1
|
Machitani M, Nomura A, Yamashita T, Yasukawa M, Ueki S, Fujita KI, Ueno T, Yamashita A, Tanzawa Y, Watanabe M, Taniguchi T, Saitoh N, Kaneko S, Kato Y, Mano H, Masutomi K. Maintenance of R-loop structures by phosphorylated hTERT preserves genome integrity. Nat Cell Biol 2024; 26:932-945. [PMID: 38806647 DOI: 10.1038/s41556-024-01427-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 04/23/2024] [Indexed: 05/30/2024]
Abstract
As aberrant accumulation of RNA-DNA hybrids (R-loops) causes DNA damage and genome instability, cells express regulators of R-loop structures. Here we report that RNA-dependent RNA polymerase (RdRP) activity of human telomerase reverse transcriptase (hTERT) regulates R-loop formation. We found that the phosphorylated form of hTERT (p-hTERT) exhibits RdRP activity in nuclear speckles both in telomerase-positive cells and telomerase-negative cells with alternative lengthening of telomeres (ALT) activity. The p-hTERT did not associate with telomerase RNA component in nuclear speckles but, instead, with TERRA RNAs to resolve R-loops. Targeting of the TERT gene in ALT cells ablated RdRP activity and impaired tumour growth. Using a genome-scale CRISPR loss-of-function screen, we identified Fanconi anaemia/BRCA genes as synthetic lethal partners of hTERT RdRP. Inactivation of RdRP and Fanconi anaemia/BRCA genes caused accumulation of R-loop structures and DNA damage. These findings indicate that RdRP activity of p-hTERT guards against genome instability by removing R-loop structures.
Collapse
Affiliation(s)
- Mitsuhiro Machitani
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo, Japan
| | - Akira Nomura
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo, Japan
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Taro Yamashita
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Mami Yasukawa
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo, Japan
| | - Saori Ueki
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo, Japan
| | - Ken-Ichi Fujita
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo, Japan
| | - Toshihide Ueno
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Akio Yamashita
- Department of Investigative Medicine, University of the Ryukyus Graduate School of Medicine, Nakagami, Japan
| | - Yoshikazu Tanzawa
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Masahiko Watanabe
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Japan
| | - Toshiyasu Taniguchi
- Department of Molecular Life Science, Tokai University School of Medicine, Isehara, Japan
| | - Noriko Saitoh
- Division of Cancer Biology, The Cancer Institute of JFCR, Tokyo, Japan
| | - Shuichi Kaneko
- Department of Gastroenterology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Kenkichi Masutomi
- Division of Cancer Stem Cell, National Cancer Center Research Institute, Tokyo, Japan.
| |
Collapse
|
2
|
Wang Y, Li C, Guo K, Hao X, Hou J, Zhang F, Jin R, Kang C, Gao L. Oxidation-responsive G-quadruplex ligand for selective inhibition of the proliferation of tumour cells. Bioorg Chem 2023; 139:106746. [PMID: 37506624 DOI: 10.1016/j.bioorg.2023.106746] [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: 03/29/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023]
Abstract
Tumour cells show a higher level of reactive oxygen species (ROS) than normal cells. On the basis of this difference, we designed an oxidation-responsive G-quadruplex proligand PDS-B by installing borolanylbenzyls on a well-known pyridostatin (PDS) ligand PDS-S to response high level ROS in tumour cells. The rapid oxidative degradation of the proligand to its active form PDS-S in the presence of H2O2 confirms the oxidation-responsive design. According to Förster resonance energy transfer (FRET) assays, circular dichroism (CD) spectra and confocal fluorescence imaging, PDS-B stabilizes telomeric G4 structures after oxidation with H2O2 or intracellular ROS. Apoptosis assays and cell cycle assays showed significant selectivity of PDS-B in inhibiting the proliferation of tumour cells over normal cells through responses to a high level of ROS in the formers. Further assays confirmed higher level of relative Caspase-3 activity in tumour cells than normal cells, consequently the enhanced apoptosis of the tumour cells induced by PDS-B. In summary, the results demonstrate a modification approach to solve the poor selectivity of the G4 ligand in tumour cells and cytotoxicity in normal cells.
Collapse
Affiliation(s)
- Yu Wang
- CAS Key Laboratory of High-Performance Synthetic Rubber and Its Composite Materials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; University of Science and Technology of China, Hefei 230026, China
| | - Chunjie Li
- CAS Key Laboratory of High-Performance Synthetic Rubber and Its Composite Materials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| | - Kai Guo
- CAS Key Laboratory of High-Performance Synthetic Rubber and Its Composite Materials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xueyu Hao
- CAS Key Laboratory of High-Performance Synthetic Rubber and Its Composite Materials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Jingwei Hou
- CAS Key Laboratory of High-Performance Synthetic Rubber and Its Composite Materials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; University of Science and Technology of China, Hefei 230026, China
| | - Feng Zhang
- CAS Key Laboratory of High-Performance Synthetic Rubber and Its Composite Materials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Rizhe Jin
- CAS Key Laboratory of High-Performance Synthetic Rubber and Its Composite Materials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Chuanqing Kang
- CAS Key Laboratory of High-Performance Synthetic Rubber and Its Composite Materials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; University of Science and Technology of China, Hefei 230026, China.
| | - Lianxun Gao
- CAS Key Laboratory of High-Performance Synthetic Rubber and Its Composite Materials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| |
Collapse
|
3
|
Liu Y, Betori RC, Pagacz J, Frost GB, Efimova EV, Wu D, Wolfgeher DJ, Bryan TM, Cohen SB, Scheidt KA, Kron SJ. Targeting telomerase reverse transcriptase with the covalent inhibitor NU-1 confers immunogenic radiation sensitization. Cell Chem Biol 2022; 29:1517-1531.e7. [PMID: 36206753 PMCID: PMC9588800 DOI: 10.1016/j.chembiol.2022.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/29/2022] [Accepted: 09/15/2022] [Indexed: 11/03/2022]
Abstract
Beyond synthesizing telomere repeats, the telomerase reverse transcriptase (TERT) also serves multiple other roles supporting cancer growth. Blocking telomerase to drive telomere erosion appears impractical, but TERT's non-canonical activities have yet to be fully explored as cancer targets. Here, we used an irreversible TERT inhibitor, NU-1, to examine impacts on resistance to conventional cancer therapies. In vitro, inhibiting TERT sensitized cells to chemotherapy and radiation. NU-1 delayed repair of double-strand breaks, resulting in persistent DNA damage signaling and cellular senescence. Although NU-1 alone did not impact growth of syngeneic CT26 tumors in BALB/c mice, it dramatically enhanced the effects of radiation, leading to immune-dependent tumor elimination. Tumors displayed persistent DNA damage, suppressed proliferation, and increased activated immune infiltrate. Our studies confirm TERT's role in limiting genotoxic effects of conventional therapy but also implicate TERT as a determinant of immune evasion and therapy resistance.
Collapse
Affiliation(s)
- Yue Liu
- Ludwig Center for Metastasis Research and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Rick C Betori
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Joanna Pagacz
- Ludwig Center for Metastasis Research and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Grant B Frost
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA
| | - Elena V Efimova
- Ludwig Center for Metastasis Research and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Ding Wu
- Ludwig Center for Metastasis Research and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Donald J Wolfgeher
- Ludwig Center for Metastasis Research and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA
| | - Tracy M Bryan
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Scott B Cohen
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Karl A Scheidt
- Department of Chemistry, Northwestern University, Evanston, IL 60208, USA.
| | - Stephen J Kron
- Ludwig Center for Metastasis Research and Department of Molecular Genetics and Cell Biology, University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
4
|
de Lima MF, Freitas MO, Hamedani MK, Rangel-Pozzo A, Zhu XD, Mai S. Consecutive Inhibition of Telomerase and Alternative Lengthening Pathway Promotes Hodgkin's Lymphoma Cell Death. Biomedicines 2022; 10:2299. [PMID: 36140400 PMCID: PMC9496562 DOI: 10.3390/biomedicines10092299] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/19/2022] [Accepted: 09/12/2022] [Indexed: 11/16/2022] Open
Abstract
Telomere maintenance is key during cancer development. Malignant cells can either use telomerase or an alternative lengthening of telomere (ALT) pathway to maintain their telomere length. In Hodgkin's Lymphoma (HL), the presence of telomerase activation is established. The activation of ALT has been reported recently. Our data confirm this notion describing co-localization of the phosphorylated form of telomeric repeat-binding factor 1 (pT371-TRF1) with ALT-associated promyelocytic leukemia bodies. Surprisingly, to our knowledge, there are no published studies targeting both telomere maintenance pathways in HL. Consequently, we investigated, for the first time, the effects of both telomerase and ALT inhibition on HL cell viability: We inhibited telomerase and/or ALT, given either individually, simultaneously, or consecutively. We report that the inhibition of telomerase using BIBR1532 followed by ALT inhibition, using trabectedin, caused a decrease of greater than 90% in cell viability in three patient-derived HL cell lines. Our results suggest that HL cells are most vulnerable to the consecutive inhibition of telomerase followed by ALT inhibition.
Collapse
Affiliation(s)
- Matheus Fabiao de Lima
- Department of Physiology and Pathophysiology, CancerCare Manitoba Research Institute, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Monique Oliveira Freitas
- Genetic Service, Institute of Paediatrics and Puericulture Martagão Gesteira (IPPMG), Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro 21941-912, Brazil
| | - Mohammad K. Hamedani
- Department of Physiology and Pathophysiology, CancerCare Manitoba Research Institute, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Aline Rangel-Pozzo
- Department of Physiology and Pathophysiology, CancerCare Manitoba Research Institute, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| | - Xu-Dong Zhu
- Department of Biology, Faculty of Science, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Sabine Mai
- Department of Physiology and Pathophysiology, CancerCare Manitoba Research Institute, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
| |
Collapse
|
5
|
Song Q, Hu Y, Yin A, Wang H, Yin Q. DNA Holliday Junction: History, Regulation and Bioactivity. Int J Mol Sci 2022; 23:9730. [PMID: 36077130 PMCID: PMC9456528 DOI: 10.3390/ijms23179730] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022] Open
Abstract
DNA Holliday junction (HJ) is a four-way stranded DNA intermediate that formed in replication fork regression, homology-dependent repair and mitosis, performing a significant role in genomic stability. Failure to remove HJ can induce an acceptable replication fork stalling and DNA damage in normal cells, leading to a serious chromosomal aberration and even cell death in HJ nuclease-deficient tumor cells. Thus, HJ is becoming an attractive target in cancer therapy. However, the development of HJ-targeting ligand faces great challenges because of flexile cavities on the center of HJs. This review introduces the discovery history of HJ, elucidates the formation and dissociation procedures of HJ in corresponding bio-events, emphasizes the importance of prompt HJ-removing in genome stability, and summarizes recent advances in HJ-based ligand discovery. Our review indicate that target HJ is a promising approach in oncotherapy.
Collapse
Affiliation(s)
- Qinqin Song
- State/Key Laboratory of Microbial Technology, Shandong University, 72 Jimo Binhai Road, Qingdao 266237, China
| | - Yuemiao Hu
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, 30 Qingquan Road, Yantai 264005, China
| | - Anqi Yin
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, 30 Qingquan Road, Yantai 264005, China
| | - Hongbo Wang
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, 30 Qingquan Road, Yantai 264005, China
| | - Qikun Yin
- Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, School of Pharmacy, Yantai University, 30 Qingquan Road, Yantai 264005, China
- Bohai Rim Advanced Research Institute for Drug Discovery, 198 Binhai East Road, Yantai 264005, China
| |
Collapse
|
6
|
Fernandez RJ, Gardner ZJG, Slovik KJ, Liberti DC, Estep KN, Yang W, Chen Q, Santini GT, Perez JV, Root S, Bhatia R, Tobias JW, Babu A, Morley MP, Frank DB, Morrisey EE, Lengner CJ, Johnson FB. GSK3 inhibition rescues growth and telomere dysfunction in dyskeratosis congenita iPSC-derived type II alveolar epithelial cells. eLife 2022; 11:64430. [PMID: 35559731 PMCID: PMC9200405 DOI: 10.7554/elife.64430] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/11/2022] [Indexed: 11/27/2022] Open
Abstract
Dyskeratosis congenita (DC) is a rare genetic disorder characterized by deficiencies in telomere maintenance leading to very short telomeres and the premature onset of certain age-related diseases, including pulmonary fibrosis (PF). PF is thought to derive from epithelial failure, particularly that of type II alveolar epithelial (AT2) cells, which are highly dependent on Wnt signaling during development and adult regeneration. We use human induced pluripotent stem cell-derived AT2 (iAT2) cells to model how short telomeres affect AT2 cells. Cultured DC mutant iAT2 cells accumulate shortened, uncapped telomeres and manifest defects in the growth of alveolospheres, hallmarks of senescence, and apparent defects in Wnt signaling. The GSK3 inhibitor, CHIR99021, which mimics the output of canonical Wnt signaling, enhances telomerase activity and rescues the defects. These findings support further investigation of Wnt agonists as potential therapies for DC-related pathologies.
Collapse
Affiliation(s)
- Rafael Jesus Fernandez
- Medical Scientist Training Program, University of Pennsylvania, Philadelphia, United States
| | - Zachary J G Gardner
- Medical Scientist Training Program, University of Pennsylvania, Philadelphia, United States
| | - Katherine J Slovik
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Derek C Liberti
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, United States
| | - Katrina N Estep
- Cell and Molecular Biology Graduate Group, University of Pennsylvania, Philadelphia, United States
| | - Wenli Yang
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Qijun Chen
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | - Garrett T Santini
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Javier V Perez
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| | - Sarah Root
- College of Arts and Sciences and Vagelos Scholars Program, University of Pennsylvania, Philadelphia, United States
| | - Ranvir Bhatia
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - John W Tobias
- Penn Genomic Analysis Core, University of Pennsylvania, Philadelphia, United States
| | - Apoorva Babu
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, United States
| | - Michael P Morley
- Penn Cardiovascular Institute, University of Pennsylvania, Philadelphia, United States
| | - David B Frank
- Penn-CHOP Lung Biology Institute, Children's Hospital of Philadelphia, Philadelphia, United States
| | - Edward E Morrisey
- Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, United States
| | - Christopher J Lengner
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, United States
| | - F Brad Johnson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
7
|
Yoon JH, Choi BJ, Nam SW, Park WS. Gastric cancer exosomes contribute to the field cancerization of gastric epithelial cells surrounding gastric cancer. Gastric Cancer 2022; 25:490-502. [PMID: 34993738 DOI: 10.1007/s10120-021-01269-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 11/14/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND A dynamic molecular interaction between cancer and the surrounding normal cells is mediated through exosomes. We investigated whether exosomes derived from gastric cancer cells affected the fate of the surrounding gastric epithelial cells. METHODS We analyzed the cell viability and immortalization of primary normal stomach epithelial cells (PNSECs) after treatment with exosomes derived from AGS gastric cancer cells and/or H. pylori CagA. Cell proliferation and apoptosis were analyzed by BrdU incorporation, flow-cytometry, and colony formation assays. We examined telomere length, expression and activity of telomerase, and expression of telomere-related genes in PNSECs treated with cancer exosomes, and in 60 gastric cancer and corresponding mucosal tissues. The differentially expressed genes and transcriptional regulation of telomere-related genes were verified using real-time qPCR and ChIP analyses, respectively. RESULTS Gastric cancer exosomes increased cell viability and the population-doubling levels but inhibited the cellular senescence and apoptosis of PNSECs. The internalization of cancer exosomes in PNSECs dramatically increased the number of surviving colonies and induced a multilayer growth and invasion into the scaffold. Treatment of PNSECs with cancer exosomes markedly increased the expression and activity of telomerase and the T/S ratio and regulated the expression of the telomere-associated genes, heat-shock genes, and hedgehog genes. Compared to gastric mucosae, gastric cancer showed increased hTERT expression, which was positively correlated with telomere length. Interestingly, seven (46.7%) of 15 non-cancerous gastric mucosae demonstrated strong telomerase activity. CONCLUSION These results suggest that gastric cancer exosomes induced the transformation and field cancerization of the surrounding non-cancerous gastric epithelial cells.
Collapse
Affiliation(s)
- Jung Hwan Yoon
- Department of Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea.,Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea
| | - Byung Joon Choi
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea
| | - Suk Woo Nam
- Department of Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea.,Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea
| | - Won Sang Park
- Department of Pathology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea. .,Functional RNomics Research Center, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, South Korea.
| |
Collapse
|
8
|
Functional interaction between compound heterozygous TERT mutations causes severe telomere biology disorder. Blood Adv 2022; 6:3779-3791. [PMID: 35477117 DOI: 10.1182/bloodadvances.2022007029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/07/2022] [Indexed: 11/20/2022] Open
Abstract
Telomere biology disorders (TBDs) are a spectrum of multisystem inherited disorders characterized by bone marrow failure, resulting from mutations in genes encoding telomerase or other proteins involved in maintaining telomere length and integrity. Pathogenicity of variants in these genes can be hard to evaluate, since TBD mutations show highly variable penetrance and genetic anticipation due to inheritance of shorter telomeres with each generation. Thus, detailed functional analysis of newly identified variants is often essential. Here we describe a patient with compound heterozygous variants in the TERT gene, which encodes the catalytic subunit of telomerase, hTERT; this patient has the extremely severe Hoyeraal-Hreidarsson form of TBD, although his heterozygous parents are clinically unaffected. Molecular dynamic modeling and detailed biochemical analyses demonstrate that 1 allele (L557P) affects association of hTERT with its cognate RNA component hTR, while the other (K1050E) affects the binding of telomerase to its DNA substrate and enzyme processivity. Unexpectedly, the data demonstrate a functional interaction between the proteins encoded by the 2 alleles, with WT hTERT able to rescue the effect of K1050E on processivity, whereas L557P hTERT cannot. These data contribute to the mechanistic understanding of telomerase, indicating that RNA binding in 1 hTERT molecule affects the processivity of telomere addition by the other molecule. This work emphasizes the importance of functional characterization of TERT variants to reach a definitive molecular diagnosis for TBD patients, and in particular it illustrates the importance of analyzing the effects of compound heterozygous variants in combination to reveal interallelic effects.
Collapse
|
9
|
Wu HY, Zheng Y, Laciak AR, Huang NN, Koszelak-Rosenblum M, Flint AJ, Carr G, Zhu G. Structure and Function of SNM1 Family Nucleases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1414:1-26. [PMID: 35708844 DOI: 10.1007/5584_2022_724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Three human nucleases, SNM1A, SNM1B/Apollo, and SNM1C/Artemis, belong to the SNM1 gene family. These nucleases are involved in various cellular functions, including homologous recombination, nonhomologous end-joining, cell cycle regulation, and telomere maintenance. These three proteins share a similar catalytic domain, which is characterized as a fused metallo-β-lactamase and a CPSF-Artemis-SNM1-PSO2 domain. SNM1A and SNM1B/Apollo are exonucleases, whereas SNM1C/Artemis is an endonuclease. This review contains a summary of recent research on SNM1's cellular and biochemical functions, as well as structural biology studies. In addition, protein structure prediction by the artificial intelligence program AlphaFold provides a different view of the proteins' non-catalytic domain features, which may be used in combination with current results from X-ray crystallography and cryo-EM to understand their mechanism more clearly.
Collapse
|
10
|
Shim HS, Horner JW, Wu CJ, Li J, Lan ZD, Jiang S, Xu X, Hsu WH, Zal T, Flores II, Deng P, Lin YT, Tsai LH, Wang YA, DePinho RA. Telomerase Reverse Transcriptase Preserves Neuron Survival and Cognition in Alzheimer's Disease Models. NATURE AGING 2021; 1:1162-1174. [PMID: 35036927 PMCID: PMC8759755 DOI: 10.1038/s43587-021-00146-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 10/29/2021] [Indexed: 12/19/2022]
Abstract
Amyloid-induced neurodegeneration plays a central role in Alzheimer's disease (AD) pathogenesis. Here, we show that telomerase reverse transcriptase (TERT) haploinsufficiency decreases BDNF and increases amyloid-β (Aβ) precursor in murine brain. Moreover, prior to disease onset, the TERT locus sustains accumulation of repressive epigenetic marks in murine and human AD neurons, implicating TERT repression in amyloid-induced neurodegeneration. To test the impact of sustained TERT expression on AD pathobiology, AD mouse models were engineered to maintain physiological levels of TERT in adult neurons, resulting in reduced Aβ accumulation, improved spine morphology, and preserved cognitive function. Mechanistically, integrated profiling revealed that TERT interacts with β-catenin and RNA polymerase II at gene promoters and upregulates gene networks governing synaptic signaling and learning processes. These TERT-directed transcriptional activities do not require its catalytic activity nor telomerase RNA. These findings provide genetic proof-of-concept for somatic TERT gene activation therapy in attenuating AD progression including cognitive decline.
Collapse
Affiliation(s)
- Hong Seok Shim
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - James W. Horner
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Chang-Jiun Wu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Jiexi Li
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Zheng D. Lan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Shan Jiang
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Xueping Xu
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Wen-Hao Hsu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Tomasz Zal
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ivonne I. Flores
- Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Pingna Deng
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Yuan-Ta Lin
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Li-Huei Tsai
- Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Y. Alan Wang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ronald A. DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
11
|
Xiang Y, Yu Y, Li Q, Jiang Z, Li J, Liang C, Chen J, Li Y, Chen X, Cao W. Mutual regulation between chicken telomerase reverse transcriptase and the Wnt/β-catenin signalling pathway inhibits apoptosis and promotes the replication of ALV-J in LMH cells. Vet Res 2021; 52:110. [PMID: 34412690 PMCID: PMC8375160 DOI: 10.1186/s13567-021-00979-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 07/10/2021] [Indexed: 12/14/2022] Open
Abstract
This study aimed to explore the mutual regulation between chicken telomerase reverse transcriptase (chTERT) and the Wnt/β-catenin signalling pathway and its effects on cell growth and avian leukosis virus subgroup J (ALV-J) replication in LMH cells. First, LMH cells stably overexpressing the chTERT gene (LMH-chTERT cells) and corresponding control cells (LMH-NC cells) were successfully constructed with a lentiviral vector expression system. The results showed that chTERT upregulated the expression of β-catenin, Cyclin D1, TCF4 and c-Myc. chTERT expression level and telomerase activity were increased when cells were treated with LiCl. When the cells were treated with ICG001 or IWP-2, the activity of the Wnt/β-catenin signalling pathway was significantly inhibited, and chTERT expression and telomerase activity were also inhibited. However, when the β-catenin gene was knocked down by small interfering RNA (siRNA), the changes in chTERT expression and telomerase activity were consistent with those in cells treated with ICG001 or IWP-2. These results indicated that chTERT and the Wnt/β-catenin signalling pathway can be mutually regulated. Subsequently, we found that chTERT not only shortened the cell cycle to promote proliferation but also inhibited apoptosis by downregulating the expression of Caspase 3, Caspase 9 and BAX; upregulating BCL-2 and BCL-X expression; and promoting autophagy. Moreover, chTERT significantly enhanced the migration ability of LMH cells, upregulated the protein and mRNA expression of ALV-J and increased the virus titre. ALV-J replication promoted chTERT expression and telomerase activity.
Collapse
Affiliation(s)
- Yong Xiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yun Yu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Qingbo Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Zeng Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jinqun Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Canxin Liang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Jian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Yu Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Xiaoyan Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Weisheng Cao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China. .,Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, South China Agricultural University, Guangzhou, 510642, China. .,Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou, 510642, China. .,National and Regional Joint Engineering Laboratory for Medicament of Zoonosis Prevention and Control, South China Agricultural University, Guangzhou, 510642, China. .,Key Laboratory of Veterinary Vaccine Innovation of the Ministry of Agriculture, Guangzhou, 510642, China.
| |
Collapse
|
12
|
Ma Y, Xia R, Ma X, Judson-Torres RL, Zeng H. Mucosal Melanoma: Pathological Evolution, Pathway Dependency and Targeted Therapy. Front Oncol 2021; 11:702287. [PMID: 34350118 PMCID: PMC8327265 DOI: 10.3389/fonc.2021.702287] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/02/2021] [Indexed: 12/16/2022] Open
Abstract
Mucosal melanoma (MM) is a rare melanoma subtype that originates from melanocytes within sun-protected mucous membranes. Compared with cutaneous melanoma (CM), MM has worse prognosis and lacks effective treatment options. Moreover, the endogenous or exogenous risk factors that influence mucosal melanocyte transformation, as well as the identity of MM precursor lesions, are ambiguous. Consequently, there remains a lack of molecular markers that can be used for early diagnosis, and therefore better management, of MM. In this review, we first summarize the main functions of mucosal melanocytes. Then, using oral mucosal melanoma (OMM) as a model, we discuss the distinct pathologic stages from benign mucosal melanocytes to metastatic MM, mapping the possible evolutionary trajectories that correspond to MM initiation and progression. We highlight key areas of ambiguity during the genetic evolution of MM from its benign lesions, and the resolution of which could aid in the discovery of new biomarkers for MM detection and diagnosis. We outline the key pathways that are altered in MM, including the MAPK pathway, the PI3K/AKT pathway, cell cycle regulation, telomere maintenance, and the RNA maturation process, and discuss targeted therapy strategies for MM currently in use or under investigation.
Collapse
Affiliation(s)
- Yanni Ma
- Department of Oncology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Institute of Precision Medicine, Shanghai, China
| | - Ronghui Xia
- Department of Oral Pathology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xuhui Ma
- Department of Oral & Maxillofacial - Head and Neck Oncology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Robert L Judson-Torres
- Department of Dermatology, University of Utah, Salt Lake City, UT, United States.,Huntsman Cancer Institute, Salt Lake City, UT, United States
| | - Hanlin Zeng
- Department of Oncology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Institute of Precision Medicine, Shanghai, China
| |
Collapse
|
13
|
Sharma S, Mukherjee AK, Roy SS, Bagri S, Lier S, Verma M, Sengupta A, Kumar M, Nesse G, Pandey DP, Chowdhury S. Human telomerase is directly regulated by non-telomeric TRF2-G-quadruplex interaction. Cell Rep 2021; 35:109154. [PMID: 34010660 PMCID: PMC7611063 DOI: 10.1016/j.celrep.2021.109154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/15/2021] [Accepted: 04/28/2021] [Indexed: 12/16/2022] Open
Abstract
Human telomerase reverse transcriptase (hTERT) remains suppressed in most normal somatic cells. Resulting erosion of telomeres leads eventually to replicative senescence. Reactivation of hTERT maintains telomeres and triggers progression of >90% of cancers. However, any direct causal link between telomeres and telomerase regulation remains unclear. Here, we show that the telomere-repeat-binding-factor 2 (TRF2) binds hTERT promoter G-quadruplexes and recruits the polycomb-repressor EZH2/PRC2 complex. This is causal for H3K27 trimethylation at the hTERT promoter and represses hTERT in cancer as well as normal cells. Two highly recurrent hTERT promoter mutations found in many cancers, including ∼83% glioblastoma multiforme, that are known to destabilize hTERT promoter G-quadruplexes, showed loss of TRF2 binding in patient-derived primary glioblastoma multiforme cells. Ligand-induced G-quadruplex stabilization restored TRF2 binding, H3K27-trimethylation, and hTERT re-suppression. These results uncover a mechanism of hTERT regulation through a telomeric factor, implicating telomere-telomerase molecular links important in neoplastic transformation, aging, and regenerative therapy.
Collapse
Affiliation(s)
- Shalu Sharma
- Integrative and Functional Biology Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India
| | - Ananda Kishore Mukherjee
- Integrative and Functional Biology Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India
| | - Shuvra Shekhar Roy
- Integrative and Functional Biology Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India
| | - Sulochana Bagri
- Integrative and Functional Biology Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India
| | - Silje Lier
- Department of Microbiology, Oslo University Hospital, Oslo, Norway; Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Meenakshi Verma
- Integrative and Functional Biology Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India; CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India
| | - Antara Sengupta
- Integrative and Functional Biology Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India
| | - Manish Kumar
- Imaging Facility, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India; CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India
| | - Gaute Nesse
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | | | - Shantanu Chowdhury
- Integrative and Functional Biology Unit, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India; GNR Knowledge Centre for Genome and Informatics, CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India; CSIR-Institute of Genomics and Integrative Biology, New Delhi 110025, India.
| |
Collapse
|
14
|
Romaniuk-Drapała A, Totoń E, Konieczna N, Machnik M, Barczak W, Kowal D, Kopczyński P, Kaczmarek M, Rubiś B. hTERT Downregulation Attenuates Resistance to DOX, Impairs FAK-Mediated Adhesion, and Leads to Autophagy Induction in Breast Cancer Cells. Cells 2021; 10:cells10040867. [PMID: 33920284 PMCID: PMC8068966 DOI: 10.3390/cells10040867] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Telomerase is known to contribute to telomere maintenance and to provide cancer cell immortality. However, numerous reports are showing that the function of the enzyme goes far beyond chromosome ends. The study aimed to explore how telomerase downregulation in MCF7 and MDA-MB-231 breast cancer cells affects their ability to survive. Consequently, sensitivity to drug resistance, proliferation, and adhesion were assessed. The lentiviral-mediated human telomerase reverse transcriptase (hTERT) downregulation efficiency was performed at gene expression and protein level using qPCR and Western blot, respectively. Telomerase activity was evaluated using the Telomeric Repeat Amplification Protocol (TRAP) assay. The study revealed that hTERT downregulation led to an increased sensitivity of breast cancer cells to doxorubicin which was demonstrated in MTT and clonogenic assays. During a long-term doubling time assessment, a decreased population doubling level was observed. Interestingly, it did not dramatically affect cell cycle distribution. hTERT downregulation was accompanied by an alteration in β1-integrin- and by focal adhesion kinase (FAK)-driven pathways together with the reduction of target proteins phosphorylation, i.e., paxillin and c-Src. Additionally, autophagy activation was observed in MDA-MB-231 cells manifested by alternations in Atg5, Beclin 1, LC3II/I ratio, and p62. These results provide new evidence supporting the possible therapeutic potential of telomerase downregulation leading to induction of autophagy and cancer cells elimination.
Collapse
Affiliation(s)
- Aleksandra Romaniuk-Drapała
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355 Poznań, Poland; (A.R.-D.); (E.T.); (N.K.); (D.K.)
| | - Ewa Totoń
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355 Poznań, Poland; (A.R.-D.); (E.T.); (N.K.); (D.K.)
| | - Natalia Konieczna
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355 Poznań, Poland; (A.R.-D.); (E.T.); (N.K.); (D.K.)
| | - Marta Machnik
- Department of Cancer Immunology, Poznan University of Medical Sciences, 60-806 Poznan, Poland;
| | - Wojciech Barczak
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, The Greater Poland Cancer Centre, 61-866 Poznan, Poland;
| | - Dagmar Kowal
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355 Poznań, Poland; (A.R.-D.); (E.T.); (N.K.); (D.K.)
| | - Przemysław Kopczyński
- Centre for Orthodontic Mini-Implants at the Department and Clinic of Maxillofacial Orthopedics and Orthodontics, Poznan University of Medical Sciences, 60-812 Poznan, Poland;
| | - Mariusz Kaczmarek
- Department of Immunology, Chair of Clinical Immunology, Poznań University of Medical Sciences, 5D Rokietnicka St., 60-806 Poznań, Poland;
| | - Błażej Rubiś
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355 Poznań, Poland; (A.R.-D.); (E.T.); (N.K.); (D.K.)
- Correspondence: ; Tel.: +48-61-869-14-27
| |
Collapse
|
15
|
Chen J, Nelson C, Wong M, Tee AE, Liu PY, La T, Fletcher JI, Kamili A, Mayoh C, Bartenhagen C, Trahair TN, Xu N, Jayatilleke N, Wong M, Peng H, Atmadibrata B, Cheung BB, Lan Q, Bryan TM, Mestdagh P, Vandesompele J, Combaret V, Boeva V, Wang JY, Janoueix-Lerosey I, Cowley MJ, MacKenzie KL, Dolnikov A, Li J, Polly P, Marshall GM, Reddel RR, Norris MD, Haber M, Fischer M, Zhang XD, Pickett HA, Liu T. Targeted Therapy of TERT-Rearranged Neuroblastoma with BET Bromodomain Inhibitor and Proteasome Inhibitor Combination Therapy. Clin Cancer Res 2020; 27:1438-1451. [PMID: 33310889 DOI: 10.1158/1078-0432.ccr-20-3044] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/23/2020] [Accepted: 12/08/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE TERT gene rearrangement with transcriptional superenhancers leads to TERT overexpression and neuroblastoma. No targeted therapy is available for clinical trials in patients with TERT-rearranged neuroblastoma. EXPERIMENTAL DESIGN Anticancer agents exerting the best synergistic anticancer effects with BET bromodomain inhibitors were identified by screening an FDA-approved oncology drug library. The synergistic effects of the BET bromodomain inhibitor OTX015 and the proteasome inhibitor carfilzomib were examined by immunoblot and flow cytometry analysis. The anticancer efficacy of OTX015 and carfilzomib combination therapy was investigated in mice xenografted with TERT-rearranged neuroblastoma cell lines or patient-derived xenograft (PDX) tumor cells, and the role of TERT reduction in the anticancer efficacy was examined through rescue experiments in mice. RESULTS The BET bromodomain protein BRD4 promoted TERT-rearranged neuroblastoma cell proliferation through upregulating TERT expression. Screening of an approved oncology drug library identified the proteasome inhibitor carfilzomib as the agent exerting the best synergistic anticancer effects with BET bromodomain inhibitors including OTX015. OTX015 and carfilzomib synergistically reduced TERT protein expression, induced endoplasmic reticulum stress, and induced TERT-rearranged neuroblastoma cell apoptosis which was blocked by TERT overexpression and endoplasmic reticulum stress antagonists. In mice xenografted with TERT-rearranged neuroblastoma cell lines or PDX tumor cells, OTX015 and carfilzomib synergistically blocked TERT expression, induced tumor cell apoptosis, suppressed tumor progression, and improved mouse survival, which was largely reversed by forced TERT overexpression. CONCLUSIONS OTX015 and carfilzomib combination therapy is likely to be translated into the first clinical trial of a targeted therapy in patients with TERT-rearranged neuroblastoma.
Collapse
Affiliation(s)
- Jingwei Chen
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Christopher Nelson
- Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Matthew Wong
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Andrew E Tee
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Pei Y Liu
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Ting La
- School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan, Australia
| | - Jamie I Fletcher
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Alvin Kamili
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Chelsea Mayoh
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Christoph Bartenhagen
- Department of Experimental Pediatric Oncology, Medical Faculty, University Hospital, University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Toby N Trahair
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Ning Xu
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Nisitha Jayatilleke
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Marie Wong
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Hui Peng
- Advanced Analytics Institute, University of Technology Sydney, Ultimo, Australia
| | | | - Belamy B Cheung
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Qing Lan
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Tracy M Bryan
- Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Pieter Mestdagh
- Center for Medical Genetics Ghent, Ghent University, Ghent, Belgium
| | - Jo Vandesompele
- Center for Medical Genetics Ghent, Ghent University, Ghent, Belgium
| | - Valerie Combaret
- Centre Léon-Bérard, Laboratoire de Recherche Translationnelle, Lyon, France
| | - Valentina Boeva
- ETH Zürich, Department of Computer Science, Institute for Machine Learning, Swiss Institute of Bioinformaticsics (SIB), Zurich, Switzerland
- Institut Cochin, Inserm U1016, CNRS UMR 8104, Université Paris Descartes UMR-S1016, Paris, France
| | - Jenny Y Wang
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Isabelle Janoueix-Lerosey
- Institut Curie, Paris Sciences et Lettres Research University, INSERM, U830, Equipe Labellisée Ligue contre le Cancer, Paris, France
- SIREDO: Care, Innovation and Research for Children, Adolescents and Young Adults with Cancer, Institut Curie, Paris, France
| | - Mark J Cowley
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Karen L MacKenzie
- Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Alla Dolnikov
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Jinyan Li
- Advanced Analytics Institute, University of Technology Sydney, Ultimo, Australia
| | - Patsie Polly
- Department of Pathology, School of Medical Sciences, UNSW Sydney, Sydney, New South Wales, Australia
| | - Glenn M Marshall
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, New South Wales, Australia
| | - Roger R Reddel
- Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Murray D Norris
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Michelle Haber
- Children's Cancer Institute, Randwick, Sydney, Australia
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Matthias Fischer
- Department of Experimental Pediatric Oncology, Medical Faculty, University Hospital, University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Xu D Zhang
- School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan, Australia
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou, China
| | - Hilda A Pickett
- Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Tao Liu
- Children's Cancer Institute, Randwick, Sydney, Australia.
- School of Women's and Children's Health, University of New South Wales Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
16
|
Engler M, Fidan M, Nandi S, Cirstea IC. Senescence in RASopathies, a possible novel contributor to a complex pathophenoype. Mech Ageing Dev 2020; 194:111411. [PMID: 33309600 DOI: 10.1016/j.mad.2020.111411] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 11/17/2020] [Accepted: 11/23/2020] [Indexed: 01/07/2023]
Abstract
Senescence is a biological process that induces a permanent cell cycle arrest and a specific gene expression program in response to various stressors. Following studies over the last few decades, the concept of senescence has evolved from an antiproliferative mechanism in cancer (oncogene-induced senescence) to a critical component of physiological processes associated with embryonic development, tissue regeneration, ageing and its associated diseases. In somatic cells, oncogenic mutations in RAS-MAPK pathway genes are associated with oncogene-induced senescence and cancer, while germline mutations in the same pathway are linked to a group of monogenic developmental disorders generally termed RASopathies. Here, we consider that in these disorders, senescence induction may result in opposing outcomes, a tumour protective effect and a possible contributor to a premature ageing phenotype identified in Costello syndrome, which belongs to the RASopathy group. In this review, we will highlight the role of senescence in organismal homeostasis and we will describe the current knowledge about senescence in RASopathies. Additionally, we provide a perspective on examples of experimentally characterised RASopathy mutations that, alone or in combination with various stressors, may also trigger an age-dependent chronic senescence, possibly contributing to the age-dependent worsening of RASopathy pathophenotype and the reduction of lifespan.
Collapse
Affiliation(s)
- Melanie Engler
- Institute of Comparative Molecular Endocrinology, Ulm University, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Miray Fidan
- Institute of Comparative Molecular Endocrinology, Ulm University, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Sayantan Nandi
- Institute of Comparative Molecular Endocrinology, Ulm University, Helmholtzstr. 8/1, 89081, Ulm, Germany
| | - Ion Cristian Cirstea
- Institute of Comparative Molecular Endocrinology, Ulm University, Helmholtzstr. 8/1, 89081, Ulm, Germany.
| |
Collapse
|
17
|
Essentiality of CTNNB1 in Malignant Transformation of Human Embryonic Stem Cells under Long-Term Suboptimal Conditions. Stem Cells Int 2020; 2020:5823676. [PMID: 33029148 PMCID: PMC7532415 DOI: 10.1155/2020/5823676] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 08/14/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022] Open
Abstract
Human embryonic stem cells (hESCs) gradually accumulate abnormal karyotypes during long-term suboptimal culture, which hinder their application in regenerative medicine. Previous studies demonstrated that the activation of CTNNB1 might be implicated in this process. Hence, the hESC line with stably silenced CTNNB1 was established to further explore the role of CTNNB1 in the malignant transformation of hESCs. It was shown to play a vital role in the maintenance of the physiological properties of stem cells, such as proliferation, migration, differentiation, and telomere regulation. Furthermore, the malignant transformation of hESCs was induced by continuous exposure to 0.001 μg/ml mitomycin C (MMC). The results showed that CTNNB1 and its target genes, including proto-oncogenes CCND1 and C-MYC, were aberrantly upregulated in hESCs after MMC treatment. Moreover, the high expression of CTNNB1 accelerated cell transition from G0/G1 phase to the S phase and stimulated the growth of cells containing breakage-fusion-bridge (BFB) cycles. Conversely, CTNNB1 silencing inhibited these effects and triggered a survival crisis. The current data indicated that CTNNB1 is intimately associated with the physiological properties of stem cells; however, the aberrant expression of CTNNB1 is involved in the malignant transformation of hESCs, which might advance the process by facilitating telomere-related unstable cell proliferation. Thus, the aberrant CTNNB1 level might serve as a potential biomarker for detecting the malignant transformation of hESCs.
Collapse
|
18
|
Pańczyszyn A, Boniewska-Bernacka E, Goc A. The role of telomeres and telomerase in the senescence of postmitotic cells. DNA Repair (Amst) 2020; 95:102956. [PMID: 32937289 DOI: 10.1016/j.dnarep.2020.102956] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
Abstract
Senescence is a process related to the stopping of divisions and changes leading the cell to the SASP phenotype. Permanent senescence of many SASP cells contributes to faster aging of the body and development of age-related diseases due to the release of pro-inflammatory factors. Both mitotically active and non-dividing cells can undergo senescence as a result of activation of different molecular pathways. Telomeres, referred to as the molecular clock, direct the dividing cell into the aging pathway when reaching a critical length. In turn, the senescence of postmitotic cells depends not on the length of telomeres, but their functionality. Dysfunctional telomeres are responsible for triggering the signaling of DNA damage response (DDR). Telomerase subunits in post-mitotic cells translocate between the nucleus, cytoplasm and mitochondria, participating in the regulation of their activity. Among other things, they contribute to the reduction of reactive oxygen species generation, which leads to telomere dysfunction and, consequently, senescence. Some proteins of the shelterin complex also play a protective role by inhibiting senescence-initiating kinases and limiting ROS production by mitochondria.
Collapse
Affiliation(s)
- Anna Pańczyszyn
- University of Opole, Institute of Medical Sciences, Department of Biology and Genetics, Opole 45-040, Pl.Kopernika 11a, Poland.
| | - Ewa Boniewska-Bernacka
- University of Opole, Institute of Medical Sciences, Department of Biology and Genetics, Opole 45-040, Pl.Kopernika 11a, Poland.
| | - Anna Goc
- University of Opole, Institute of Medical Sciences, Department of Biology and Genetics, Opole 45-040, Pl.Kopernika 11a, Poland.
| |
Collapse
|
19
|
Guterres AN, Villanueva J. Targeting telomerase for cancer therapy. Oncogene 2020; 39:5811-5824. [PMID: 32733068 PMCID: PMC7678952 DOI: 10.1038/s41388-020-01405-w] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 07/02/2020] [Accepted: 07/23/2020] [Indexed: 12/20/2022]
Abstract
Telomere maintenance via telomerase reactivation is a nearly universal hallmark of cancer cells which enables replicative immortality. In contrast, telomerase activity is silenced in most adult somatic cells. Thus, telomerase represents an attractive target for highly selective cancer therapeutics. However, development of telomerase inhibitors has been challenging and thus far there are no clinically approved strategies exploiting this cancer target. The discovery of prevalent mutations in the TERT promoter region in many cancers and recent advances in telomerase biology has led to a renewed interest in targeting this enzyme. Here we discuss recent efforts targeting telomerase, including immunotherapies and direct telomerase inhibitors, as well as emerging approaches such as targeting TERT gene expression driven by TERT promoter mutations. We also address some of the challenges to telomerase-directed therapies including potential therapeutic resistance and considerations for future therapeutic applications and translation into the clinical setting. Although much work remains to be done, effective strategies targeting telomerase will have a transformative impact for cancer therapy and the prospect of clinically effective drugs is boosted by recent advances in structural models of human telomerase.
Collapse
Affiliation(s)
- Adam N Guterres
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Jessie Villanueva
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA.
- Melanoma Research Center, The Wistar Institute, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Bryan TM. G-Quadruplexes at Telomeres: Friend or Foe? Molecules 2020; 25:molecules25163686. [PMID: 32823549 PMCID: PMC7464828 DOI: 10.3390/molecules25163686] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 12/28/2022] Open
Abstract
Telomeres are DNA-protein complexes that cap and protect the ends of linear chromosomes. In almost all species, telomeric DNA has a G/C strand bias, and the short tandem repeats of the G-rich strand have the capacity to form into secondary structures in vitro, such as four-stranded G-quadruplexes. This has long prompted speculation that G-quadruplexes play a positive role in telomere biology, resulting in selection for G-rich tandem telomere repeats during evolution. There is some evidence that G-quadruplexes at telomeres may play a protective capping role, at least in yeast, and that they may positively affect telomere maintenance by either the enzyme telomerase or by recombination-based mechanisms. On the other hand, G-quadruplex formation in telomeric DNA, as elsewhere in the genome, can form an impediment to DNA replication and a source of genome instability. This review summarizes recent evidence for the in vivo existence of G-quadruplexes at telomeres, with a focus on human telomeres, and highlights some of the many unanswered questions regarding the location, form, and functions of these structures.
Collapse
Affiliation(s)
- Tracy M Bryan
- Children's Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| |
Collapse
|
21
|
Jones RJ, Singh RK, Shirazi F, Wan J, Wang H, Wang X, Ha MJ, Baljevic M, Kuiatse I, Davis RE, Orlowski RZ. Intravenous Immunoglobulin G Suppresses Heat Shock Protein (HSP)-70 Expression and Enhances the Activity of HSP90 and Proteasome Inhibitors. Front Immunol 2020; 11:1816. [PMID: 32903557 PMCID: PMC7438474 DOI: 10.3389/fimmu.2020.01816] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/07/2020] [Indexed: 12/11/2022] Open
Abstract
Intravenous immunoglobulin G (IVIgG) is approved for primary immunodeficiency syndromes but may induce anti-cancer effects, and while this has been attributed to its anti-inflammatory properties, IgG against specific tumor targets may play a role. We evaluated IVIgG alone, and with a Heat shock protein (HSP)-90 or proteasome inhibitor, using multiple myeloma and mantle cell lymphoma (MCL) cells in vitro, and with the proteasome inhibitor bortezomib in vivo. IVIgG inhibited the growth of all cell lines tested, induced G1 cell cycle arrest, and suppressed pro-tumor cytokines including Interleukin (IL)-6, IL-8, and IL-10. Genomic and proteomic studies showed that IVIgG reduced tumor cell HSP70-1 levels by suppressing the ability of extracellular HSP70-1 to stimulate endogenous HSP70-1 promoter activity, and reduced extracellular vesicle uptake. Preparations of IVIgG were found to contain high titers of anti-HSP70-1 IgG, and recombinant HSP70-1 reduced the efficacy of IVIgG to suppress HSP70-1 levels. Combining IVIgG with the HSP90 inhibitor AUY922 produced superior cell growth inhibition and correlated with HSP70-1 suppression. Also, IVIgG with bortezomib or carfilzomib was superior to each single agent, and enhanced bortezomib's activity in bortezomib-resistant myeloma cells. Moreover, IVIgG reduced transfer of extracellular vesicles (EVs) to cells, and blocked transfer of bortezomib resistance through EVs. Finally, IVIgG with bortezomib were superior to the single agents in an in vivo myeloma model. These studies support the possibility that anti-HSP70-1 IgG contained in IVIgG can inhibit myeloma and MCL growth by interfering with a novel mechanism involving uptake of exogenous HSP70-1 which then induces its own promoter.
Collapse
Affiliation(s)
- Richard J Jones
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ram K Singh
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Fazal Shirazi
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jie Wan
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hua Wang
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xiaobin Wang
- The Urology Department, ShengJing Hospital, China Medical University, ShenYang, China
| | - Min Jin Ha
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Muhamed Baljevic
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States
| | - Isere Kuiatse
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Richard E Davis
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Robert Z Orlowski
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, TX, United States.,Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
22
|
Abstract
Several decades elapsed between the first descriptions of G-quadruplex nucleic acid structures (G4s) assembled in vitro and the emergence of experimental findings indicating that such structures can form and function in living systems. A large body of evidence now supports roles for G4s in many aspects of nucleic acid biology, spanning processes from transcription and chromatin structure, mRNA processing, protein translation, DNA replication and genome stability, and telomere and mitochondrial function. Nonetheless, it must be acknowledged that some of this evidence is tentative, which is not surprising given the technical challenges associated with demonstrating G4s in biology. Here I provide an overview of evidence for G4 biology, focusing particularly on the many potential pitfalls that can be encountered in its investigation, and briefly discuss some of broader biological processes that may be impacted by G4s including cancer.
Collapse
Affiliation(s)
- F. Brad Johnson
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
23
|
Telomerase Biogenesis and Activities from the Perspective of Its Direct Interacting Partners. Cancers (Basel) 2020; 12:cancers12061679. [PMID: 32599885 PMCID: PMC7352425 DOI: 10.3390/cancers12061679] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 12/15/2022] Open
Abstract
Telomerase reverse transcriptase (TERT)—the catalytic subunit of telomerase—is reactivated in up to 90% of all human cancers. TERT is observed in heterogenous populations of protein complexes, which are dynamically regulated in a cell type- and cell cycle-specific manner. Over the past two decades, in vitro protein–protein interaction detection methods have discovered a number of endogenous TERT binding partners in human cells that are responsible for the biogenesis and functionalization of the telomerase holoenzyme, including the processes of TERT trafficking between subcellular compartments, assembly into telomerase, and catalytic action at telomeres. Additionally, TERT have been found to interact with protein species with no known telomeric functions, suggesting that these complexes may contribute to non-canonical activities of TERT. Here, we survey TERT direct binding partners and discuss their contributions to TERT biogenesis and functions. The goal is to review the comprehensive spectrum of TERT pro-malignant activities, both telomeric and non-telomeric, which may explain the prevalence of its upregulation in cancer.
Collapse
|
24
|
Betori RC, Liu Y, Mishra RK, Cohen SB, Kron SJ, Scheidt KA. Targeted Covalent Inhibition of Telomerase. ACS Chem Biol 2020; 15:706-717. [PMID: 32017522 DOI: 10.1021/acschembio.9b00945] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Telomerase is a ribonuceloprotein complex responsible for maintaining telomeres and protecting chromosomal integrity. The human telomerase reverse transcriptase (hTERT) is expressed in ∼90% of cancer cells where it confers the capacity for limitless proliferation. Along with its established role in telomere lengthening, telomerase also serves noncanonical extra-telomeric roles in oncogenic signaling, resistance to apoptosis, and enhanced DNA damage response. We report a new class of natural-product-inspired covalent inhibitors of telomerase that target the catalytic active site.
Collapse
Affiliation(s)
- Rick C. Betori
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Yue Liu
- Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois 60637, United States
| | - Rama K. Mishra
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, United States
| | - Scott B. Cohen
- Children’s Medical Research Institute, University of Sydney, Westmead, New South Wales 2145, Australia
| | - Stephen J. Kron
- Ludwig Center for Metastasis Research, The University of Chicago, Chicago, Illinois 60637, United States
| | - Karl A. Scheidt
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
- Department of Pharmacology, Northwestern University, Chicago, Illinois 60611, United States
- Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, Illinois 60208, United States
| |
Collapse
|