1
|
Salgado S, Abreu PL, Moleirinho B, Guedes DS, Larcombe L, Azzalin CM. Human PC4 supports telomere stability and viability in cells utilizing the alternative lengthening of telomeres mechanism. EMBO Rep 2024; 25:5294-5315. [PMID: 39468351 PMCID: PMC11624207 DOI: 10.1038/s44319-024-00295-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/30/2024] Open
Abstract
Cancer cells with an activated Alternative Lengthening of Telomeres (ALT) mechanism elongate telomeres via homology-directed repair. Sustained telomeric replication stress is an essential trigger of ALT activity; however, it can lead to cell death if not properly restricted. By analyzing publicly available data from genome-wide CRISPR KO screenings, we have identified the multifunctional protein PC4 as a novel factor essential for ALT cell viability. Depletion of PC4 results in rapid ALT cell death, while telomerase-positive cells show minimal effects. PC4 depletion induces replication stress and telomere fragility primarily in ALT cells, and increases ALT activity. PC4 binds to telomeric DNA in cells, and its binding can be enhanced by telomeric replication stress. Finally, a mutant PC4 with partly impaired single stranded DNA binding activity is capable to localize to telomeres and suppress ALT activity and telomeric replication stress. We propose that PC4 supports ALT cell viability, at least partly, by averting telomere dysfunction. Further studies of PC4 interactions at ALT telomeres may hold promise for innovative therapies to eradicate ALT cancers.
Collapse
Affiliation(s)
- Sara Salgado
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035, Lisbon, Portugal
| | - Patricia L Abreu
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035, Lisbon, Portugal
| | - Beatriz Moleirinho
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035, Lisbon, Portugal
| | - Daniela S Guedes
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035, Lisbon, Portugal
| | - Lee Larcombe
- Apexomic, Stevenage Bioscience Catalyst, Hertfordshire, SG1 2FX, UK
- TessellateBio Ltd, Stevenage Bioscience Catalyst, Hertfordshire, SG1 2FX, UK
| | - Claus M Azzalin
- GIMM - Gulbenkian Institute for Molecular Medicine, 1649-035, Lisbon, Portugal.
| |
Collapse
|
2
|
Clatterbuck Soper SF, Walker RL, Pineda MA, Zhu YJ, Dalgleish JLT, Wang J, Meltzer PS. Cancer-associated DAXX mutations reveal a critical role for ATRX localization in ALT suppression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.18.624165. [PMID: 39803578 PMCID: PMC11722231 DOI: 10.1101/2024.11.18.624165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
To maintain genome stability, proliferating cells must enact a program of telomere maintenance. While most tumors maintain telomeres through the action of telomerase, a subset of tumors utilize a DNA-templated process termed Alternative Lengthening of Telomeres or ALT. ALT is associated with mutations in the ATRX/DAXX/H3.3 histone chaperone complex, which is responsible for deposition of non-replicative histone variant H3.3 at heterochromatic regions of the genome including telomeres. We wished to better understand the role DAXX plays in ALT suppression, and to determine which disease-associated DAXX mutations are unable to suppress ALT. To answer this question, we have leveraged the G292 cell line, in which ATRX is wild type but DAXX has undergone a fusion event with the non-canonical kinesin KIFC3. Restoration of wild-type DAXX in G292 localizes ATRX and abrogates ALT. Using this model system, we tested the ability of a panel of disease-associated DAXX missense variants to suppress ALT. Missense mutations in the ATRX binding domain, the histone binding domain, and the C-terminal SUMO interaction motif reduce the ability of DAXX to suppress ALT. Unexpectedly, we find that mutations in the DAXX histone binding domain lead to failure of ATRX localization. We conclude that a key function of DAXX in ALT suppression is the localization of ATRX to nuclear foci.
Collapse
Affiliation(s)
- Sarah F. Clatterbuck Soper
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Robert L. Walker
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Marbin A. Pineda
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Yuelin J. Zhu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - James L. T. Dalgleish
- Current address: MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS
| | - Jasmine Wang
- Current address: Sidney Kimmel Medical College, Thomas Jefferson University, 1025 Walnut St. #100, Philadelphia, PA 19107
| | - Paul S. Meltzer
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
3
|
Mu Y, Liu H, Luo A, Zhang Q. KIFC3 promotes the progression of non-small cell lung cancer cells through the PI3K/Akt pathway. Thorac Cancer 2024; 15:2356-2364. [PMID: 39390964 PMCID: PMC11586134 DOI: 10.1111/1759-7714.15465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND Kinesin family member C3 (KIFC3), as reported, plays important roles in several tumor types. Nevertheless, it is unknown whether KIFC3 has effects on non-small cell lung cancer (NSCLC) development. MATERIALS AND METHODS KIFC3 expression was detected by RT-PCR, and its correlation with prognosis was analyzed by GEPIA website. Small interfering RNA against KIFC3 were adopted for modulating KIFC3 expression in NSCLC cells. KIFC3 effects on NSCLC cell proliferation were determined using the MTT and clone formation assay. Matrigel invasion and wound healing assays were adopted for measuring the invasion and migration capability of NSCLC cells. Western blot was applied for measuring the levels of proteins associated with the phosphatidylinositol-3-kinase/protein kinase B (PI3K/Akt) pathway in NSCLC cells. RESULTS KIFC3 was markedly increased in NSCLC samples and cells. KIFC3 knockdown suppressed the proliferation, invasion, and migration in NSCLC. Mechanically, KIFC3 silencing suppressed NSCLC progression through inhibiting the PI3K/Akt pathway. CONCLUSIONS KIFC3 lack suppressed the proliferation, invasion, and migration which works, at least partially, by the PI3K/Akt pathway. These findings suggest that targeting KIFC3 via the PI3K/Akt pathway may offer a novel therapeutic strategy for NSCLC.
Collapse
Affiliation(s)
- Yu Mu
- School of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Haoxiang Liu
- School of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Anni Luo
- School of Traditional Chinese MedicineTexas Health and Science UniversityAustinTexasUSA
| | - Qingxiang Zhang
- School of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
4
|
Ma Y, Zhang Y, Jiang X, Guan J, Wang H, Zhang J, Tong Y, Qiu X, Zhou R. KIFC3 promotes the proliferation, migration and invasion of non-small cell lung cancer through the PI3K/AKT signaling pathway. Sci Rep 2024; 14:20471. [PMID: 39227687 PMCID: PMC11372156 DOI: 10.1038/s41598-024-71602-0] [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: 03/22/2024] [Accepted: 08/29/2024] [Indexed: 09/05/2024] Open
Abstract
KIFC3 is a member of the Kinesin superfamily proteins (KIFs). The role of KIFC3 in non-small cell lung cancer (NSCLC) is unknown. This study aimed to elucidate the function of KIFC3 in NSCLC and the underlying mechanism. Immunohistochemistry indicated that KIFC3 was highly expressed in NSCLC tissues and correlated with the degree of differentiation, tumor size, lymph node metastasis and TNM stage. MTT, colony formation and Transwell assays demonstrated that KIFC3 overexpression promoted the proliferation, migration and invasion of NSCLC cells in vitro, while KIFC3 knockdown led to the opposite results. The protein expression levels of PI3Kp85α and p-Akt were increased after KIFC3 overexpression, meanwhile the downstream protein expression levels such as cyclin D1, CDK4, CDK6, RhoA, RhoC and MMP2 were increased. This promotion effect could be inhibited by a specific inhibitor of the PI3K/Akt pathway, LY294002. Co-immunoprecipitation assays confirmed the interaction between endogenous/exogenous KIFC3 and PI3Kp85α. Tumor formation experiments in nude mice confirmed that KIFC3 overexpression promoted the proliferation, migration and invasion of NSCLC cells in vivo and performed its biological function through the PI3K/Akt signaling pathway.In conclusion, KIFC3 promotes the malignant behavior of NSCLC cells through the PI3K/Akt signaling pathway.
Collapse
Affiliation(s)
- Yue Ma
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yao Zhang
- Department of Pathology, China Medical University, 77 Puhe Road, North Shenyang New Area, Shenyang, 110122, Liaoning, China
| | - Xizi Jiang
- Department of Pathology, China Medical University, 77 Puhe Road, North Shenyang New Area, Shenyang, 110122, Liaoning, China
| | - Jingqian Guan
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Huanxi Wang
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jiameng Zhang
- Department of Pathology, China Medical University, 77 Puhe Road, North Shenyang New Area, Shenyang, 110122, Liaoning, China
| | - Yue Tong
- Department of Pathology, China Medical University, 77 Puhe Road, North Shenyang New Area, Shenyang, 110122, Liaoning, China
| | - Xueshan Qiu
- Department of Pathology, China Medical University, 77 Puhe Road, North Shenyang New Area, Shenyang, 110122, Liaoning, China.
| | - Renyi Zhou
- Department of Orthopedics, First Hospital of China Medical University, No.155 Nan Jing North Street, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
5
|
Tlemsani C, Heske CM, Elloumi F, Pongor L, Khandagale P, Varma S, Luna A, Meltzer PS, Khan J, Reinhold WC, Pommier Y. Sarcoma_CellminerCDB: A tool to interrogate the genomic and functional characteristics of a comprehensive collection of sarcoma cell lines. iScience 2024; 27:109781. [PMID: 38868205 PMCID: PMC11167437 DOI: 10.1016/j.isci.2024.109781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/28/2023] [Accepted: 04/15/2024] [Indexed: 06/14/2024] Open
Abstract
Sarcomas are a diverse group of rare malignancies composed of multiple different clinical and molecular subtypes. Due to their rarity and heterogeneity, basic, translational, and clinical research in sarcoma has trailed behind that of other cancers. Outcomes for patients remain generally poor due to an incomplete understanding of disease biology and a lack of novel therapies. To address some of the limitations impeding preclinical sarcoma research, we have developed Sarcoma_CellMinerCDB, a publicly available interactive tool that merges publicly available sarcoma cell line data and newly generated omics data to create a comprehensive database of genomic, transcriptomic, methylomic, proteomic, metabolic, and pharmacologic data on 133 annotated sarcoma cell lines. The reproducibility, functionality, biological relevance, and therapeutic applications of Sarcoma_CellMinerCDB described herein are powerful tools to address and generate biological questions and test hypotheses for translational research. Sarcoma_CellMinerCDB (https://discover.nci.nih.gov/SarcomaCellMinerCDB) aims to contribute to advancing the preclinical study of sarcoma.
Collapse
Affiliation(s)
- Camille Tlemsani
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Department of Medical Oncology, Cochin Hospital, Paris Cancer Institute CARPEM, Université Paris Cité, APHP. Centre, Paris, France
- Institut Cochin, INSERM U1016, CNRS UMR8104, Paris Cancer Institute CARPEM, Université Paris Cité, Paris, France
| | - Christine M. Heske
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Fathi Elloumi
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Lorinc Pongor
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Hungarian Centre of Excellence for Molecular Medicine, Cancer Genomics and Epigenetics Core Group, Szeged, Hungary
| | - Prashant Khandagale
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Sudhir Varma
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Augustin Luna
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
- Computational Biology Branch, National Library of Medicine, NIH, Bethesda, Maryland 20892, USA
| | - Paul S. Meltzer
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - William C. Reinhold
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yves Pommier
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
6
|
Gambelli A, Ferrando A, Boncristiani C, Schoeftner S. Regulation and function of R-loops at repetitive elements. Biochimie 2023; 214:141-155. [PMID: 37619810 DOI: 10.1016/j.biochi.2023.08.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 08/13/2023] [Accepted: 08/19/2023] [Indexed: 08/26/2023]
Abstract
R-loops are atypical, three-stranded nucleic acid structures that contain a stretch of RNA:DNA hybrids and an unpaired, single stranded DNA loop. R-loops are physiological relevant and can act as regulators of gene expression, chromatin structure, DNA damage repair and DNA replication. However, unscheduled and persistent R-loops are mutagenic and can mediate replication-transcription conflicts, leading to DNA damage and genome instability if left unchecked. Detailed transcriptome analysis unveiled that 85% of the human genome, including repetitive regions, hold transcriptional activity. This anticipates that R-loops management plays a central role for the regulation and integrity of genomes. This function is expected to have a particular relevance for repetitive sequences that make up to 75% of the human genome. Here, we review the impact of R-loops on the function and stability of repetitive regions such as centromeres, telomeres, rDNA arrays, transposable elements and triplet repeat expansions and discuss their relevance for associated pathological conditions.
Collapse
Affiliation(s)
- Alice Gambelli
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Via E. Weiss 2, 34127, Trieste, Italy
| | - Alessandro Ferrando
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Via E. Weiss 2, 34127, Trieste, Italy
| | - Chiara Boncristiani
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Via E. Weiss 2, 34127, Trieste, Italy
| | - Stefan Schoeftner
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, Via E. Weiss 2, 34127, Trieste, Italy.
| |
Collapse
|
7
|
Liu H, Xu C, Diplas BH, Brown A, Strickland LM, Yao H, Ling J, McLendon RE, Keir ST, Ashley DM, He Y, Waitkus MS. Cancer-associated SMARCAL1 loss-of-function mutations promote alternative lengthening of telomeres and tumorigenesis in telomerase-negative glioblastoma cells. Neuro Oncol 2023; 25:1563-1575. [PMID: 36689342 PMCID: PMC10484176 DOI: 10.1093/neuonc/noad022] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Telomere maintenance mechanisms are required to enable the replicative immortality of malignant cells. While most cancers activate the enzyme telomerase, a subset of cancers uses telomerase-independent mechanisms termed alternative lengthening of telomeres (ALT). ALT occurs via homology-directed-repair mechanisms and is frequently associated with ATRX mutations. We previously showed that a subset of adult glioblastoma (GBM) patients with ATRX-expressing ALT-positive tumors harbored loss-of-function mutations in the SMARCAL1 gene, which encodes an annealing helicase involved in replication fork remodeling and the resolution of replication stress. However, the causative relationship between SMARCAL1 deficiency, tumorigenesis, and de novo telomere synthesis is not understood. METHODS We used a patient-derived ALT-positive GBM cell line with native SMARCAL1 deficiency to investigate the role of SMARCAL1 in ALT-mediated de novo telomere synthesis, replication stress, and gliomagenesis in vivo. RESULTS Inducible rescue of SMARCAL1 expression suppresses ALT indicators and inhibits de novo telomere synthesis in GBM and osteosarcoma cells, suggesting that SMARCAL1 deficiency plays a functional role in ALT induction in cancers that natively lack SMARCAL1 function. SMARCAL1-deficient ALT-positive cells can be serially propagated in vivo in the absence of detectable telomerase activity, demonstrating that the SMARCAL1-deficient ALT phenotype maintains telomeres in a manner that promotes tumorigenesis. CONCLUSIONS SMARCAL1 deficiency is permissive to ALT and promotes gliomagenesis. Inducible rescue of SMARCAL1 in ALT-positive cell lines permits the dynamic modulation of ALT activity, which will be valuable for future studies aimed at understanding the mechanisms of ALT and identifying novel anticancer therapeutics that target the ALT phenotype.
Collapse
Affiliation(s)
- Heng Liu
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Cheng Xu
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Bill H Diplas
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Alexandrea Brown
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Laura M Strickland
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Haipei Yao
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Jinjie Ling
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Roger E McLendon
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Stephen T Keir
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - David M Ashley
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Yiping He
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Pathology, Duke University Medical Center, Durham, North Carolina, USA
| | - Matthew S Waitkus
- The Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina, USA
- Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
8
|
Reiss M, Keegan J, Aldrich A, Lyons SM, Flynn RL. The exoribonuclease XRN2 mediates degradation of the long non-coding telomeric RNA TERRA. FEBS Lett 2023; 597:1818-1836. [PMID: 37191774 PMCID: PMC10524182 DOI: 10.1002/1873-3468.14639] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/01/2023] [Accepted: 05/05/2023] [Indexed: 05/17/2023]
Abstract
The telomeric repeat-containing RNA, TERRA, associates with both telomeric DNA and telomeric proteins, often forming RNA:DNA hybrids (R-loops). TERRA is most abundant in cancer cells utilizing the alternative lengthening of telomeres (ALT) pathway for telomere maintenance, suggesting that persistent TERRA R-loops may contribute to activation of the ALT mechanism. Therefore, we sought to identify the enzyme(s) that regulate TERRA metabolism in mammalian cells. Here, we identify that the 5'-3' exoribonuclease XRN2 regulates the stability of TERRA RNA. Moreover, while stabilization of TERRA alone was insufficient to drive ALT, depletion of XRN2 in ALT-positive cells led to a significant increase in TERRA R-loops and exacerbated ALT activity. Together, our findings highlight XRN2 as a key determinant of TERRA metabolism and telomere stability in cancer cells that rely on the ALT pathway.
Collapse
Affiliation(s)
- Matthew Reiss
- Departments of Pharmacology and Experimental Therapeutics, and Medicine, Cancer Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Joshua Keegan
- Departments of Pharmacology and Experimental Therapeutics, and Medicine, Cancer Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Anne Aldrich
- Departments of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Shawn M. Lyons
- Departments of Biochemistry and Cell Biology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Rachel Litman Flynn
- Departments of Pharmacology and Experimental Therapeutics, and Medicine, Cancer Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
9
|
Li F, Wang Y, Hwang I, Jang JY, Xu L, Deng Z, Yu EY, Cai Y, Wu C, Han Z, Huang YH, Huang X, Zhang L, Yao J, Lue NF, Lieberman PM, Ying H, Paik J, Zheng H. Histone demethylase KDM2A is a selective vulnerability of cancers relying on alternative telomere maintenance. Nat Commun 2023; 14:1756. [PMID: 36991019 PMCID: PMC10060224 DOI: 10.1038/s41467-023-37480-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
Telomere length maintenance is essential for cellular immortalization and tumorigenesis. 5% - 10% of human cancers rely on a recombination-based mechanism termed alternative lengthening of telomeres (ALT) to sustain their replicative immortality, yet there are currently no targeted therapies. Through CRISPR/Cas9-based genetic screens in an ALT-immortalized isogenic cellular model, here we identify histone lysine demethylase KDM2A as a molecular vulnerability selectively for cells contingent on ALT-dependent telomere maintenance. Mechanistically, we demonstrate that KDM2A is required for dissolution of the ALT-specific telomere clusters following recombination-directed telomere DNA synthesis. We show that KDM2A promotes de-clustering of ALT multitelomeres through facilitating isopeptidase SENP6-mediated SUMO deconjugation at telomeres. Inactivation of KDM2A or SENP6 impairs post-recombination telomere de-SUMOylation and thus dissolution of ALT telomere clusters, leading to gross chromosome missegregation and mitotic cell death. These findings together establish KDM2A as a selective molecular vulnerability and a promising drug target for ALT-dependent cancers.
Collapse
Affiliation(s)
- Fei Li
- Department of Neurosurgery, Southwest Hospital, Chongqing, 400038, China
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Yizhe Wang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Inah Hwang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Ja-Young Jang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Libo Xu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
- Key Laboratory of Pathobiology, Ministry of Education, and Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Zhong Deng
- The Wistar Institute, Philadelphia, PA, 19104, USA
| | - Eun Young Yu
- Department of Microbiology and Immunology, W. R. Hearst Microbiology Research Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Yiming Cai
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Caizhi Wu
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Zhenbo Han
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yu-Han Huang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Xiangao Huang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Ling Zhang
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
- Key Laboratory of Pathobiology, Ministry of Education, and Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Jun Yao
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Neal F Lue
- Department of Microbiology and Immunology, W. R. Hearst Microbiology Research Center, Weill Cornell Medicine, New York, NY, 10065, USA
| | | | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jihye Paik
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
| | - Hongwu Zheng
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, 10065, USA.
| |
Collapse
|
10
|
Clatterbuck Soper SF, Meltzer PS. ATRX/DAXX: Guarding the Genome against the Hazards of ALT. Genes (Basel) 2023; 14:genes14040790. [PMID: 37107548 PMCID: PMC10137841 DOI: 10.3390/genes14040790] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Proliferating cells must enact a telomere maintenance mechanism to ensure genomic stability. In a subset of tumors, telomeres are maintained not by telomerase, but through a homologous recombination-based mechanism termed Alternative Lengthening of Telomeres or ALT. The ALT process is linked to mutations in the ATRX/DAXX/H3.3 histone chaperone complex. This complex is responsible for depositing non-replicative histone variant H3.3 at pericentric and telomeric heterochromatin but has also been found to have roles in ameliorating replication in repeat sequences and in promoting DNA repair. In this review, we will discuss ways in which ATRX/DAXX helps to protect the genome, and how loss of this complex allows ALT to take hold.
Collapse
|
11
|
Li S, Zhang H, Liu J, Shang G. Targeted therapy for osteosarcoma: a review. J Cancer Res Clin Oncol 2023:10.1007/s00432-023-04614-4. [PMID: 36807762 DOI: 10.1007/s00432-023-04614-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 01/27/2023] [Indexed: 02/21/2023]
Abstract
BACKGROUND Osteosarcoma is a common primary malignant tumour of the bone that usually occurs in children and adolescents. It is characterised by difficult treatment, recurrence and metastasis, and poor prognosis. Currently, the treatment of osteosarcoma is mainly based on surgery and auxiliary chemotherapy. However, for recurrent and some primary osteosarcoma cases, owing to the rapid progression of disease and chemotherapy resistance, the effects of chemotherapy are poor. With the rapid development of tumour-targeted therapy, molecular-targeted therapy for osteosarcoma has shown promise. PURPOSE In this paper, we review the molecular mechanisms, related targets, and clinical applications of targeted osteosarcoma therapy. In doing this, we provide a summary of recent literature on the characteristics of targeted osteosarcoma therapy, the advantages of its clinical application, and development of targeted therapy in future. We aim to provide new insights into the treatment of osteosarcoma. CONCLUSION Targeted therapy shows potential in the treatment of osteosarcoma and may offer an important means of precise and personalised treatment in the future, but drug resistance and adverse effects may limit its application.
Collapse
Affiliation(s)
- Shizhe Li
- Department of Bone and Soft Tissue Oncology, Shengjing Hospital Affiliated to China Medical University, Shenyang, 110022, Liaoning Province, China.,Graduate School, Jinzhou Medical University, Jinzhou, 121001, Liaoning Province, China
| | - He Zhang
- Department of Bone and Soft Tissue Oncology, Shengjing Hospital Affiliated to China Medical University, Shenyang, 110022, Liaoning Province, China
| | - Jinxin Liu
- Department of Bone and Soft Tissue Oncology, Shengjing Hospital Affiliated to China Medical University, Shenyang, 110022, Liaoning Province, China
| | - Guanning Shang
- Department of Bone and Soft Tissue Oncology, Shengjing Hospital Affiliated to China Medical University, Shenyang, 110022, Liaoning Province, China.
| |
Collapse
|
12
|
Chung YC, Bisht M, Thuma J, Tu LC. Single-chromosome dynamics reveals locus-dependent dynamics and chromosome territory orientation. J Cell Sci 2023; 136:jcs260137. [PMID: 36718642 PMCID: PMC10022681 DOI: 10.1242/jcs.260137] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 01/19/2023] [Indexed: 02/01/2023] Open
Abstract
Dynamic chromatin organization instantly influences DNA accessibility through modulating local macromolecular density and interactions, driving changes in transcription activities. Chromatin dynamics have been reported to be locally confined but contribute to coherent chromatin motion across the entire nucleus. However, the regulation of dynamics, nuclear orientation and compaction of subregions along a single chromosome are not well-understood. We used CRISPR-based real-time single-particle tracking and polymer models to characterize the dynamics of specific genomic loci and determine compaction levels of large human chromosomal domains. Our studies showed that chromosome compaction changed during interphase and that compactions of two arms on chromosome 19 were different. The dynamics of genomic loci were subdiffusive and dependent on chromosome regions and transcription states. Surprisingly, the correlation between locus-dependent nuclear localization and mobility was negligible. Strong tethering interactions detected at the pericentromeric region implies local condensation or associations with organelles within local nuclear microenvironments, such as chromatin-nuclear body association. Based on our findings, we propose a 'guided radial model' for the nuclear orientation of the long arm of chromosome 19.
Collapse
Affiliation(s)
- Yu-Chieh Chung
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
| | - Madhoolika Bisht
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
- Department of Molecular Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Jenna Thuma
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, USA
| | - Li-Chun Tu
- Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH 43210, USA
- Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH 43210, USA
- Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
13
|
Histone demethylase KDM2A is a selective vulnerability of cancers relying on alternative telomere maintenance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.10.528023. [PMID: 36798426 PMCID: PMC9934630 DOI: 10.1101/2023.02.10.528023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Telomere length maintenance is essential for cellular immortalization and tumorigenesis. 5% - 10% of human cancers rely on a recombination-based mechanism termed alternative lengthening of telomeres (ALT) to sustain their replicative immortality, yet there are currently no targeted therapies. Through CRISPR/Cas9-based genetic screens in an ALT-immortalized isogenic cellular model, here we identify histone lysine demethylase KDM2A as a molecular vulnerability selectively for cells contingent on ALT-dependent telomere maintenance. Mechanistically, we demonstrate that KDM2A is required for dissolution of the ALT-specific telomere clusters following homology-directed telomere DNA synthesis. We show that KDM2A promotes de-clustering of ALT multitelomeres through facilitating isopeptidase SENP6-mediated SUMO deconjugation at telomeres. Inactivation of KDM2A or SENP6 impairs post-recombination telomere de-SUMOylation and thus dissolution of ALT telomere clusters, leading to gross chromosome missegregation and mitotic cell death. These findings together establish KDM2A as a selective molecular vulnerability and a promising drug target for ALT-dependent cancers.
Collapse
|
14
|
Sun H, Chen G, Guo B, Lv S, Yuan G. Potential clinical treatment prospects behind the molecular mechanism of alternative lengthening of telomeres (ALT). J Cancer 2023; 14:417-433. [PMID: 36860927 PMCID: PMC9969575 DOI: 10.7150/jca.80097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/25/2022] [Indexed: 02/04/2023] Open
Abstract
Normal somatic cells inevitably experience replicative stress and senescence during proliferation. Somatic cell carcinogenesis can be prevented in part by limiting the reproduction of damaged or old cells and removing them from the cell cycle [1, 2]. However, Cancer cells must overcome the issues of replication pressure and senescence as well as preserve telomere length in order to achieve immortality, in contrast to normal somatic cells [1, 2]. Although telomerase accounts for the bulk of telomere lengthening methods in human cancer cells, there is a non-negligible portion of telomere lengthening pathways that depend on alternative lengthening of telomeres (ALT) [3]. For the selection of novel possible therapeutic targets for ALT-related disorders, a thorough understanding of the molecular biology of these diseases is crucial [4]. The roles of ALT, typical ALT tumor cell traits, the pathophysiology and molecular mechanisms of ALT tumor disorders, such as adrenocortical carcinoma (ACC), are all summarized in this work. Additionally, this research compiles as many of its hypothetically viable but unproven treatment targets as it can (ALT-associated PML bodies (APB), etc.). This review is intended to contribute as much as possible to the development of research, while also trying to provide a partial information for prospective investigations on ALT pathways and associated diseases.
Collapse
Affiliation(s)
- Haolu Sun
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230011, China
| | - Guijuan Chen
- School of Environment and Chemical Engineering, Anhui Vocational and Technical College, Hefei, 230011, China
| | - Baochang Guo
- Rehabilitation Department of Traditional Chinese Medicine, 969 Hospital of the Joint Support Force of the Chinese People's Liberation Army, Hohhot, 010000, China
| | - Shushu Lv
- Department of Pathology, The First Affiliated Hospital of Huzhou University, Huzhou 313000, China
| | - Guojun Yuan
- School of Environment and Chemical Engineering, Anhui Vocational and Technical College, Hefei, 230011, China
| |
Collapse
|
15
|
Kent T, Clynes D. Alternative Lengthening of Telomeres: Lessons to Be Learned from Telomeric DNA Double-Strand Break Repair. Genes (Basel) 2021; 12:1734. [PMID: 34828344 PMCID: PMC8619803 DOI: 10.3390/genes12111734] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 10/22/2021] [Accepted: 10/28/2021] [Indexed: 12/25/2022] Open
Abstract
The study of the molecular pathways underlying cancer has given us important insights into how breaks in our DNA are repaired and the dire consequences that can occur when these processes are perturbed. Extensive research over the past 20 years has shown that the key molecular event underpinning a subset of cancers involves the deregulated repair of DNA double-strand breaks (DSBs) at telomeres, which in turn leads to telomere lengthening and the potential for replicative immortality. Here we discuss, in-depth, recent major breakthroughs in our understanding of the mechanisms underpinning this pathway known as the alternative lengthening of telomeres (ALT). We explore how this gives us important insights into how DSB repair at telomeres is regulated, with relevance to the cell-cycle-dependent regulation of repair, repair of stalled replication forks and the spatial regulation of DSB repair.
Collapse
Affiliation(s)
- Thomas Kent
- Molecular Haematology Unit, Radcliffe Department of Medicine, The MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK;
| | - David Clynes
- Department of Oncology, The MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DS, UK
| |
Collapse
|
16
|
MacKenzie D, Watters AK, To JT, Young MW, Muratori J, Wilkoff MH, Abraham RG, Plummer MM, Zhang D. ALT Positivity in Human Cancers: Prevalence and Clinical Insights. Cancers (Basel) 2021; 13:2384. [PMID: 34069193 PMCID: PMC8156225 DOI: 10.3390/cancers13102384] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 02/08/2023] Open
Abstract
Many exciting advances in cancer-related telomere biology have been made in the past decade. Of these recent advances, great progress has also been made with respect to the Alternative Lengthening of Telomeres (ALT) pathway. Along with a better understanding of the molecular mechanism of this unique telomere maintenance pathway, many studies have also evaluated ALT activity in various cancer subtypes. We first briefly review and assess a variety of commonly used ALT biomarkers. Then, we provide both an update on ALT-positive (ALT+) tumor prevalence as well as a systematic clinical assessment of the presently studied ALT+ malignancies. Additionally, we discuss the pathogenetic alterations in ALT+ cancers, for example, the mutation status of ATRX and DAXX, and their correlations with the activation of the ALT pathway. Finally, we highlight important ALT+ clinical associations within each cancer subtype and subdivisions within, as well as their prognoses. We hope this alternative perspective will allow scientists, clinicians, and drug developers to have greater insight into the ALT cancers so that together, we may develop more efficacious treatments and improved management strategies to meet the urgent needs of cancer patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Maria M. Plummer
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY 11568, USA; (D.M.J.); (A.K.W.); (J.T.T.); (M.W.Y.); (J.M.); (M.H.W.); (R.G.A.)
| | - Dong Zhang
- Department of Biomedical Sciences, College of Osteopathic Medicine, New York Institute of Technology, Old Westbury, NY 11568, USA; (D.M.J.); (A.K.W.); (J.T.T.); (M.W.Y.); (J.M.); (M.H.W.); (R.G.A.)
| |
Collapse
|
17
|
Abstract
Cancer is a complex disease characterized by loss of cellular homeostasis through genetic and epigenetic alterations. Emerging evidence highlights a role for histone variants and their dedicated chaperones in cancer initiation and progression. Histone variants are involved in processes as diverse as maintenance of genome integrity, nuclear architecture and cell identity. On a molecular level, histone variants add a layer of complexity to the dynamic regulation of transcription, DNA replication and repair, and mitotic chromosome segregation. Because these functions are critical to ensure normal proliferation and maintenance of cellular fate, cancer cells are defined by their capacity to subvert them. Hijacking histone variants and their chaperones is emerging as a common means to disrupt homeostasis across a wide range of cancers, particularly solid tumours. Here we discuss histone variants and histone chaperones as tumour-promoting or tumour-suppressive players in the pathogenesis of cancer.
Collapse
Affiliation(s)
| | - Dan Filipescu
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | |
Collapse
|
18
|
Goncalves T, Zoumpoulidou G, Alvarez-Mendoza C, Mancusi C, Collopy LC, Strauss SJ, Mittnacht S, Tomita K. Selective Elimination of Osteosarcoma Cell Lines with Short Telomeres by Ataxia Telangiectasia and Rad3-Related Inhibitors. ACS Pharmacol Transl Sci 2020; 3:1253-1264. [PMID: 33344901 PMCID: PMC7737214 DOI: 10.1021/acsptsci.0c00125] [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: 08/31/2020] [Indexed: 12/12/2022]
Abstract
![]()
To
avoid replicative senescence or telomere-induced apoptosis,
cancers employ telomere maintenance mechanisms (TMMs) involving either
the upregulation of telomerase or the acquisition of recombination-based
alternative telomere lengthening (ALT). The choice of TMM may differentially
influence cancer evolution and be exploitable in targeted therapies.
Here, we examine TMMs in a panel of 17 osteosarcoma-derived cell lines,
defining three separate groups according to TMM and the length of
telomeres maintained. Eight were ALT-positive, including the previously
uncharacterized lines, KPD and LM7. While ALT-positive lines all showed
excessive telomere length, ALT-negative cell lines fell into two groups
according to their telomere length: HOS-MNNG, OHSN, SJSA-1, HAL, 143b,
and HOS displayed subnormally short telomere length, while MG-63,
MHM, and HuO-3N1 displayed long telomeres. Hence, we further subcategorized
ALT-negative TMM into long-telomere (LT) and short-telomere (ST) maintenance groups.
Importantly, subnormally short telomeres were significantly associated
with hypersensitivity to three different therapeutics targeting the
protein kinase ataxia telangiectasia and Rad3-related (ATR) (AZD-6738/Ceralasertib,
VE-822/Berzoserib, and BAY-1895344) compared to long telomeres maintained
via ALT or telomerase. Within 24 h of ATR inhibition, cells with short
but not long telomeres displayed chromosome bridges and underwent
cell death, indicating a selective dependency on ATR for chromosome
stability. Collectively, our work provides a resource to identify
links between the mode of telomere maintenance and drug sensitivity
in osteosarcoma and indicates that telomere length predicts ATR inhibitor
sensitivity in cancer.
Collapse
Affiliation(s)
- Tomas Goncalves
- Centre for Genome Engineering and Maintenance, College of Health, Medicine and Life Sciences, Brunel University London, London UB8 3PH, United Kingdom.,Chromosome Maintenance Group, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Georgia Zoumpoulidou
- Cancer Cell Signalling, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Carlos Alvarez-Mendoza
- Cancer Cell Signalling, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Caterina Mancusi
- Cancer Cell Signalling, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Laura C Collopy
- Chromosome Maintenance Group, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Sandra J Strauss
- Department of Oncology, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom.,London Sarcoma Service, University College London Hospitals Foundation Trust, London WC1E 6DD, United Kingdom
| | - Sibylle Mittnacht
- Cancer Cell Signalling, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Kazunori Tomita
- Centre for Genome Engineering and Maintenance, College of Health, Medicine and Life Sciences, Brunel University London, London UB8 3PH, United Kingdom.,Chromosome Maintenance Group, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| |
Collapse
|
19
|
Telomere length alterations and ATRX/DAXX loss in pituitary adenomas. Mod Pathol 2020; 33:1475-1481. [PMID: 32203094 PMCID: PMC8867890 DOI: 10.1038/s41379-020-0523-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/05/2020] [Accepted: 03/05/2020] [Indexed: 01/03/2023]
Abstract
Telomeres are nucleoprotein complexes located at the termini of eukaryotic chromosomes that prevent exonucleolytic degradation and end-to-end chromosomal fusions. Cancers often have critically shortened, dysfunctional telomeres contributing to genomic instability. Telomere shortening has been reported in a wide range of precancerous lesions and invasive carcinomas. However, the role of telomere alterations, including the presence of alternative lengthening of telomeres (ALT), has not been studied in pituitary adenomas. Telomere length and the presence of ALT were assessed directly at the single cell level using a telomere-specific fluorescence in situ hybridization assay in tissue microarrays. Tumors were characterized as either ALT-positive or having short, normal, or long telomere lengths and then these categories were compared with clinicopathological characteristics. ATRX and DAXX expression was studied through immunohistochemistry. We characterized a discovery set of 106 pituitary adenomas including both functional and nonfunctional subsets (88 primary, 18 recurrent). Telomere lengths were estimated and we observed 64 (59.4%) cases with short, 39 (36.8%) cases with normal, and 0 (0%) cases with long telomeres. We did not observe significant differences in the clinicopathological characteristics of the group with abnormally shortened telomeres compared to the group with normal telomeres. However, three pituitary adenomas were identified as ALT-positive of which two were recurrent tumors. Two of these three ALT-positive cases had alterations in either of the chromatin remodeling proteins, ATRX and DAXX, which are routinely altered in other ALT-positive tumor subtypes. In a second cohort of 32 recurrent pituitary adenomas from 22 patients, we found that the tumors from 36% of patients (n = 8) were ALT-positive. This study demonstrates that short telomere lengths are prevalent in pituitary adenomas and that ALT-positive pituitary adenomas are enriched in recurrent disease.
Collapse
|
20
|
Batchu S, Gold JL. Analysis of Transcriptomic Similarity between Osteosarcoma Cell Lines and Primary Tumors. Oncology 2020; 98:814-816. [PMID: 32702705 DOI: 10.1159/000508720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 05/13/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Osteosarcoma (OS) cell lines are commonly used to mimic tumors for in vitro experiments. The present study explores the resemblance of OS cell lines to OS primary tumors in regard to gene expression. METHODS Transcriptomic data were retrieved from published data sets for 18 primary tumor samples and 13 commonly used OS cell lines. Tumor purity was accounted for when correlating tumor and cell line gene expression. Differentially expressed genes between tumors and cell lines were discovered and gene ontology analysis was performed. RESULTS Certain commonly used cell lines, including NY, NOS1, and U2OS, display less resemblance to OS tumors than do other cell lines. For genes overexpressed in tumors, and consequently underexpressed in cell lines, gene ontology analysis enriched pathways related to cell-cell adhesion and stimulus detection. CONCLUSION The pathways dysregulated between cell lines and tumors have been implicated in OS pathogenesis. Therefore, the findings suggest that the transcriptome of OS cell lines may not be completely representative of OS primary tumors' gene expression and the disease process.
Collapse
Affiliation(s)
- Sai Batchu
- National Heart, Lung, and Blood Institute (NHLBI), Bethesda, Maryland, USA,
| | | |
Collapse
|
21
|
Mason-Osann E, Terranova K, Lupo N, Lock YJ, Carson LM, Flynn RL. RAD54 promotes alternative lengthening of telomeres by mediating branch migration. EMBO Rep 2020; 21:e49495. [PMID: 32337843 PMCID: PMC7271314 DOI: 10.15252/embr.201949495] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 03/19/2020] [Accepted: 03/31/2020] [Indexed: 12/15/2022] Open
Abstract
Cancer cells can activate the alternative lengthening of telomeres (ALT) pathway to promote replicative immortality. The ALT pathway promotes telomere elongation through a homologous recombination pathway known as break‐induced replication (BIR), which is often engaged to repair single‐ended double‐stranded breaks (DSBs). Single‐ended DSBs are resected to promote strand invasion and facilitate the formation of a local displacement loop (D‐loop), which can trigger DNA synthesis, and ultimately promote telomere elongation. However, the exact proteins involved in the maturation, migration, and resolution of D‐loops at ALT telomeres are unclear. In vitro, the DNA translocase RAD54 both binds D‐loops and promotes branch migration suggesting that RAD54 may function to promote ALT activity. Here, we demonstrate that RAD54 is enriched at ALT telomeres and promotes telomeric DNA synthesis through its ATPase‐dependent branch migration activity. Loss of RAD54 leads to the formation of unresolved recombination intermediates at telomeres that form ultra‐fine anaphase bridges in mitosis. These data demonstrate an important role for RAD54 in promoting ALT‐mediated telomere synthesis.
Collapse
Affiliation(s)
- Emily Mason-Osann
- Departments of Pharmacology & Experimental Therapeutics, Medicine Cancer Center, Boston University School of Medicine, Boston, MA, USA
| | - Katherine Terranova
- Departments of Pharmacology & Experimental Therapeutics, Medicine Cancer Center, Boston University School of Medicine, Boston, MA, USA
| | - Nicholas Lupo
- Departments of Pharmacology & Experimental Therapeutics, Medicine Cancer Center, Boston University School of Medicine, Boston, MA, USA
| | - Ying Jie Lock
- Departments of Pharmacology & Experimental Therapeutics, Medicine Cancer Center, Boston University School of Medicine, Boston, MA, USA
| | - Lisa M Carson
- Departments of Pharmacology & Experimental Therapeutics, Medicine Cancer Center, Boston University School of Medicine, Boston, MA, USA
| | - Rachel Litman Flynn
- Departments of Pharmacology & Experimental Therapeutics, Medicine Cancer Center, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
22
|
Xiong W, Zhang Y, Yu H. Comprehensive characterization of circular RNAs in osteosarcoma cell lines. Cell Signal 2020; 71:109603. [PMID: 32199934 DOI: 10.1016/j.cellsig.2020.109603] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 03/04/2020] [Accepted: 03/14/2020] [Indexed: 12/21/2022]
Abstract
Circular RNA (circRNA) is a looped noncoding RNA with a stable structure and tissue-specific expression and widely reported to regulate cancer initiation and progression. However, the circRNA expression patterns and their roles in osteosarcoma initiation and progression are still poorly understood. In this study, we characterized the landscape of circRNAs in osteosarcoma (OS) cell lines, and calculated the epithelial-mesenchymal transition (EMT) scores for OS cell lines. The differential expression analysis revealed that the EMT-related genes were significantly upregulated in the OS cell lines with higher metastatic potentials, and some inflammation-related pathways and pathways involved in cell-cell communications were enriched by these upregulated genes. Furthermore, we constructed a circRNA-based competing endogenous RNA (ceRNA) network, which consisted of 5 circRNAs, 17 miRNAs, and 73 mRNAs. Particularly, hsa_circ_0085360, which had the highest correlation with TRPS1, were characterized by some cancer-related pathways, and TRPS1 and its target gene FGFR3 were closely associated with both event-free survival and overall survival of OS, indicating that hsa_circ_0085360 might have the potential to predict the OS prognosis. In summary, we profiled the circRNA expression patterns in OS, predicted their functionality, and explored the underlying mechanism and prognostic values, which might provide some evidences for OS-related circRNA researches.
Collapse
Affiliation(s)
- Wen Xiong
- Department of Orthopaedics, The First People's Hospital of Tianmen, Tianmen, Hubei, China
| | - Yun Zhang
- Department of Clinical Laboratory Center, Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China
| | - Huaixi Yu
- Department of Orthopedics,the Affiliated Huai'an Hospital of Xuzhou Medical University, the Second People's Hospital of Huai'an, No.62, Huaihai Road(S.), Huai'an 223002, China.
| |
Collapse
|
23
|
Amato R, Valenzuela M, Berardinelli F, Salvati E, Maresca C, Leone S, Antoccia A, Sgura A. G-quadruplex Stabilization Fuels the ALT Pathway in ALT-positive Osteosarcoma Cells. Genes (Basel) 2020; 11:genes11030304. [PMID: 32183119 PMCID: PMC7140816 DOI: 10.3390/genes11030304] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/20/2020] [Accepted: 03/09/2020] [Indexed: 12/20/2022] Open
Abstract
Most human tumors maintain telomere lengths by telomerase, whereas a portion of them (10–15%) uses a mechanism named alternative lengthening of telomeres (ALT). The telomeric G-quadruplex (G4) ligand RHPS4 is known for its potent antiproliferative effect, as shown in telomerase-positive cancer models. Moreover, RHPS4 is also able to reduce cell proliferation in ALT cells, although the influence of G4 stabilization on the ALT mechanism has so far been poorly investigated. Here we show that sensitivity to RHPS4 is comparable in ALT-positive (U2OS; SAOS-2) and telomerase-positive (HOS) osteosarcoma cell lines, unlinking the telomere maintenance mechanism and RHPS4 responsiveness. To investigate the impact of G4 stabilization on ALT, the cardinal ALT hallmarks were analyzed. A significant induction of telomeric doublets, telomeric clusterized DNA damage, ALT-associated Promyelocytic Leukaemia-bodies (APBs), telomere sister chromatid exchanges (T-SCE) and c-circles was found exclusively in RHPS4-treated ALT cells. We surmise that RHPS4 affects ALT mechanisms through the induction of replicative stress that in turn is converted in DNA damage at telomeres, fueling recombination. In conclusion, our work indicates that RHPS4-induced telomeric DNA damage promotes overactivation of telomeric recombination in ALT cells, opening new questions on the therapeutic employment of G4 ligands in the treatment of ALT positive tumors.
Collapse
Affiliation(s)
- Roberta Amato
- Department of Science, Roma Tre University, I-00146 Rome, Italy; (R.A.); (M.V.); (S.L.); (A.A.); (A.S.)
| | - Martina Valenzuela
- Department of Science, Roma Tre University, I-00146 Rome, Italy; (R.A.); (M.V.); (S.L.); (A.A.); (A.S.)
| | - Francesco Berardinelli
- Department of Science, Roma Tre University, I-00146 Rome, Italy; (R.A.); (M.V.); (S.L.); (A.A.); (A.S.)
- Correspondence: ; Tel.: +39-0657-33-6330
| | - Erica Salvati
- BPM-CNR Institute of Molecular Biology and Pathology, National Research Council, 00185 Rome, Italy;
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Carmen Maresca
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Stefano Leone
- Department of Science, Roma Tre University, I-00146 Rome, Italy; (R.A.); (M.V.); (S.L.); (A.A.); (A.S.)
| | - Antonio Antoccia
- Department of Science, Roma Tre University, I-00146 Rome, Italy; (R.A.); (M.V.); (S.L.); (A.A.); (A.S.)
| | - Antonella Sgura
- Department of Science, Roma Tre University, I-00146 Rome, Italy; (R.A.); (M.V.); (S.L.); (A.A.); (A.S.)
| |
Collapse
|
24
|
Hoang SM, O'Sullivan RJ. Alternative Lengthening of Telomeres: Building Bridges To Connect Chromosome Ends. Trends Cancer 2020; 6:247-260. [PMID: 32101727 PMCID: PMC7199893 DOI: 10.1016/j.trecan.2019.12.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 12/15/2022]
Abstract
Alternative lengthening of telomeres (ALT) is a mechanism of telomere maintenance that is observed in many of the most recalcitrant cancer subtypes. Telomeres in ALT cancer cells exhibit a distinctive nucleoprotein architecture shaped by the mismanagement of chromatin that fosters cycles of DNA damage and replicative stress that activate homology-directed repair (HDR). Mutations in specific chromatin-remodeling factors appear to be key determinants of the emergence and survival of ALT cancer cells. However, these may represent vulnerabilities for the targeted elimination of ALT cancer cells that infiltrate tissues and organs to become devastating tumors. In this review we examine recent findings that provide new insights into the factors and mechanisms that mediate telomere length maintenance and survival of ALT cancer cells.
Collapse
Affiliation(s)
- Song My Hoang
- Department of Pharmacology and Chemical Biology, Hillman Cancer Center, University of Pittsburgh School of Medicine (UPMC), Pittsburgh, PA, USA
| | - Roderick J O'Sullivan
- Department of Pharmacology and Chemical Biology, Hillman Cancer Center, University of Pittsburgh School of Medicine (UPMC), Pittsburgh, PA, USA.
| |
Collapse
|
25
|
Srinivas N, Rachakonda S, Kumar R. Telomeres and Telomere Length: A General Overview. Cancers (Basel) 2020; 12:E558. [PMID: 32121056 PMCID: PMC7139734 DOI: 10.3390/cancers12030558] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 02/06/2023] Open
Abstract
Telomeres are highly conserved tandem nucleotide repeats that include proximal double-stranded and distal single-stranded regions that in complex with shelterin proteins afford protection at chromosomal ends to maintain genomic integrity. Due to the inherent limitations of DNA replication and telomerase suppression in most somatic cells, telomeres undergo age-dependent incremental attrition. Short or dysfunctional telomeres are recognized as DNA double-stranded breaks, triggering cells to undergo replicative senescence. Telomere shortening, therefore, acts as a counting mechanism that drives replicative senescence by limiting the mitotic potential of cells. Telomere length, a complex hereditary trait, is associated with aging and age-related diseases. Epidemiological data, in general, support an association with varying magnitudes between constitutive telomere length and several disorders, including cancers. Telomere attrition is also influenced by oxidative damage and replicative stress caused by genetic, epigenetic, and environmental factors. Several single nucleotide polymorphisms at different loci, identified through genome-wide association studies, influence inter-individual variation in telomere length. In addition to genetic factors, environmental factors also influence telomere length during growth and development. Telomeres hold potential as biomarkers that reflect the genetic predisposition together with the impact of environmental conditions and as targets for anti-cancer therapies.
Collapse
Affiliation(s)
| | | | - Rajiv Kumar
- Division of Functional Genome Analysis, German Cancer Research Center, Im Neunheimer Feld 580, 69120 Heidelberg, Germany; (N.S.); (S.R.)
| |
Collapse
|
26
|
Mahmud I, Liao D. DAXX in cancer: phenomena, processes, mechanisms and regulation. Nucleic Acids Res 2019; 47:7734-7752. [PMID: 31350900 PMCID: PMC6735914 DOI: 10.1093/nar/gkz634] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/05/2019] [Accepted: 07/12/2019] [Indexed: 12/13/2022] Open
Abstract
DAXX displays complex biological functions. Remarkably, DAXX overexpression is a common feature in diverse cancers, which correlates with tumorigenesis, disease progression and treatment resistance. Structurally, DAXX is modular with an N-terminal helical bundle, a docking site for many DAXX interactors (e.g. p53 and ATRX). DAXX's central region folds with the H3.3/H4 dimer, providing a H3.3-specific chaperoning function. DAXX has two functionally critical SUMO-interacting motifs. These modules are connected by disordered regions. DAXX's structural features provide a framework for deciphering how DAXX mechanistically imparts its functions and how its activity is regulated. DAXX modulates transcription through binding to transcription factors, epigenetic modifiers, and chromatin remodelers. DAXX's localization in the PML nuclear bodies also plays roles in transcriptional regulation. DAXX-regulated genes are likely important effectors of its biological functions. Deposition of H3.3 and its interactions with epigenetic modifiers are likely key events for DAXX to regulate transcription, DNA repair, and viral infection. Interactions between DAXX and its partners directly impact apoptosis and cell signaling. DAXX's activity is regulated by posttranslational modifications and ubiquitin-dependent degradation. Notably, the tumor suppressor SPOP promotes DAXX degradation in phase-separated droplets. We summarize here our current understanding of DAXX's complex functions with a focus on how it promotes oncogenesis.
Collapse
Affiliation(s)
- Iqbal Mahmud
- Department of Anatomy and Cell Biology, UF Health Cancer Center, University of Florida College of Medicine, 1333 Center Drive, Gainesville, FL 32610-0235, USA
| | - Daiqing Liao
- Department of Anatomy and Cell Biology, UF Health Cancer Center, University of Florida College of Medicine, 1333 Center Drive, Gainesville, FL 32610-0235, USA
| |
Collapse
|
27
|
Graham MK, Kim J, Da J, Brosnan-Cashman JA, Rizzo A, Baena Del Valle JA, Chia L, Rubenstein M, Davis C, Zheng Q, Cope L, Considine M, Haffner MC, De Marzo AM, Meeker AK, Heaphy CM. Functional Loss of ATRX and TERC Activates Alternative Lengthening of Telomeres (ALT) in LAPC4 Prostate Cancer Cells. Mol Cancer Res 2019; 17:2480-2491. [PMID: 31611308 DOI: 10.1158/1541-7786.mcr-19-0654] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/06/2019] [Accepted: 10/07/2019] [Indexed: 01/06/2023]
Abstract
A key hallmark of cancer, unlimited replication, requires cancer cells to evade both replicative senescence and potentially lethal chromosomal instability induced by telomere dysfunction. The majority of cancers overcome these critical barriers by upregulating telomerase, a telomere-specific reverse transcriptase. However, a subset of cancers maintains telomere lengths by the telomerase-independent Alternative Lengthening of Telomeres (ALT) pathway. The presence of ALT is strongly associated with recurrent cancer-specific somatic inactivating mutations in the ATRX-DAXX chromatin-remodeling complex. Here, we generate an ALT-positive adenocarcinoma cell line following functional inactivation of ATRX and telomerase in a telomerase-positive adenocarcinoma cell line. Inactivating mutations in ATRX were introduced using CRISPR-cas9 nickase into two prostate cancer cell lines, LAPC-4 (derived from a lymph node metastasis) and CWR22Rv1 (sourced from a xenograft established from a primary prostate cancer). In LAPC-4, but not CWR22Rv1, abolishing ATRX was sufficient to induce multiple ALT-associated hallmarks, including the presence of ALT-associated promyelocytic leukemia bodies (APB), extrachromosomal telomere C-circles, and dramatic telomere length heterogeneity. However, telomerase activity was still present in these ATRXKO cells. Telomerase activity was subsequently crippled in these LAPC-4 ATRXKO cells by introducing mutations in the TERC locus, the essential RNA component of telomerase. These LAPC-4 ATRXKO TERCmut cells continued to proliferate long-term and retained ALT-associated hallmarks, thereby demonstrating their reliance on the ALT mechanism for telomere maintenance. IMPLICATIONS: These prostate cancer cell line models provide a unique system to explore the distinct molecular alterations that occur upon induction of ALT, and may be useful tools to screen for ALT-specific therapies.
Collapse
Affiliation(s)
- Mindy K Graham
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jiyoung Kim
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joseph Da
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Anthony Rizzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Lionel Chia
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael Rubenstein
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, Maryland
| | - Christine Davis
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qizhi Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Leslie Cope
- Department of Oncology Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael Considine
- Department of Oncology Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael C Haffner
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Angelo M De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Oncology Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alan K Meeker
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Oncology Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christopher M Heaphy
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland. .,Department of Oncology Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
28
|
Panier S, Maric M, Hewitt G, Mason-Osann E, Gali H, Dai A, Labadorf A, Guervilly JH, Ruis P, Segura-Bayona S, Belan O, Marzec P, Gaillard PHL, Flynn RL, Boulton SJ. SLX4IP Antagonizes Promiscuous BLM Activity during ALT Maintenance. Mol Cell 2019; 76:27-43.e11. [PMID: 31447390 PMCID: PMC6863466 DOI: 10.1016/j.molcel.2019.07.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 06/03/2019] [Accepted: 07/08/2019] [Indexed: 12/18/2022]
Abstract
Cancer cells acquire unlimited proliferative capacity by either re-expressing telomerase or inducing alternative lengthening of telomeres (ALT), which relies on telomere recombination. Here, we show that ALT recombination requires coordinate regulation of the SMX and BTR complexes to ensure the appropriate balance of resolution and dissolution activities at recombining telomeres. Critical to this control is SLX4IP, which accumulates at ALT telomeres and interacts with SLX4, XPF, and BLM. Loss of SLX4IP increases ALT-related phenotypes, which is incompatible with cell growth following concomitant loss of SLX4. Inactivation of BLM is sufficient to rescue telomere aggregation and the synthetic growth defect in this context, suggesting that SLX4IP favors SMX-dependent resolution by antagonizing promiscuous BLM activity during ALT recombination. Finally, we show that SLX4IP is inactivated in a subset of ALT-positive osteosarcomas. Collectively, our findings uncover an SLX4IP-dependent regulatory mechanism critical for telomere maintenance in ALT cancer cells.
Collapse
Affiliation(s)
| | - Marija Maric
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Graeme Hewitt
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Emily Mason-Osann
- Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Himabindu Gali
- Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Anqi Dai
- Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Adam Labadorf
- Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Jean-Hugues Guervilly
- Centre de Recherche en Cancérologie de Marseille, CRCM, CNRS, Aix Marseille Université, INSERM, Institut Paoli-Calmettes, 27 Boulevard Leï Roure, 13009 Marseille, France
| | - Philip Ruis
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | | | - Ondrej Belan
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Paulina Marzec
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Pierre-Henri L Gaillard
- Centre de Recherche en Cancérologie de Marseille, CRCM, CNRS, Aix Marseille Université, INSERM, Institut Paoli-Calmettes, 27 Boulevard Leï Roure, 13009 Marseille, France
| | - Rachel L Flynn
- Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, USA
| | - Simon J Boulton
- The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| |
Collapse
|
29
|
Yost KE, Clatterbuck Soper SF, Walker RL, Pineda MA, Zhu YJ, Ester CD, Showman S, Roschke AV, Waterfall JJ, Meltzer PS. Rapid and reversible suppression of ALT by DAXX in osteosarcoma cells. Sci Rep 2019; 9:4544. [PMID: 30872698 PMCID: PMC6418139 DOI: 10.1038/s41598-019-41058-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 02/07/2019] [Indexed: 01/19/2023] Open
Abstract
Many tumors maintain chromosome-ends through a telomerase-independent, DNA-templated mechanism called alternative lengthening of telomeres (ALT). While ALT occurs in only a subset of tumors, it is strongly associated with mutations in the genes ATRX and DAXX, which encode components of an H3.3 histone chaperone complex. The role of ATRX and DAXX mutations in potentiating the mechanism of ALT remains incompletely understood. Here we characterize an osteosarcoma cell line, G292, with wild-type ATRX but a unique chromosome translocation resulting in loss of DAXX function. While ATRX and DAXX form a complex in G292, this complex fails to localize to nuclear PML bodies. We demonstrate that introduction of wild type DAXX suppresses the ALT phenotype and restores the localization of ATRX/DAXX to PML bodies. Using an inducible system, we show that ALT-associated PML bodies are disrupted rapidly following DAXX induction and that ALT is again restored following withdrawal of DAXX.
Collapse
Affiliation(s)
- Kathryn E Yost
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.,Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Sarah F Clatterbuck Soper
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Robert L Walker
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marbin A Pineda
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yuelin J Zhu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Corbin D Ester
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.,Harvard Medical School, Boston, MA, 02115, USA
| | - Soyeon Showman
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Anna V Roschke
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Joshua J Waterfall
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA. .,Translational Research Department & INSERM U830, Institut Curie, Paris, France.
| | - Paul S Meltzer
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
30
|
Brosnan-Cashman JA, Graham MK, Heaphy CM. Genetic alterations associated with ALTered telomeres. Oncotarget 2018; 9:33739-33740. [PMID: 30333904 PMCID: PMC6173459 DOI: 10.18632/oncotarget.26111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/04/2018] [Indexed: 01/01/2023] Open
Affiliation(s)
- Jacqueline A Brosnan-Cashman
- Christopher M. Heaphy: Departments of Pathology and Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mindy K Graham
- Christopher M. Heaphy: Departments of Pathology and Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher M Heaphy
- Christopher M. Heaphy: Departments of Pathology and Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|