is known about its variation at the species level. The recent publication of the genomes of over 1,000 S. cerevisiae strains enabled us to explore telomere DNA length variation at an unprecedented scale. Here, we developed a bioinformatic pipeline (YeaISTY) to estimate telomere DNA length from whole-genome sequences and applied it to the sequenced S. cerevisiae collection. Our results revealed broad natural telomere DNA length variation among the isolates. Notably, telomere DNA length is shorter in those derived from wild rather than domesticated environments. Moreover, telomere DNA length variation is associated with mitochondrial metabolism, and this association is driven by wild strains. Overall, these findings reveal broad variation in budding yeast's telomere DNA length regulation, which might be shaped by its different ecological life-styles.
Collapse
Affiliation(s)
- Melania D’Angiolo
- Institute for Research on Cancer and Aging (IRCAN), Université Côte d’Azur, 28 Avenue de Valombrose, 06107 Nice, France
| | - Jia-Xing Yue
- Institute for Research on Cancer and Aging (IRCAN), Université Côte d’Azur, 28 Avenue de Valombrose, 06107 Nice, France
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-sen University Cancer Center (SYSUCC), 651 Dongfeng Road East, China
| | - Matteo De Chiara
- Institute for Research on Cancer and Aging (IRCAN), Université Côte d’Azur, 28 Avenue de Valombrose, 06107 Nice, France
| | - Benjamin P Barré
- Institute for Research on Cancer and Aging (IRCAN), Université Côte d’Azur, 28 Avenue de Valombrose, 06107 Nice, France
| | - Marie-Josèphe Giraud Panis
- Institute for Research on Cancer and Aging (IRCAN), Université Côte d’Azur, 28 Avenue de Valombrose, 06107 Nice, France
| | - Eric Gilson
- Institute for Research on Cancer and Aging (IRCAN), Université Côte d’Azur, 28 Avenue de Valombrose, 06107 Nice, France
- Department of Genetics, CHU, 06107 Nice, France
| | - Gianni Liti
- Institute for Research on Cancer and Aging (IRCAN), Université Côte d’Azur, 28 Avenue de Valombrose, 06107 Nice, France
| |
Collapse
|
6
|
Martin H, Doumic M, Teixeira MT, Xu Z. Telomere shortening causes distinct cell division regimes during replicative senescence in Saccharomyces cerevisiae. Cell Biosci 2021; 11:180. [PMID: 34627377 PMCID: PMC8502270 DOI: 10.1186/s13578-021-00693-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/24/2021] [Indexed: 11/21/2022] Open
Abstract
Background Telomerase-negative cells have limited proliferation potential. In these cells, telomeres shorten until they reach a critical length and induce a permanently arrested state. This process called replicative senescence is associated with genomic instability and participates in tissue and organismal ageing. Experimental data using single-cell approaches in the budding yeast model organism show that telomerase-negative cells often experience abnormally long cell cycles, which can be followed by cell cycles of normal duration, before reaching the terminal senescent state. These series of non-terminal cell cycle arrests contribute to the heterogeneity of senescence and likely magnify its genomic instability. Due to their apparent stochastic nature, investigating the dynamics and the molecular origins of these arrests has been difficult. In particular, whether the non-terminal arrests series stem from a mechanism similar to the one that triggers terminal senescence is not known. Results Here, we provide a mathematical description of sequences of non-terminal arrests to understand how they appear. We take advantage of an experimental data set of cell cycle duration measurements performed in individual telomerase-negative yeast cells that keep track of the number of generations since telomerase inactivation. Using numerical simulations, we show that the occurrence of non-terminal arrests is a generation-dependent process that can be explained by the shortest telomere reaching a probabilistic threshold length. While the onset of senescence is also triggered by telomere shortening, we highlight differences in the laws that describe the number of consecutive arrests in non-terminal arrests compared to senescence arrests, suggesting distinct underlying mechanisms and cellular states. Conclusions Replicative senescence is a complex process that affects cell divisions earlier than anticipated, as exemplified by the frequent occurrence of non-terminal arrests early after telomerase inactivation. The present work unravels two kinetically and mechanistically distinct generation-dependent processes underlying non-terminal and terminal senescence arrests. We suggest that these two processes are responsible for two consequences of senescence at the population level, the increase of genome instability on the one hand, and the limitation of proliferation capacity on the other hand. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00693-3.
Collapse
Affiliation(s)
- Hugo Martin
- JL Lions Laboratory, Sorbonne Université, 75005, Paris, France
| | - Marie Doumic
- JL Lions Laboratory, Sorbonne Université, 75005, Paris, France.
| | - Maria Teresa Teixeira
- Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, Institut de Biologie Physico-Chimique, PSL, CNRS, UMR8226, Sorbonne Université, 75005, Paris, France
| | - Zhou Xu
- Laboratory of Computational and Quantitative Biology, Institut de Biologie Paris-Seine, CNRS, UMR7238, Sorbonne Université, 75005, Paris, France.
| |
Collapse
|
7
|
Chromatin modifiers and recombination factors promote a telomere fold-back structure, that is lost during replicative senescence. PLoS Genet 2020; 16:e1008603. [PMID: 33370275 PMCID: PMC7793543 DOI: 10.1371/journal.pgen.1008603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 01/08/2021] [Accepted: 11/03/2020] [Indexed: 12/27/2022] Open
Abstract
Telomeres have the ability to adopt a lariat conformation and hence, engage in long and short distance intra-chromosome interactions. Budding yeast telomeres were proposed to fold back into subtelomeric regions, but a robust assay to quantitatively characterize this structure has been lacking. Therefore, it is not well understood how the interactions between telomeres and non-telomeric regions are established and regulated. We employ a telomere chromosome conformation capture (Telo-3C) approach to directly analyze telomere folding and its maintenance in S. cerevisiae. We identify the histone modifiers Sir2, Sin3 and Set2 as critical regulators for telomere folding, which suggests that a distinct telomeric chromatin environment is a major requirement for the folding of yeast telomeres. We demonstrate that telomeres are not folded when cells enter replicative senescence, which occurs independently of short telomere length. Indeed, Sir2, Sin3 and Set2 protein levels are decreased during senescence and their absence may thereby prevent telomere folding. Additionally, we show that the homologous recombination machinery, including the Rad51 and Rad52 proteins, as well as the checkpoint component Rad53 are essential for establishing the telomere fold-back structure. This study outlines a method to interrogate telomere-subtelomere interactions at a single unmodified yeast telomere. Using this method, we provide insights into how the spatial arrangement of the chromosome end structure is established and demonstrate that telomere folding is compromised throughout replicative senescence.
Collapse
|
8
|
Cavinato M, Madreiter-Sokolowski CT, Büttner S, Schosserer M, Zwerschke W, Wedel S, Grillari J, Graier WF, Jansen-Dürr P. Targeting cellular senescence based on interorganelle communication, multilevel proteostasis, and metabolic control. FEBS J 2020; 288:3834-3854. [PMID: 33200494 PMCID: PMC7611050 DOI: 10.1111/febs.15631] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 11/02/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
Abstract
Cellular senescence, a stable cell division arrest caused by severe damage and stress, is a hallmark of aging in vertebrates including humans. With progressing age, senescent cells accumulate in a variety of mammalian tissues, where they contribute to tissue aging, identifying cellular senescence as a major target to delay or prevent aging. There is an increasing demand for the discovery of new classes of small molecules that would either avoid or postpone cellular senescence by selectively eliminating senescent cells from the body (i.e., ‘senolytics’) or inactivating/switching damage‐inducing properties of senescent cells (i.e., ‘senostatics/senomorphics’), such as the senescence‐associated secretory phenotype. Whereas compounds with senolytic or senostatic activity have already been described, their efficacy and specificity has not been fully established for clinical use yet. Here, we review mechanisms of senescence that are related to mitochondria and their interorganelle communication, and the involvement of proteostasis networks and metabolic control in the senescent phenotype. These cellular functions are associated with cellular senescence in in vitro and in vivo models but have not been fully exploited for the search of new compounds to counteract senescence yet. Therefore, we explore possibilities to target these mechanisms as new opportunities to selectively eliminate and/or disable senescent cells with the aim of tissue rejuvenation. We assume that this research will provide new compounds from the chemical space which act as mimetics of caloric restriction, modulators of calcium signaling and mitochondrial physiology, or as proteostasis optimizers, bearing the potential to counteract cellular senescence, thereby allowing healthy aging.
Collapse
Affiliation(s)
- Maria Cavinato
- Institute for Biomedical Aging Research, Leopold-Franzens Universität Innsbruck, Austria.,Center for Molecular Biosciences Innsbruck (CMBI), Leopold-Franzens Universität Innsbruck, Austria
| | - Corina T Madreiter-Sokolowski
- Department of Health Sciences and Technology, Institute of Translational Medicine, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland.,Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Sabrina Büttner
- Institute of Molecular Biosciences, University of Graz, Austria.,Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Sweden
| | - Markus Schosserer
- Christian Doppler Laboratory for Skin Multimodal Analytical Imaging of Aging and Senescence, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Medical University of Vienna, Austria
| | - Werner Zwerschke
- Institute for Biomedical Aging Research, Leopold-Franzens Universität Innsbruck, Austria.,Center for Molecular Biosciences Innsbruck (CMBI), Leopold-Franzens Universität Innsbruck, Austria
| | - Sophia Wedel
- Institute for Biomedical Aging Research, Leopold-Franzens Universität Innsbruck, Austria.,Center for Molecular Biosciences Innsbruck (CMBI), Leopold-Franzens Universität Innsbruck, Austria
| | - Johannes Grillari
- Christian Doppler Laboratory for Skin Multimodal Analytical Imaging of Aging and Senescence, Institute of Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Medical University of Vienna, Austria.,Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria
| | - Wolfgang F Graier
- Molecular Biology and Biochemistry, Gottfried Schatz Research Center, Medical University of Graz, Austria.,BioTechMed Graz, Austria
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research, Leopold-Franzens Universität Innsbruck, Austria.,Center for Molecular Biosciences Innsbruck (CMBI), Leopold-Franzens Universität Innsbruck, Austria
| |
Collapse
|
9
|
Phung HTT, Tran DH, Nguyen TX. The cruciform DNA-binding protein Crp1 stimulates the endonuclease activity of Mus81-Mms4 in Saccharomyces cerevisiae. FEBS Lett 2020; 594:4320-4337. [PMID: 32936932 DOI: 10.1002/1873-3468.13931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/03/2020] [Accepted: 09/06/2020] [Indexed: 11/07/2022]
Abstract
The Saccharomyces cerevisiae Mus81-Mms4 complex is a highly conserved DNA structure-specific endonuclease that plays essential roles in the processing of recombination intermediates that arise during the repair of stalled replication forks and double-stranded breaks. To identify novel factors functioning conjointly with Mus81-Mms4, we performed a biochemical screen and found that Crp1, a cruciform DNA-recognizing protein that specifically binds to DNA four-way junction structures, could stimulate the Mus81-Mms4 endonuclease. The specific protein interaction between Mus81-Mms4 and Crp1 was responsible for the stimulation observed. Multicopy expression of Crp1 could partially rescue the sensitivity to DNA-damaging agents of the sgs1∆mus81∆21-24N mutant. Our results provide insight into the functional role and interaction of Crp1 with other proteins involved in DNA repair.
Collapse
Affiliation(s)
- Huong Thi Thu Phung
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam
| | - Diem Hong Tran
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam
| | - Ta Xuan Nguyen
- NTT Hi-Tech Institute, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam
| |
Collapse
|
10
|
Song S, Perez JV, Svitko W, Ricketts MD, Dean E, Schultz D, Marmorstein R, Johnson FB. Rap1-mediated nucleosome displacement can regulate gene expression in senescent cells without impacting the pace of senescence. Aging Cell 2020; 19:e13061. [PMID: 31742863 PMCID: PMC6974733 DOI: 10.1111/acel.13061] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 09/19/2019] [Accepted: 10/13/2019] [Indexed: 01/18/2023] Open
Abstract
Cell senescence is accompanied, and in part mediated, by changes in chromatin, including histone losses, but underlying mechanisms are not well understood. We reported previously that during yeast cell senescence driven by telomere shortening, the telomeric protein Rap1 plays a major role in reprogramming gene expression by relocalizing hundreds of new target genes (called NRTS, for new Rap1 targets at senescence) to the promoters. This leads to two types of histone loss: Rap1 lowers histone level globally by repressing histone gene expression, and it also causes local nucleosome displacement at the promoters of upregulated NRTS. Here, we present evidence of direct binding between Rap1 and histone H3/H4 heterotetramers, and map amino acids involved in the interaction within the Rap1 SANT domain to amino acids 392-394 (SHY). Introduction of a point mutation within the native RAP1 locus that converts these residues to alanines (RAP1SHY ), and thus disrupts Rap1-H3/H4 interaction, does not interfere with Rap1 relocalization to NRTS at senescence, but prevents full nucleosome displacement and gene upregulation, indicating direct Rap1-H3/H4 contacts are involved in nucleosome displacement. Consistent with this, the histone H3/H4 chaperone Asf1 is similarly unnecessary for Rap1 localization to NRTS but is required for full Rap1-mediated nucleosome displacement and gene activation. Remarkably, RAP1SHY does not affect the pace of senescence-related cell cycle arrest, indicating that some changes in gene expression at senescence are not coupled to this arrest.
Collapse
Affiliation(s)
- Shufei Song
- Department of Biochemistry and Molecular Biophysics University of Pennsylvania Philadelphia PA USA
- Graduate Group in Biochemistry and Molecular Biophysics University of Pennsylvania Philadelphia PA USA
- Department of Pathology and Laboratory Medicine University of Pennsylvania Philadelphia PA USA
| | - Javier V. Perez
- Department of Pathology and Laboratory Medicine University of Pennsylvania Philadelphia PA USA
| | - William Svitko
- Department of Pathology and Laboratory Medicine University of Pennsylvania Philadelphia PA USA
| | - M. Daniel Ricketts
- Department of Biochemistry and Molecular Biophysics University of Pennsylvania Philadelphia PA USA
- Abramson Family Cancer Research Institute University of Pennsylvania Philadelphia PA USA
| | - Elliot Dean
- High‐Throughput Screening Core University of Pennsylvania Philadelphia PA USA
| | - David Schultz
- High‐Throughput Screening Core University of Pennsylvania Philadelphia PA USA
| | - Ronen Marmorstein
- Department of Biochemistry and Molecular Biophysics University of Pennsylvania Philadelphia PA USA
- Abramson Family Cancer Research Institute University of Pennsylvania Philadelphia PA USA
| | - F. Brad Johnson
- Department of Biochemistry and Molecular Biophysics University of Pennsylvania Philadelphia PA USA
- Department of Pathology and Laboratory Medicine University of Pennsylvania Philadelphia PA USA
- Institute on Aging University of Pennsylvania Philadelphia PA USA
| |
Collapse
|
11
|
Xu Z, Teixeira MT. The many types of heterogeneity in replicative senescence. Yeast 2019; 36:637-648. [PMID: 31306505 PMCID: PMC6900063 DOI: 10.1002/yea.3433] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 07/04/2019] [Accepted: 07/07/2019] [Indexed: 11/10/2022] Open
Abstract
Replicative senescence, which is induced by telomere shortening, underlies the loss of regeneration capacity of organs and is ultimately detrimental to the organism. At the same time, it is required to protect organisms from unlimited cell proliferation that may arise from numerous stimuli or deregulations. One important feature of replicative senescence is its high level of heterogeneity and asynchrony, which promote genome instability and senescence escape. Characterizing this heterogeneity and investigating its sources are thus critical to understanding the robustness of replicative senescence. Here we review the different aspects of senescence driven by telomere attrition that are subject to variation in Saccharomyces cerevisiae, the current understanding of the molecular processes at play, and the consequences of heterogeneity in replicative senescence.
Collapse
Affiliation(s)
- Zhou Xu
- CNRS, UMR7238, Institut de Biologie Paris‐Seine, Laboratory of Computational and Quantitative BiologySorbonne UniversitéParisFrance
| | - Maria Teresa Teixeira
- CNRS, UMR8226, Institut de Biologie Physico‐Chimique, Laboratory of Molecular and Cell Biology of EukaryotesSorbonne Université, PSL Research UniversityParisFrance
| |
Collapse
|
12
|
Červenák F, Juríková K, Devillers H, Kaffe B, Khatib A, Bonnell E, Sopkovičová M, Wellinger RJ, Nosek J, Tzfati Y, Neuvéglise C, Tomáška Ľ. Identification of telomerase RNAs in species of the Yarrowia clade provides insights into the co-evolution of telomerase, telomeric repeats and telomere-binding proteins. Sci Rep 2019; 9:13365. [PMID: 31527614 PMCID: PMC6746865 DOI: 10.1038/s41598-019-49628-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 07/29/2019] [Indexed: 12/17/2022] Open
Abstract
Telomeric repeats in fungi of the subphylum Saccharomycotina exhibit great inter- and intra-species variability in length and sequence. Such variations challenged telomeric DNA-binding proteins that co-evolved to maintain their functions at telomeres. Here, we compare the extent of co-variations in telomeric repeats, encoded in the telomerase RNAs (TERs), and the repeat-binding proteins from 13 species belonging to the Yarrowia clade. We identified putative TER loci, analyzed their sequence and secondary structure conservation, and predicted functional elements. Moreover, in vivo complementation assays with mutant TERs showed the functional importance of four novel TER substructures. The TER-derived telomeric repeat unit of all species, except for one, is 10 bp long and can be represented as 5′-TTNNNNAGGG-3′, with repeat sequence variations occuring primarily outside the vertebrate telomeric motif 5′-TTAGGG-3′. All species possess a homologue of the Yarrowia lipolytica Tay1 protein, YlTay1p. In vitro, YlTay1p displays comparable DNA-binding affinity to all repeat variants, suggesting a conserved role among these species. Taken together, these results add significant insights into the co-evolution of TERs, telomeric repeats and telomere-binding proteins in yeasts.
Collapse
Affiliation(s)
- Filip Červenák
- Departments of Genetics and Biochemistry, Comenius University in Bratislava, Faculty of Natural Sciences, Ilkovičova 6, Mlynská dolina, 84215, Bratislava, Slovakia
| | - Katarína Juríková
- Departments of Genetics and Biochemistry, Comenius University in Bratislava, Faculty of Natural Sciences, Ilkovičova 6, Mlynská dolina, 84215, Bratislava, Slovakia
| | - Hugo Devillers
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Binyamin Kaffe
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Safra Campus, Jerusalem, 91904, Israel
| | - Areej Khatib
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Safra Campus, Jerusalem, 91904, Israel
| | - Erin Bonnell
- Department of Microbiology and Infectiology, RNA Group, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, J1E 4K8, Canada
| | - Martina Sopkovičová
- Departments of Genetics and Biochemistry, Comenius University in Bratislava, Faculty of Natural Sciences, Ilkovičova 6, Mlynská dolina, 84215, Bratislava, Slovakia
| | - Raymund J Wellinger
- Department of Microbiology and Infectiology, RNA Group, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, J1E 4K8, Canada
| | - Jozef Nosek
- Departments of Genetics and Biochemistry, Comenius University in Bratislava, Faculty of Natural Sciences, Ilkovičova 6, Mlynská dolina, 84215, Bratislava, Slovakia
| | - Yehuda Tzfati
- Department of Genetics, The Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Safra Campus, Jerusalem, 91904, Israel.
| | - Cécile Neuvéglise
- Micalis Institute, INRA, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France.
| | - Ľubomír Tomáška
- Departments of Genetics and Biochemistry, Comenius University in Bratislava, Faculty of Natural Sciences, Ilkovičova 6, Mlynská dolina, 84215, Bratislava, Slovakia.
| |
Collapse
|
13
|
Jolivet P, Serhal K, Graf M, Eberhard S, Xu Z, Luke B, Teixeira MT. A subtelomeric region affects telomerase-negative replicative senescence in Saccharomyces cerevisiae. Sci Rep 2019; 9:1845. [PMID: 30755624 PMCID: PMC6372760 DOI: 10.1038/s41598-018-38000-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 12/04/2018] [Indexed: 11/21/2022] Open
Abstract
In eukaryotes, telomeres determine cell proliferation potential by triggering replicative senescence in the absence of telomerase. In Saccharomyces cerevisiae, senescence is mainly dictated by the first telomere that reaches a critically short length, activating a DNA-damage-like response. How the corresponding signaling is modulated by the telomeric structure and context is largely unknown. Here we investigated how subtelomeric elements of the shortest telomere in a telomerase-negative cell influence the onset of senescence. We found that a 15 kb truncation of the 7L subtelomere widely used in studies of telomere biology affects cell growth when combined with telomerase inactivation. This effect is likely not explained by (i) elimination of sequence homology at chromosome ends that would compromise homology-directed DNA repair mechanisms; (ii) elimination of the conserved subtelomeric X-element; (iii) elimination of a gene that would become essential in the absence of telomerase; and (iv) heterochromatinization of inner genes, causing the silencing of an essential gene in replicative senescent cells. This works contributes to better delineate subtelomere functions and their impact on telomere biology.
Collapse
Affiliation(s)
- Pascale Jolivet
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005, Paris, France
| | - Kamar Serhal
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005, Paris, France.,Institut de Génétique Humaine, CNRS, Université Montpellier, Montpellier, France
| | - Marco Graf
- Institute of Molecular Biology (IMB), 55128, Mainz, Germany
| | - Stephan Eberhard
- Sorbonne Université, PSL, CNRS, UMR7141, Institut de Biologie Physico-Chimique, Laboratoire de Physiologie Moléculaire et Membranaire du Chloroplaste, F-75005, Paris, France
| | - Zhou Xu
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005, Paris, France
| | - Brian Luke
- Institute of Neurobiology and Developmental Biology, JGU Mainz, Ackermannweg 4, 55128, Mainz, Germany.,Institute of Molecular Biology (IMB), 55128, Mainz, Germany
| | - Maria Teresa Teixeira
- Sorbonne Université, PSL, CNRS, UMR8226, Institut de Biologie Physico-Chimique, Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, F-75005, Paris, France.
| |
Collapse
|
14
|
Romaniuk A, Paszel-Jaworska A, Totoń E, Lisiak N, Hołysz H, Królak A, Grodecka-Gazdecka S, Rubiś B. The non-canonical functions of telomerase: to turn off or not to turn off. Mol Biol Rep 2018; 46:1401-1411. [PMID: 30448892 DOI: 10.1007/s11033-018-4496-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/12/2018] [Indexed: 12/19/2022]
Abstract
Telomerase is perceived as an immortality enzyme that enables passing the Hayflick limit. Its main function is telomere restoration but only in a limited group of cells, including cancer cells. Since it is found in a vast majority of cancer cells, it became a natural target for cancer therapy. However, it has much more functions than just altering the metabolism of telomeres-it also reveals numerous so-called non-canonical functions. Thus, a question arises whether it is always beneficial to turn it off when planning a cancer strategy and considering potential side effects? The purpose of this review is to discuss some of the recent discoveries about telomere-independent functions of telomerase in the context of cancer therapy and potential side effects.
Collapse
Affiliation(s)
- Aleksandra Romaniuk
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland
| | - Anna Paszel-Jaworska
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland
| | - Ewa Totoń
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland
| | - Natalia Lisiak
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland
| | - Hanna Hołysz
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland
| | - Anna Królak
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland
| | | | - Błażej Rubiś
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355, Poznań, Poland.
| |
Collapse
|
15
|
Wanat JJ, Logsdon GA, Driskill JH, Deng Z, Lieberman PM, Johnson FB. TERRA and the histone methyltransferase Dot1 cooperate to regulate senescence in budding yeast. PLoS One 2018; 13:e0195698. [PMID: 29649255 PMCID: PMC5896980 DOI: 10.1371/journal.pone.0195698] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 03/27/2018] [Indexed: 01/27/2023] Open
Abstract
The events underlying senescence induced by critical telomere shortening are not fully understood. Here we provide evidence that TERRA, a non-coding RNA transcribed from subtelomeres, contributes to senescence in yeast lacking telomerase (tlc1Δ). Levels of TERRA expressed from multiple telomere ends appear elevated at senescence, and expression of an artificial RNA complementary to TERRA (anti-TERRA) binds TERRA in vivo and delays senescence. Anti-TERRA acts independently from several other mechanisms known to delay senescence, including those elicited by deletions of EXO1, TEL1, SAS2, and genes encoding RNase H enzymes. Further, it acts independently of the senescence delay provided by RAD52-dependent recombination. However, anti-TERRA delays senescence in a fashion epistatic to inactivation of the conserved histone methyltransferase Dot1. Dot1 associates with TERRA, and anti-TERRA disrupts this interaction in vitro and in vivo. Surprisingly, the anti-TERRA delay is independent of the C-terminal methyltransferase domain of Dot1 and instead requires only its N-terminus, which was previously found to facilitate release of telomeres from the nuclear periphery. Together, these data suggest that TERRA and Dot1 cooperate to drive senescence.
Collapse
Affiliation(s)
- Jennifer J. Wanat
- University of Pennsylvania School of Medicine, Pathology and Laboratory Medicine, Philadelphia, Pennsylvania, United States of America
- Washington College, Department of Biology, Chestertown, Maryland, United States of America
| | - Glennis A. Logsdon
- University of Pennsylvania School of Medicine, Pathology and Laboratory Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jordan H. Driskill
- University of Pennsylvania School of Medicine, Pathology and Laboratory Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Zhong Deng
- The Wistar Institute, Gene Expression and Regulation, Philadelphia, Pennsylvania, United States of America
| | - Paul M. Lieberman
- The Wistar Institute, Gene Expression and Regulation, Philadelphia, Pennsylvania, United States of America
| | - F. Brad Johnson
- University of Pennsylvania School of Medicine, Pathology and Laboratory Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
16
|
Simoneau A, Ricard É, Wurtele H. An interplay between multiple sirtuins promotes completion of DNA replication in cells with short telomeres. PLoS Genet 2018; 14:e1007356. [PMID: 29659581 PMCID: PMC5919697 DOI: 10.1371/journal.pgen.1007356] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 04/26/2018] [Accepted: 04/09/2018] [Indexed: 01/08/2023] Open
Abstract
The evolutionarily-conserved sirtuin family of histone deacetylases regulates a multitude of DNA-associated processes. A recent genome-wide screen conducted in the yeast Saccharomyces cerevisiae identified Yku70/80, which regulate nonhomologous end-joining (NHEJ) and telomere structure, as being essential for cell proliferation in the presence of the pan-sirtuin inhibitor nicotinamide (NAM). Here, we show that sirtuin-dependent deacetylation of both histone H3 lysine 56 and H4 lysine 16 promotes growth of yku70Δ and yku80Δ cells, and that the NAM sensitivity of these mutants is not caused by defects in DNA double-strand break repair by NHEJ, but rather by their inability to maintain normal telomere length. Indeed, our results indicate that in the absence of sirtuin activity, cells with abnormally short telomeres, e.g., yku70/80Δ or est1/2Δ mutants, present striking defects in S phase progression. Our data further suggest that early firing of replication origins at short telomeres compromises the cellular response to NAM- and genotoxin-induced replicative stress. Finally, we show that reducing H4K16ac in yku70Δ cells limits activation of the DNA damage checkpoint kinase Rad53 in response to replicative stress, which promotes usage of translesion synthesis and S phase progression. Our results reveal a novel interplay between sirtuin-mediated regulation of chromatin structure and telomere-regulating factors in promoting timely completion of S phase upon replicative stress.
Collapse
Affiliation(s)
- Antoine Simoneau
- Centre de recherche de l’Hôpital Maisonneuve-Rosemont, boulevard de l’Assomption, Montréal, Canada
- Programme de Biologie Moléculaire, Université de Montréal, Montréal, Canada
| | - Étienne Ricard
- Centre de recherche de l’Hôpital Maisonneuve-Rosemont, boulevard de l’Assomption, Montréal, Canada
- Programme de Biologie Moléculaire, Université de Montréal, Montréal, Canada
| | - Hugo Wurtele
- Centre de recherche de l’Hôpital Maisonneuve-Rosemont, boulevard de l’Assomption, Montréal, Canada
- Département de Médecine, Université de Montréal, Montréal, Canada
| |
Collapse
|
17
|
Induction and relocalization of telomeric repeat-containing RNAs during diauxic shift in budding yeast. Curr Genet 2018; 64:1117-1127. [PMID: 29569051 DOI: 10.1007/s00294-018-0829-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/09/2018] [Accepted: 03/20/2018] [Indexed: 02/03/2023]
Abstract
Telomeres are maintained in a heterochromatic state that represses transcription of subtelomeric genes, a phenomenon known as telomere position effect. Nevertheless, telomeric DNA is actively transcribed, leading to the synthesis of telomeric repeat-containing noncoding RNA or TERRA. This nuclear noncoding RNA has been proposed to play important roles at telomeres, regulating their silencing, capping, repair and elongation by telomerase. In the budding yeast Saccharomyces cerevisiae, TERRA accumulation is repressed by telomeric silencing and the Rat1 exonuclease. On the other hand, telomere shortening promotes expression of TERRA. So far, little is known about the biological processes that induce TERRA expression in yeast. Understanding the dynamics of TERRA expression and localization is essential to define its function in telomere biology. Here, we aim to study the dynamics of TERRA expression during yeast cell growth. Using live-cell imaging, RNA-FISH and quantitative RT-PCR, we show that TERRA expression is induced as yeast cells undergo diauxic shift, a lag phase during which yeast cells switch their metabolism from anaerobic fermentation to oxidative respiration. This induction is transient as TERRA levels decrease during post-diauxic shift. The increased expression of TERRA is not due to the shortening of telomeres or increased stability of this transcript. Surprisingly, this induction is coincident with a cytoplasmic accumulation of TERRA molecules. Our results suggest that TERRA transcripts may play extranuclear functions with important implications in telomere biology and add a novel layer of complexity in the interplay between telomere biology, metabolism and stress response.
Collapse
|
18
|
Beletsky AV, Malyavko AN, Sukhanova MV, Mardanova ES, Zvereva MI, Petrova OA, Parfenova YY, Rubtsova MP, Mardanov AV, Lavrik OI, Dontsova OA, Ravin NV. The genome-wide transcription response to telomerase deficiency in the thermotolerant yeast Hansenula polymorpha DL-1. BMC Genomics 2017; 18:492. [PMID: 28659185 PMCID: PMC5490237 DOI: 10.1186/s12864-017-3889-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Accepted: 06/21/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND In the course of replication of eukaryotic chromosomes, the telomere length is maintained due to activity of telomerase, the ribonucleoprotein reverse transcriptase. Abolishing telomerase function causes progressive shortening of telomeres and, ultimately, cell cycle arrest and replicative senescence. To better understand the cellular response to telomerase deficiency, we performed a transcriptomic study for the thermotolerant methylotrophic yeast Hansenula polymorpha DL-1 lacking telomerase activity. RESULTS Mutant strain of H. polymorpha carrying a disrupted telomerase RNA gene was produced, grown to senescence and analyzed by RNA-seq along with wild type strain. Telomere shortening induced a transcriptional response involving genes relevant to telomere structure and maintenance, DNA damage response, information processing, and some metabolic pathways. Genes involved in DNA replication and repair, response to environmental stresses and intracellular traffic were up-regulated in senescent H. polymorpha cells, while strong down-regulation was observed for genes involved in transcription and translation, as well as core histones. CONCLUSIONS Comparison of the telomerase deletion transcription responses by Saccharomyces cerevisiae and H. polymorpha demonstrates that senescence makes different impact on the main metabolic pathways of these yeast species but induces similar changes in processes related to nucleic acids metabolism and protein synthesis. Up-regulation of a subunit of the TORC1 complex is clearly relevant for both types of yeast.
Collapse
Affiliation(s)
- Alexey V Beletsky
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld 2, Moscow, 119071, Russia
| | - Alexander N Malyavko
- Faculty of Chemistry, Moscow State University, Leninskie Gory 1, bld. 3, Moscow, 119991, Russia.,Center of Functional Genomics, Skolkovo Institute of Science and Technology, Moscow, 143026, Russia
| | - Maria V Sukhanova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave. 8, Novosibirsk, 630090, Russia
| | - Eugenia S Mardanova
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld 2, Moscow, 119071, Russia
| | - Maria I Zvereva
- Faculty of Chemistry, Moscow State University, Leninskie Gory 1, bld. 3, Moscow, 119991, Russia
| | - Olga A Petrova
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Leninskie Gory 1, bld. 40, Moscow, 119992, Russia
| | - Yulia Yu Parfenova
- Faculty of Chemistry, Moscow State University, Leninskie Gory 1, bld. 3, Moscow, 119991, Russia
| | - Maria P Rubtsova
- Faculty of Chemistry, Moscow State University, Leninskie Gory 1, bld. 3, Moscow, 119991, Russia
| | - Andrey V Mardanov
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld 2, Moscow, 119071, Russia
| | - Olga I Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave. 8, Novosibirsk, 630090, Russia.,Novosibirsk State University, Novosibirsk, 630090, Russia
| | - Olga A Dontsova
- Faculty of Chemistry, Moscow State University, Leninskie Gory 1, bld. 3, Moscow, 119991, Russia.,Belozersky Institute of Physico-Chemical Biology, Moscow State University, Leninskie Gory 1, bld. 40, Moscow, 119992, Russia.,Center of Functional Genomics, Skolkovo Institute of Science and Technology, Moscow, 143026, Russia
| | - Nikolai V Ravin
- Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bld 2, Moscow, 119071, Russia.
| |
Collapse
|
19
|
Lue NF, Yu EY. Telomere recombination pathways: tales of several unhappy marriages. Curr Genet 2016; 63:401-409. [PMID: 27666406 DOI: 10.1007/s00294-016-0653-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 09/19/2016] [Accepted: 09/21/2016] [Indexed: 01/29/2023]
Abstract
All happy families are alike; each unhappy family is unhappy in its own way.-Leo Tolstoy, Anna Karenina.
Collapse
Affiliation(s)
- Neal F Lue
- Department of Microbiology and Immunology, W. R. Hearst Microbiology Research Center, Weill Cornell Medicine, New York, NY, USA. .,Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
| | - Eun Young Yu
- Department of Microbiology and Immunology, W. R. Hearst Microbiology Research Center, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
20
|
Early Loss of Telomerase Action in Yeast Creates a Dependence on the DNA Damage Response Adaptor Proteins. Mol Cell Biol 2016; 36:1908-19. [PMID: 27161319 PMCID: PMC4936065 DOI: 10.1128/mcb.00943-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 04/07/2016] [Indexed: 02/06/2023] Open
Abstract
Telomeres cap the ends of chromosomes, protecting them from degradation and inappropriate DNA repair processes that can lead to genomic instability. A short telomere elicits increased telomerase action on itself that replenishes telomere length, thereby stabilizing the telomere. In the prolonged absence of telomerase activity in dividing cells, telomeres eventually become critically short, inducing a permanent cell cycle arrest (senescence). We recently showed that even early after telomerase inactivation (ETI), yeast cells have accelerated mother cell aging and mildly perturbed cell cycles. Here, we show that the complete disruption of DNA damage response (DDR) adaptor proteins in ETI cells causes severe growth defects. This synthetic-lethality phenotype was as pronounced as that caused by extensive DNA damage in wild-type cells but showed genetic dependencies distinct from such damage and was completely alleviated by SML1 deletion, which increases deoxynucleoside triphosphate (dNTP) pools. Our results indicated that these deleterious effects in ETI cells cannot be accounted for solely by the slow erosion of telomeres due to incomplete replication that leads to senescence. We propose that normally occurring telomeric DNA replication stress is resolved by telomerase activity and the DDR in two parallel pathways and that deletion of Sml1 prevents this stress.
Collapse
|
21
|
Lustig AJ. Hypothesis: Paralog Formation from Progenitor Proteins and Paralog Mutagenesis Spur the Rapid Evolution of Telomere Binding Proteins. Front Genet 2016; 7:10. [PMID: 26904098 PMCID: PMC4748036 DOI: 10.3389/fgene.2016.00010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 01/22/2016] [Indexed: 12/31/2022] Open
Abstract
Through elegant studies in fungal cells and complex organisms, we propose a unifying paradigm for the rapid evolution of telomere binding proteins (TBPs) that associate with either (or both) telomeric DNA and telomeric proteins. TBPs protect and regulate telomere structure and function. Four critical factors are involved. First, TBPs that commonly bind to telomeric DNA include the c-Myb binding proteins, OB-fold single-stranded binding proteins, and G-G base paired Hoogsteen structure (G4) binding proteins. Each contributes independently or, in some cases, cooperatively, to provide a minimum level of telomere function. As a result of these minimal requirements and the great abundance of homologs of these motifs in the proteome, DNA telomere-binding activity may be generated more easily than expected. Second, telomere dysfunction gives rise to genome instability, through the elevation of recombination rates, genome ploidy, and the frequency of gene mutations. The formation of paralogs that diverge from their progenitor proteins ultimately can form a high frequency of altered TBPs with altered functions. Third, TBPs that assemble into complexes (e.g., mammalian shelterin) derive benefits from the novel emergent functions. Fourth, a limiting factor in the evolution of TBP complexes is the formation of mutually compatible interaction surfaces amongst the TBPs. These factors may have different degrees of importance in the evolution of different phyla, illustrated by the apparently simpler telomeres in complex plants. Selective pressures that can utilize the mechanisms of paralog formation and mutagenesis to drive TBP evolution along routes dependent on the requisite physiologic changes.
Collapse
Affiliation(s)
- Arthur J Lustig
- Department of Biochemistry and Molecular Biology, Tulane University, New Orleans LA, USA
| |
Collapse
|
22
|
Niederer RO, Papadopoulos N, Zappulla DC. Identification of novel noncoding transcripts in telomerase-negative yeast using RNA-seq. Sci Rep 2016; 6:19376. [PMID: 26786024 PMCID: PMC4726298 DOI: 10.1038/srep19376] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 12/07/2015] [Indexed: 12/20/2022] Open
Abstract
Telomerase is a ribonucleoprotein that maintains the ends of linear chromosomes in most eukaryotes. Loss of telomerase activity results in shortening of telomeric DNA and eventually a specific G2/M cell-cycle arrest known as senescence. In humans, telomere shortening occurs during aging, while inappropriate activation of telomerase is associated with approximately 90% of cancers. Previous studies have identified several classes of noncoding RNAs (ncRNA) also associated with aging-related senescence and cancer, but whether ncRNAs are also involved in short-telomere-induced senescence in yeast is unknown. Here, we report 112 putative novel lncRNAs in the yeast Saccharomyces cerevisiae, 41 of which are only expressed in telomerase-negative yeast. Expression of approximately half of the lncRNAs is strongly correlated with that of adjacent genes, suggesting this subset may influence transcription of neighboring genes. Our results reveal a new potential mechanism governing adaptive changes in senescing and post-senescent survivor yeast cells.
Collapse
Affiliation(s)
- Rachel O Niederer
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218 USA
| | - Nickolas Papadopoulos
- Ludwig Center for Cancer Genetics and Therapeutics, The Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21231 USA
| | - David C Zappulla
- Department of Biology, Johns Hopkins University, Baltimore, MD, 21218 USA
| |
Collapse
|
23
|
Xie Z, Jay KA, Smith DL, Zhang Y, Liu Z, Zheng J, Tian R, Li H, Blackburn EH. Early telomerase inactivation accelerates aging independently of telomere length. Cell 2015; 160:928-939. [PMID: 25723167 DOI: 10.1016/j.cell.2015.02.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 10/20/2014] [Accepted: 01/28/2015] [Indexed: 10/23/2022]
Abstract
Telomerase is required for long-term telomere maintenance and protection. Using single budding yeast mother cell analyses we found that, even early after telomerase inactivation (ETI), yeast mother cells show transient DNA damage response (DDR) episodes, stochastically altered cell-cycle dynamics, and accelerated mother cell aging. The acceleration of ETI mother cell aging was not explained by increased reactive oxygen species (ROS), Sir protein perturbation, or deprotected telomeres. ETI phenotypes occurred well before the population senescence caused late after telomerase inactivation (LTI). They were morphologically distinct from LTI senescence, were genetically uncoupled from telomere length, and were rescued by elevating dNTP pools. Our combined genetic and single-cell analyses show that, well before critical telomere shortening, telomerase is continuously required to respond to transient DNA replication stress in mother cells and that a lack of telomerase accelerates otherwise normal aging.
Collapse
Affiliation(s)
- Zhengwei Xie
- Center for Quantitative Biology, School of Physics and The Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Kyle A Jay
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Dana L Smith
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yi Zhang
- Center for Quantitative Biology, School of Physics and The Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Zairan Liu
- Center for Quantitative Biology, School of Physics and The Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jiashun Zheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Ruilin Tian
- Center for Quantitative Biology, School of Physics and The Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Hao Li
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA; California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Elizabeth H Blackburn
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
24
|
Bui VN, Nguyen TTH, Bettarel Y, Nguyen THT, Pham TL, Hoang TY, Nguyen VTT, Nghiem NM, Wölfl S. Genotoxicity of Chemical Compounds Identification and Assessment by Yeast Cells Transformed With GFP Reporter Constructs Regulated by the PLM2 or DIN7 Promoter. Int J Toxicol 2015; 34:31-43. [DOI: 10.1177/1091581814566870] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Yeast cells transformed with high-copy number plasmids comprising a green fluorescent protein (GFP)-encoding gene optimized for yeast under the control of the new DIN7 or PLM2 and the established RNR2 and RAD54 promoters were used to assess the genotoxic potential of chemical compounds. The activity of potential DNA-damaging agents was investigated by genotoxicity assays and by OxoPlate assay in the presence of various test compounds. The fluorescence signal generated by GFP in response to DNA damage was related to the different concentrations of analytes and the analyte-dependent GFP synthesis. The use of distinct DNA damage-inducible promoters presents alternative genotoxicity testing strategies by selective induction of promoters in response to DNA damage. The new DIN7 and PLM2 systems show higher sensitivity than the RNR2 and RAD54 systems in detecting 4-nitroquinoline- N-oxide and actinomycin D. Both DIN7 and PLM2 systems are able to detect camptothecin while RNR2 and RAD54 systems are not. Automated laboratory systems with assay performance on 384-well microplates provide for cost-effective high-throughput screening of DNA-damaging agents, reducing compound consumption to about 53% as compared with existing eukaryotic genotoxicity bioassays.
Collapse
Affiliation(s)
- Van Ngoc Bui
- Institute of Biotechnology, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Thi Thu Huyen Nguyen
- Thai Nguyen University of Sciences, Thai Nguyen University, Thai Nguyen, Hanoi, Vietnam
| | - Yvan Bettarel
- Institute of Research and Development, UMR ECOSYM, Montpellier, France
| | - Thi Hoai Thu Nguyen
- Institute of Biotechnology, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Thuy Linh Pham
- Institute of Biotechnology, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Thi Yen Hoang
- Institute of Biotechnology, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Vu Thanh Thanh Nguyen
- Thai Nguyen University of Sciences, Thai Nguyen University, Thai Nguyen, Hanoi, Vietnam
| | - Ngoc Minh Nghiem
- Institute of Biotechnology, Vietnam Academy of Science and Technology (VAST), Hanoi, Vietnam
| | - Stefan Wölfl
- Institute of Pharmacy and Molecular Biotechnology (IPMB), Heidelberg University, Heidelberg, Germany
| |
Collapse
|
25
|
Brown NA, Ries LNA, Goldman GH. How nutritional status signalling coordinates metabolism and lignocellulolytic enzyme secretion. Fungal Genet Biol 2014; 72:48-63. [PMID: 25011009 DOI: 10.1016/j.fgb.2014.06.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Revised: 06/26/2014] [Accepted: 06/28/2014] [Indexed: 11/30/2022]
Abstract
The utilisation of lignocellulosic plant biomass as an abundant, renewable feedstock for green chemistries and biofuel production is inhibited by its recalcitrant nature. In the environment, lignocellulolytic fungi are naturally capable of breaking down plant biomass into utilisable saccharides. Nonetheless, within the industrial context, inefficiencies in the production of lignocellulolytic enzymes impede the implementation of green technologies. One of the primary causes of such inefficiencies is the tight transcriptional control of lignocellulolytic enzymes via carbon catabolite repression. Fungi coordinate metabolism, protein biosynthesis and secretion with cellular energetic status through the detection of intra- and extra-cellular nutritional signals. An enhanced understanding of the signals and signalling pathways involved in regulating the transcription, translation and secretion of lignocellulolytic enzymes is therefore of great biotechnological interest. This comparative review describes how nutrient sensing pathways regulate carbon catabolite repression, metabolism and the utilisation of alternative carbon sources in Saccharomyces cerevisiae and ascomycete fungi.
Collapse
Affiliation(s)
- Neil Andrew Brown
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil.
| | | | - Gustavo Henrique Goldman
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil; Laboratório Nacional de Ciência e Tecnologia do Bioetanol (CTBE), Campinas, Brazil.
| |
Collapse
|
26
|
Abstract
Telomeres protect chromosome ends from degradation and inappropriate DNA damage response activation through their association with specific factors. Interestingly, these telomeric factors are able to localize outside telomeric regions, where they can regulate the transcription of genes involved in metabolism, immunity and differentiation. These findings delineate a signalling pathway by which telomeric changes control the ability of their associated factors to regulate transcription. This mechanism is expected to enable a greater diversity of cellular responses that are adapted to specific cell types and telomeric changes, and may therefore represent a pivotal aspect of development, ageing and telomere-mediated diseases.
Collapse
|
27
|
Platt JM, Ryvkin P, Wanat JJ, Donahue G, Ricketts MD, Barrett SP, Waters HJ, Song S, Chavez A, Abdallah KO, Master SR, Wang LS, Johnson FB. Rap1 relocalization contributes to the chromatin-mediated gene expression profile and pace of cell senescence. Genes Dev 2013; 27:1406-20. [PMID: 23756653 DOI: 10.1101/gad.218776.113] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cellular senescence is accompanied by dramatic changes in chromatin structure and gene expression. Using Saccharomyces cerevisiae mutants lacking telomerase (tlc1Δ) to model senescence, we found that with critical telomere shortening, the telomere-binding protein Rap1 (repressor activator protein 1) relocalizes to the upstream promoter regions of hundreds of new target genes. The set of new Rap1 targets at senescence (NRTS) is preferentially activated at senescence, and experimental manipulations of Rap1 levels indicate that it contributes directly to NRTS activation. A notable subset of NRTS includes the core histone-encoding genes; we found that Rap1 contributes to their repression and that histone protein levels decline at senescence. Rap1 and histones also display a target site-specific antagonism that leads to diminished nucleosome occupancy at the promoters of up-regulated NRTS. This antagonism apparently impacts the rate of senescence because underexpression of Rap1 or overexpression of the core histones delays senescence. Rap1 relocalization is not a simple consequence of lost telomere-binding sites, but rather depends on the Mec1 checkpoint kinase. Rap1 relocalization is thus a novel mechanism connecting DNA damage responses (DDRs) at telomeres to global changes in chromatin and gene expression while driving the pace of senescence.
Collapse
Affiliation(s)
- Jesse M Platt
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Schroeder EA, Raimundo N, Shadel GS. Epigenetic silencing mediates mitochondria stress-induced longevity. Cell Metab 2013; 17:954-964. [PMID: 23747251 PMCID: PMC3694503 DOI: 10.1016/j.cmet.2013.04.003] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 02/27/2013] [Accepted: 04/01/2013] [Indexed: 11/30/2022]
Abstract
Reactive oxygen species (ROS) play complex roles in aging, having both damaging effects and signaling functions. Transiently elevating mitochondrial stress, including mitochondrial ROS (mtROS), elicits beneficial responses that extend lifespan. However, these adaptive, longevity-signaling pathways remain poorly understood. We show here that Tel1p and Rad53p, homologs of the mammalian DNA damage response kinases ATM and Chk2, mediate a hormetic mtROS longevity signal that extends yeast chronological lifespan. This pathway senses mtROS in a manner distinct from the nuclear DNA damage response and ultimately imparts longevity by inactivating the histone demethylase Rph1p specifically at subtelomeric heterochromatin, enhancing binding of the silencing protein Sir3p, and repressing subtelomeric transcription. These results demonstrate the existence of conserved mitochondria-to-nucleus stress-signaling pathways that regulate aging through epigenetic modulation of nuclear gene expression.
Collapse
Affiliation(s)
- Elizabeth A Schroeder
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Nuno Raimundo
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Gerald S Shadel
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Genetics, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
29
|
Teixeira MT. Saccharomyces cerevisiae as a Model to Study Replicative Senescence Triggered by Telomere Shortening. Front Oncol 2013; 3:101. [PMID: 23638436 PMCID: PMC3636481 DOI: 10.3389/fonc.2013.00101] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 04/11/2013] [Indexed: 01/22/2023] Open
Abstract
In many somatic human tissues, telomeres shorten progressively because of the DNA-end replication problem. Consequently, cells cease to proliferate and are maintained in a metabolically viable state called replicative senescence. These cells are characterized by an activation of DNA damage checkpoints stemming from eroded telomeres, which are bypassed in many cancer cells. Hence, replicative senescence has been considered one of the most potent tumor suppressor pathways. However, the mechanism through which short telomeres trigger this cellular response is far from being understood. When telomerase is removed experimentally in Saccharomyces cerevisiae, telomere shortening also results in a gradual arrest of population growth, suggesting that replicative senescence also occurs in this unicellular eukaryote. In this review, we present the key steps that have contributed to the understanding of the mechanisms underlying the establishment of replicative senescence in budding yeast. As in mammals, signals stemming from short telomeres activate the DNA damage checkpoints, suggesting that the early cellular response to the shortest telomere(s) is conserved in evolution. Yet closer analysis reveals a complex picture in which the apparent single checkpoint response may result from a variety of telomeric alterations expressed in the absence of telomerase. Accordingly, the DNA replication of eroding telomeres appears as a critical challenge for senescing budding yeast cells and the easy manipulation of S. cerevisiae is providing insights into the way short telomeres are integrated into their chromatin and nuclear environments. Finally, the loss of telomerase in budding yeast triggers a more general metabolic alteration that remains largely unexplored. Thus, telomerase-deficient S. cerevisiae cells may have more common points than anticipated with somatic cells, in which telomerase depletion is naturally programed, thus potentially inspiring investigations in mammalian cells.
Collapse
Affiliation(s)
- M Teresa Teixeira
- Laboratoire de Biologie Moléculaire et Cellulaire des Eucaryotes, FRE3354 Centre National de la Recherche Scientifique, Université Pierre et Marie Curie, Institut de Biologie Physico-Chimique Paris, France
| |
Collapse
|
30
|
Holysz H, Lipinska N, Paszel-Jaworska A, Rubis B. Telomerase as a useful target in cancer fighting-the breast cancer case. Tumour Biol 2013; 34:1371-80. [PMID: 23558965 PMCID: PMC3661921 DOI: 10.1007/s13277-013-0757-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 03/20/2013] [Indexed: 12/17/2022] Open
Abstract
Telomerase was initially considered as a relevant factor distinguishing cancer from normal cells. During detailed studies, it appeared that its expression and activity is not only limited to cancer cells however, but in this particular cells, the telomerase is much more abundant. Thus, it has become a very promising target for an anticancer therapy. It was revealed in many studies that regulation of telomerase is a multifactorial process in mammalian cells, involving regulation of expression of telomerase subunits coding genes, post-translational protein–protein interactions, and protein phosphorylation. Numerous proto-oncogenes and tumor suppressor genes are engaged in this mechanism, and the complexity of telomerase control is studied in the context of tumor development as well as aging. Additionally, since numerous studies reveal a correlation between short telomeres and increased genome instability or cell mortality, the telomerase control appears to be one of the crucial factors to study in order to improve the cancer diagnostics and therapy or prevention. Interestingly, almost 100 % of adenocarcinoma, including breast cancer cells, expresses telomerase which makes it a good target for telomerase-related therapy. Additionally, telomerase is also supposed to be associated with drug resistance. Thus, targeting the enzyme might result in attenuation of this phenomenon. Moreover, since stem cells existence was reported, it must be considered whether targeting telomerase can bring some serious side effects and result in stem cells viability or their regenerative potential decrease. Thus, we review some molecular mechanisms engaged in therapy based on targeting telomerase in breast cancer cells.
Collapse
Affiliation(s)
- Hanna Holysz
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, ul. Przybyszewskiego 49, 60-355, Poznan, Poland
| | | | | | | |
Collapse
|
31
|
Xu J, McEachern MJ. Long telomeres produced by telomerase-resistant recombination are established from a single source and are subject to extreme sequence scrambling. PLoS Genet 2012; 8:e1003017. [PMID: 23133400 PMCID: PMC3486848 DOI: 10.1371/journal.pgen.1003017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 07/30/2012] [Indexed: 11/18/2022] Open
Abstract
Considerable evidence now supports the idea that the moderate telomere lengthening produced by recombinational telomere elongation (RTE) in a Kluyveromyces lactis telomerase deletion mutant occurs through a roll-and-spread mechanism. However, it is unclear whether this mechanism can account for other forms of RTE that produce much longer telomeres such as are seen in human alternative lengthening of telomere (ALT) cells or in the telomerase-resistant type IIR “runaway” RTE such as occurs in the K. lactis stn1-M1 mutant. In this study we have used mutationally tagged telomeres to examine the mechanism of RTE in an stn1-M1 mutant both with and without telomerase. Our results suggest that the establishment stage of the mutant state in newly generated stn1-M1 ter1-Δ mutants surprisingly involves a first stage of sudden telomere shortening. Our data also show that, as predicted by the roll-and-spread mechanism, all lengthened telomeres in a newly established mutant cell commonly emerge from a single telomere source. However, in sharp contrast to the RTE of telomerase deletion survivors, we show that the RTE of stn1-M1 ter1-Δ cells produces telomeres whose sequences undergo continuous intense scrambling via recombination. While telomerase was not necessary for the long telomeres in stn1-M1 cells, its presence during their establishment was seen to interfere with the amplification of repeats via recombination, a result consistent with telomerase retaining its ability to add repeats during active RTE. Finally, we observed that the presence of active mismatch repair or telomerase had important influences on telomeric amplification and/or instability. Indefinite growth of tumor cells requires a mechanism to maintain telomeres. While most cancers use telomerase for this, some maintain long and heterogeneous telomeres using a recombination-dependent mechanism termed alternative lengthening of telomeres (ALT). What causes ALT and how their long and heterogeneous telomeres form and are maintained are not well understood. In this study, we use mutationally tagged telomeric repeats to probe the mechanisms by which highly elongated telomeres are generated by recombination in an ALT–like yeast mutant. Our data show that most or all lengthened telomeres in a newly established mutant cell are commonly generated by amplifying sequence from a single telomere source. This is consistent with the roll-and-spread model, which proposes that a single circle of telomeric DNA can be the ultimate source of all newly amplified telomeres. Other evidence showed that the telomeres of the mutant are exceptionally dynamic. Rapid terminal deletions preceded telomere elongation at the establishment of the mutant state. Also, patterns of telomeric repeats present in long telomeres became rapidly scrambled. These findings may have implications for the establishment and maintenance of long telomeres in human ALT cells.
Collapse
Affiliation(s)
| | - Michael J. McEachern
- Department of Genetics, Fred Davision Life Science Complex, University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
32
|
Abstract
There has been mounting evidence of a causal role for telomere dysfunction in a number of degenerative disorders. Their manifestations encompass common disease states such as idiopathic pulmonary fibrosis and bone marrow failure. Although these disorders seem to be clinically diverse, collectively they comprise a single syndrome spectrum defined by the short telomere defect. Here we review the manifestations and unique genetics of telomere syndromes. We also discuss their underlying molecular mechanisms and significance for understanding common age-related disease processes.
Collapse
Affiliation(s)
- Mary Armanios
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA.
| | | |
Collapse
|
33
|
Abstract
There has been mounting evidence of a causal role for telomere dysfunction in a number of degenerative disorders. Their manifestations encompass common disease states such as idiopathic pulmonary fibrosis and bone marrow failure. Although these disorders seem to be clinically diverse, collectively they comprise a single syndrome spectrum defined by the short telomere defect. Here we review the manifestations and unique genetics of telomere syndromes. We also discuss their underlying molecular mechanisms and significance for understanding common age-related disease processes.
Collapse
|
34
|
Abstract
There has been mounting evidence of a causal role for telomere dysfunction in a number of degenerative disorders. Their manifestations encompass common disease states such as idiopathic pulmonary fibrosis and bone marrow failure. Although these disorders seem to be clinically diverse, collectively they comprise a single syndrome spectrum defined by the short telomere defect. Here we review the manifestations and unique genetics of telomere syndromes. We also discuss their underlying molecular mechanisms and significance for understanding common age-related disease processes.
Collapse
Affiliation(s)
- Mary Armanios
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA.
| | | |
Collapse
|
35
|
Abstract
Progressive DNA damage and mitochondrial decline are both considered to be prime instigators of natural ageing. Traditionally, these two pathways have been viewed largely in isolation. However, recent studies have revealed a molecular circuit that directly links DNA damage to compromised mitochondrial biogenesis and function via p53. This axis of ageing may account for both organ decline and disease development associated with advanced age and could illuminate a path for the development of relevant therapeutics.
Collapse
|
36
|
Marshall PA, Netzel N, Guintchev JW. Assessing compensation for loss of vacuolar function in Saccharomyces cerevisiae. Can J Microbiol 2012; 58:132-44. [PMID: 22260156 DOI: 10.1139/w11-114] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We analyzed how Saccharomyces cerevisiae cells compensate for the lack of a functional vacuole, an acidic membrane-bound degradative and ion storage compartment. We hypothesized that cells lacking a functional vacuole would compensate for the loss of the functions of the vacuole by altering gene expression and (or) metabolic flux. We used gene expression profiling and Biolog phenotype microarray analysis to determine the compensatory mechanisms of cells lacking vacuolar function. In steady state, vps33 and vps41 cells changed the transcriptional profile of some genes, but no complete pathways were upregulated or downregulated. We treated vps41 cells with calcium to tease out cellular compensation for loss of vacuole function under ionic stress; however, changes in gene expression were not utilized to compensate for loss of vacuole function under stress either, as genes whose transcriptional profiles were changed did not function together in any one cellular process. Phenotype microarray analysis indicated that logarithmically growing vps33 or vps41 cells did not seem to compensate for loss of vacuolar function but instead demonstrated additional pleiotropic phenotypes due to the function of the vacuole. Under rich media conditions, yeast utilize the vacuole to regulate stress, ion response, and peptide degradation. However, loss of the vacuole does not lead to observable compensation mechanisms.
Collapse
Affiliation(s)
- Pamela A Marshall
- Division of Mathematical and Natural Sciences, New College of Interdisciplinary Arts and Sciences, Arizona State University, Phoenix, AZ 85069, USA.
| | | | | |
Collapse
|
37
|
Grandin N, Bailly A, Charbonneau M. Activation of Mrc1, a mediator of the replication checkpoint, by telomere erosion. Biol Cell 2012; 97:799-814. [PMID: 15760303 DOI: 10.1042/bc20040526] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND INFORMATION In budding yeast, the loss of either telomere sequences (in telomerase-negative cells) or telomere capping (in mutants of two telomere end-protection proteins, Cdc13 and Yku) lead, by distinct pathways, to telomeric senescence. After DNA damage, activation of Rad53, which together with Chk1 represents a protein kinase central to all checkpoint pathways, normally requires Rad9, a checkpoint adaptor. RESULTS We report that in telomerase-negative (tlc1Delta) cells, activation of Rad53, although diminished, could still take place in the absence of Rad9. In contrast, Rad9 was essential for Rad53 activation in cells that entered senescence in the presence of functional telomerase, namely in senescent cells bearing mutations in telomere end-protection proteins (cdc13-1 yku70Delta). In telomerase-negative cells deleted for RAD9, Mrc1, another checkpoint adaptor previously implicated in the DNA replication checkpoint, mediated Rad53 activation. Rad9 and Rad53, as well as other DNA damage checkpoint proteins (Mec1, Mec3, Chk1 and Dun1), were required for complete DNA-damage-induced cell-cycle arrest after loss of telomerase function. However, unexpectedly, given the formation of an active Rad53-Mrc1 complex in tlc1Delta rad9Delta cells, Mrc1 did not mediate the cell-cycle arrest elicited by telomerase loss. Finally, we report that Rad9, Mrc1, Dun1 and Chk1 are activated by phosphorylation after telomerase inactivation. CONCLUSIONS These results indicate that loss of telomere capping and loss of telomere sequences, both of which provoke telomeric senescence, are perceived as two distinct types of damages. In contrast with the Rad53-Rad9-mediated cell-cycle arrest that functions in a similar way in both types of telomeric senescence, activation of Rad53-Mrc1 might represent a specific response to telomerase inactivation and/or telomere shortening, the functional significance of which has yet to be uncovered.
Collapse
Affiliation(s)
- Nathalie Grandin
- IFR128 BioSciences Gerland, UMR CNRS no. 5161, Ecole Normale Supérieure de Lyon, 46 allée d'Italie, 69364 Lyon, France
| | | | | |
Collapse
|
38
|
Genome-wide analysis to identify pathways affecting telomere-initiated senescence in budding yeast. G3-GENES GENOMES GENETICS 2011; 1:197-208. [PMID: 22384331 PMCID: PMC3276134 DOI: 10.1534/g3.111.000216] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 06/01/2011] [Indexed: 12/23/2022]
Abstract
In telomerase-deficient yeast cells, like equivalent mammalian cells, telomeres shorten over many generations until a period of senescence/crisis is reached. After this, a small fraction of cells can escape senescence, principally using recombination-dependent mechanisms. To investigate the pathways that affect entry into and recovery from telomere-driven senescence, we combined a gene deletion disrupting telomerase (est1Δ) with the systematic yeast deletion collection and measured senescence characteristics in high-throughput assays. As expected, the vast majority of gene deletions showed no strong effects on entry into/exit from senescence. However, around 200 gene deletions behaving similarly to a rad52Δest1Δ archetype (rad52Δ affects homologous recombination) accelerated entry into senescence, and such cells often could not recover growth. A smaller number of strains similar to a rif1Δest1Δ archetype (rif1Δ affects proteins that bind telomeres) accelerated entry into senescence but also accelerated recovery from senescence. Our genome-wide analysis identifies genes that affect entry into and/or exit from telomere-initiated senescence and will be of interest to those studying telomere biology, replicative senescence, cancer, and ageing. Our dataset is complementary to other high-throughput studies relevant to telomere biology, genetic stability, and DNA damage responses.
Collapse
|
39
|
Recombination can either help maintain very short telomeres or generate longer telomeres in yeast cells with weak telomerase activity. EUKARYOTIC CELL 2011; 10:1131-42. [PMID: 21666075 DOI: 10.1128/ec.05079-11] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Yeast mutants lacking telomerase are able to elongate their telomeres through processes involving homologous recombination. In this study, we investigated telomeric recombination in several mutants that normally maintain very short telomeres due to the presence of a partially functional telomerase. The abnormal colony morphology present in some mutants was correlated with especially short average telomere length and with a requirement for RAD52 for indefinite growth. Better-growing derivatives of some of the mutants were occasionally observed and were found to have substantially elongated telomeres. These telomeres were composed of alternating patterns of mutationally tagged telomeric repeats and wild-type repeats, an outcome consistent with amplification occurring via recombination rather than telomerase. Our results suggest that recombination at telomeres can produce two distinct outcomes in the mutants we studied. In occasional cells, recombination generates substantially longer telomeres, apparently through the roll-and-spread mechanism. However, in most cells, recombination appears limited to helping to maintain very short telomeres. The latter outcome likely represents a simplified form of recombinational telomere maintenance that is independent of the generation and copying of telomeric circles.
Collapse
|
40
|
Abstract
In the budding yeast Saccharomyces cerevisiae, the structure and function of telomeres are maintained by binding proteins, such as Cdc13-Stn1-Ten1 (CST), Yku, and the telomerase complex. Like CST and Yku, telomerase also plays a role in telomere protection or capping. Unlike CST and Yku, however, the underlying molecular mechanism of telomerase-mediated telomere protection remains unclear. In this study, we employed both the CDC13-EST1 fusion gene and the separation-of-function allele est1-D514A to elucidate that Est1 provided a telomere protection pathway that was independent of both the CST and Yku pathways. Est1's ability to convert single-stranded telomeric DNA into a G quadruplex was required for telomerase-mediated telomere protection function. Additionally, Est1 maintained the integrity of telomeres by suppressing the recombination of subtelomeric Y' elements. Our results demonstrate that one major functional role that Est1 brings to the telomerase complex is the capping or protection of telomeres.
Collapse
|
41
|
Cell size regulation during telomere-directed senescence in Saccharomyces cerevisiae. Biosci Biotechnol Biochem 2010; 74:195-8. [PMID: 20057141 DOI: 10.1271/bbb.90627] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
DNA replication without telomerase leads to telomere shortening and induces replicative senescence. We found that in a telomerase-deficient budding yeast mutant, the volume of each telomere-shortened cell increased as its growth capacity decreased, and that this process was associated with changes in vacuolar morphology. Senescence-induced cell expansion required Mec1, a DNA damage-responsive kinase, but not vacuolar SNARE Vam3.
Collapse
|
42
|
Lin YH, Chang CC, Wong CW, Teng SC. Recruitment of Rad51 and Rad52 to short telomeres triggers a Mec1-mediated hypersensitivity to double-stranded DNA breaks in senescent budding yeast. PLoS One 2009; 4:e8224. [PMID: 20011546 PMCID: PMC2790616 DOI: 10.1371/journal.pone.0008224] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2009] [Accepted: 11/16/2009] [Indexed: 01/05/2023] Open
Abstract
Telomere maintenance is required for chromosome stability, and telomeres are typically replicated by the action of telomerase. In both mammalian tumor and yeast cells that lack telomerase, telomeres are maintained by an alternative recombination mechanism. Here we demonstrated that the budding yeast Saccharomyces cerevisiae type I survivors derived from telomerase-deficient cells were hypersensitive to DNA damaging agents. Assays to track telomere lengths and drug sensitivity of telomerase-deficient cells from spore colonies to survivors suggested a correlation between telomere shortening and bleomycin sensitivity. Our genetic studies demonstrated that this sensitivity depends on Mec1, which signals checkpoint activation, leading to prolonged cell-cycle arrest in senescent budding yeasts. Moreover, we also observed that when cells equipped with short telomeres, recruitments of homologous recombination proteins, Rad51 and Rad52, were reduced at an HO-endonuclease-catalyzed double-strand break (DSB), while their associations were increased at chromosome ends. These results suggested that the sensitive phenotype may be attributed to the sequestration of repair proteins to compromised telomeres, thus limiting the repair capacity at bona fide DSB sites.
Collapse
Affiliation(s)
- Yi-Hsuan Lin
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Ching Chang
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chui-Wei Wong
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Shu-Chun Teng
- Department of Microbiology, College of Medicine, National Taiwan University, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
43
|
Kozak ML, Chavez A, Dang W, Berger SL, Ashok A, Guo X, Johnson FB. Inactivation of the Sas2 histone acetyltransferase delays senescence driven by telomere dysfunction. EMBO J 2009; 29:158-70. [PMID: 19875981 DOI: 10.1038/emboj.2009.314] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2009] [Accepted: 09/24/2009] [Indexed: 01/28/2023] Open
Abstract
Changes in telomere chromatin have been linked to cellular senescence, but the underlying mechanisms and impact on lifespan are unclear. We found that inactivation of the Sas2 histone acetyltransferase delays senescence in Saccharomyces cerevisiae telomerase (tlc1) mutants through a homologous recombination-dependent mechanism. Sas2 acetylates histone H4 lysine 16 (H4K16), and telomere shortening in tlc1 mutants was accompanied by a selective and Sas2-dependent increase in subtelomeric H4K16 acetylation. Further, mutation of H4 lysine 16 to arginine, which mimics constitutively deacetylated H4K16, delayed senescence and was epistatic to sas2 deletion, indicating that deacetylated H4K16 mediates the delay caused by sas2 deletion. Sas2 normally prevents the Sir2/3/4 heterochromatin complex from leaving the telomere and spreading to internal euchromatic loci. Senescence was delayed by sir3 deletion, but not sir2 deletion, indicating that senescence delay is mediated by release of Sir3 specifically from the telomere repeats. In contrast, sir4 deletion sped senescence and blocked the delay conferred by sas2 or sir3 deletion. We thus show that manipulation of telomere chromatin modulates senescence caused by telomere shortening.
Collapse
Affiliation(s)
- Marina L Kozak
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104-6100, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Lydall D. Taming the tiger by the tail: modulation of DNA damage responses by telomeres. EMBO J 2009; 28:2174-87. [PMID: 19629039 PMCID: PMC2722249 DOI: 10.1038/emboj.2009.176] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Accepted: 06/03/2009] [Indexed: 11/09/2022] Open
Abstract
Telomeres are by definition stable and inert chromosome ends, whereas internal chromosome breaks are potent stimulators of the DNA damage response (DDR). Telomeres do not, as might be expected, exclude DDR proteins from chromosome ends but instead engage with many DDR proteins. However, the most powerful DDRs, those that might induce chromosome fusion or cell-cycle arrest, are inhibited at telomeres. In budding yeast, many DDR proteins that accumulate most rapidly at double strand breaks (DSBs), have important functions in physiological telomere maintenance, whereas DDR proteins that arrive later tend to have less important functions. Considerable diversity in telomere structure has evolved in different organisms and, perhaps reflecting this diversity, different DDR proteins seem to have distinct roles in telomere physiology in different organisms. Drawing principally on studies in simple model organisms such as budding yeast, in which many fundamental aspects of the DDR and telomere biology have been established; current views on how telomeres harness aspects of DDR pathways to maintain telomere stability and permit cell-cycle division are discussed.
Collapse
Affiliation(s)
- David Lydall
- Centre for Integrated Systems Biology of Ageing and Nutrition, Institute for Ageing and Health, HenryWellcome Laboratory, Newcastle University, Tyne and Wear, UK.
| |
Collapse
|
45
|
A genomewide suppressor and enhancer analysis of cdc13-1 reveals varied cellular processes influencing telomere capping in Saccharomyces cerevisiae. Genetics 2008; 180:2251-66. [PMID: 18845848 DOI: 10.1534/genetics.108.092577] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
In Saccharomyces cerevisiae, Cdc13 binds telomeric DNA to recruit telomerase and to "cap" chromosome ends. In temperature-sensitive cdc13-1 mutants telomeric DNA is degraded and cell-cycle progression is inhibited. To identify novel proteins and pathways that cap telomeres, or that respond to uncapped telomeres, we combined cdc13-1 with the yeast gene deletion collection and used high-throughput spot-test assays to measure growth. We identified 369 gene deletions, in eight different phenotypic classes, that reproducibly demonstrated subtle genetic interactions with the cdc13-1 mutation. As expected, we identified DNA damage checkpoint, nonsense-mediated decay and telomerase components in our screen. However, we also identified genes affecting casein kinase II activity, cell polarity, mRNA degradation, mitochondrial function, phosphate transport, iron transport, protein degradation, and other functions. We also identified a number of genes of previously unknown function that we term RTC, for restriction of telomere capping, or MTC, for maintenance of telomere capping. It seems likely that many of the newly identified pathways/processes that affect growth of budding yeast cdc13-1 mutants will play evolutionarily conserved roles at telomeres. The high-throughput spot-testing approach that we describe is generally applicable and could aid in understanding other aspects of eukaryotic cell biology.
Collapse
|
46
|
Greenall A, Lei G, Swan DC, James K, Wang L, Peters H, Wipat A, Wilkinson DJ, Lydall D. A genome wide analysis of the response to uncapped telomeres in budding yeast reveals a novel role for the NAD+ biosynthetic gene BNA2 in chromosome end protection. Genome Biol 2008; 9:R146. [PMID: 18828915 PMCID: PMC2760873 DOI: 10.1186/gb-2008-9-10-r146] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2008] [Revised: 09/23/2008] [Accepted: 10/01/2008] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Telomeres prevent the ends of eukaryotic chromosomes from being recognized as damaged DNA and protect against cancer and ageing. When telomere structure is perturbed, a co-ordinated series of events promote arrest of the cell cycle so that cells carrying damaged telomeres do not divide. In order to better understand the eukaryotic response to telomere damage, budding yeast strains harboring a temperature sensitive allele of an essential telomere capping gene (cdc13-1) were subjected to a transcriptomic study. RESULTS The genome-wide response to uncapped telomeres in yeast cdc13-1 strains, which have telomere capping defects at temperatures above approximately 27 degrees C, was determined. Telomere uncapping in cdc13-1 strains is associated with the differential expression of over 600 transcripts. Transcripts affecting responses to DNA damage and diverse environmental stresses were statistically over-represented. BNA2, required for the biosynthesis of NAD+, is highly and significantly up-regulated upon telomere uncapping in cdc13-1 strains. We find that deletion of BNA2 and NPT1, which is also involved in NAD+ synthesis, suppresses the temperature sensitivity of cdc13-1 strains, indicating that NAD+ metabolism may be linked to telomere end protection. CONCLUSIONS Our data support the hypothesis that the response to telomere uncapping is related to, but distinct from, the response to non-telomeric double-strand breaks. The induction of environmental stress responses may be a conserved feature of the eukaryotic response to telomere damage. BNA2, which is involved in NAD+ synthesis, plays previously unidentified roles in the cellular response to telomere uncapping.
Collapse
Affiliation(s)
- Amanda Greenall
- Aging Research Laboratories, Institute for Aging and Health, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
- Centre for Integrated Systems Biology of Aging and Nutrition, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Guiyuan Lei
- Centre for Integrated Systems Biology of Aging and Nutrition, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
- School of Mathematics & Statistics, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Daniel C Swan
- Bioinformatics Support Unit, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Katherine James
- Centre for Integrated Systems Biology of Aging and Nutrition, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
- Institute of Human Genetics, International Centre for Life, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Liming Wang
- School of Computing Science, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Heiko Peters
- School of Computing Science, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Anil Wipat
- Centre for Integrated Systems Biology of Aging and Nutrition, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
- Institute of Human Genetics, International Centre for Life, Newcastle University, Newcastle upon Tyne, NE1 3BZ, UK
| | - Darren J Wilkinson
- Centre for Integrated Systems Biology of Aging and Nutrition, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
- School of Mathematics & Statistics, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - David Lydall
- Aging Research Laboratories, Institute for Aging and Health, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
- Centre for Integrated Systems Biology of Aging and Nutrition, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
- Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| |
Collapse
|
47
|
Carlson ME, Silva HS, Conboy IM. Aging of signal transduction pathways, and pathology. Exp Cell Res 2008; 314:1951-61. [PMID: 18474281 PMCID: PMC2572856 DOI: 10.1016/j.yexcr.2008.03.017] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Revised: 03/26/2008] [Accepted: 03/29/2008] [Indexed: 12/15/2022]
Abstract
The major cell signaling pathways, and their specific mechanisms of transduction, have been a subject of investigation for many years. As our understanding of these pathways advances, we find that they are evolutionarily well-conserved not only individually, but also at the level of their crosstalk and signal integration. Productive interactions within the key signal transduction networks determine success in embryonic organogenesis, and postnatal tissue repair throughout adulthood. However, aside from clues revealed through examining age-related degenerative diseases, much remains uncertain about imbalances within these pathways during normal aging. Further, little is known about the molecular mechanisms by which alterations in the major cell signal transduction networks cause age-related pathologies. The aim of this review is to describe the complex interplay between the Notch, TGFbeta, WNT, RTK-Ras and Hh signaling pathways, with a specific focus on the changes introduced within these networks by the aging process, and those typical of age-associated human pathologies.
Collapse
Affiliation(s)
- Morgan E Carlson
- Department of Bioengineering, University of California, Berkeley, Berkeley, California 94720-1762, USA.
| | | | | |
Collapse
|
48
|
|
49
|
The telotype defines the telomere state in Saccharomyces cerevisiae and is inherited as a dominant non-Mendelian characteristic in cells lacking telomerase. Genetics 2008; 178:245-57. [PMID: 18202371 DOI: 10.1534/genetics.107.083030] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Telomeres are an unusual component of the genome because they do not encode genes, but their structure and cellular maintenance machinery (which we define as "telotype") are essential for chromosome stability. Cells can switch between different phenotypic states. One such example is when they switch from maintenance mediated by telomerase (TERT telotype) to one of the two alternative mechanisms of telomere preservation (ALT I and ALT II telotype). The nature of this switch is largely unknown. Reintroduction of telomerase into ALT II, but not ALT I, yeast led to the loss of their ability to survive a second round of telomerase withdrawal. Mating-based genetic analysis of ALT I and II revealed that both types of telomerase-independent telomere maintenance are inherited as a non-Mendelian trait dominant over senescence (SEN telotype). Additionally, inheritance of ALT I and ALT II did not depend on either the mitochondrial genome or a prion-based mechanism. Type I, but not type II, survivor cells exhibited impaired gene silencing, potentially connecting the switch to the ALT telotype epigenetic changes. These data provide evidence that nonprion epigenetic-like mechanisms confer flexibility on cells as a population to adjust to the life-threatening situation of telomerase loss, allowing cells to switch from TERT to ALT telotypes that can sustain viable populations.
Collapse
|
50
|
Ahmed S, Passos JF, Birket MJ, Beckmann T, Brings S, Peters H, Birch-Machin MA, von Zglinicki T, Saretzki G. Telomerase does not counteract telomere shortening but protects mitochondrial function under oxidative stress. J Cell Sci 2008; 121:1046-53. [PMID: 18334557 DOI: 10.1242/jcs.019372] [Citation(s) in RCA: 349] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Telomerase is a ribonucleoprotein that counteracts telomere shortening and can immortalise human cells. There is also evidence for a telomere-independent survival function of telomerase. However, its mechanism is not understood. We show here that TERT, the catalytic subunit of human telomerase, protects human fibroblasts against oxidative stress. While TERT maintains telomere length under standard conditions, telomeres under increased stress shorten as fast as in cells without active telomerase. This is because TERT is reversibly excluded from the nucleus under stress in a dose- and time-dependent manner. Extranuclear telomerase colocalises with mitochondria. In TERT-overexpressing cells, mtDNA is protected, mitochondrial membrane potential is increased and mitochondrial superoxide production and cell peroxide levels are decreased, all indicating improved mitochondrial function and diminished retrograde response. We propose protection of mitochondria under mild stress as a novel function of TERT.
Collapse
Affiliation(s)
- Shaheda Ahmed
- Crucible Laboratory, Institute for Ageing and Health, Newcastle University, Newcastle upon Tyne, NE4 6BE, UK
| | | | | | | | | | | | | | | | | |
Collapse
|