1
|
Lanna A. Unexpected links between cancer and telomere state. Semin Cancer Biol 2025; 110:46-55. [PMID: 39952372 DOI: 10.1016/j.semcancer.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 01/05/2025] [Accepted: 01/22/2025] [Indexed: 02/17/2025]
Abstract
Eukaryotes possess chromosome ends known as telomeres. As telomeres shorten, organisms age, a process defined as senescence. Although uncontrolled telomere lengthening has been naturally connected with cancer developments and immortalized state, many cancers are instead characterized by extremely short, genomically unstable telomeres that may hide cancer cells from immune attack. By contrast, other malignancies feature extremely long telomeres due to absence of 'shelterin' end cap protecting factors. The reason for rampant telomere extension in these cancers had remained elusive. Hence, while telomerase supports tumor progression and escape in cancers with very short telomeres, it is possible that different - transfer based or alternative - lengthening pathways be involved in the early stage of tumorigenesis, when telomere length is intact. In this Review, I hereby discuss recent discoveries in the field of telomeres and highlight unexpected links connecting cancer and telomere state. We hope these parallelisms may inform new therapies to eradicate cancers.
Collapse
Affiliation(s)
- Alessio Lanna
- Sentcell UK laboratories, Tuscany Life Sciences, GSK Vaccine Campus, Siena, Italy; University College London, Division of Medicine, London, United Kingdom; Monte-Carlo, Principality of Monaco, France.
| |
Collapse
|
2
|
Trastus LA, d'Adda di Fagagna F. The complex interplay between aging and cancer. NATURE AGING 2025; 5:350-365. [PMID: 40038418 DOI: 10.1038/s43587-025-00827-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/17/2025] [Indexed: 03/06/2025]
Abstract
Cancer is an age-related disease, but the interplay between cancer and aging is complex and their shared molecular drivers are deeply intertwined. This Review provides an overview of how different biological pathways affect cancer and aging, leveraging evidence mainly derived from animal studies. We discuss how genome maintenance and accumulation of DNA mutations affect tumorigenesis and tissue homeostasis during aging. We describe how age-related telomere dysfunction and cellular senescence intricately modulate tumor development through mechanisms involving genomic instability and inflammation. We examine how an aged immune system and chronic inflammation shape tumor immunosurveillance, fueling DNA damage and cellular senescence. Finally, as animal models are important to untangling the relative contributions of these aging-modulated pathways to cancer progression and to test interventions, we discuss some of the limitations of physiological and accelerated aging models, aiming to improve experimental designs and enhance translation.
Collapse
Affiliation(s)
| | - Fabrizio d'Adda di Fagagna
- IFOM ETS-the AIRC Institute of Molecular Oncology, Milan, Italy.
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche, Pavia, Italy.
| |
Collapse
|
3
|
Dunn PL, Logeswaran D, Chen JJL. Telomerase-Mediated Anti-Ageing Interventions. Subcell Biochem 2024; 107:1-20. [PMID: 39693017 DOI: 10.1007/978-3-031-66768-8_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
The ageing process involves a gradual decline of chromosome integrity throughout an organism's lifespan. Telomeres are protective DNA-protein complexes that cap the ends of linear chromosomes in eukaryotic organisms. Telomeric DNA consists of long stretches of short "TTAGGG" repeats that are conserved across most eukaryotes including humans. Telomeres shorten progressively with each round of DNA replication due to the inability of conventional DNA polymerase to completely replicate the chromosome ends, known as the "end-replication problem". The telomerase enzyme counteracts the telomeric DNA loss by de novo addition of telomeric repeats onto chromosomal ends. Germline and stem cells maintain significant levels of telomerase activity to maintain telomere length and can divide almost indefinitely. However, the differentiation of stem cells accompanies the inactivation of telomerase gene expression, resulting in the progressive shortening of telomeres in somatic cells over successive divisions. Critically short telomeres elicit and sustain a persistent DNA damage response leading to permanent growth arrest of cells known as cellular senescence, a hallmark of cellular ageing. The accumulation of senescent cells in tissues and organs contributes to organismal ageing. Thus, the prevention of telomere shortening is a promising means to delay or even reverse cellular ageing. In this chapter, we summarize potential anti-ageing interventions that mitigate telomere shortening through increasing telomerase level or activity and discuss these strategies' risks, benefits, and future outlooks.
Collapse
Affiliation(s)
- Phoebe L Dunn
- School of Life Sciences, Arizona State University, Tempe, Arizona, USA
| | | | - Julian J-L Chen
- School of Molecular Sciences, Arizona State University, Tempe, Arizona, USA.
| |
Collapse
|
4
|
Borges G, Criqui M, Harrington L. Tieing together loose ends: telomere instability in cancer and aging. Mol Oncol 2022; 16:3380-3396. [PMID: 35920280 PMCID: PMC9490142 DOI: 10.1002/1878-0261.13299] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/27/2022] [Accepted: 08/02/2022] [Indexed: 11/29/2022] Open
Abstract
Telomere maintenance is essential for maintaining genome integrity in both normal and cancer cells. Without functional telomeres, chromosomes lose their protective structure and undergo fusion and breakage events that drive further genome instability, including cell arrest or death. One means by which this loss can be overcome in stem cells and cancer cells is via re-addition of G-rich telomeric repeats by the telomerase reverse transcriptase (TERT). During aging of somatic tissues, however, insufficient telomerase expression leads to a proliferative arrest called replicative senescence, which is triggered when telomeres reach a critically short threshold that induces a DNA damage response. Cancer cells express telomerase but do not entirely escape telomere instability as they often possess short telomeres; hence there is often selection for genetic alterations in the TERT promoter that result in increased telomerase expression. In this review, we discuss our current understanding of the consequences of telomere instability in cancer and aging, and outline the opportunities and challenges that lie ahead in exploiting the reliance of cells on telomere maintenance for preserving genome stability.
Collapse
Affiliation(s)
- Gustavo Borges
- Molecular Biology Programme, Institute for Research in Immunology and CancerUniversity of MontrealQCCanada
| | - Mélanie Criqui
- Molecular Biology Programme, Institute for Research in Immunology and CancerUniversity of MontrealQCCanada
| | - Lea Harrington
- Molecular Biology Programme, Institute for Research in Immunology and CancerUniversity of MontrealQCCanada
- Departments of Medicine and Biochemistry and Molecular MedicineUniversity of MontrealQCCanada
| |
Collapse
|
5
|
Jalali A, Yu K, Beechar V, Bosquez Huerta NA, Grichuk A, Mehra D, Lozzi B, Kong K, Scott KL, Rao G, Bainbridge MN, Bondy ML, Deneen B. POT1 Regulates Proliferation and Confers Sexual Dimorphism in Glioma. Cancer Res 2021; 81:2703-2713. [PMID: 33782098 DOI: 10.1158/0008-5472.can-20-3755] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/10/2021] [Accepted: 03/25/2021] [Indexed: 11/16/2022]
Abstract
Germline POT1 mutations are found in a spectrum of cancers and confer increased risk. Recently, we identified a series of novel germline POT1 mutations that predispose carrier families to the development of glioma. Despite these strong associations, how these glioma-associated POT1 mutations contribute to glioma tumorigenesis remains undefined. Here we show that POT1-G95C increases proliferation in glioma-initiating cells in vitro and in progenitor populations in the developing brain. In a native mouse model of glioma, loss of Pot1a/b resulted in decreased survival in females compared with males. These findings were corroborated in human glioma, where low POT1 expression correlated with decreased survival in females. Transcriptomic and IHC profiling of Pot1a/b-deficient glioma revealed that tumors in females exhibited decreased expression of immune markers and increased expression of cell-cycle signatures. Similar sex-dependent trends were observed in human gliomas that had low expression of POT1. Together, our studies demonstrate context-dependent functions for POT1 mutation or loss in driving progenitor proliferation in the developing brain and sexual dimorphism in glioma. SIGNIFICANCE: This study shows that manipulation of POT1 expression in glioma has sex-specific effects on tumorigenesis and associated immune signatures.
Collapse
Affiliation(s)
- Ali Jalali
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Kwanha Yu
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas.,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Vivek Beechar
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Navish A Bosquez Huerta
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas.,Program in Developmental Biology, Baylor College of Medicine, Houston, Texas
| | - Anthony Grichuk
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Deepika Mehra
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Brittney Lozzi
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Kathleen Kong
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas
| | - Kenneth L Scott
- Department of Human and Molecular Genetics, Baylor College of Medicine, Houston, Texas
| | - Ganesh Rao
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas
| | - Matthew N Bainbridge
- Rady Children's Institute of Genomic Medicine, Rady Children's Hospital-San Diego, California
| | - Melissa L Bondy
- Department of Epidemiology and Population Health, Stanford University, Palo Alto, California
| | - Benjamin Deneen
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas. .,Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas.,Program in Developmental Biology, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
6
|
Schmutz I, Mensenkamp AR, Takai KK, Haadsma M, Spruijt L, de Voer RM, Choo SS, Lorbeer FK, van Grinsven EJ, Hockemeyer D, Jongmans MCJ, de Lange T. TINF2 is a haploinsufficient tumor suppressor that limits telomere length. eLife 2020; 9:e61235. [PMID: 33258446 PMCID: PMC7707837 DOI: 10.7554/elife.61235] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/11/2020] [Indexed: 12/20/2022] Open
Abstract
Telomere shortening is a presumed tumor suppressor pathway that imposes a proliferative barrier (the Hayflick limit) during tumorigenesis. This model predicts that excessively long somatic telomeres predispose to cancer. Here, we describe cancer-prone families with two unique TINF2 mutations that truncate TIN2, a shelterin subunit that controls telomere length. Patient lymphocyte telomeres were unusually long. We show that the truncated TIN2 proteins do not localize to telomeres, suggesting that the mutations create loss-of-function alleles. Heterozygous knock-in of the mutations or deletion of one copy of TINF2 resulted in excessive telomere elongation in clonal lines, indicating that TINF2 is haploinsufficient for telomere length control. In contrast, telomere protection and genome stability were maintained in all heterozygous clones. The data establish that the TINF2 truncations predispose to a tumor syndrome. We conclude that TINF2 acts as a haploinsufficient tumor suppressor that limits telomere length to ensure a timely Hayflick limit.
Collapse
Affiliation(s)
- Isabelle Schmutz
- Laboratory for Cell Biology and Genetics, Rockefeller UniversityNew YorkUnited States
| | - Arjen R Mensenkamp
- Department of Human Genetics, Radboud University Medical CenterNijmegenNetherlands
| | - Kaori K Takai
- Laboratory for Cell Biology and Genetics, Rockefeller UniversityNew YorkUnited States
| | - Maaike Haadsma
- Department of Human Genetics, Radboud University Medical CenterNijmegenNetherlands
| | - Liesbeth Spruijt
- Department of Human Genetics, Radboud University Medical CenterNijmegenNetherlands
| | - Richarda M de Voer
- Department of Human Genetics, Radboud University Medical CenterNijmegenNetherlands
| | - Seunga Sara Choo
- Department of Molecular and Cellular Biology, University of California, BerkeleyBerkeleyUnited States
| | - Franziska K Lorbeer
- Department of Molecular and Cellular Biology, University of California, BerkeleyBerkeleyUnited States
| | - Emma J van Grinsven
- Department of Molecular and Cellular Biology, University of California, BerkeleyBerkeleyUnited States
| | - Dirk Hockemeyer
- Department of Molecular and Cellular Biology, University of California, BerkeleyBerkeleyUnited States
- Chan Zuckerberg BiohubSan FranciscoUnited States
| | | | - Titia de Lange
- Laboratory for Cell Biology and Genetics, Rockefeller UniversityNew YorkUnited States
| |
Collapse
|
7
|
Gong Y, Stock AJ, Liu Y. The enigma of excessively long telomeres in cancer: lessons learned from rare human POT1 variants. Curr Opin Genet Dev 2020; 60:48-55. [PMID: 32155570 DOI: 10.1016/j.gde.2020.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 01/17/2020] [Accepted: 02/02/2020] [Indexed: 01/10/2023]
Abstract
The discovery that rare POT1 variants are associated with extremely long telomeres and increased cancer predisposition has provided a framework to revisit the relationship between telomere length and cancer development. Telomere shortening is linked with increased risk for cancer. However, over the past decade, there is increasing evidence to show that extremely long telomeres caused by mutations in shelterin components (POT1, TPP1, and RAP1) also display an increased risk of cancer. Here, we will review current knowledge on germline mutations of POT1 identified from cancer-prone families. In particular, we will discuss some common features presented by the mutations through structure-function studies. We will further provide an overview of how POT1 mutations affect telomere length regulation and tumorigenesis.
Collapse
Affiliation(s)
- Yi Gong
- Biomedical Research Center, National Institute on Aging/National Institutes of Health, 251 Bayview Blvd, Baltimore, MD, USA.
| | - Amanda J Stock
- Biomedical Research Center, National Institute on Aging/National Institutes of Health, 251 Bayview Blvd, Baltimore, MD, USA
| | - Yie Liu
- Biomedical Research Center, National Institute on Aging/National Institutes of Health, 251 Bayview Blvd, Baltimore, MD, USA.
| |
Collapse
|
8
|
Claude E, Decottignies A. Telomere maintenance mechanisms in cancer: telomerase, ALT or lack thereof. Curr Opin Genet Dev 2020; 60:1-8. [PMID: 32114293 DOI: 10.1016/j.gde.2020.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/10/2020] [Accepted: 01/17/2020] [Indexed: 12/31/2022]
Abstract
Cancer cells acquire replicative immortality by activating a telomere maintenance mechanism (TMM), either the telomerase or the Alternative Lengthening of Telomeres (ALT) mechanism. ALT is frequently activated in tumors derived from mesenchymal cells, which are more frequent in childhood cancers. Recent studies showed that, occasionally, cancer cells can arise without any TMM activation. Here, we discuss the challenge in assessing which TMM is activated in tumors. We also evaluate the prevalence of ALT mechanism in pediatric cancers and review the associated survival prognosis in different tumor types. Finally, we discuss about possible anti-TMM therapies for new emerging cancer treatments.
Collapse
|
9
|
miR-10a suppresses colorectal cancer metastasis by modulating the epithelial-to-mesenchymal transition and anoikis. Cell Death Dis 2017; 8:e2739. [PMID: 28383561 PMCID: PMC5477594 DOI: 10.1038/cddis.2017.61] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 12/28/2016] [Accepted: 01/03/2017] [Indexed: 12/16/2022]
Abstract
MicroRNAs (miRNAs) have a critical role in tumorigenesis and metastasis, which are major obstacles of cancer therapy. However, the role of miRNAs in colorectal cancer (CRC) metastasis remains poorly understood. Here, we found that miRNA-10a (miR-10a) was upregulated in primary CRC tissues and cell line (SW480) derived from primary CRC compared with metastatic cancer tissues in lymph node and cell line (SW620). The differential expression of miR-10a was inversely correlated with distant metastasis and invasion depth. miR-10a promoted migration and invasion in vitro but inhibited metastasis in vivo by regulating the epithelial-to-mesenchymal transition and anoikis. Furthermore, matrix metalloproteinase 14 (MMP14) and actin gamma 1 (ACTG1) were validated as target genes of miR-10a in CRC cells. Ectopic expression of MMP14 and ACTG1 counteracted the decreased cell adhesion and anoikis resistance activities induced by miR-10a. These findings not only describe the mechanism by which miR-10a suppresses CRC metastasis but also suggest the potential prognostic and therapeutic value of miR-10a in CRC patients.
Collapse
|
10
|
Ait-Aissa K, Ebben JD, Kadlec AO, Beyer AM. Friend or foe? Telomerase as a pharmacological target in cancer and cardiovascular disease. Pharmacol Res 2016; 111:422-433. [PMID: 27394166 PMCID: PMC5026584 DOI: 10.1016/j.phrs.2016.07.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/01/2016] [Accepted: 07/02/2016] [Indexed: 12/20/2022]
Abstract
Aging, cancer, and chronic disease have remained at the forefront of basic biological research for decades. Within this context, significant attention has been paid to the role of telomerase, the enzyme responsible for lengthening telomeres, the nucleotide sequences located at the end of chromosomes found in the nucleus. Alterations in telomere length and telomerase activity are a common denominator to the underlying pathology of these diseases. While nuclear-specific, telomere-lengthening effects of telomerase impact cellular/organismal aging and cancer development, non-canonical, extra-nuclear, and non-telomere-lengthening contributions of telomerase have only recently been described and their exact physiological implications are ill defined. Although the mechanism remains unclear, recent reports reveal that the catalytic subunit of telomerase, telomerase reverse transcriptase (TERT), regulates levels of mitochondrial-derived reactive oxygen species (mtROS), independent of its established role in the nucleus. Telomerase inhibition has been the target of chemotherapy (directed or indirectly) for over a decade now, yet no telomerase inhibitor is FDA approved and few are currently in late-stage clinical trials, possibly due to underappreciation of the distinct extra-nuclear functions of telomerase. Moreover, evaluation of telomerase-specific therapies is largely limited to the context of chemotherapy, despite reports of the beneficial effects of telomerase activation in the cardiovascular system in relation to such processes as endothelial dysfunction and myocardial infarction. Thus, there is a need for better understanding of telomerase-focused cell and organism physiology, as well as development of telomerase-specific therapies in relation to cancer and extension of these therapies to cardiovascular pathologies. This review will detail findings related to telomerase and evaluate its potential to serve as a therapeutic target.
Collapse
Affiliation(s)
- Karima Ait-Aissa
- Department of Medicine
- Department of Physiology, Cardiovascular Center
| | - Johnathan D. Ebben
- Department of Pharmacology & Toxicology
- Cancer Center, Medical College of Wisconsin
| | - Andrew O. Kadlec
- Department of Medicine
- Department of Physiology, Cardiovascular Center
| | - Andreas M. Beyer
- Department of Medicine
- Department of Physiology, Cardiovascular Center
| |
Collapse
|
11
|
Yin J, Wan B, Sarkar J, Horvath K, Wu J, Chen Y, Cheng G, Wan K, Chin P, Lei M, Liu Y. Dimerization of SLX4 contributes to functioning of the SLX4-nuclease complex. Nucleic Acids Res 2016; 44:4871-80. [PMID: 27131364 PMCID: PMC4889959 DOI: 10.1093/nar/gkw354] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 04/05/2016] [Accepted: 04/21/2016] [Indexed: 11/16/2022] Open
Abstract
The Fanconi anemia protein SLX4 assembles a genome and telomere maintenance toolkit, consisting of the nucleases SLX1, MUS81 and XPF. Although it is known that SLX4 acts as a scaffold for building this complex, the molecular basis underlying this function of SLX4 remains unclear. Here, we report that functioning of SLX4 is dependent on its dimerization via an oligomerization motif called the BTB domain. We solved the crystal structure of the SLX4BTB dimer, identifying key contacts (F681 and F708) that mediate dimerization. Disruption of BTB dimerization abrogates nuclear foci formation and telomeric localization of not only SLX4 but also of its associated nucleases. Furthermore, dimerization-deficient SLX4 mutants cause defective cellular response to DNA interstrand crosslinking agent and telomere maintenance, underscoring the contribution of BTB domain-mediated dimerization of SLX4 in genome and telomere maintenance.
Collapse
Affiliation(s)
- Jinhu Yin
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21044, USA
| | - Bingbing Wan
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 333 Haike Road, Shanghai 200031, China Shanghai Research Center, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jaya Sarkar
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21044, USA
| | - Kent Horvath
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21044, USA
| | - Jian Wu
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 333 Haike Road, Shanghai 200031, China
| | - Yong Chen
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 333 Haike Road, Shanghai 200031, China
| | - Guangjuan Cheng
- Shanghai Research Center, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ke Wan
- Shanghai Research Center, Chinese Academy of Sciences, Shanghai 200031, China
| | - Peiju Chin
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21044, USA
| | - Ming Lei
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 333 Haike Road, Shanghai 200031, China Department of Biological Chemistry, University of Michigan Medical School, 1150 W. Medical Center Drive, Ann Arbor, MI 48109, USA Shanghai Research Center, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yie Liu
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21044, USA
| |
Collapse
|
12
|
Sarkar J, Liu Y. Fanconi anemia proteins in telomere maintenance. DNA Repair (Amst) 2016; 43:107-12. [PMID: 27118469 DOI: 10.1016/j.dnarep.2016.02.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 02/03/2016] [Indexed: 11/15/2022]
Abstract
Mammalian chromosome ends are protected by nucleoprotein structures called telomeres. Telomeres ensure genome stability by preventing chromosome termini from being recognized as DNA damage. Telomere length homeostasis is inevitable for telomere maintenance because critical shortening or over-lengthening of telomeres may lead to DNA damage response or delay in DNA replication, and hence genome instability. Due to their repetitive DNA sequence, unique architecture, bound shelterin proteins, and high propensity to form alternate/secondary DNA structures, telomeres are like common fragile sites and pose an inherent challenge to the progression of DNA replication, repair, and recombination apparatus. It is conceivable that longer the telomeres are, greater is the severity of such challenges. Recent studies have linked excessively long telomeres with increased tumorigenesis. Here we discuss telomere abnormalities in a rare recessive chromosomal instability disorder called Fanconi Anemia and the role of the Fanconi Anemia pathway in telomere biology. Reports suggest that Fanconi Anemia proteins play a role in maintaining long telomeres, including processing telomeric joint molecule intermediates. We speculate that ablation of the Fanconi Anemia pathway would lead to inadequate aberrant structural barrier resolution at excessively long telomeres, thereby causing replicative burden on the cell.
Collapse
Affiliation(s)
- Jaya Sarkar
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21044, USA
| | - Yie Liu
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institutes of Health, 251 Bayview Blvd, Baltimore, MD 21044, USA.
| |
Collapse
|
13
|
Liu J, Yu C, Hu X, Kim JK, Bierma JC, Jun HI, Rychnovsky SD, Huang L, Qiao F. Dissecting Fission Yeast Shelterin Interactions via MICro-MS Links Disruption of Shelterin Bridge to Tumorigenesis. Cell Rep 2015; 12:2169-80. [PMID: 26365187 PMCID: PMC4591219 DOI: 10.1016/j.celrep.2015.08.043] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/12/2015] [Accepted: 08/11/2015] [Indexed: 11/16/2022] Open
Abstract
Shelterin, a six-member complex, protects telomeres from nucleolytic attack and regulates their elongation by telomerase. Here, we have developed a strategy, called MICro-MS (Mapping Interfaces via Crosslinking-Mass Spectrometry), that combines crosslinking-mass spectrometry and phylogenetic analysis to identify contact sites within the complex. This strategy allowed identification of separation-of-function mutants of fission yeast Ccq1, Poz1, and Pot1 that selectively disrupt their respective interactions with Tpz1. The various telomere dysregulation phenotypes observed in these mutants further emphasize the critical regulatory roles of Tpz1-centered shelterin interactions in telomere homeostasis. Furthermore, the conservation between fission yeast Tpz1-Pot1 and human TPP1-POT1 interactions led us to map a human melanoma-associated POT1 mutation (A532P) to the TPP1-POT1 interface. Diminished TPP1-POT1 interaction caused by hPOT1-A532P may enable unregulated telomere extension, which, in turn, helps cancer cells to achieve replicative immortality. Therefore, our study reveals a connection between shelterin connectivity and tumorigenicity.
Collapse
Affiliation(s)
- Jinqiang Liu
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697-1700, USA
| | - Clinton Yu
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA 92697-4560, USA
| | - Xichan Hu
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697-1700, USA
| | - Jin-Kwang Kim
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697-1700, USA
| | - Jan C Bierma
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697-1700, USA
| | - Hyun-Ik Jun
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697-1700, USA
| | - Scott D Rychnovsky
- Department of Chemistry, University of California, Irvine, Irvine, CA 92697-2025, USA
| | - Lan Huang
- Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, Irvine, CA 92697-4560, USA
| | - Feng Qiao
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA 92697-1700, USA.
| |
Collapse
|
14
|
Sarkar J, Wan B, Yin J, Vallabhaneni H, Horvath K, Kulikowicz T, Bohr VA, Zhang Y, Lei M, Liu Y. SLX4 contributes to telomere preservation and regulated processing of telomeric joint molecule intermediates. Nucleic Acids Res 2015; 43:5912-23. [PMID: 25990736 PMCID: PMC4499145 DOI: 10.1093/nar/gkv522] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 05/07/2015] [Indexed: 11/14/2022] Open
Abstract
SLX4 assembles a toolkit of endonucleases SLX1, MUS81 and XPF, which is recruited to telomeres via direct interaction of SLX4 with TRF2. Telomeres present an inherent obstacle for DNA replication and repair due to their high propensity to form branched DNA intermediates. Here we provide novel insight into the mechanism and regulation of the SLX4 complex in telomere preservation. SLX4 associates with telomeres throughout the cell cycle, peaking in late S phase and under genotoxic stress. Disruption of SLX4's interaction with TRF2 or SLX1 and SLX1's nuclease activity independently causes telomere fragility, suggesting a requirement of the SLX4 complex for nucleolytic resolution of branched intermediates during telomere replication. Indeed, the SLX1-SLX4 complex processes a variety of telomeric joint molecules in vitro. The nucleolytic activity of SLX1-SLX4 is negatively regulated by telomeric DNA-binding proteins TRF1 and TRF2 and is suppressed by the RecQ helicase BLM in vitro. In vivo, in the presence of functional BLM, telomeric circle formation and telomere sister chromatid exchange, both arising out of nucleolytic processing of telomeric homologous recombination intermediates, are suppressed. We propose that the SLX4-toolkit is a telomere accessory complex that, in conjunction with other telomere maintenance proteins, ensures unhindered, but regulated telomere maintenance.
Collapse
Affiliation(s)
- Jaya Sarkar
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institute of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Bingbing Wan
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 333 Haike Road, Shanghai 200031, China Department of Biological Chemistry, University of Michigan Medical School, 1150 W. Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Jinhu Yin
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institute of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Haritha Vallabhaneni
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institute of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Kent Horvath
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institute of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Tomasz Kulikowicz
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institute of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institute of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| | - Yanbin Zhang
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Ming Lei
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institute of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 333 Haike Road, Shanghai 200031, China
| | - Yie Liu
- Laboratory of Molecular Gerontology, National Institute on Aging/National Institute of Health, 251 Bayview Blvd, Baltimore, MD 21224, USA
| |
Collapse
|
15
|
Vallabhaneni H, Zhou F, Maul RW, Sarkar J, Yin J, Lei M, Harrington L, Gearhart PJ, Liu Y. Defective repair of uracil causes telomere defects in mouse hematopoietic cells. J Biol Chem 2015; 290:5502-11. [PMID: 25572391 DOI: 10.1074/jbc.m114.607101] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Uracil in the genome can result from misincorporation of dUTP instead of dTTP during DNA synthesis, and is primarily removed by uracil DNA glycosylase (UNG) during base excision repair. Telomeres contain long arrays of TTAGGG repeats and may be susceptible to uracil misincorporation. Using model telomeric DNA substrates, we showed that the position and number of uracil substitutions of thymine in telomeric DNA decreased recognition by the telomere single-strand binding protein, POT1. In primary mouse hematopoietic cells, uracil was detectable at telomeres, and UNG deficiency further increased uracil loads and led to abnormal telomere lengthening. In UNG-deficient cells, the frequencies of sister chromatid exchange and fragility in telomeres also significantly increased in the absence of telomerase. Thus, accumulation of uracil and/or UNG deficiency interferes with telomere maintenance, thereby underscoring the necessity of UNG-initiated base excision repair for the preservation of telomere integrity.
Collapse
Affiliation(s)
| | - Fang Zhou
- From the Laboratory of Molecular Gerontology
| | - Robert W Maul
- Laboratory of Molecular Biology and Immunology, NIA, National Institutes of Health, Baltimore, Maryland 21224
| | - Jaya Sarkar
- From the Laboratory of Molecular Gerontology
| | - Jinhu Yin
- From the Laboratory of Molecular Gerontology
| | - Ming Lei
- National Center for Protein Science Shanghai, State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031 China
| | - Lea Harrington
- Department of Medicine, Institute for Research in Immunology and Cancer, University of Montréal, Montréal, Québec H3C 3J7, Canada
| | - Patricia J Gearhart
- Laboratory of Molecular Biology and Immunology, NIA, National Institutes of Health, Baltimore, Maryland 21224,
| | - Yie Liu
- From the Laboratory of Molecular Gerontology,
| |
Collapse
|
16
|
Fairlie J, Harrington L. Enforced telomere elongation increases the sensitivity of human tumour cells to ionizing radiation. DNA Repair (Amst) 2014; 25:54-9. [PMID: 25484304 PMCID: PMC4286114 DOI: 10.1016/j.dnarep.2014.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 11/13/2014] [Accepted: 11/17/2014] [Indexed: 10/29/2022]
Abstract
More than 85% of all human cancers possess the ability to maintain chromosome ends, or telomeres, by virtue of telomerase activity. Loss of functional telomeres is incompatible with survival, and telomerase inhibition has been established in several model systems to be a tractable target for cancer therapy. As human tumour cells typically maintain short equilibrium telomere lengths, we wondered if enforced telomere elongation would positively or negatively impact cell survival. We found that telomere elongation beyond a certain length significantly decreased cell clonogenic survival after gamma irradiation. Susceptibility to irradiation was dosage-dependent and increased at telomere lengths exceeding 17kbp despite the fact that all chromosome ends retained telomeric DNA. These data suggest that an optimal telomere length may promote human cancer cell survival in the presence of genotoxic stress.
Collapse
Affiliation(s)
- Jennifer Fairlie
- Wellcome Trust Centre for Cell Biology and Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Mayfield Road, EH9 3JR Edinburgh, UK
| | - Lea Harrington
- Wellcome Trust Centre for Cell Biology and Institute of Evolutionary Biology, School of Biological Sciences, University of Edinburgh, Mayfield Road, EH9 3JR Edinburgh, UK; Institute for Research in Immunology and Cancer, Department of Medicine, University of Montreal, 2950 chemin de Polytechnique, Montreal, Canada H3T 1J4.
| |
Collapse
|
17
|
Zhang Y, Calado R, Rao M, Hong JA, Meeker AK, Dumitriu B, Atay S, McCormick PJ, Garfield SH, Wangsa D, Padilla-Nash HM, Burkett S, Zhang M, Kunst TF, Peterson NR, Xi S, Inchauste S, Altorki NK, Casson AG, Beer DG, Harris CC, Ried T, Young NS, Schrump DS. Telomerase variant A279T induces telomere dysfunction and inhibits non-canonical telomerase activity in esophageal carcinomas. PLoS One 2014; 9:e101010. [PMID: 24983628 PMCID: PMC4077737 DOI: 10.1371/journal.pone.0101010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Accepted: 06/02/2014] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Although implicated in the pathogenesis of several chronic inflammatory disorders and hematologic malignancies, telomerase mutations have not been thoroughly characterized in human cancers. The present study was performed to examine the frequency and potential clinical relevance of telomerase mutations in esophageal carcinomas. METHODS Sequencing techniques were used to evaluate mutational status of telomerase reverse transcriptase (TERT) and telomerase RNA component (TERC) in neoplastic and adjacent normal mucosa from 143 esophageal cancer (EsC) patients. MTS, flow cytometry, time lapse microscopy, and murine xenograft techniques were used to assess proliferation, apoptosis, chemotaxis, and tumorigenicity of EsC cells expressing either wtTERT or TERT variants. Immunoprecipitation, immunoblot, immunofluorescence, promoter-reporter and qRT-PCR techniques were used to evaluate interactions of TERT and several TERT variants with BRG-1 and β-catenin, and to assess expression of cytoskeletal proteins, and cell signaling. Fluorescence in-situ hybridization and spectral karyotyping techniques were used to examine telomere length and chromosomal stability. RESULTS Sequencing analysis revealed one deletion involving TERC (TERC del 341-360), and two non-synonymous TERT variants [A279T (2 homozygous, 9 heterozygous); A1062T (4 heterozygous)]. The minor allele frequency of the A279T variant was five-fold higher in EsC patients compared to healthy blood donors (p<0.01). Relative to wtTERT, A279T decreased telomere length, destabilized TERT-BRG-1-β-catenin complex, markedly depleted β-catenin, and down-regulated canonical Wnt signaling in cancer cells; these phenomena coincided with decreased proliferation, depletion of additional cytoskeletal proteins, impaired chemotaxis, increased chemosensitivity, and significantly decreased tumorigenicity of EsC cells. A279T expression significantly increased chromosomal aberrations in mouse embryonic fibroblasts (MEFs) following Zeocin™ exposure, as well as Li Fraumeni fibroblasts in the absence of pharmacologically-induced DNA damage. CONCLUSIONS A279T induces telomere dysfunction and inhibits non-canonical telomerase activity in esophageal cancer cells. These findings warrant further analysis of A279T expression in esophageal cancers and premalignant esophageal lesions.
Collapse
Affiliation(s)
- Yuwei Zhang
- Thoracic Surgery Section, Thoracic and GI Oncology Branch; National Cancer Institute, Bethesda, Maryland, United States of America
| | - Rodrigo Calado
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, United States of America
| | - Mahadev Rao
- Thoracic Surgery Section, Thoracic and GI Oncology Branch; National Cancer Institute, Bethesda, Maryland, United States of America
| | - Julie A. Hong
- Thoracic Surgery Section, Thoracic and GI Oncology Branch; National Cancer Institute, Bethesda, Maryland, United States of America
| | - Alan K. Meeker
- Departments of Pathology and Oncology, Johns Hopkins University of Medicine, Baltimore, Maryland, United States of America
| | - Bogdan Dumitriu
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, United States of America
| | - Scott Atay
- Thoracic Surgery Section, Thoracic and GI Oncology Branch; National Cancer Institute, Bethesda, Maryland, United States of America
| | - Peter J. McCormick
- Laboratory of Cellular Oncology, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Susan H. Garfield
- Laboratory of Experimental Carcinogenesis, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Danny Wangsa
- Section of Cancer Genomics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Hesed M. Padilla-Nash
- Section of Cancer Genomics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Sandra Burkett
- Comparative Molecular Cytogenetics Core Facility, National Cancer Institute, Frederick, Maryland, United States of America
| | - Mary Zhang
- Thoracic Surgery Section, Thoracic and GI Oncology Branch; National Cancer Institute, Bethesda, Maryland, United States of America
| | - Tricia F. Kunst
- Thoracic Surgery Section, Thoracic and GI Oncology Branch; National Cancer Institute, Bethesda, Maryland, United States of America
| | - Nathan R. Peterson
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, United States of America
| | - Sichuan Xi
- Thoracic Surgery Section, Thoracic and GI Oncology Branch; National Cancer Institute, Bethesda, Maryland, United States of America
| | - Suzanne Inchauste
- Thoracic Surgery Section, Thoracic and GI Oncology Branch; National Cancer Institute, Bethesda, Maryland, United States of America
| | - Nasser K. Altorki
- Department of Thoracic Surgery, Weill Cornell Medical Center, New York, New York, United States of America
| | - Alan G. Casson
- Department of Surgery, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - David G. Beer
- Section of Thoracic Surgery, University of Michigan Medical Center, Ann Arbor, Michigan, United States of America
| | - Curtis C. Harris
- Laboratory of Human Carcinogenesis, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Thomas Ried
- Section of Cancer Genomics, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Neal S. Young
- National Heart, Lung, and Blood Institute, Bethesda, Maryland, United States of America
| | - David S. Schrump
- Thoracic Surgery Section, Thoracic and GI Oncology Branch; National Cancer Institute, Bethesda, Maryland, United States of America
| |
Collapse
|
18
|
Shi J, Yang XR, Ballew B, Rotunno M, Calista D, Fargnoli MC, Ghiorzo P, Bressac-de Paillerets B, Nagore E, Avril MF, Caporaso NE, McMaster ML, Cullen M, Wang Z, Zhang X, Bruno W, Pastorino L, Queirolo P, Banuls-Roca J, Garcia-Casado Z, Vaysse A, Mohamdi H, Riazalhosseini Y, Foglio M, Jouenne F, Hua X, Hyland PL, Yin J, Vallabhaneni H, Chai W, Minghetti P, Pellegrini C, Ravichandran S, Eggermont A, Lathrop M, Peris K, Scarra GB, Landi G, Savage SA, Sampson JN, He J, Yeager M, Goldin LR, Demenais F, Chanock SJ, Tucker MA, Goldstein AM, Liu Y, Landi MT. Rare missense variants in POT1 predispose to familial cutaneous malignant melanoma. Nat Genet 2014; 46:482-486. [PMID: 24686846 PMCID: PMC4056593 DOI: 10.1038/ng.2941] [Citation(s) in RCA: 248] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 03/07/2014] [Indexed: 12/15/2022]
Abstract
Although CDKN2A is the most frequent high-risk melanoma susceptibility gene, the underlying genetic factors for most melanoma-prone families remain unknown. Using whole-exome sequencing, we identified a rare variant that arose as a founder mutation in the telomere shelterin gene POT1 (chromosome 7, g.124493086C>T; p.Ser270Asn) in five unrelated melanoma-prone families from Romagna, Italy. Carriers of this variant had increased telomere lengths and numbers of fragile telomeres, suggesting that this variant perturbs telomere maintenance. Two additional rare POT1 variants were identified in all cases sequenced in two separate Italian families, one variant per family, yielding a frequency for POT1 variants comparable to that for CDKN2A mutations in this population. These variants were not found in public databases or in 2,038 genotyped Italian controls. We also identified two rare recurrent POT1 variants in US and French familial melanoma cases. Our findings suggest that POT1 is a major susceptibility gene for familial melanoma in several populations.
Collapse
Affiliation(s)
- Jianxin Shi
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2]
| | - Xiaohong R Yang
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2]
| | - Bari Ballew
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Melissa Rotunno
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Donato Calista
- Department of Dermatology, Maurizio Bufalini Hospital, Cesena, Italy
| | | | - Paola Ghiorzo
- 1] Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy. [2] Genetics of Rare Hereditary Cancers, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | | | - Eduardo Nagore
- 1] Department of Dermatology, Instituto Valenciano de Oncología, Valencia, Spain. [2] Department of Dermatology, Universidad Católica de Valencia, Valencia, Spain
| | - Marie Francoise Avril
- Université Paris Descartes, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Cochin, Paris, France
| | - Neil E Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Mary L McMaster
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Michael Cullen
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2] Cancer Genomics Research Laboratory, NCI-Frederick, SAIC-Frederick, Inc., Frederick, Maryland, USA
| | - Zhaoming Wang
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2] Cancer Genomics Research Laboratory, NCI-Frederick, SAIC-Frederick, Inc., Frederick, Maryland, USA
| | - Xijun Zhang
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2] Cancer Genomics Research Laboratory, NCI-Frederick, SAIC-Frederick, Inc., Frederick, Maryland, USA
| | - William Bruno
- 1] Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy. [2] Genetics of Rare Hereditary Cancers, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Lorenza Pastorino
- 1] Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy. [2] Genetics of Rare Hereditary Cancers, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Paola Queirolo
- Genetics of Rare Hereditary Cancers, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Jose Banuls-Roca
- Department of Dermatology, Hospital General Universitario de Alicante, Alicante, Spain
| | - Zaida Garcia-Casado
- Laboratory of Molecular Biology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Amaury Vaysse
- 1] INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, France. [2] Université Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
| | - Hamida Mohamdi
- 1] INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, France. [2] Université Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
| | - Yasser Riazalhosseini
- 1] McGill University and Génome Québec Innovation Centre, Montreal, Quebec, Canada. [2] Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | | | | | - Xing Hua
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Paula L Hyland
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Jinhu Yin
- Laboratory of Molecular Gerontology, National Institute on Aging, US National Institutes of Health, US Department of Health and Human Services, Baltimore, Maryland, USA
| | - Haritha Vallabhaneni
- Laboratory of Molecular Gerontology, National Institute on Aging, US National Institutes of Health, US Department of Health and Human Services, Baltimore, Maryland, USA
| | - Weihang Chai
- Section of Medical Sciences, School of Molecular Biosciences, Washington State University, Spokane, Washington, USA
| | - Paola Minghetti
- Department of Dermatology, Maurizio Bufalini Hospital, Cesena, Italy
| | - Cristina Pellegrini
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Sarangan Ravichandran
- SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Simulation, Analysis and Mathematical Modeling Group, Advanced Biomedical Computing Center, Frederick, Maryland, USA
| | - Alexander Eggermont
- 1] Service de Génétique, Gustave Roussy, Villejuif, France. [2] Université Paris-Sud, Kremlin Bicêtre France, Gustave Roussy, Villejuif, France
| | - Mark Lathrop
- 1] McGill University and Génome Québec Innovation Centre, Montreal, Quebec, Canada. [2] Department of Human Genetics, McGill University, Montreal, Quebec, Canada. [3] Fondation Jean Dausset-Centre d'Etude du Polymorphisme Humain (CEPH), Paris, France
| | - Ketty Peris
- Department of Dermatology, University of L'Aquila, L'Aquila, Italy
| | | | - Giorgio Landi
- Department of Dermatology, Maurizio Bufalini Hospital, Cesena, Italy
| | - Sharon A Savage
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Joshua N Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Ji He
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2] Cancer Genomics Research Laboratory, NCI-Frederick, SAIC-Frederick, Inc., Frederick, Maryland, USA
| | - Meredith Yeager
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2] Cancer Genomics Research Laboratory, NCI-Frederick, SAIC-Frederick, Inc., Frederick, Maryland, USA
| | - Lynn R Goldin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Florence Demenais
- 1] INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, France. [2] Université Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Margaret A Tucker
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Alisa M Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Yie Liu
- Laboratory of Molecular Gerontology, National Institute on Aging, US National Institutes of Health, US Department of Health and Human Services, Baltimore, Maryland, USA
| | - Maria Teresa Landi
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2]
| |
Collapse
|
19
|
Cheung HH, Liu X, Canterel-Thouennon L, Li L, Edmonson C, Rennert OM. Telomerase protects werner syndrome lineage-specific stem cells from premature aging. Stem Cell Reports 2014; 2:534-46. [PMID: 24749076 PMCID: PMC3986587 DOI: 10.1016/j.stemcr.2014.02.006] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Revised: 02/20/2014] [Accepted: 02/20/2014] [Indexed: 12/12/2022] Open
Abstract
Werner syndrome (WS) patients exhibit premature aging predominantly in mesenchyme-derived tissues, but not in neural lineages, a consequence of telomere dysfunction and accelerated senescence. The cause of this lineage-specific aging remains unknown. Here, we document that reprogramming of WS fibroblasts to pluripotency elongated telomere length and prevented telomere dysfunction. To obtain mechanistic insight into the origin of tissue-specific aging, we differentiated iPSCs to mesenchymal stem cells (MSCs) and neural stem/progenitor cells (NPCs). We observed recurrence of premature senescence associated with accelerated telomere attrition and defective synthesis of the lagging strand telomeres in MSCs, but not in NPCs. We postulate this “aging” discrepancy is regulated by telomerase. Expression of hTERT or p53 knockdown ameliorated the accelerated aging phenotypein MSC, whereas inhibition of telomerase sensitized NPCs to DNA damage. Our findings unveil a role for telomerase in the protection of accelerated aging in a specific lineage of stem cells. Prevention of premature senescence with corrected telomeres in reprogrammed WS iPSCs Recurrence of premature senescence and telomere dysfunction in WS iPSC-derived MSCs Rescue of premature senescence in WS MSCs by hTERT overexpression or p53 depletion Telomerase protects and prevents NPCs from DNA damage
Collapse
Affiliation(s)
- Hoi-Hung Cheung
- Laboratory of Clinical and Developmental Genomics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA ; School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, N.T., 852 Hong Kong S.A.R
| | - Xiaozhuo Liu
- Laboratory of Clinical and Developmental Genomics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA ; School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, N.T., 852 Hong Kong S.A.R
| | - Lucile Canterel-Thouennon
- Laboratory of Clinical and Developmental Genomics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lu Li
- School of Biomedical Sciences, Faculty of Medicine, the Chinese University of Hong Kong, Shatin, N.T., 852 Hong Kong S.A.R
| | - Catherine Edmonson
- Laboratory of Clinical and Developmental Genomics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Owen M Rennert
- Laboratory of Clinical and Developmental Genomics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
20
|
Abstract
Limitless reproductive potential is one of the hallmarks of cancer cells. This ability is due to the maintenance of telomeres, erosion of which causes cellular senescence or death. While most cancer cells activate telomerase, a telomere-elongating enzyme, it remains elusive as to why cancer cells often maintain shorter telomeres than the cells in the surrounding normal tissues. Here, we show that forced telomere elongation in cancer cells promotes their differentiation in vivo. We elongated the telomeres of human prostate cancer cells that possess short telomeres by enhancing their telomerase activity. The resulting cells had long telomeres and retained the ability to form tumors in nude mice. Strikingly, these tumors exhibited many duct-like structures and reduced N-cadherin expression, reminiscent of well-differentiated adenocarcinoma. These changes were caused by telomere elongation and not by enhanced telomerase activity. Gene expression profiling revealed that tumor formation was accompanied by the expression of innate immune system-related genes, which have been implicated in maintaining tumor cells in an undifferentiated state and poor-prognosis cancers. In tumors derived from the telomere-elongated cells, upregulation of such gene sets is not observed. Our observations suggest a functional contribution of short telomeres to tumor malignancy by regulation of cancer cell differentiation.
Collapse
|
21
|
Gan P, Zha Y, Yao Q, Chen Z, Tan J. miRNA interference-mediated Rap1 gene silencing increases apoptosis in hepatocellular carcinoma cell line HepG2. Shijie Huaren Xiaohua Zazhi 2013; 21:894-898. [DOI: 10.11569/wcjd.v21.i10.894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of repressor/activator protein 1 (Rap1) in nuclear factor-κB (NF-κB)-mediated apoptosis of liver cancer cells.
METHODS: Hepatocellular carcinoma HepG2 cells were divided into three groups: control group, empty plasmid-transfected group, and Rap1 miRNA group. Apoptosis was determined by flow cytometry 72 h after transfection. The expression of Rap1 protein was measured by Western blot, and the expression of NF-κB p65 mRNA was measured by RT-PCR.
RESULTS: Compared to the control group and empty plasmid-transfected group, the level of Rap1 was significantly decreased and apoptosis rate (33.0% ± 5.8% vs 8.2% ± 2.5%, 0.6% ± 0.2%, both P < 0.05) was significantly increased 72 h after miRNA transfection. Cells in the Rap1 miRNA group had a significant reduction in the level of NF-κB p65 mRNA compared to controls.
CONCLUSION: Rap1 miRNA increases apoptosis in hepatocellular carcinoma cell line HepG2 by decreasing the expression of NF-κB p65.
Collapse
|
22
|
Wong L, Unciti-Broceta A, Spitzer M, White R, Tyers M, Harrington L. A yeast chemical genetic screen identifies inhibitors of human telomerase. CHEMISTRY & BIOLOGY 2013; 20:333-40. [PMID: 23521791 PMCID: PMC3650558 DOI: 10.1016/j.chembiol.2012.12.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 12/11/2012] [Accepted: 12/20/2012] [Indexed: 01/13/2023]
Abstract
Telomerase comprises a reverse transcriptase and an internal RNA template that maintains telomeres in many eukaryotes, and it is a well-validated cancer target. However, there is a dearth of small molecules with efficacy against human telomerase in vivo. We developed a surrogate yeast high-throughput assay to identify human telomerase inhibitors. The reversibility of growth arrest induced by active human telomerase was assessed against a library of 678 compounds preselected for bioactivity in S. cerevisiae. Four of eight compounds identified reproducibly restored growth to strains expressing active human telomerase, and three of these four compounds also specifically inhibited purified human telomerase in vitro. These compounds represent probes for human telomerase function, and potential entry points for development of lead compounds against telomerase-positive cancers.
Collapse
Affiliation(s)
- Lai Hong Wong
- Wellcome Trust Centre for Cell Biology, King’s Buildings, University of Edinburgh, Mayfield Road, Edinburgh, EH9 3JR, UK
| | - Asier Unciti-Broceta
- Edinburgh Cancer Research UK Centre, Medical Research Council Institute of Genetics and Molecular Medicine, University of Edinburgh, Crewe Road South, Edinburgh, EH4 2XR, UK
| | - Michaela Spitzer
- Wellcome Trust Centre for Cell Biology, King’s Buildings, University of Edinburgh, Mayfield Road, Edinburgh, EH9 3JR, UK
| | - Rachel White
- Wellcome Trust Centre for Cell Biology, King’s Buildings, University of Edinburgh, Mayfield Road, Edinburgh, EH9 3JR, UK
| | - Mike Tyers
- Wellcome Trust Centre for Cell Biology, King’s Buildings, University of Edinburgh, Mayfield Road, Edinburgh, EH9 3JR, UK
- Faculty of Medicine, University of Montreal, Institute for Research in Immunology and Cancer, Chemin de Polytechnique, Montreal, Quebec, H3T 1J4 Canada
| | - Lea Harrington
- Wellcome Trust Centre for Cell Biology, King’s Buildings, University of Edinburgh, Mayfield Road, Edinburgh, EH9 3JR, UK
- Faculty of Medicine, University of Montreal, Institute for Research in Immunology and Cancer, Chemin de Polytechnique, Montreal, Quebec, H3T 1J4 Canada
| |
Collapse
|