1
|
Lee S, Chang TC, Schreiner P, Fan Y, Agarwal N, Owens C, Dummer R, Kirkwood JM, Barnhill RL, Theodorescu D, Wu G, Bahrami A. Targeted Long-Read Bisulfite Sequencing Identifies Differences in the TERT Promoter Methylation Profiles between TERT Wild-Type and TERT Mutant Cancer Cells. Cancers (Basel) 2022; 14:4018. [PMID: 36011010 PMCID: PMC9406525 DOI: 10.3390/cancers14164018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/06/2022] [Accepted: 08/10/2022] [Indexed: 11/17/2022] Open
Abstract
Background: TERT promoter methylation, located several hundred base pairs upstream of the transcriptional start site, is cancer specific and correlates with increased TERT mRNA expression and poorer patient outcome. Promoter methylation, however, is not mutually exclusive to TERT activating genetic alterations, as predicted for functionally redundant mechanisms. To annotate the altered patterns of TERT promoter methylation and their relationship with gene expression, we applied a Pacific Biosciences-based, long-read, bisulfite-sequencing technology and compared the differences in the methylation marks between wild-type and mutant cancers in an allele-specific manner. Results: We cataloged TERT genetic alterations (i.e., promoter point mutations or structural variations), allele-specific promoter methylation patterns, and allele-specific expression levels in a cohort of 54 cancer cell lines. In heterozygous mutant cell lines, the mutant alleles were significantly less methylated than their silent, mutation-free alleles (p < 0.05). In wild-type cell lines, by contrast, both epialleles were equally methylated to high levels at the TERT distal promoter, but differentially methylated in the proximal regions. ChIP analysis showed that epialleles with the hypomethylated proximal and core promoter were enriched in the active histone mark H3K4me2/3, whereas epialleles that were methylated in those regions were enriched in the repressive histone mark H3K27me3. Decitabine therapy induced biallelic expression in the wild-type cancer cells, whereas the mutant cell lines were unaffected. Conclusions: Long-read bisulfite sequencing analysis revealed differences in the methylation profiles and responses to demethylating agents between TERT wild-type and genetically altered cancer cell lines. The causal relation between TERT promoter methylation and gene expression remains to be established.
Collapse
Affiliation(s)
- Seungjae Lee
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Ti-Cheng Chang
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN 38015, USA
| | - Patrick Schreiner
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN 38015, USA
| | - Yiping Fan
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN 38015, USA
| | - Neeraj Agarwal
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA 90048, USA
| | - Charles Owens
- Department of Surgery, University of Colorado-Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - John M. Kirkwood
- Department of Pathology, University of Pittsburgh Cancer Center, Pittsburgh, PA 15232, USA
| | | | - Dan Theodorescu
- Cedars-Sinai Samuel Oschin Comprehensive Cancer Institute, Los Angeles, CA 90048, USA
- Department of Surgery (Urology), Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gang Wu
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN 38015, USA
| | - Armita Bahrami
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30307, USA
| |
Collapse
|
2
|
Alidousty C, Duerbaum N, Wagener-Ryczek S, Baar T, Martelotto LG, Heydt C, Siemanowski J, Holz B, Binot E, Fassunke J, Merkelbach-Bruse S, Wolf J, Kron A, Buettner R, Schultheis AM. Prevalence and potential biological role of TERT amplifications in ALK translocated adenocarcinoma of the lung. Histopathology 2020; 78:578-585. [PMID: 32946634 DOI: 10.1111/his.14256] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/11/2020] [Indexed: 12/20/2022]
Abstract
AIMS The advent of specific ALK-targeting drugs has radically changed the outcome of patients with ALK translocated non-small-cell lung cancer (NSCLC). However, emerging resistance to treatment with ALK inhibitors in these patients remains a major concern. In previous studies, we analysed two ALK+ patient cohorts (TP53 wild-type/TP53 mutated) in terms of copy number alterations. All patients belonging to the TP53 wild-type group had mainly genetically stable genomes, with one exception showing chromosomal instability and amplifications of several gene loci, including TERT. Here, we aimed to determine the prevalence of TERT amplifications in these ALK+ lung cancer patients by analysing an independent cohort of 109 ALK translocated cases. We further analysed the copy numbers of numerous cancer-relevant genes and other genetic aberrations. METHODS AND RESULTS The prevalence of TERT amplifications was determined by means of FISH analyses. Copy numbers of 87 cancer-relevant genes were determined by NanoString nCounter® technology, FoundationOne® and lung-specific NGS panels in some of these TERT-amplified samples, and clinical data on patients with TERT-amplified tumours were collected. Our data revealed that five (4.6%) of all 109 analysed ALK+ patients harboured amplification of TERT and that these patients had genetically unstable genomes. CONCLUSIONS Our preliminary study shows that ALK+ adenocarcinomas should be evaluated in the context of their genomic background in order to more clearly understand and predict patients' individual course of disease.
Collapse
Affiliation(s)
| | - Nicolai Duerbaum
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | | - Till Baar
- Faculty of Medicine, Institute of Medical Statistics and Computational Biology, University of Cologne, Cologne, Germany
| | | | - Carina Heydt
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Janna Siemanowski
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Barbara Holz
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Elke Binot
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Jana Fassunke
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | | - Jürgen Wolf
- Network Genomic Medicine, Cologne, Germany.,Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany.,Center for Integrated Oncology Koeln Bonn, Cologne, Germany
| | - Anna Kron
- Network Genomic Medicine, Cologne, Germany.,Lung Cancer Group Cologne, Department I of Internal Medicine, University Hospital of Cologne, Cologne, Germany.,Center for Integrated Oncology Koeln Bonn, Cologne, Germany
| | - Reinhard Buettner
- Institute of Pathology, University Hospital Cologne, Cologne, Germany.,Network Genomic Medicine, Cologne, Germany.,Center for Integrated Oncology Koeln Bonn, Cologne, Germany
| | - Anne M Schultheis
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| |
Collapse
|
3
|
Rowland TJ, Bonham AJ, Cech TR. Allele-specific proximal promoter hypomethylation of the telomerase reverse transcriptase gene (TERT) associates with TERT expression in multiple cancers. Mol Oncol 2020; 14:2358-2374. [PMID: 33245585 PMCID: PMC7530785 DOI: 10.1002/1878-0261.12786] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/04/2020] [Accepted: 08/13/2020] [Indexed: 12/14/2022] Open
Abstract
Telomerase reverse transcriptase (TERT) is pathologically expressed in the vast majority of human cancers, but the epigenetic regulation of its expression is only beginning to be understood. In particular, the active TERT gene in cancer cells has been characterized as having a hypermethylated CpG island, opposite to the general association of DNA methylation with gene repression. Here, we analyzed TERT promoter CpG methylation in 833 human cancer cell lines representing 23 different tissue types and found hypermethylation of the upstream portion of the CpG island and more conserved hypomethylation of a region including the proximal TERT promoter and exon 1. In cell lines with monoallelic expression of TERT, we found allelic methylation of the proximal TERT promoter. This included cell lines with the -124 or -146 activating promoter mutation as well as wild-type TERT cancer lines. In these cell line types, decreased proximal promoter methylation is associated with the active allele. Compared to cells with monoallelic expression of TERT, lines with biallelic expression of TERT had even lower methylation in the proximal TERT promoter. Thus, in cell lines from cancers of many different tissues, the TERT proximal promoter has canonical DNA methylation, with low methylation correlating with increased TERT expression.
Collapse
Affiliation(s)
- Teisha J. Rowland
- Department of BiochemistryBioFrontiers InstituteUniversity of Colorado BoulderBoulderCOUSA
- Howard Hughes Medical InstituteUniversity of Colorado BoulderBoulderCOUSA
| | - Andrew J. Bonham
- Department of Chemistry & BiochemistryMetropolitan State University of DenverDenverCOUSA
| | - Thomas R. Cech
- Department of BiochemistryBioFrontiers InstituteUniversity of Colorado BoulderBoulderCOUSA
- Howard Hughes Medical InstituteUniversity of Colorado BoulderBoulderCOUSA
| |
Collapse
|
4
|
Yuan T, Zhao W, Niu Y, Fu Y, Lu L, Niu D. Exploration of the temporal-spatial expression pattern and DNA methylation-related regulation of the duck telomerase reverse transcriptase gene. Poult Sci 2019; 98:3257-3267. [PMID: 31064004 DOI: 10.3382/ps/pez240] [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: 10/27/2018] [Accepted: 04/09/2019] [Indexed: 12/14/2022] Open
Abstract
Telomerase reverse transcriptase (TERT) is a catalytic subunit of telomerase that adds TTAGGG repeats to the 3'-overhang of telomeres. In the present study, we detected that the duck TERT (dTERT) gene was highly expressed in small intestine and kidney, followed by heart, leg muscle, spleen, pancreas, gonad, and liver at neonatal stage. From embryonic to neonatal stage, the highest dTERT mRNA in liver appeared at stage E19 (19 days at embryonic stage), while for the leg muscle the maximum expression occurred at E26. We also measured the relative telomerase activity (RTA) and relative telomere length (RTL) in the examined tissues and found that the changed tendency of RTA and RTL was not very consistent with that of TERT. In silico analysis revealed that there were three CpG islands (S1, S2, and S3) within the 5' regulatory region of the dTERT gene. Bisulfite sequencing PCR (BSP) assay showed that liver (D7, 7 days after birth) which expressed significantly lower dTERT mRNA had an obviously higher methylation level of S1 compared with small intestine (D7) or liver (E19). Quantitative real-time PCR analysis revealed that the expression of DNA methyltransferase DNMT1 in liver (D7) was significantly higher than that in small intestine (D7) or in liver (E19). In vitro, dTERT expression was upregulated and the methylation status of S1 decreased in both duck embryonic fibroblasts and small intestinal epithelial cells following treatment with the demethylation reagent, 5-aza-2'-deoxycytidine (5-aza-dC), further suggesting that dTERT is epigenetically regulated by DNA methylation. This work lays a solid foundation for further study of TERT function and regulation in avian species.
Collapse
Affiliation(s)
- Taoyan Yuan
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.,Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, 145 Shiqiao Road, Hangzhou 310021, China
| | - Wanqiu Zhao
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China.,Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, 145 Shiqiao Road, Hangzhou 310021, China
| | - Yifan Niu
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou 450002, Henan, China
| | - Yan Fu
- College of Animal Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Lizhi Lu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, 145 Shiqiao Road, Hangzhou 310021, China.,Key Laboratory of Information Traceability for Agricultural Products, Ministry of Agriculture of China, Hangzhou 310021, Zhejiang, China
| | - Dong Niu
- College of Animal Science and Technology, Zhejiang A&F University
| |
Collapse
|
5
|
Jie MM, Chang X, Zeng S, Liu C, Liao GB, Wu YR, Liu CH, Hu CJ, Yang SM, Li XZ. Diverse regulatory manners of human telomerase reverse transcriptase. Cell Commun Signal 2019; 17:63. [PMID: 31186051 PMCID: PMC6560729 DOI: 10.1186/s12964-019-0372-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 05/17/2019] [Indexed: 12/22/2022] Open
Abstract
Human telomerase reverse transcriptase (hTERT) is the core subunit of human telomerase and plays important roles in human cancers. Aberrant expression of hTERT is closely associated with tumorigenesis, cancer cell stemness maintaining, cell proliferation, apoptosis inhibition, senescence evasion and metastasis. The molecular basis of hTERT regulation is highly complicated and consists of various layers. A deep and full-scale comprehension of the regulatory mechanisms of hTERT is pivotal in understanding the pathogenesis and searching for therapeutic approaches. In this review, we summarize the recent advances regarding the diverse regulatory mechanisms of hTERT, including the transcriptional (promoter mutation, promoter region methylation and histone acetylation), post-transcriptional (mRNA alternative splicing and non-coding RNAs) and post-translational levels (phosphorylation and ubiquitination), which may provide novel perspectives for further translational diagnosis or therapeutic strategies targeting hTERT.
Collapse
Affiliation(s)
- Meng-Meng Jie
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Xing Chang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Shuo Zeng
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Cheng Liu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Guo-Bin Liao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Ya-Ran Wu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Chun-Hua Liu
- Teaching evaluation center of Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Chang-Jiang Hu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China.
| | - Xin-Zhe Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China.
| |
Collapse
|
6
|
ALV Integration-Associated Hypomethylation at the TERT Promoter Locus. Viruses 2018; 10:v10020074. [PMID: 29439385 PMCID: PMC5850381 DOI: 10.3390/v10020074] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 02/06/2018] [Accepted: 02/08/2018] [Indexed: 12/13/2022] Open
Abstract
Avian leukosis virus (ALV) is a simple retrovirus that can induce B-cell lymphoma in chicken(s) and other birds by insertional mutagenesis. The promoter region of telomerase reverse transcriptase (TERT) has been identified as an important integration site for tumorigenesis. Tumors with TERT promoter integrations are associated with increased TERT expression. The mechanism of this activation is still under investigation. We asked whether insertion of proviral DNA perturbs the epigenome of the integration site and, subsequently, impacts the regulation of neighboring genes. DNA cytosine methylation, which generally acts to suppress transcription, is one major form of epigenetic regulation. In this study, we examine allele-specific methylation patterns of genomic DNA from chicken tumors by bisulfite sequencing. We observed that alleles with TERT promoter integrations are associated with decreased methylation in the host genome near the site of integration. Our observations suggest that insertion of ALV in the TERT promoter region may induce expression of TERT through inhibition of maintenance methylation in the TERT promoter region.
Collapse
|
7
|
Haraguchi K, Sato S, Habu M, Yada N, Hayakawa M, Takahashi O, Yoshioka I, Matsuo K, Tominaga K, Takenaka S. Oral Cancer Screening Based on Methylation Frequency Detection inhTERTGene Using Electrochemical Hybridization Assay via a Multi-electrode Chip Coupled with Ferrocenylnaphthalene Diimide. ELECTROANAL 2017. [DOI: 10.1002/elan.201700028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Kazuya Haraguchi
- Department of Science of Physical Functions; Division of Oral and Maxillofacial Surgery; Kyushu Dental University; 2-6-1 Manazuru, Kokurakitaku, Kitakyushu Fukuoka 803-8580 Japan
| | - Shinobu Sato
- Department of Applied Chemistry and Research Center for Bio-microsensing Technology; Kyushu Institute of Technology; 1-1 Sensui, Tobataku, Kitakyushu Fukuoka 804-8550 Japan
| | - Manabu Habu
- Department of Science of Physical Functions; Division of Oral and Maxillofacial Surgery; Kyushu Dental University; 2-6-1 Manazuru, Kokurakitaku, Kitakyushu Fukuoka 803-8580 Japan
| | - Naomi Yada
- Department of Health Promotion, Division of Oral Pathology; Kyushu Dental University; 2-6-1 Manazuru, Kokurakitaku, Kitakyushu Fukuoka 803-8580 Japan
| | - Mana Hayakawa
- Department of Science of Physical Functions; Division of Oral and Maxillofacial Surgery; Kyushu Dental University; 2-6-1 Manazuru, Kokurakitaku, Kitakyushu Fukuoka 803-8580 Japan
| | - Osamu Takahashi
- Department of Science of Physical Functions; Division of Oral and Maxillofacial Surgery; Kyushu Dental University; 2-6-1 Manazuru, Kokurakitaku, Kitakyushu Fukuoka 803-8580 Japan
| | - Izumi Yoshioka
- Department of Science of Physical Functions, Division of Oral Medicine; Kyushu Dental University; 2-6-1 Manazuru, Kokurakitaku, Kitakyushu Fukuoka 803-8580 Japan
| | - Kou Matsuo
- Department of Health Promotion, Division of Oral Pathology; Kyushu Dental University; 2-6-1 Manazuru, Kokurakitaku, Kitakyushu Fukuoka 803-8580 Japan
| | - Kazuhiro Tominaga
- Department of Science of Physical Functions; Division of Oral and Maxillofacial Surgery; Kyushu Dental University; 2-6-1 Manazuru, Kokurakitaku, Kitakyushu Fukuoka 803-8580 Japan
| | - Shigeori Takenaka
- Department of Applied Chemistry and Research Center for Bio-microsensing Technology; Kyushu Institute of Technology; 1-1 Sensui, Tobataku, Kitakyushu Fukuoka 804-8550 Japan
| |
Collapse
|
8
|
Molano M, Moreno-Acosta P, Morales N, Burgos M, Buitrago L, Gamboa O, Alvarez R, Garland SM, Tabrizi SN, Steenbergen RDM, Mejía JC. Association Between Type-specific HPV Infections and hTERT DNA Methylation in Patients with Invasive Cervical Cancer. Cancer Genomics Proteomics 2017; 13:483-491. [PMID: 27807071 DOI: 10.21873/cgp.20011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Accepted: 09/21/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There exists limited information on the role of hTERT methylation, and its association with type-specific HPV infections in cervical cancer. MATERIALS AND METHODS Eighty-seven frozen samples were analyzed for type-specific HPV infection using a GP5+/GP6+ PCR-RLB assay (RLB). hTERT DNA methylation analysis was performed using a newly developed PCR-RLB-hTERT. RESULTS Ninety-three percent of samples were HPV-positive and fifteen different types were detected. hTERT methylation analysis of region 1 revealed no methylation in 78.8% of the samples and partial methylation in 21.2%. In region two, 68.2% showed no methylation and 31.8% showed a pattern of partial methylation. An association between the alpha 9 and alpha 7 species with a pattern of no methylation of hTERT in the region 1 was established (p=0.02 and p=0.03, respectively). CONCLUSION Differences in patterns of methylation of the hTERT core promoter [region 1 (nt -208 to -1) and region 2 (nt +1 to +104) relative to first ATG] are related to the HPV species present.
Collapse
Affiliation(s)
- Mónica Molano
- Research Group in Cancer Biology, Research Branch, National Cancer Institute, Bogotá, Colombia.,Research Group in Radiobiology Clinical, Molecular and Cellular, National Cancer Institute, Bogotá, Colombia.,Microbiology and Infection Diseases, The Royal Women´s Hospital, Melbourne, VIC, Australia
| | - Pablo Moreno-Acosta
- Research Group in Cancer Biology, Research Branch, National Cancer Institute, Bogotá, Colombia .,Research Group in Radiobiology Clinical, Molecular and Cellular, National Cancer Institute, Bogotá, Colombia
| | - Nicolás Morales
- Research Group in Cancer Biology, Research Branch, National Cancer Institute, Bogotá, Colombia
| | - Marcela Burgos
- Research Group in Cancer Biology, Research Branch, National Cancer Institute, Bogotá, Colombia
| | - Lina Buitrago
- Unit Group of Analysis, Research Branch, National Cancer Institute, Bogotá, Colombia
| | - Oscar Gamboa
- Research Group in Radiobiology Clinical, Molecular and Cellular, National Cancer Institute, Bogotá, Colombia.,Unit Group of Analysis, Research Branch, National Cancer Institute, Bogotá, Colombia
| | - Rayner Alvarez
- Research Group in Cancer Biology, Research Branch, National Cancer Institute, Bogotá, Colombia.,Research Group in Radiobiology Clinical, Molecular and Cellular, National Cancer Institute, Bogotá, Colombia
| | - Suzanne M Garland
- Microbiology and Infection Diseases, The Royal Women´s Hospital, Melbourne, VIC, Australia.,Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
| | - Sepehr N Tabrizi
- Microbiology and Infection Diseases, The Royal Women´s Hospital, Melbourne, VIC, Australia.,Murdoch Children's Research Institute, Parkville, VIC, Australia.,Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC, Australia
| | | | - Juan Carlos Mejía
- Oncological Pathology Group, National Cancer Institute, Bogotá, Colombia
| |
Collapse
|
9
|
Telomere Length Maintenance and Cardio-Metabolic Disease Prevention Through Exercise Training. Sports Med 2016; 46:1213-37. [DOI: 10.1007/s40279-016-0482-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
10
|
Heeg S. Variations in telomere maintenance and the role of telomerase inhibition in gastrointestinal cancer. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2015; 8:171-80. [PMID: 26675332 PMCID: PMC4675635 DOI: 10.2147/pgpm.s52808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Immortalization is an important step toward the malignant transformation of human cells and is critically dependent upon telomere maintenance. There are two known mechanisms to maintain human telomeres. The process of telomere maintenance is either mediated through activation of the enzyme telomerase or through an alternative mechanism of telomere lengthening called ALT. While 85% of all human tumors show reactivation of telomerase, the remaining 15% are able to maintain telomeres via ALT. The therapeutic potential of telomerase inhibitors is currently investigated in a variety of human cancers. Gastrointestinal tumors are highly dependent on telomerase as a mechanism of telomere maintenance, rendering telomeres as well as telomerase potential targets for cancer therapy. This article focuses on the molecular mechanisms of telomere biology and telomerase activation in gastrointestinal cancers and reviews strategies of telomerase inhibition and their potential therapeutic use in these tumor entities.
Collapse
Affiliation(s)
- Steffen Heeg
- Department of Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, Medical Center - University of Freiburg, Freiburg, Germany
| |
Collapse
|
11
|
Aberrant methylation of the TERT promoter in esophageal squamous cell carcinoma. Cancer Genet 2015; 208:602-9. [PMID: 26669682 DOI: 10.1016/j.cancergen.2015.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 10/20/2015] [Accepted: 10/26/2015] [Indexed: 02/07/2023]
Abstract
A recent study indicated that upstream of the transcription start site (UTSS) hypermethylation of the telomerase reverse transcriptase (TERT) gene was associated with tumor progression and poor prognosis in pediatric brain tumors. The potential for methylation-mediated regulation of the UTSS region of the TERT gene in esophageal squamous cell carcinoma (ESCC) has not yet been investigated. Here, TERT methylation was investigated in tumor and paired adjacent non-cancerous tissues (ANT) from 185 ESCC patients, and the expression of TERT was investigated in 26 tumors paired with ANTs selected from the same cohort. The methylation level of TERT was analyzed in three different regions: region 1, region 2, and the UTSS region. Comparison and correlation of methylation level and clinical features were analyzed in the abovementioned regions. The results showed that the methylation level of TERT was significantly elevated in the tumor relative to the ANT in ESCC. TERT RNA expression was significantly reduced in primary tumors. Tumor stage was the major determinant of survival. The UTSS region may not be an accessible biomarker for ESCC.
Collapse
|
12
|
Karsli-Ceppioglu S, Ngollo M, Adjakly M, Dagdemir A, Judes G, Lebert A, Boiteux JP, Penault-LLorca F, Bignon YJ, Guy L, Bernard-Gallon D. Genome-wide DNA methylation modified by soy phytoestrogens: role for epigenetic therapeutics in prostate cancer? OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2015; 19:209-19. [PMID: 25831061 DOI: 10.1089/omi.2014.0142] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In prostate cancer, DNA methylation is significantly associated with tumor initiation, progression, and metastasis. Previous studies have suggested that soy phytoestrogens might regulate DNA methylation at individual candidate gene loci and that they play a crucial role as potential therapeutic agents for prostate cancer. The purpose of our study was to examine the modulation effects of phytoestrogens on a genome-wide scale in regards to DNA methylation in prostate cancer. Prostate cancer cell lines DU-145 and LNCaP were treated with 40 μM of genistein and 110 μM of daidzein. DNMT inhibitor 5-azacytidine (2 μM) and the methylating agent budesonide (2 μM) were used to compare their demethylation/methylation effects with phytoestrogens. The regulatory effects of phytoestrogens on DNA methylation were analyzed by using a methyl-DNA immunoprecipitation method coupled with Human DNA Methylation Microarrays (MeDIP-chip). We observed that the methylation profiles of 58 genes were altered by genistein and daidzein treatments in DU-145 and LNCaP prostate cancer cells. In addition, the methylation frequencies of the MAD1L1, TRAF7, KDM4B, and hTERT genes were remarkably modified by genistein treatment. Our results suggest that the modulation effects of phytoestrogens on DNA methylation essentially lead to inhibition of cell growth and induction of apoptosis. Genome-wide methylation profiling reported here suggests that epigenetic regulation mechanisms and, by extension, epigenetics-driven novel therapeutic candidates warrant further consideration in future "omics" studies of prostate cancer.
Collapse
Affiliation(s)
- Seher Karsli-Ceppioglu
- 1 Department of Oncogenetics, Centre Jean Perrin-CBRV , Dunant, Clermont-Ferrand, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Zhang ZX, Wang Y, Tao ZZ, Chen SM, Xiao BK, Zhou T. Subtelomeric Demethylation Deregulated hTERT Expression, Telomerase Activity, and Telomere Length in Four Nasopharyngeal Carcinoma Cell Lines. Cancer Biother Radiopharm 2014; 29:289-94. [PMID: 25153197 DOI: 10.1089/cbr.2013.1581] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Zi-Xiong Zhang
- Department of Otolaryngology, Head and Neck Surgery, Renmin Hospital of Wuhan University , Wuhan, People's Republic of China
| | | | | | | | | | | |
Collapse
|
14
|
Qin Y, Guo H, Tang B, Yang SM. The non-reverse transcriptase activity of the human telomerase reverse transcriptase promotes tumor progression (review). Int J Oncol 2014; 45:525-31. [PMID: 24888567 DOI: 10.3892/ijo.2014.2470] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 04/17/2014] [Indexed: 11/05/2022] Open
Abstract
In human cancer, high expression of telomerase is correlated with tumor aggressiveness and metastatic potential. Human telomerase reverse transcriptase (hTERT), which regulates telomere length, can promote tumor development. Most research on hTERT has been focused on its crucial function of telomere maintenance. However, there are many phenomena that cannot be explained by its reverse transcriptase activity. Accumulating evidence suggests that hTERT has functions independent of its protective function at the telomere ends, such as increasing the anti-apoptotic capacity of cells, enhancing DNA repair, maintaining stem cells and regulating gene expression. This review will provide an update on the non-reverse transcriptase activity of hTERT and its contribution to tumor formation, metastasis and cancer stem cell maintenance. Repression of the non-reverse transcriptase activity of hTERT may be a new strategy for tumor therapy.
Collapse
Affiliation(s)
- Yong Qin
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Hong Guo
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing 400037, P.R. China
| |
Collapse
|
15
|
Liu T, Ullenbruch M, Young Choi Y, Yu H, Ding L, Xaubet A, Pereda J, Feghali-Bostwick CA, Bitterman PB, Henke CA, Pardo A, Selman M, Phan SH. Telomerase and telomere length in pulmonary fibrosis. Am J Respir Cell Mol Biol 2013; 49:260-8. [PMID: 23526226 DOI: 10.1165/rcmb.2012-0514oc] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
In addition to its expression in stem cells and many cancers, telomerase activity is transiently induced in murine bleomycin (BLM)-induced pulmonary fibrosis with increased levels of telomerase transcriptase (TERT) expression, which is essential for fibrosis. To extend these observations to human chronic fibrotic lung disease, we investigated the expression of telomerase activity in lung fibroblasts from patients with interstitial lung diseases (ILDs), including idiopathic pulmonary fibrosis (IPF). The results showed that telomerase activity was induced in more than 66% of IPF lung fibroblast samples, in comparison with less than 29% from control samples, some of which were obtained from lung cancer resections. Less than 4% of the human IPF lung fibroblast samples exhibited shortened telomeres, whereas less than 6% of peripheral blood leukocyte samples from patients with IPF or hypersensitivity pneumonitis demonstrated shortened telomeres. Moreover, shortened telomeres in late-generation telomerase RNA component knockout mice did not exert a significant effect on BLM-induced pulmonary fibrosis. In contrast, TERT knockout mice exhibited deficient fibrosis that was independent of telomere length. Finally, TERT expression was up-regulated by a histone deacetylase inhibitor, while the induction of TERT in lung fibroblasts was associated with the binding of acetylated histone H3K9 to the TERT promoter region. These findings indicate that significant telomerase induction was evident in fibroblasts from fibrotic murine lungs and a majority of IPF lung samples, whereas telomere shortening was not a common finding in the human blood and lung fibroblast samples. Notably, the animal studies indicated that the pathogenesis of pulmonary fibrosis was independent of telomere length.
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109-2200, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Sui X, Kong N, Wang Z, Pan H. Epigenetic regulation of the human telomerase reverse transciptase gene: A potential therapeutic target for the treatment of leukemia (Review). Oncol Lett 2013; 6:317-322. [PMID: 24137323 PMCID: PMC3789043 DOI: 10.3892/ol.2013.1367] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Accepted: 05/17/2013] [Indexed: 12/21/2022] Open
Abstract
Telomerase activation is a critical step in human carcinogenesis through the maintenance of telomeres. Telomerase activity is primarily regulated by the human telomerase reverse transcriptase gene (hTERT), thus, an improved understanding of the transcriptional control of hTERT may provide potential therapeutic targets for the treatment of leukemia and other forms of cancer. Epigenetic modulation, a significant regulatory process in cell biology, has recently been shown to be involved in the regulation of the hTERT gene. Moreover, several epigenetic modifiers, including DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors, are now in pre- and early clinical trials of leukemia as monotherapies or in combination with other drugs, and have achieved significant clinical success. In the present review, the epigenetic mechanisms associated with telomerase activity in leukemia, and the therapeutic potential of an antitelomerase strategy that combines epigenetic modifiers with telomerase hTR subunit small molecule inhibitors are discussed.
Collapse
Affiliation(s)
- Xinbing Sui
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, P.R. China
| | | | | | | |
Collapse
|
17
|
hTERT: Another brick in the wall of cancer cells. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2013; 752:119-128. [DOI: 10.1016/j.mrrev.2012.12.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 11/28/2012] [Accepted: 12/10/2012] [Indexed: 01/06/2023]
|
18
|
The Intersection of Genetics and Epigenetics: Reactivation of Mammalian LINE-1 Retrotransposons by Environmental Injury. ENVIRONMENTAL EPIGENOMICS IN HEALTH AND DISEASE 2013. [DOI: 10.1007/978-3-642-23380-7_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
19
|
Jiang J, Zhao LJ, Zhao C, Zhang G, Zhao Y, Li JR, Li XP, Wei LH. Hypomethylated CpG around the transcription start site enables TERT expression and HPV16 E6 regulates TERT methylation in cervical cancer cells. Gynecol Oncol 2012; 124:534-541. [PMID: 22108635 DOI: 10.1016/j.ygyno.2011.11.023] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Revised: 11/06/2011] [Accepted: 11/14/2011] [Indexed: 01/08/2023]
Abstract
OBJECTIVE The human papillomavirus (HPV) oncoprotein, E6, activates telomerase reverse transcriptase (TERT) expression and causes cellular immortalization. It remains unclear whether E6 affects TERT transcription by altering DNA methylation profiles. In this study, we explored the methylation status of the TERT promoter in cervical cancer cell lines and its variations after E6 was silenced by RNAi. METHODS Three kinds of cervical cell lines (HPV16 positive: CaSki and SiHa; HPV18 positive: HeLa), were taken to analyze the methylation status of the TERT promoter by methylation-specific polymerase chain reaction (MSP) and bisulfite sequencing (BS). Stealth RNAi was transiently transfected to these cell lines to silence the expression of HPV16/18 E6, and the subsequent changes of TERT mRNA levels and TERT promoter DNA methylation were examined. RESULTS Hypomethylation of the DNA around the TERT transcription start site (-156 to +162 bp) was functionally related to its transcription. After transfection with Stealth RNAi, the levels of HPV16/18 E6 and TERT mRNA were greatly decreased. The methylated CpG around the transcription start sites in CaSki and SiHa cells were statistically increased (respectively P=0.016, P=0.000). However, there was no significant difference in HeLa cells (P=0.128). CONCLUSION Hypomethylated CpG around the transcription start site enables the expression of TERT in cervical cancer cells. Our results show for the first time that HPV16 E6 can promote TERT transcription through demethylating the DNA sequence around the TERT transcription start site in cervical squamous cancer cells.
Collapse
Affiliation(s)
- Jing Jiang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, China
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Regulation of the human catalytic subunit of telomerase (hTERT). Gene 2012; 498:135-46. [PMID: 22381618 DOI: 10.1016/j.gene.2012.01.095] [Citation(s) in RCA: 205] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 01/29/2012] [Accepted: 01/30/2012] [Indexed: 12/12/2022]
Abstract
Over the past decade, there has been much interest in the regulation of telomerase, the enzyme responsible for maintaining the integrity of chromosomal ends, and its crucial role in cellular immortalization, tumorigenesis, and the progression of cancer. Telomerase activity is characterized by the expression of the telomerase reverse transcriptase (TERT) gene, suggesting that TERT serves as the major limiting agent for telomerase activity. Recent discoveries have led to characterization of various interactants that aid in the regulation of human TERT (hTERT), including numerous transcription factors; further supporting the pivotal role that transcription plays in both the expression and repression of telomerase. Several studies have suggested that epigenetic modulation of the hTERT core promoter region may provide an additional level of regulation. Although these studies have provided essential information on the regulation of hTERT, there has been ambiguity of the role of methylation within the core promoter region and the subsequent binding of various activating and repressive agents. As a result, we found it necessary to consolidate and summarize these recent developments and elucidate these discrepancies. In this review, we focus on the co-regulation of hTERT via transcriptional regulation, the presence or absence of various activators and repressors, as well as the epigenetic pathways of DNA methylation and histone modifications.
Collapse
|
21
|
Gerhauser C. Cancer chemoprevention and nutriepigenetics: state of the art and future challenges. Top Curr Chem (Cham) 2012; 329:73-132. [PMID: 22955508 DOI: 10.1007/128_2012_360] [Citation(s) in RCA: 96] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The term "epigenetics" refers to modifications in gene expression caused by heritable, but potentially reversible, changes in DNA methylation and chromatin structure. Epigenetic alterations have been identified as promising new targets for cancer prevention strategies as they occur early during carcinogenesis and represent potentially initiating events for cancer development. Over the past few years, nutriepigenetics - the influence of dietary components on mechanisms influencing the epigenome - has emerged as an exciting new field in current epigenetic research. During carcinogenesis, major cellular functions and pathways, including drug metabolism, cell cycle regulation, potential to repair DNA damage or to induce apoptosis, response to inflammatory stimuli, cell signalling, and cell growth control and differentiation become deregulated. Recent evidence now indicates that epigenetic alterations contribute to these cellular defects, for example epigenetic silencing of detoxifying enzymes, tumor suppressor genes, cell cycle regulators, apoptosis-inducing and DNA repair genes, nuclear receptors, signal transducers and transcription factors by promoter methylation, and modifications of histones and non-histone proteins such as p53, NF-κB, and the chaperone HSP90 by acetylation or methylation.The present review will summarize the potential of natural chemopreventive agents to counteract these cancer-related epigenetic alterations by influencing the activity or expression of DNA methyltransferases and histone modifying enzymes. Chemopreventive agents that target the epigenome include micronutrients (folate, retinoic acid, and selenium compounds), butyrate, polyphenols from green tea, apples, coffee, black raspberries, and other dietary sources, genistein and soy isoflavones, curcumin, resveratrol, dihydrocoumarin, nordihydroguaiaretic acid (NDGA), lycopene, anacardic acid, garcinol, constituents of Allium species and cruciferous vegetables, including indol-3-carbinol (I3C), diindolylmethane (DIM), sulforaphane, phenylethyl isothiocyanate (PEITC), phenylhexyl isothiocyanate (PHI), diallyldisulfide (DADS) and its metabolite allyl mercaptan (AM), cambinol, and relatively unexplored modulators of histone lysine methylation (chaetocin, polyamine analogs). So far, data are still mainly derived from in vitro investigations, and results of animal models or human intervention studies are limited that demonstrate the functional relevance of epigenetic mechanisms for health promoting or cancer preventive efficacy of natural products. Also, most studies have focused on single candidate genes or mechanisms. With the emergence of novel technologies such as next-generation sequencing, future research has the potential to explore nutriepigenomics at a genome-wide level to understand better the importance of epigenetic mechanisms for gene regulation in cancer chemoprevention.
Collapse
Affiliation(s)
- Clarissa Gerhauser
- Division Epigenomics and Cancer Risk Factors, German Cancer Research Center, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
22
|
Maritz MF, Napier CE, Wen VW, MacKenzie KL. Targeting telomerase in hematologic malignancy. Future Oncol 2010; 6:769-89. [PMID: 20465390 DOI: 10.2217/fon.10.42] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Over the past two decades, it has become increasingly apparent that telomerase-mediated telomere maintenance plays a crucial role in hematopoiesis. Supporting evidence is underscored by recent findings of mutations in genes involved in telomerase-mediated telomere maintenance that contribute to the pathogenesis of bone marrow failure syndromes. More recently described telomere-independent functions of telomerase are also likely to contribute to both normal hematopoiesis and hematologic diseases. The high levels of telomerase detected in aggressive leukemias have fueled fervent investigation into diverse approaches to targeting telomerase in hematologic malignancies. Successful preclinical investigations that employed genetic strategies, oligonucleotides, small-molecule inhibitors and immunotherapy have resulted in a rapid translation to clinical trials. Further investigation of telomere-independent functions of telomerase and detailed preclinical studies of telomerase inhibition in both normal and malignant hematopoiesis will be invaluable for refining treatments to effectively and safely exploit telomerase as a therapeutic target in hematologic malignancies.
Collapse
Affiliation(s)
- Michelle F Maritz
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, New South Wales, Australia
| | | | | | | |
Collapse
|
23
|
Salvatico J, Kim JH, Chung IK, Muller MT. Differentiation linked regulation of telomerase activity by Makorin-1. Mol Cell Biochem 2010; 342:241-50. [PMID: 20473778 DOI: 10.1007/s11010-010-0490-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Accepted: 05/04/2010] [Indexed: 01/23/2023]
Abstract
To understand telomere homeostasis, a significant aspect of cancer and growth control, it is important to examine telomerase induction as well as mechanisms of regulated elimination. Makorin-1 (MKRN1) was previously shown to be an E3 ubiquitin ligase that targets the telomerase catalytic subunit (hTERT) for proteasome processing (Kim et al., Genes Dev 19:776-781, 2005). In this study we examined expression and regulation of endogenous MKRN1 during the cell cycle and terminal differentiation. When WI-38 cells transition from active growth into a resting G1 state, basal levels of MKRN1 were found to increase by sixfold. In contrast, cancer cells typically contained low or in some cases undetectable levels of MKRN1 protein. HL-60 cells growing exponentially in culture contain no detectable MKRN1; however, following terminal differentiation, MKRN1 mRNA and protein levels are strongly up-regulated while hTERT mRNA, hTERC, and telomerase are shut down. The initial decrease in telomerase activity is due to a gradual reduction in transcription of the hTERT gene that occurs during the first 12 h of terminal differentiation. MKRN1 protein appears between 12 and 24 h and is attended by a more rapid loss of telomerase activity. As more MKRN1 protein accumulates, significantly less telomerase activity is seen. Addition of the proteasome inhibitor, MG132, reverses the loss of telomerase activity; therefore, reductions in telomerase activity are dynamic, ongoing, and correlated with robust up-regulation of MKRN1 as the cells terminally differentiate. The data are consistent with the idea that MKRN1 represents a telomerase elimination pathway to rapidly draw down the activity during differentiation or cell cycle arrest when telomerase action at chromosome ends is no longer necessary.
Collapse
Affiliation(s)
- Jose Salvatico
- Department of Molecular and Microbiology, College of Medicine, University of Central Florida, Orlando, FL 32826, USA
| | | | | | | |
Collapse
|
24
|
Mathew R, Jia W, Sharma A, Zhao Y, Clarke LE, Cheng X, Wang H, Salli U, Vrana KE, Robertson GP, Zhu J, Wang S. Robust activation of the human but not mouse telomerase gene during the induction of pluripotency. FASEB J 2010; 24:2702-15. [PMID: 20354136 DOI: 10.1096/fj.09-148973] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pluripotent stem cells (PSCs) express telomerase and have unlimited proliferative potential. To study telomerase activation during reprogramming, 3 classes of embryonic stem cell (ESC)-like clones were isolated from mouse fibroblasts containing a transgenic hTERT reporter. Class I expressed few pluripotency markers, whereas class II contained many, but not Oct4, Nanog, and Sox2. Neither class of cells differentiated efficiently. Class III cells, the fully reprogrammed induced PSCs (iPSCs), expressed all pluripotency markers, formed teratomas indistinguishable from those of mESCs, and underwent efficient osteogenic differentiation in vitro. Interestingly, whereas the endogenous mTERT gene expression was only moderately increased during reprogramming, the hTERT promoter was strongly activated in class II cells and was further elevated in class III cells. Treatment of class II cells with chemical inhibitors of MEKs and glycogen synthase kinase 3 resulted in their further reprogramming into class III cells, accompanied by a strong activation of hTERT promoter. In reprogrammed human cells, the endogenous telomerase level, although variable among different clones, was dramatically elevated. Only in cells with the highest telomerase were telomeres restored to the lengths in hESCs. Our data, for the first time, demonstrated that the hTERT promoter was strongly activated in discrete steps, revealing a critical difference in human and mouse cell reprogramming. Because telomere elongation is crucial for self-renewal of hPSCs and replicative aging of their differentiated progeny, these findings have important implications in the generation and applications of iPSCs.
Collapse
Affiliation(s)
- Renjith Mathew
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Epigenetic plasticity of hTERT gene promoter determines retinoid capacity to repress telomerase in maturation-resistant acute promyelocytic leukemia cells. Leukemia 2010; 24:613-22. [PMID: 20072159 DOI: 10.1038/leu.2009.283] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The expression of hTERT gene, encoding the catalytic subunit of telomerase, is a feature of most cancer cells. Changes in the chromatin environment of its promoter and binding of transcriptional factors have been reported in differentiating cells when its transcription is repressed. However, it is not clear whether these changes are directly involved in this repression or only linked to differentiation. In a maturation-resistant acute promyelocytic leukemia (APL) cell line (NB4-LR1), we have previously identified a new pathway of retinoid-induced hTERT repression independent of differentiation. Using a variant of this cell line (NB4-LR1(SFD)), which resists to this repression, we show that although distinct patterns of histone modifications and transcription factor binding at the proximal domain of hTERT gene promoter could concur to modulate its expression, this region is not sufficient to the on/off switch of hTERT by retinoids. DNA methylation analysis of the hTERT promoter led to the identification of two distinct functional domains, a proximal one, fully unmethylated in both cell lines, and a distal one, significantly methylated in NB4-LR1(SFD) cells, whose methylation was further re-enforced by retinoid treatment. Interestingly, we showed that the binding to this distal domain of a known hTERT repressor, WT1, was defective only in NB4-LR1(SFD) cells. We propose that epigenetic modifications targeting this distal region could modulate the binding of hTERT repressors and account either for hTERT reactivation and resistance to retinoid-induced hTERT downregulation.
Collapse
|
26
|
Gigek CO, Leal MF, Silva PNO, Lisboa LCF, Lima EM, Calcagno DQ, Assumpção PP, Burbano RR, Smith MDAC. hTERTmethylation and expression in gastric cancer. Biomarkers 2009; 14:630-6. [DOI: 10.3109/13547500903225912] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
27
|
Li Y, Liu L, Andrews LG, Tollefsbol TO. Genistein depletes telomerase activity through cross-talk between genetic and epigenetic mechanisms. Int J Cancer 2009; 125:286-96. [PMID: 19358274 PMCID: PMC2995334 DOI: 10.1002/ijc.24398] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Genistein, a natural isoflavone found in soybean products, has been reported to down-regulate telomerase activity and that this prevents cancer and contributes to the apoptosis of cancer cells. However, the precise molecular mechanism by which genistein represses telomerase is not clear. Here, we show that genistein inhibits the transcription of hTERT (human telomerase reverse transcriptase), the catalytic subunit of the human telomerase enzyme, in breast MCF10AT benign cells and MCF-7 cancer cells in a time- and dose-dependent manner. Three major DNA methyltransferases (DNMT1, 3a and 3b) were also decreased in genistein-treated breast cancer cells suggesting that genistein may repress hTERT by impacting epigenetic pathways. Sequential depletion of the hTERT promoter revealed that the hTERT core promoter region is responsible for the genistein-induced repression of hTERT transcription. Using a newly developed technique of chromatin immunoprecipitation (ChIP)-related bifulfite sequencing analysis, we found an increased binding of E2F-1 to the hTERT promoter is due to the site-specific hypomethylation of the E2F-1 recognition site. In addition, we found that genistein can remodel chromatin structures of the hTERT promoter by increasing trimethyl-H3K9 but decreasing dimethyl-H3K4 in the hTERT promoter. The repression of hTERT was enhanced by combination with genistein and the DNMT inhibitor, 5-aza-2'-deoxycytidine (5-aza-dCyd). These findings collectively show that genistein is working, at least in part, through epigenetic mechanisms of telomerase inhibition in breast benign and cancer cells and may facilitate approaches to breast cancer prevention and treatment using an epigenetic modulator combined with genistein.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Liang Liu
- Center for Aging, University of Alabama at Birmingham, Birmingham, AL
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Lucy G. Andrews
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL
| | - Trygve O. Tollefsbol
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL
- Center for Aging, University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL
- Clinical Nutrition Research Center, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
28
|
Wang S, Zhao Y, Hu C, Zhu J. Differential repression of human and mouse TERT genes during cell differentiation. Nucleic Acids Res 2009; 37:2618-29. [PMID: 19270068 PMCID: PMC2677880 DOI: 10.1093/nar/gkp125] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Differential regulation of telomerase reverse transcriptase (TERT) contributes to the distinct aging and tumorigenic processes in humans and mice. Here, we report that the hTERT gene was strongly repressed during differentiation of human cells, whereas modest mTERT expression was detected in terminally differentiated and post-mitotic cells. The stringent hTERT repression depended on the native chromatin environment because transiently transfected hTERT promoters were not repressed in differentiated cells. Conversely, the transiently transfected mTERT core promoter was repressed during cell differentiation, suggesting that the repression of mTERT promoter did not require its endogenous chromatin structures. To understand the mechanisms of this differential regulation, we examined chromatin structures of the endogenous TERT loci during cell differentiation. In both human and mouse cells, repression was accompanied by the loss of multiple DNase I hypersensitive sites at the TERT promoters and their upstream regions, revealing positions of potential regulatory elements. Interestingly, the hTERT locus was located within a nuclease-resistant chromatin domain in human cells, whereas a corresponding chromatin domain was not detected for the mTERT locus. Taken together, our study indicated that, unlike the repression of mTERT gene, the condensed native chromatin environment of hTERT locus was central to its silencing during cell differentiation.
Collapse
Affiliation(s)
- Shuwen Wang
- Department of Cellular and Molecular Physiology, Pennsylvania State University, College of Medicine, Hershey, PA 17033, USA
| | | | | | | |
Collapse
|
29
|
Phipps SMO, Love WK, Mott TE, Andrews LG, Tollefsbol TO. Differential expression of epigenetic modulators during human embryonic stem cell differentiation. Mol Biotechnol 2009; 41:201-7. [PMID: 18953677 PMCID: PMC2629501 DOI: 10.1007/s12033-008-9118-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Accepted: 10/04/2008] [Indexed: 10/21/2022]
Abstract
Although the progression of aging and the diseases associated with it are extensively studied, little is known about the initiation of the aging process. Telomerase is down-regulated early in embryonic differentiation, thereby contributing to telomeric attrition and aging. The mechanisms underlying this inhibition remain elusive, but epigenetic studies in differentiating human embryonic stem (hES) cells could give clues about how and when DNA methylation and histone deacetylation work together to contribute to the inactivation of hTERT, the catalytic subunit of telomerase, at the onset of the aging process. We have confirmed the differentiation status of cultured hES colonies with morphological assessment and immunohistochemical stainings for pluripotent stem cells. In hES cells with varying degrees of differentiation, we have shown a stronger association between hES differentiation and expression of the epigenetic regulators DNMT3A and DNMT3B than between genetic modulators of differentiation such as c-MYC. We also propose a new model system for analyses of stem cell regions, which are differentially down-regulating the expression of hTERT and the actions of epigenetic modulators such as the DNMTs and histone methyltransferases.
Collapse
Affiliation(s)
- Sharla M. O. Phipps
- Department of Biology, University of Alabama at Birmingham, 175 Campbell Hall, 1300 University Boulevard, Birmingham, AL 35294-1170, USA
| | - William K. Love
- Department of Biology, University of Alabama at Birmingham, 175 Campbell Hall, 1300 University Boulevard, Birmingham, AL 35294-1170, USA
| | - Troy E. Mott
- Department of Biology, University of Alabama at Birmingham, 175 Campbell Hall, 1300 University Boulevard, Birmingham, AL 35294-1170, USA
| | - Lucy G. Andrews
- Department of Biology, University of Alabama at Birmingham, 175 Campbell Hall, 1300 University Boulevard, Birmingham, AL 35294-1170, USA
| | - Trygve O. Tollefsbol
- Department of Biology, University of Alabama at Birmingham, 175 Campbell Hall, 1300 University Boulevard, Birmingham, AL 35294-1170, USA
- Center for Aging, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Clinical Nutrition Research Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
30
|
Krunic D, Moshir S, Greulich-Bode KM, Figueroa R, Cerezo A, Stammer H, Stark HJ, Gray SG, Nielsen KV, Hartschuh W, Boukamp P. Tissue context-activated telomerase in human epidermis correlates with little age-dependent telomere loss. Biochim Biophys Acta Mol Basis Dis 2009; 1792:297-308. [PMID: 19419690 DOI: 10.1016/j.bbadis.2009.02.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2008] [Revised: 02/06/2009] [Accepted: 02/06/2009] [Indexed: 10/21/2022]
Abstract
Telomerase- and telomere length regulation in normal human tissues is still poorly understood. We show here that telomerase is expressed in the epidermis in situ independent of age but was repressed upon the passaging of keratinocytes in monolayer culture. However, when keratinocytes were grown in organotypic cultures (OTCs), telomerase was re-established, indicating that telomerase activity is not merely proliferation-associated but is regulated in a tissue context-dependent manner in human keratinocytes. While not inducible by growth factors, treatment with the histone deacetylation inhibitor FK228 restored telomerase activity in keratinocytes grown in monolayer cultures. Accordingly, CHIP analyses demonstrated an acetylated, active hTERT promoter in the epidermis in situ and in the epidermis of OTCs but a deacetylated, silenced hTERT promoter with subsequent propagation in monolayer culture suggesting that histone acetylation is part of the regulatory program to guarantee hTERT expression/telomerase activity in the epidermis. In agreement with the loss of telomerase activity, telomeres shortened during continuous propagation in monolayer culture by an average of approximately 70 base pairs (bp) per population doubling (pd). However, telomere erosion varied strongly between different keratinocyte strains and even between individual cells within the same culture, thereby arguing against a defined rate of telomere loss per replication cycle. In the epidermis in situ, as determined from early-passage keratinocytes and tissue sections from different age donors, we calculated a telomere loss of only approximately 25 bp per year. Since we determined the same rate for the non-regenerating melanocytes and dermal fibroblasts, our data suggest that in human epidermis telomerase is a protective mechanism against excessive telomere loss during the life-long regeneration.
Collapse
Affiliation(s)
- Damir Krunic
- Division of Genetics of Skin Carcinogenesis, German Cancer Research Center, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Kyo S, Takakura M, Fujiwara T, Inoue M. Understanding and exploiting hTERT promoter regulation for diagnosis and treatment of human cancers. Cancer Sci 2008; 99:1528-38. [PMID: 18754863 PMCID: PMC11158053 DOI: 10.1111/j.1349-7006.2008.00878.x] [Citation(s) in RCA: 271] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Telomerase activation is a critical step for human carcinogenesis through the maintenance of telomeres, but the activation mechanism during carcinogenesis remains unclear. Transcriptional regulation of the human telomerase reverse transcriptase (hTERT) gene is the major mechanism for cancer-specific activation of telomerase, and a number of factors have been identified to directly or indirectly regulate the hTERT promoter, including cellular transcriptional activators (c-Myc, Sp1, HIF-1, AP2, ER, Ets, etc.) as well as the repressors, most of which comprise tumor suppressor gene products, such as p53, WT1, and Menin. Nevertheless, none of them can clearly account for the cancer specificity of hTERT expression. The chromatin structure via the DNA methylation or modulation of nucleosome histones has recently been suggested to be important for regulation of the hTERT promoter. DNA unmethylation or histone methylation around the transcription start site of the hTERT promoter triggers the recruitment of histone acetyltransferase (HAT) activity, allowing hTERT transcription. These facts prompted us to apply these regulatory mechanisms to cancer diagnostics and therapeutics. Telomerase-specific replicative adenovirus (Telomelysin, OBP-301), in which E1A and E1B genes are driven by the hTERT promoter, has been developed as an oncolytic virus that replicates specifically in cancer cells and causes cell death via viral toxicity. Direct administration of Telomelysin was proved to effectively eradicate solid tumors in vivo, without apparent adverse effects. Clinical trials using Telomelysin for cancer patients with progressive stages are currently ongoing. Furthermore, we incorporated green fluorescent protein gene (GFP) into Telomelysin (TelomeScan, OBP-401). Administration of TelomeScan into the primary tumor enabled the visualization of cancer cells under the cooled charged-coupled device (CCD) camera, not only in primary tumors but also the metastatic foci. This technology can be applied to intraoperative imaging of metastatic lymphnodes. Thus, we found novel tools for cancer diagnostics and therapeutics by utilizing the hTERT promoter.
Collapse
Affiliation(s)
- Satoru Kyo
- Department of Obstetrics and Gynecology, Kanazawa University, Graduate School of Medical Science, 13-1 Takaramachi, Kanazawa, Ishikawa 920-8641, Japan.
| | | | | | | |
Collapse
|
32
|
Genome-wide identification of aberrantly methylated promoter associated CpG islands in acute lymphocytic leukemia. Leukemia 2008; 22:1529-38. [PMID: 18528427 DOI: 10.1038/leu.2008.130] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We performed a genome-wide analysis of promoter associated CpG island methylation using methylated CpG island amplification (MCA) coupled to representational differential analysis (RDA) or a DNA promoter microarray in acute lymphoblastic leukemia (ALL). We identified 65 potential targets of methylation with the MCA/RDA approach, and 404 with the MCA/array. Thirty-six (77%) of the genes identified by MCA/RDA were shared by the MCA/array approach. Chromosomal location of these genes was evenly distributed in all autosomes. Functionally, 303 of these genes clustered in 18 molecular pathways. Of the 36 shared genes, 31 were validated and 26 were confirmed as being hypermethylated in leukemia cell lines. Expression analysis of eight of these genes was epigenetically modulated by hypomethylating agents and/or HDAC inhibitors in leukemia cell lines. Subsequently, DNA methylation of 15 of these genes (GIPC2, RSPO1, MAGI1, CAST1, ADCY5, HSPA4L, OCLN, EFNA5, MSX2, GFPT2, GNA14, SALL1, MYO5B, ZNF382 and MN1) was validated in primary ALL samples. Patients with methylation of multiple CpG islands had a worse overall survival. This is the largest published list of potential methylation target genes in human leukemia offering the possibility of performing rational unbiased methylation studies in ALL.
Collapse
|
33
|
Berletch JB, Liu C, Love WK, Andrews LG, Katiyar SK, Tollefsbol TO. Epigenetic and genetic mechanisms contribute to telomerase inhibition by EGCG. J Cell Biochem 2008; 103:509-19. [PMID: 17570133 PMCID: PMC2435482 DOI: 10.1002/jcb.21417] [Citation(s) in RCA: 150] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The ends of human chromosomes are protected from the degradation associated with cell division by 15-20 kb long segments of hexameric repeats of 5'-TTAGGG-3' termed telomeres. In normal cells telomeres lose up to 300 bp of DNA per cell division that ultimately leads to senescence; however, most cancer cells bypass this lifespan restriction through the expression of telomerase. hTERT, the catalytic subunit essential for the proper function of telomerase, has been shown to be expressed in approximately 90% of all cancers. In this study we investigated the hTERT inhibiting effects of (-)-epigallocatechin-3-gallate (EGCG), the major polyphenol found in green tea catechins, in MCF-7 breast cancers cells and HL60 promyelocytic leukemia cells. Exposure to EGCG reduced cellular proliferation and induced apoptosis in both MCF-7 and HL60 cells in vitro, although hTERT mRNA expression was decreased only in MCF-7 cells when treated with EGCG. Furthermore, down-regulation of hTERT gene expression in MCF-7 cells appeared to be largely due to epigenetic alterations. Treatment of MCF-7 cells with EGCG resulted in a time-dependent decrease in hTERT promoter methylation and ablated histone H3 Lys9 acetylation. In conjunction with demethylation, further analysis showed an increase in hTERT repressor E2F-1 binding at the promoter. From these findings, we propose that EGCG is effective in causing cell death in both MCF-7 and HL60 cancer cell lines and may work through different pathways involving both anti-oxidant effects and epigenetic modulation.
Collapse
Affiliation(s)
- Joel B Berletch
- Department of Biology, University of Alabama Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | |
Collapse
|
34
|
Liu C, Fang X, Ge Z, Jalink M, Kyo S, Björkholm M, Gruber A, Sjöberg J, Xu D. The telomerase reverse transcriptase (hTERT) gene is a direct target of the histone methyltransferase SMYD3. Cancer Res 2007; 67:2626-31. [PMID: 17363582 DOI: 10.1158/0008-5472.can-06-4126] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Recent evidence has accumulated that the dynamic histone methylation mediated by histone methyltransferases and demethylases plays key roles in regulation of chromatin structure and transcription. In the present study, we show that SET and MYND domain-containing protein 3 (SMYD3), a histone methyltransferase implicated in oncogenesis, directly trans-activates the telomerase reverse transcriptase (hTERT) gene that is essential for cellular immortalization and transformation. SMYD3 occupies its binding motifs on the hTERT promoter and is required for maintenance of histone H3-K4 trimethylation, thereby contributing to inducible and constitutive hTERT expression in normal and malignant human cells. Knocking down SMYD3 in tumor cells abolished trimethylation of H3-K4, attenuated the occupancy by the trans-activators c-MYC and Sp1, and led to diminished histone H3 acetylation in the hTERT promoter region, which was coupled with down-regulation of hTERT mRNA and telomerase activity. These results suggest that SMYD3-mediated trimethylation of H3-K4 functions as a licensing element for subsequent transcription factor binding to the hTERT promoter. The present findings provide significant insights into regulatory mechanisms of hTERT/telomerase expression; moreover, identification of the hTERT gene as a direct target of SMYD3 contributes to a better understanding of SMYD3-mediated cellular transformation.
Collapse
Affiliation(s)
- Cheng Liu
- Department of Medicine, Division of Hematology, Karolinska University Hospital Solna, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Zinn RL, Pruitt K, Eguchi S, Baylin SB, Herman JG. hTERT is expressed in cancer cell lines despite promoter DNA methylation by preservation of unmethylated DNA and active chromatin around the transcription start site. Cancer Res 2007; 67:194-201. [PMID: 17210699 DOI: 10.1158/0008-5472.can-06-3396] [Citation(s) in RCA: 158] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
hTERT, which encodes the catalytic subunit of telomerase and is expressed in most immortalized and cancer cells, has been reported to have increased DNA methylation in its promoter region in many cancers. This pattern is inconsistent with observations that DNA methylation of promoter CpG islands is typically associated with gene silencing. Here, we provide a comprehensive analysis of promoter DNA methylation, chromatin patterns, and expression of hTERT in cancer and immortalized cells. Methylation-specific PCR and bisulfite sequencing of the hTERT promoter in breast, lung, and colon cancer cells show that all cancer cell lines retain alleles with little or no methylation around the transcription start site despite being densely methylated in a region 600 bp upstream of the transcription start site. By real-time reverse transcription-PCR, all cancer cell lines express hTERT. Chromatin immunoprecipitation (ChIP) analysis reveals that both active (acetyl-H3K9 and dimethyl-H3K4) and inactive (trimethyl-H3K9 and trimethyl-H3K27) chromatin marks are present across the hTERT promoter. However, using a novel approach combining methylation analysis of ChIP DNA, we show that active chromatin marks are associated with unmethylated DNA, whereas inactive marks of chromatin are associated with methylated DNA in the region around the transcription start site. These results show that DNA methylation patterns of the hTERT promoter (-150 to +150 around the transcription start) are consistent with the usual dynamics of gene expression in that the absence of methylation in this region and the association with active chromatin marks allow for the continued expression of hTERT.
Collapse
Affiliation(s)
- Rebekah L Zinn
- Sidney Kimmel Comprehensive Cancer Center and Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | | | | |
Collapse
|
36
|
Wang S, Hu C, Zhu J. Transcriptional silencing of a novel hTERT reporter locus during in vitro differentiation of mouse embryonic stem cells. Mol Biol Cell 2006; 18:669-77. [PMID: 17151355 PMCID: PMC1783791 DOI: 10.1091/mbc.e06-09-0840] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The human telomerase reverse transcriptase hTERT is highly expressed in undifferentiated embryonic cells and silenced in the majority of somatic cells. To investigate the mechanisms of hTERT silencing, we have developed a novel reporter using a bacterial artificial chromosome (BAC) that contained the entire hTERT gene and its neighboring loci, hCRR9 and hXtrp2. Firefly and Renilla luciferases were used to monitor transcription from the hTERT and hCRR9 promoters, respectively. In mouse embryonic stem cells stably integrated with the BAC reporter, both hTERT and hCRR9 promoters were highly expressed. Upon differentiation into embryoid bodies and further into mineral-producing osteogenic cells, the hTERT promoter activity decreased progressively, whereas the hCRR9 promoter remained highly active, both resembling their endogenous counterparts. In fully differentiated cells, the hTERT promoter was completely silenced and adopted a chromatin structure that was similar to its native counterpart in human cells. Inhibition of histone deacetylases led to the opening of the hTERT promoter and partially relieved repression, suggesting that histone deacetylation was necessary but not sufficient for hTERT silencing. Thus, our result demonstrated that developmental silencing of the human TERT locus could be recapitulated in a chromosomal position-independent manner during the differentiation of mouse embryonic stem cells.
Collapse
MESH Headings
- Acetylation
- Animals
- Cell Differentiation/genetics
- Cells, Cultured
- Chromosomes, Artificial, Bacterial/genetics
- Embryonic Stem Cells/cytology
- Embryonic Stem Cells/enzymology
- Gene Expression Regulation, Developmental
- Gene Silencing
- Genes, Reporter
- Histones/metabolism
- Humans
- Mice
- Mice, Transgenic
- Models, Biological
- Promoter Regions, Genetic
- Receptors, CCR
- Receptors, Chemokine/genetics
- Telomerase/genetics
- Transcription, Genetic
- Transgenes
Collapse
Affiliation(s)
- Shuwen Wang
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Chunguang Hu
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Jiyue Zhu
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| |
Collapse
|
37
|
Abstract
Ageing is often defined in the context of telomerase activity and telomere length regulation. Most somatic cells have limited replication ability and undergo senescence eventually. Stem cells are unique as they possess more abundant telomerase activity and are able to maintain telomere lengths for a longer period. Embryonic stem cells are particularly resistant to ageing and can be propagated indefinitely. Remarkably, adult somatic cells can be reprogrammed to an ESC-like state by various means including cell fusion, exposure to ESC cell-free extracts, enforced expression of specific molecules, and somatic cell nuclear transfer. Thus, the rejuvenation of an 'aged' state can be effected by the activation of specific key molecules in the cell. Here, we argue that cellular ageing is a reversible process, and this is determined by the balance of biological molecules which directly or indirectly control telomere length and telomerase activity, either through altering gene expression and/or modulating the epigenetic state of the chromatin.
Collapse
Affiliation(s)
- Wai-Leong Tam
- Stem Cell & Developmental Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | | | | |
Collapse
|
38
|
Andollo N, Boyano MD, Andrade R, Aréchaga JM. Epigenetic regulation of the imprinted U2af1-rs1 gene during retinoic acid-induced differentiation of embryonic stem cells. Dev Growth Differ 2006; 48:349-60. [PMID: 16872448 DOI: 10.1111/j.1440-169x.2006.00873.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Epigenetic modifications such as DNA methylation and changes in chromatin structure are changes in the chemical composition or structure of DNA that work by regulating gene expression. Their mechanisms of action have been generally studied in imprinted genes. The present work analyzes the involvement of these mechanisms in the expression of the U2af1-rs1 imprinted gene during the differentiation process of embryonic stem (ES) cells induced by retinoic acid. By DNA digestion with methylation-dependent or independent restriction enzymes and consecutive Southern blot, we have found that methylation of the U2af1-rs1 gene increases in differentiated ES cells and in embryoid bodies. However, northern blot and real-time reverse transcription-polymerase chain reaction analysis showed a higher expression of the U2af1-rs1 gene in differentiated ES cells and in embryoid bodies than in undifferentiated ones. On the other hand, the sensitivity to DNase-I assay demonstrated an open chromatin conformation for differentiated cells with regard to undifferentiated ES cells. Our results suggest that the expression of the U2af1-rs1 gene would be regulated by changes in chromatin structure rather than by DNA methylation during the RA-induced process of differentiation of ES cells.
Collapse
Affiliation(s)
- Noelia Andollo
- Department of Cell Biology and Histology, Faculty of Medicine and Dentistry, University of the Basque Country, Leioa, Vizcaya, Spain
| | | | | | | |
Collapse
|
39
|
Akiyama M, Yamada O, Yanagisawa T, Fujisawa K, Eto Y, Yamada H. Analysis of telomerase activity and RNA expression in a patient with acute promyelocytic leukemia treated with all-trans retinoic acid. Pediatr Blood Cancer 2006; 46:506-11. [PMID: 15770638 DOI: 10.1002/pbc.20392] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In this study, we show that all-trans retinoic acid (ATRA) treatment leads to a rapid decrease in telomerase activity, which was associated with the reduction in myeloblasts and occurs before the appearance of myelocytes, in a patient with acute promyelocytic leukemia (APL). Microarray analysis by ATRA treatment for 48 hr in peripheral blood mononuclear cells (in vivo) and in cultured bone marrow mononuclear cells (in vitro) from a patient with APL revealed upregulation of CD11b, CD11c, CCAAT enhancer binding protein epsilon, Rb1, Mad, and tumor necrosis factor-related genes; and downregulation of hTERT, c-Myc, WT1, bcl-2, and eukaryotic translation elongation factor 1alpha2. The results might offer the potential to define the molecular mechanism underlying ATRA-induced granulocytic differentiation in patients with APL, and provide clues to identify novel molecular therapeutic targets.
Collapse
MESH Headings
- Antigens, Differentiation/drug effects
- Antigens, Differentiation/genetics
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Child
- Disseminated Intravascular Coagulation/drug therapy
- Enzyme Activation/drug effects
- Female
- Gene Expression Profiling
- Gene Expression Regulation, Leukemic/drug effects
- Gene Expression Regulation, Leukemic/genetics
- Granulocytes/drug effects
- Humans
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/enzymology
- Leukemia, Promyelocytic, Acute/genetics
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/enzymology
- Oligonucleotide Array Sequence Analysis
- RNA/genetics
- Remission Induction
- Reverse Transcriptase Polymerase Chain Reaction
- Telomerase/drug effects
- Telomerase/metabolism
- Tretinoin/pharmacology
- Tretinoin/therapeutic use
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Masaharu Akiyama
- Department of Pediatrics, Jikei University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
40
|
Ge Z, Liu C, Björkholm M, Gruber A, Xu D. Mitogen-activated protein kinase cascade-mediated histone H3 phosphorylation is critical for telomerase reverse transcriptase expression/telomerase activation induced by proliferation. Mol Cell Biol 2006; 26:230-7. [PMID: 16354694 PMCID: PMC1317632 DOI: 10.1128/mcb.26.1.230-237.2006] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Telomerase activity and telomerase reverse transcriptase (hTERT), the key component of the telomerase complex, are tightly proliferation regulated in normal and malignant cells both in vitro and in vivo; however, underlying mechanisms are unclear. In the present study, we identified mitogen-activated protein kinase (MAPK) cascade-mediated histone H3 ser10 phosphorylation to be a molecular link between proliferation and induction of hTERT/telomerase activity. In normal human T lymphocytes and fibroblasts, growth or stress stimuli known to drive H3 phosphorylation through the MAPK signaling induce hTERT expression and/or telomerase activity that was preceded by phosphorylated histone H3 (ser10) at the hTERT promoter. Blockade of the MAPK-triggered H3 phosphorylation significantly abrogates hTERT induction and ser10 phosphorylation at this promoter. However, H3 ser10 phosphorylation alone resulted in low, transient hTERT induction, as seen in fibroblasts, whereas H3 phosphorylation followed by its acetylation at lys14 robustly trans-activated the hTERT gene accompanying constitutive telomerase activity in normal and malignant T cells. H3 acetylation without phosphorylation similarly exerted weak effects on hTERT expression. These results define H3 phosphorylation as a key to hTERT transactivation induced by proliferation and reveal a fundamental mechanism for telomerase regulation in both normal human cells and transformed T cells.
Collapse
Affiliation(s)
- Zheng Ge
- Division of Hematology, Deaprtment of Medicine, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | | | | | | | | |
Collapse
|
41
|
Bibikova M, Lin Z, Zhou L, Chudin E, Garcia EW, Wu B, Doucet D, Thomas NJ, Wang Y, Vollmer E, Goldmann T, Seifart C, Jiang W, Barker DL, Chee MS, Floros J, Fan JB. High-throughput DNA methylation profiling using universal bead arrays. Genes Dev 2006; 16:383-93. [PMID: 16449502 PMCID: PMC1415217 DOI: 10.1101/gr.4410706] [Citation(s) in RCA: 489] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2005] [Accepted: 11/01/2005] [Indexed: 12/13/2022]
Abstract
We have developed a high-throughput method for analyzing the methylation status of hundreds of preselected genes simultaneously and have applied it to the discovery of methylation signatures that distinguish normal from cancer tissue samples. Through an adaptation of the GoldenGate genotyping assay implemented on a BeadArray platform, the methylation state of 1536 specific CpG sites in 371 genes (one to nine CpG sites per gene) was measured in a single reaction by multiplexed genotyping of 200 ng of bisulfite-treated genomic DNA. The assay was used to obtain a quantitative measure of the methylation level at each CpG site. After validating the assay in cell lines and normal tissues, we analyzed a panel of lung cancer biopsy samples (N = 22) and identified a panel of methylation markers that distinguished lung adenocarcinomas from normal lung tissues with high specificity. These markers were validated in a second sample set (N = 24). These results demonstrate the effectiveness of the method for reliably profiling many CpG sites in parallel for the discovery of informative methylation markers. The technology should prove useful for DNA methylation analyses in large populations, with potential application to the classification and diagnosis of a broad range of cancers and other diseases.
Collapse
|
42
|
Abstract
Telomeres are complex structures which serve to protect chromosome ends. Telomere shortening occurs in normal somatic cells reaching a point in which cells senesce. Senescence can be counteracted by activating telomerase. Telomerase activity is present in a majority of cancer cells and requires the upregulation of the reverse transcriptase component called hTERT. Because telomerase activity is essential for proliferation of most cancer cells, therapeutic strategies have been developed to inhibit its activity. These strategies centre on targeting the active site, hTERT and hTERC expression, core enzyme stability and telomeric DNA. Successful approaches involve a combination of traditional drugs with telomerase inhibitors. Disrupting the functional expression of hTERT is particularly effective in agreement with evidence that hTERT is an antiapoptotic factor in some cancer cells. In addition, approaches that stabilise DNA secondary structures may disrupt telomere maintenance through a variety of routes making them, potentially, very potent in attack-ing cancer cells.
Collapse
Affiliation(s)
- Terace M Fletcher
- Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, 1011 NW 15 Street, Miami, FL 33136, USA.
| |
Collapse
|
43
|
Liu L, Lai S, Andrews LG, Tollefsbol TO. Genetic and epigenetic modulation of telomerase activity in development and disease. Gene 2004; 340:1-10. [PMID: 15556289 DOI: 10.1016/j.gene.2004.06.011] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2004] [Revised: 05/18/2004] [Accepted: 06/03/2004] [Indexed: 11/22/2022]
Abstract
Telomerase activity is one of the most important factors that have been linked to multiple developmental processes, including cell proliferation, differentiation, aging and senescence. Dysregulation of telomerase has often been found in developmental abnormalities, such as cancer, loss of function in the hematopoietic system, and low success rate of somatic cloning. A comprehensive network of transcription factors has been shown to be involved in the genetic control of telomerase expression and activity. Epigenetic mechanisms have recently been shown to provide an additional level of regulation, and may be responsible for the diverse expression status of telomerase that is manifested in a tissue and cell-type-dependent manner. This article summarizes the recent developments in the field of telomerase research with a focus on the coregulation of the telomerase gene by both genetic and epigenetic pathways. Developmental consequences of aberrant telomerase activity will also be summarized and discussed.
Collapse
Affiliation(s)
- Liang Liu
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL 35294-1170, USA
| | | | | | | |
Collapse
|
44
|
Liu L, Berletch JB, Green JG, Pate MS, Andrews LG, Tollefsbol TO. Telomerase inhibition by retinoids precedes cytodifferentiation of leukemia cells and may contribute to terminal differentiation. Mol Cancer Ther 2004. [DOI: 10.1158/1535-7163.1003.3.8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Human promyelocytic leukemia HL60 cells display high telomerase activity, a phenotype related to their immortal status. All-trans retinoic acid (ATRA) is a clinically effective cytodifferentiating agent. To understand the mechanism underlying ATRA-induced cytodifferentiation, we did a kinetic analysis of the role of ATRA in inhibiting telomerase in HL60 cells. Our studies indicate that telomerase inhibition by ATRA occurred relatively early after treatment of HL60 cells due to a rapid decrease in hTERT gene expression. More importantly, however, we found through monitoring the expression of CD11b, a marker for granulocytic differentiation of HL60 cells, that down-regulation of telomerase preceded the differentiation of HL60 cells. These observations suggest that the hTERT gene may be a primary target of ATRA regulation of cellular differentiation and the antileukemia activity of ATRA may be mediated by its ability to induce the differentiation of the promyelocytic leukemia cells through down-regulation of the hTERT gene.
Collapse
Affiliation(s)
| | | | | | | | | | - Trygve O. Tollefsbol
- 1Department of Biology,
- 2Center for Aging, and
- 3Comprehensive Cancer Center, University of Alabama-Birmingham, Birmingham, Alabama
| |
Collapse
|