1
|
He X, Cao L, Fu X, Wu Y, Wen H, Gao Y, Huo W, Wang M, Liu M, Su Y, Liu G, Zhang M, Hu F, Hu D, Zhao Y. The Association Between Telomere Length and Diabetes Mellitus: Accumulated Evidence From Observational Studies. J Clin Endocrinol Metab 2024; 110:e177-e185. [PMID: 39087945 DOI: 10.1210/clinem/dgae536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 07/11/2024] [Accepted: 07/31/2024] [Indexed: 08/02/2024]
Abstract
OBJECTIVE In order to assess the associations between telomere length (TL) and diabetes mellitus (DM), especially type 2 diabetes (T2DM), we performed this systematic review and meta-analysis. METHODS PubMed, Embase, and Web of Science were thoroughly searched up to July 11, 2023. The pooled standardized mean difference (SMD) and the 95% confidence interval (CI) were evaluated using the random-effects model. Age, sex, study design, duration of diabetes, region, sample size, and body mass index (BMI) were used to stratify subgroup analyses. RESULTS A total of 37 observational studies involving 18 181 participants from 14 countries were included in the quantitative meta-analysis. In this study, patients with diabetes had shorter TL than the non-diabetic, whether those patients had T1DM (-2.70; 95% CI: -4.47, -0.93; P < .001), T2DM (-3.70; 95% CI: -4.20, -3.20; P < .001), or other types of diabetes (-0.71; 95% CI: -1.10, -0.31; P < .001). Additionally, subgroup analysis of T2DM showed that TL was significantly correlated with age, sex, study design, diabetes duration, sample size, detection method, region, and BMI. CONCLUSION A negative correlation was observed between TL and DM. To validate this association in the interim, more extensive, superior prospective investigations and clinical trials are required.
Collapse
Affiliation(s)
- Xinxin He
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Lu Cao
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Xueru Fu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Yuying Wu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Hongwei Wen
- Department of Public Health, Zhengzhou Shuqing Medical College, Zhengzhou, Henan 450000, People's Republic of China
| | - Yajuan Gao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Weifeng Huo
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Mengdi Wang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Mengna Liu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Yijia Su
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Ge Liu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Ming Zhang
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong 518060, People's Republic of China
| | - Fulan Hu
- Department of Biostatistics and Epidemiology, School of Public Health, Shenzhen University Medical School, Shenzhen, Guangdong 518060, People's Republic of China
| | - Dongsheng Hu
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| | - Yang Zhao
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, People's Republic of China
| |
Collapse
|
2
|
Murugan AK, Kannan S, Alzahrani AS. TERT promoter mutations in gliomas: Molecular roles in tumorigenesis, metastasis, diagnosis, prognosis, therapeutic targeting, and drug resistance. Biochim Biophys Acta Rev Cancer 2024; 1880:189243. [PMID: 39674418 DOI: 10.1016/j.bbcan.2024.189243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Telomerase reverse transcriptase (TERT), a critical player in cellular immortalization, has emerged as a focal point of investigation due to its frequent promoter mutations in various human malignancies. TERT promoter mutations exhibit a significant role in tumorigenesis, fostering unbridled cellular proliferation and survival. This comprehensive review delves into the landscape of TERT promoter mutations and their profound implications in cancer, particularly within the context of gliomas. This article meticulously examines the intricate interplay between TERT promoter mutations and the metastatic cascade, shedding light on their capacity to orchestrate invasive behavior in gliomas. Moreover, this review describes the recent trends in therapeutic targeting of the TERT and dissects the evolving landscape of drug resistance associated with TERT mutations, providing insights into potential therapeutic challenges. In addition, the diagnostic and prognostic implications of TERT promoter mutations in gliomas are scrutinized, unraveling their potential as robust biomarkers. It also discusses the recent advancements in molecular diagnostics, illustrating the promise of TERT mutations as diagnostic tools and prognostic indicators. This review collectively aims to contribute to a deeper understanding of TERT promoter mutations in gliomas, offering a foundation for future research endeavors and paving the way for innovative strategies in glioma management.
Collapse
Affiliation(s)
- Avaniyapuram Kannan Murugan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia.
| | - Siddarth Kannan
- School of Medicine, University of Central Lancashire, Preston PR1 2HE, UK
| | - Ali S Alzahrani
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia; Department of Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
3
|
Sadr Z, Ghasemi M, Jafarpour S, Seyfi R, Ghasemi A, Boustanipour E, Khorshid HRK, Ehtesham N. Beginning at the ends: telomere and telomere-based cancer therapeutics. Mol Genet Genomics 2024; 300:1. [PMID: 39638969 DOI: 10.1007/s00438-024-02206-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024]
Abstract
Telomeres, which are situated at the terminal ends of chromosomes, undergo a reduction in length with each cellular division, ultimately reaching a critical threshold that triggers cellular senescence. Cancer cells circumvent this senescence by utilizing telomere maintenance mechanisms (TMMs) that grant them a form of immortality. These mechanisms can be categorized into two primary processes: the reactivation of telomerase reverse transcriptase and the alternative lengthening of telomeres (ALT) pathway, which is dependent on homologous recombination (HR). Various strategies have been developed to inhibit telomerase activation in 85-95% of cancers, including the use of antisense oligonucleotides such as small interfering RNAs and endogenous microRNAs, agents that simulate telomere uncapping, expression modulators, immunotherapeutic vaccines targeting telomerase, reverse transcriptase inhibitors, stabilization of G-quadruplex structures, and gene therapy approaches. Conversely, in the remaining 5-15% of human cancers that rely on ALT, mechanisms involve modifications in the chromatin environment surrounding telomeres, upregulation of TERRA long non-coding RNA, enhanced activation of the ataxia telangiectasia and Rad-3-related protein kinase signaling pathway, increased interactions with nuclear receptors, telomere repositioning driven by HR, and recombination events between non-sister chromatids, all of which present potential targets for therapeutic intervention. Additionally, combinatorial therapy has emerged as a strategy that employs selective agents to simultaneously target both telomerase and ALT, aiming for optimal clinical outcomes. Given the critical role of anti-TMM strategies in cancer treatment, this review provides an overview of the latest insights into the structure and function of telomeres, their involvement in tumorigenesis, and the advancements in TMM-based cancer therapies.
Collapse
Affiliation(s)
- Zahra Sadr
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoumeh Ghasemi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Soheyla Jafarpour
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reyhaneh Seyfi
- Department of Stem Cells Technology and Tissue Regeneration, Faculty of Interdisciplinary Science and Technologies, Tarbiat Modares University, Tehran, Iran
| | - Aida Ghasemi
- Neuromuscular Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Boustanipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Naeim Ehtesham
- Department of Medical Genetics, School of Medicine, Iranshahr University of Medical Sciences, Iranshahr, Iran.
| |
Collapse
|
4
|
Murillo-Ortiz BO, García-Corrales K, Martínez-Garza S, Romero-Vázquez MJ, Agustín-Godínez E, Escareño-Gómez A, Silva-Guerrero DG, Mendoza-Ramírez S, Murguia-Perez M. Association of hTERT expression, Her2Neu, estrogen receptors, progesterone receptors, with telomere length before and at the end of treatment in breast cancer patients. Front Med (Lausanne) 2024; 11:1450147. [PMID: 39188883 PMCID: PMC11345256 DOI: 10.3389/fmed.2024.1450147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 07/29/2024] [Indexed: 08/28/2024] Open
Abstract
Background Breast cancer shows significant clinical, morphologic, and molecular variation. Telomeres are nucleoprotein complexes composed of hexanucleotide repeat DNA sequence, TTAGGG, and numerous telomere-associated proteins. The maintenance of telomere length is carried out by a ribonucleoprotein called telomerase, which consists of two main components: a catalytic subunit called hTERT (human telomerase reverse transcriptase) and an RNA template called hTR (human telomerase RNA). The importance of evaluating hTERT expression lies in its potential therapeutic application, being an attractive target due to its almost non-existent expression in normal somatic cells. It is also expected that the anti-neoplastic effect would appear earlier in neoplastic cells with shorter telomeres. Additionally, a significant relationship has been observed between Her2-Neu overexpression and Her2-Neu positivity, which could suggest new combined therapies.The aim of this study was to detect the expression of hTERT, estrogen receptor (ER), progesterone receptor (PR), and HER2-Neu in neoplastic breast tissue embedded in paraffin before treatment and to investigate the relationship between them and with baseline and post-treatment telomere length, as well as with various clinicopathological parameters. Materials and methods A cross-sectional-correlational, 21 women diagnosed with breast cancer at the Oncology Service of the High Specialty Medical Unit No. 1 of Bajio of the Mexican Institute of Social Security. The study complies with the Helsinki Declaration and was approved by the Institutional Ethical Committee of the Mexican Institute of Social Security (R-2019-1001-127). A peripheral blood sample was obtained before oncological treatment and at the end of oncological treatment for the measurement of telomere length by extracting DNA from leukocytes, was performed by the quantitative polymerase chain reaction (PCR) method described by Cawthon. Tumor samples were collected from each patient at the oncology department for immunohistochemical determination of biomarker expression (ER, PR, Her2/neu) and hTERT. Results Of the 21 cases included in the study, the median age was 57.57 years. Eighteen cases were classified as invasive ductal carcinoma NOS (85.71%), 10 were histologic grade 2 (47.61%), 16 cases were hormone receptor positive (76.19%), 7 were Her2Neu positive (33.33%), and only 2 cases were triple negative (9.52%). Positive hTERT expression was detected in 11 cases (52.38%) and was negative in the remaining cases. A significant association was identified between hTERT-positive cases and Her2-Neu positive cases (p = 0.04). Baseline and post-treatment telomere lengths showed a significant difference using the non-parametric Wilcoxon t-test (p = 0.002). In hTERT-positive cases, there was significant telomere shortening at the end of oncological treatment (6.14 ± 1.54 vs. 4.75 ± 1.96 Kb, p = 0.007). Conclusion Positive hTERT immunostaining cases were associated with poor prognostic factors, such as Her2-Neu overexpression and post-treatment telomere shortening. In the future, hTERT immunostaining could be used to select patients for therapies with antagonistic effects on hTERT, as well as in the selection of more appropriate chemotherapy regimens for patients who express it.
Collapse
Affiliation(s)
- Blanca Olivia Murillo-Ortiz
- Unidad de Investigación en Epidemiología Clínica, OOAD Guanajuato, Instituto Mexicano del Seguro Social, León, Mexico
| | - Kenia García-Corrales
- Servicio de Anatomía Patológica, Hospital General de Zona No. 33, Instituto Mexicano del Seguro Social, Bahía de Banderas, Mexico
| | - Sandra Martínez-Garza
- Unidad de Investigación en Epidemiología Clínica, OOAD Guanajuato, Instituto Mexicano del Seguro Social, León, Mexico
| | - Marcos Javier Romero-Vázquez
- Unidad de Investigación en Epidemiología Clínica, OOAD Guanajuato, Instituto Mexicano del Seguro Social, León, Mexico
| | - Eduardo Agustín-Godínez
- Laboratorio de Anatomía Patológica e Inmunohistoquímica Especializada DIME, Hospital Médica Campestre, León, Mexico
| | - Andrea Escareño-Gómez
- Departamento de Patología Quirúrgica, UMAE Hospital de Especialidades No. 1, Centro Médico Nacional Bajío, Instituto Mexicano del Seguro Social, León, Mexico
| | | | | | - Mario Murguia-Perez
- Laboratorio de Anatomía Patológica e Inmunohistoquímica Especializada DIME, Hospital Médica Campestre, León, Mexico
- Departamento de Patología Quirúrgica, UMAE Hospital de Especialidades No. 1, Centro Médico Nacional Bajío, Instituto Mexicano del Seguro Social, León, Mexico
| |
Collapse
|
5
|
Jian X, Sun W, Zhang J, Zhang Q, Meng X, Lu H, Zheng D, Wu L, Wang Y. Frailty mediating the causality between leucocyte telomere length and mortality: a cohort study of 440,551 UK Biobank participants. EPMA J 2024; 15:99-110. [PMID: 38463625 PMCID: PMC10923753 DOI: 10.1007/s13167-024-00355-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/02/2024] [Indexed: 03/12/2024]
Abstract
Introduction Previous studies reported leucocyte telomere length (LTL) and frailty were associated with mortality, but it remains unclear whether frailty serves as a mediator in the relationship between leucocyte telomere length and mortality risk. This study aimed to evaluate how measuring LTL and frailty can support early monitoring and prevention of risk of mortality from the prospective of predictive, preventive, and personalized medicine (PPPM/3PM). Methods We included 440,551 participants from the UK Biobank between the baseline visit (2006-2010) and November 30, 2022. The time-dependent Cox proportional hazards model was conducted to assess the association between LTL and frailty index with the risk of mortality. Furthermore, we conducted causal mediation analyses to examine the extent to which frailty mediated the association between LTL and mortality. Results During a median follow-up of 13.74 years, each SD increase in LTL significantly decreased the risk of all-cause [hazard ratio (HR): 0.94, 95% confidence interval (CI): 0.93-0.95] and CVD-specific mortality (HR: 0.92, 95% CI: 0.90-0.95). The SD increase in FI elevated the risk of all-cause (HR: 1.35, 95% CI: 1.34-1.36), CVD-specific (HR: 1.47, 95% CI: 1.44-1.50), and cancer-specific mortality (HR: 1.22, 95% CI: 1.20-1.24). Frailty mediated approximately 10% of the association between LTL and all-cause and CVD-specific mortality. Conclusions Our results indicate that frailty mediates the effect of LTL on all-cause and CVD-specific mortality. There findings might be valuable to predict, prevent, and reduce mortality through primary prevention and healthcare in context of PPPM. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00355-7.
Collapse
Affiliation(s)
- Xuening Jian
- School of Public Health, Capital Medical University, Beijing, 100069 China
| | - Wenxin Sun
- School of Public Health, Capital Medical University, Beijing, 100069 China
| | - Jie Zhang
- School of Public Health, Capital Medical University, Beijing, 100069 China
| | - Qiaoyun Zhang
- Department of Anaesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiaoni Meng
- School of Public Health, Capital Medical University, Beijing, 100069 China
| | - Huimin Lu
- School of Public Health, Capital Medical University, Beijing, 100069 China
| | - Deqiang Zheng
- School of Public Health, Capital Medical University, Beijing, 100069 China
| | - Lijuan Wu
- School of Public Health, Capital Medical University, Beijing, 100069 China
| | - Youxin Wang
- School of Public Health, Capital Medical University, Beijing, 100069 China
- School of Public Health, North China University of Science and Technology, Tangshan, 063210 China
- Beijing Key Laboratory of Clinical Epidemiology, Capital Medical University, Beijing, 100069 China
| |
Collapse
|
6
|
Benites-Zapata VA, Ulloque-Badaracco JR, Alarcón-Braga EA, Fernández-Alonso AM, López-Baena MT, Pérez-López FR. Telomerase activity and telomere length in women with breast cancer or without malignancy: A systematic review and meta-analysis. Maturitas 2024; 180:107882. [PMID: 38029511 DOI: 10.1016/j.maturitas.2023.107882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/02/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023]
Abstract
AIM We performed a systematic review and meta-analysis to assess whether telomerase activity and telomere length are associated with breast cancer. METHODS PubMed, Web of Science, Embase, LILACS, Scielo, Embase, and CNKI databases were searched to obtain relevant articles published through May 10, 2023, following PRISMA guidelines and a registered PROSPERO protocol (CRD42022335402). We included observational studies reporting telomerase activity or telomere length in patients with breast cancer compared with women with benign lesions or normal tissue (control women). The Newcastle-Ottawa Scale was used to evaluate the quality of studies. Data were expressed as odds ratios (OR) and 95 % confidence intervals (CI). Random effects and inverse variance methods were used to meta-analyze associations. The I2 test was used to assess heterogeneity. RESULTS The meta-analysis of telomerase shows significantly greater activity in patients with breast cancer than in those without malignancies (OR = 23.46, 95 % CI 14.07-39.11, p < 0.00001, I2 = 72 %). There were non-significant differences in relative telomere length (OR = 1.16, 95 % CI = 0.90-1.49, p = 0.26, I2 = 86 %) and leukocyte telomere length (OR = 2.32, 95 % CI = 0.89-6.08, p = 0.09, I2 = 98 %) between women with and without breast cancer. In subgroup analyses by world regions of studies, both telomerase activity and telomere length displayed the same trends as in their respective meta-analyses. In sensitivity analyses, variables showed their respective same trends. CONCLUSION Telomerase activity is higher in patients with breast cancer than in women without malignancies. There were no significant differences in either relative telomere length or leukocyte telomere length in women with and without breast cancer. PROSPERO protocol CRD42022335402.
Collapse
Affiliation(s)
- Vicente A Benites-Zapata
- Unidad de Investigación para la Generación y Síntesis de Evidencias en Salud, Vicerrectorado de Investigación, Universidad San Ignacio de Loyola, Lima, Peru.
| | | | | | | | | | - Faustino R Pérez-López
- Instituto Aragonés de Ciencias de la Salud, Zaragoza, Spain; Facultad de Medicina, Universidad de Zaragoza, Zaragoza, Spain
| |
Collapse
|
7
|
Chen Z, Shen Y, He J, Shen Y, Zhu W, Wu X, Xiao M. Longer leukocyte telomere length increases cardiovascular mortality in type 2 diabetes patients. J Diabetes 2023; 15:325-331. [PMID: 36890680 PMCID: PMC10101828 DOI: 10.1111/1753-0407.13376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/05/2023] [Accepted: 02/15/2023] [Indexed: 03/10/2023] Open
Abstract
AIMS Leukocyte telomere length (LTL), as a biomarker of biological aging, is associated with the prevalence and complications of diabetes. This study aims to investigate the associations between LTL and all-cause and cause-specific mortality in patients with type 2 diabetes. METHODS All participants with baseline LTL records were included from the National Health and Nutrition Examination Survey 1999-2002. Death status and its causes were ascertained for National Death Index based on International Classification of Diseases, Tenth Revision code. Cox proportional hazards regression models were established to estimate the hazard ratios (HRs) of LTL associating with all-cause and cause-specific mortality. RESULTS The study enrolled 804 diabetic patients with the mean follow-up of 14.9 ± 2.59 years. There were 367 (45.6%) all-cause deaths, 80 (10.0%) cardiovascular deaths, and 42 (5.2%) cancer-related deaths. Longer LTL was associated with reduced all-cause mortality, whereas this association disappeared after adjusting for other variables. Compared with the lowest tertiles of LTL, the multivariable-adjusted hazard ratio of cardiovascular mortality was 2.11 (95% confidence interval [CI] 1.31-3.39; p < .05) in the highest tertiles. In terms of cancer mortality, the highest tertile was negatively correlated with the risk of cancer mortality (HR 0.58 [95% CI 0.37, 0.91], p < .05). CONCLUSION In conclusion, LTL was independently associated with the risk of cardiovascular mortality in patients with type 2 diabetes and was negatively correlated with the risk of cancer mortality. Telomere length may be a predictor of cardiovascular mortality in diabetes.
Collapse
Affiliation(s)
- Ziwei Chen
- Department of CardiologyAffiliated Hospital of Nantong UniversityNantongChina
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongChina
| | - Yao Shen
- Eye InstituteAffiliated Hospital of Nantong UniversityNantongChina
| | - Jing He
- Department of ChemotherapyAffiliated Hospital of Nantong UniversityNantongChina
| | - Yang Shen
- Department of CardiologyAffiliated Hospital of Nantong UniversityNantongChina
| | - Weida Zhu
- Department of CardiologyAffiliated Hospital of Nantong UniversityNantongChina
| | - Xiaohui Wu
- Department of CardiologyAffiliated Hospital of Nantong UniversityNantongChina
| | - Mingbing Xiao
- Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongChina
| |
Collapse
|
8
|
Ueno M. Exploring Genetic Interactions with Telomere Protection Gene pot1 in Fission Yeast. Biomolecules 2023; 13:biom13020370. [PMID: 36830739 PMCID: PMC9953254 DOI: 10.3390/biom13020370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
The regulation of telomere length has a significant impact on cancer risk and aging in humans. Circular chromosomes are found in humans and are often unstable during mitosis, resulting in genome instability. Some types of cancer have a high frequency of a circular chromosome. Fission yeast is a good model for studying the formation and stability of circular chromosomes as deletion of pot1 (encoding a telomere protection protein) results in rapid telomere degradation and chromosome fusion. Pot1 binds to single-stranded telomere DNA and is conserved from fission yeast to humans. Loss of pot1 leads to viable strains in which all three fission yeast chromosomes become circular. In this review, I will introduce pot1 genetic interactions as these inform on processes such as the degradation of uncapped telomeres, chromosome fusion, and maintenance of circular chromosomes. Therefore, exploring genes that genetically interact with pot1 contributes to finding new genes and/or new functions of genes related to the maintenance of telomeres and/or circular chromosomes.
Collapse
Affiliation(s)
- Masaru Ueno
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima 739-8530, Japan; ; Tel.: +81-82-424-7768
- Hiroshima Research Center for Healthy Aging (HiHA), Hiroshima University, Higashi-Hiroshima 739-8530, Japan
| |
Collapse
|
9
|
Zia S, Khan N, Tehreem K, Rehman N, Sami R, Baty RS, Tayeb FJ, Almashjary MN, Alsubhi NH, Alrefaei GI, Shahid R. Transcriptomic Analysis of Conserved Telomere Maintenance Component 1 (CTC1) and Its Association with Leukemia. J Clin Med 2022; 11:jcm11195780. [PMID: 36233645 PMCID: PMC9571731 DOI: 10.3390/jcm11195780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/16/2022] Open
Abstract
Telomere length (TEL) regulation is important for genome stability and is governed by the coordinated role of shelterin proteins, telomerase (TERT), and CST (CTC1/OBFC1/TEN1) complex. Previous studies have shown the association of telomerase expression with the risk of acute lymphoblastic leukemia (ALL). However, no data are available for CST association with the ALL. The current pilot study was designed to evaluate the CST expression levels in ALL. In total, 350 subjects were recruited, including 250 ALL cases and 100 controls. The subjects were stratified by age and categorized into pediatrics (1–18 years) and adults (19–54 years). TEL and expression patterns of CTC1, OBFC1, and TERT genes were determined by qPCR. The univariable logistic regression analysis was performed to determine the association of gene expression with ALL, and the results were adjusted for age and sex in multivariable analyses. Pediatric and adult cases did not reflect any change in telomere lengths relative to controls. However, expression of CTC1, OBFC1, and TERT genes were induced among ALL cases. Multivariable logistic regression analyses showed association of CTC1 with ALL in pediatric [β estimate (standard error (SE)= −0.013 (0.007), p = 0.049, and adults [0.053 (0.023), p = 0.025]. The association of CTC1 remained significant when taken together with OBFC1 and TERT in a multivariable model. Furthermore, CTC1 showed significant association with B-cell ALL [−0.057(0.017), p = 0.002) and T-cell ALL [−0.050 (0.018), p = 0.008] in pediatric group while no such association was noted in adults. Together, our findings demonstrated that telomere modulating genes, particularly CTC1, are strongly associated with ALL. Therefore, CTC1 can potentially be used as a risk biomarker for the identification of ALL in both pediatrics and adults.
Collapse
Affiliation(s)
- Saadiya Zia
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 45550, Pakistan
- Department of Biochemistry, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Netasha Khan
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 45550, Pakistan
| | - Komal Tehreem
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 45550, Pakistan
| | - Nazia Rehman
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 45550, Pakistan
| | - Rokayya Sami
- Department of Food Science and Nutrition, College of Sciences, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Roua S. Baty
- Department of Biotechnology, College of Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Faris J. Tayeb
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 47713, Saudi Arabia
| | - Majed N. Almashjary
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| | - Nouf H. Alsubhi
- Biological Sciences Department, College of Science and Arts, King Abdulaziz University, Rabigh 21911, Saudi Arabia
| | - Ghadeer I. Alrefaei
- Department of Biology, College of Science, University of Jeddah, P.O. Box 80327, Jeddah 21589, Saudi Arabia
| | - Ramla Shahid
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 45550, Pakistan
- Correspondence:
| |
Collapse
|
10
|
CRISPR/Cas: A New Tool in the Research of Telomeres and Telomerase as Well as a Novel Form of Cancer Therapy. Int J Mol Sci 2022; 23:ijms23063002. [PMID: 35328421 PMCID: PMC8953708 DOI: 10.3390/ijms23063002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 02/28/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
Due to their close connection with senescence, aging, and disease, telomeres and telomerase provide a unique and vital research route for boosting longevity and health span. Despite significant advances during the last three decades, earlier studies into these two biological players were impeded by the difficulty of achieving real-time changes inside living cells. As a result of the clustered regularly interspaced short palindromic repeats (CRISPR)-associated system’s (Cas) method, targeted genetic studies are now underway to change telomerase, the genes that govern it as well as telomeres. This review will discuss studies that have utilized CRISPR-related technologies to target and modify genes relevant to telomeres and telomerase as well as to develop targeted anti-cancer therapies. These studies greatly improve our knowledge and understanding of cellular and molecular mechanisms that underlie cancer development and aging.
Collapse
|
11
|
Sharma V, Letson J, Furuta S. Fibrous stroma: Driver and passenger in cancer development. Sci Signal 2022; 15:eabg3449. [PMID: 35258999 DOI: 10.1126/scisignal.abg3449] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cumulative evidence shows that fibrogenic stroma and stiff extracellular matrix (ECM) not only result from tumor growth but also play pivotal roles in cellular transformation and tumor initiation. This emerging concept may largely account for the increased cancer risk associated with environmental fibrogenic agents, such as asbestos and silica, and with chronic conditions that are fibrogenic, such as obesity and diabetes. It may also contribute to poor outcomes in patients treated with certain chemotherapeutics that can promote fibrosis, such as bleomycin and methotrexate. Although the mechanistic details of this phenomenon are still being unraveled, we provide an overview of the experimental evidence linking fibrogenic stroma and tumor initiation. In this Review, we will summarize the causes and consequences of fibrous stroma and how this stromal cue is transmitted to the nuclei of parenchymal cells through a physical continuum from the ECM to chromatin, as well as ECM-dependent biochemical signaling that contributes to cellular transformation.
Collapse
Affiliation(s)
- Vandana Sharma
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA
| | - Joshua Letson
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA
| | - Saori Furuta
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, 3000 Arlington Ave., Toledo, OH 43614, USA
| |
Collapse
|
12
|
Zinc Finger E-Box Binding Homeobox 2 as a Prognostic Biomarker in Various Cancers and Its Correlation with Infiltrating Immune Cells in Ovarian Cancer. Curr Issues Mol Biol 2022; 44:1203-1214. [PMID: 35723302 PMCID: PMC8947672 DOI: 10.3390/cimb44030079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 02/25/2022] [Accepted: 02/27/2022] [Indexed: 11/17/2022] Open
Abstract
This study investigated the expression of zinc finger E-box binding homeobox 2 (ZEB2), its prognostic significance in various cancers, and the correlation between ZEB2 and infiltrating immune cells and ZEB2-related proteins in ovarian cancer (OV). The Gene Expression Profiling Interactive Analysis tool was used to analyze RNA sequencing data and cancer survival rates, based on normal and tumor tissue data available in The Cancer Genome Atlas (TCGA) database. The Kaplan–Meier plotter and PrognoScan databases were used to analyze the prognostic value of ZEB2 in OV (n = 1144). The Tumor Immune Estimation Resource was used to investigate the correlation between ZEB2 and infiltrating immune cells in various cancers, including OV. High ZEB2 expression was associated with a poorer prognosis in OV. In OV, ZEB2 is positively correlated with CD8+T cells, neutrophils, macrophages, and dendritic cell invasion; and ZEB2 is negatively correlated with tumor-infiltrating B cells. The STRING database was used to investigate the correlations with ZEB2-related proteins. The results reveal that ZEB2 was positively correlated with SMAD1 and SMAD2 in OV. Our findings may serve as a potential prognostic biomarker, and provide novel insights into the tumor immunology in OV. Thus, ZEB2 may be a potential diagnostic and therapeutic target in OV.
Collapse
|
13
|
El Azzouzi M, El Ahanidi H, Hafidi Alaoui C, Chaoui I, Benbacer L, Tetou M, Hassan I, Bensaid M, Oukabli M, Ameur A, Al Bouzidi A, El Mzibri M, Attaleb M. Evaluation of DNA methylation in promoter regions of hTERT, TWIST1, VIM and NID2 genes in Moroccan bladder cancer patients. Cancer Genet 2021; 260-261:41-45. [PMID: 34922269 DOI: 10.1016/j.cancergen.2021.12.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 11/08/2021] [Accepted: 12/05/2021] [Indexed: 12/11/2022]
Abstract
Promoter hypermethylation have been reported to play a key role in bladder cancer development and progression. The aim of this study is to evaluate the methylation status of hTERT, TWIST1, VIM and NID2 genes in bladder cancer. The methylation status was evaluated using the Methylation-Specific PCR (MSP) approach on 70 tumour biopsies from Moroccan bladder cancer patients. Overall, methylation frequencies of hTERT, TWIST1, VIM and NID2 genes, were 90%, 85.71%, 67.14% and 67.14%, respectively. Hypermethylation of all studied genes was found in all pathological grades and stages of bladder cancer. Nevertheless, statistical analysis showed no significant association between promoter methylation of hTERT, TWIST1, VIM and NID2 genes and tumours stage/grade (p value >0.05). Moreover, we have investigated the association between the methylation pattern of selected genes and the treatment outcome in a sub-group of non-muscle-invasive bladder cancer cases (52/70). Hypermethylation of hTERT, TWIST1, VIM and NID2 was detected in 83.34%; 66.67%; 83.34% and 58.34% of recurrent cases, respectively, and in 80%; 80%; 80% and 60% of progressive cases, respectively. Statistical analysis highlighted a significant association between TWIST1 hypermethylation and tumour recurrence (p = 0.041<0.05). Our results indicate that hypermethylation of hTERT, TWIST1, VIM and NID2 genes is a frequent epigenetic event in bladder cancer and could be a promising therapeutic target to prevent bladder cancer progression and metastasis.
Collapse
Affiliation(s)
- Meryem El Azzouzi
- Biology and Medical Research Unit, CNESTEN, Rabat, Morocco; Faculty of Medicine and Pharmacy of Rabat. Mohammed V University in Rabat, Rabat, Morocco
| | - Hajar El Ahanidi
- Biology and Medical Research Unit, CNESTEN, Rabat, Morocco; Faculty of Medicine and Pharmacy of Rabat. Mohammed V University in Rabat, Rabat, Morocco
| | - Chaimae Hafidi Alaoui
- Biology and Medical Research Unit, CNESTEN, Rabat, Morocco; Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Imane Chaoui
- Biology and Medical Research Unit, CNESTEN, Rabat, Morocco
| | - Laila Benbacer
- Biology and Medical Research Unit, CNESTEN, Rabat, Morocco
| | | | | | | | - Mohamed Oukabli
- Faculty of Medicine and Pharmacy of Rabat. Mohammed V University in Rabat, Rabat, Morocco; Mohammed V Military Hospital, Rabat, Morocco
| | - Ahmed Ameur
- Faculty of Medicine and Pharmacy of Rabat. Mohammed V University in Rabat, Rabat, Morocco; Mohammed V Military Hospital, Rabat, Morocco
| | | | | | | |
Collapse
|
14
|
Kim HR, Seo CW, Yoo K, Han SJ, Kim J. Yes-associated protein 1 as a prognostic biomarker and its correlation with telomerase in various cancers. Osong Public Health Res Perspect 2021; 12:324-332. [PMID: 34719224 PMCID: PMC8561022 DOI: 10.24171/j.phrp.2021.0207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 09/02/2021] [Indexed: 11/06/2022] Open
Abstract
Objectives The aims of this study were to investigate the expression of Yes-associated protein 1 (YAP1), its prognostic significance, and the correlation between YAP1 and telomerase in various cancers. Methods The Gene Expression Profiling Interactive Analysis database was used to analyze RNA sequencing data and the survival rate of patients with various cancers in The Cancer Genome Atlas (TCGA) database. PrognoScan was used to analyze the prognostic value of YAP1 expression in various cancers. Tumor Immune Estimation Resource was used to determine the correlation between YAP1 expression and telomerase in various cancer types based on TCGA data. Results The analysis suggested that YAP1 was differentially expressed between tissues of various cancers and non-tumor tissues. High YAP1 expression was also related to a poor prognosis in adrenocortical carcinoma, bladder urothelial carcinoma, and pancreatic adenocarcinoma. Moreover, YAP1 expression was correlated with the expression of telomerase reverse transcriptase and telomerase RNA component in various cancer types. Conclusion These results suggest that YAP1 is a potential biomarker with prognostic significance and relevance for oncogene research in various cancer types. The correlation between the expression of YAP1 and telomere-associated genes will help to understand their cancer-promoting mechanisms and interactions.
Collapse
Affiliation(s)
- Hye-Ran Kim
- Department of Biomedical Laboratory Science, Dong-Eui Institute of Technology, Busan, Korea
| | - Choong-Won Seo
- Department of Biomedical Laboratory Science, Dong-Eui Institute of Technology, Busan, Korea
| | - Keunje Yoo
- Department of Environmental Engineering, Korea Maritime and Ocean University, Busan, Korea
| | - Sang Jun Han
- Department of Biotechnology, College of Fisheries Sciences, Pukyong National University, Busan, Korea
| | - Jongwan Kim
- Department of Biomedical Laboratory Science, Dong-Eui Institute of Technology, Busan, Korea
| |
Collapse
|
15
|
Analysis of Telomere Maintenance Related Genes Reveals NOP10 as a New Metastatic-Risk Marker in Pheochromocytoma/Paraganglioma. Cancers (Basel) 2021; 13:cancers13194758. [PMID: 34638246 PMCID: PMC8507560 DOI: 10.3390/cancers13194758] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Telomere maintenance involving TERT and ATRX genes has been recently described in metastatic pheochromocytoma and paraganglioma, reinforcing the importance of immortalization mechanisms in the progression of these tumors. Thus, the aim of this study was to analyze additional telomere-related genes to uncover potential new markers capable of identifying metastatic-risk patients more accurately. After analyzing 29 telomere-related genes, we were able to validate the predictive value of TERT and ATRX in mPPGL progression. In addition, we were able to identify NOP10 as a novel prognostic risk marker of mPPGLs, which also facilitates telomerase-dependent telomere length maintenance in these tumors. Interestingly, NOP10 overexpression assessment by IHC could be easily included within the current battery of markers for stratifying PPGL patients to fine-tune their clinical diagnoses. Abstract One of the main problems we face with PPGL is the lack of molecular markers capable of predicting the development of metastases in patients. Telomere-related genes, such as TERT and ATRX, have been recently described in PPGL, supporting the association between the activation of immortalization mechanisms and disease progression. However, the contribution of other genes involving telomere preservation machinery has not been previously investigated. In this work, we aimed to analyze the prognostic value of a comprehensive set of genes involved in telomere maintenance. For this study, we collected 165 PPGL samples (97 non-metastatic/63 metastatic), genetically characterized, in which the expression of 29 genes of interest was studied by NGS. Three of the 29 genes studied, TERT, ATRX and NOP10, showed differential expression between metastatic and non-metastatic cases, and alterations in these genes were associated with a shorter time to progression, independent of SDHB-status. We studied telomere length by Q-FISH in patient samples and in an in vitro model. NOP10 overexpressing tumors displayed an intermediate-length telomere phenotype without ALT, and in vitro results suggest that NOP10 has a role in telomerase-dependent telomere maintenance. We also propose the implementation of NOP10 IHC to better stratify PPGL patients.
Collapse
|
16
|
Sun L, Arbesman J. Canonical Signaling Pathways in Melanoma. Clin Plast Surg 2021; 48:551-560. [PMID: 34503716 DOI: 10.1016/j.cps.2021.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Melanoma is the most lethal type of skin cancer, originating from the uncontrolled proliferation of melanocytes. The transformation of normal melanocytes into malignant tumor cells has been a focus of research seeking to better understand melanoma's pathogenesis and develop new therapeutic targets. Over the past few decades, a conglomeration of studies has pinpointed several driver mutations and their associated signaling pathways. In this review, we summarize the key signaling pathways and the driver mutations involved in melanoma tumorigenesis and also discuss the potential underlying mechanisms.
Collapse
Affiliation(s)
- Lillian Sun
- Cleveland Clinic, Lerner College of Medicine at Case Western Reserve University, 9501 Euclid Avenue, Cleveland, OH 44106, USA
| | - Joshua Arbesman
- Department of Dermatology, Cleveland Clinic, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, Cleveland, OH 44195, USA.
| |
Collapse
|
17
|
Functional characterization of miR-708 microRNA in telomerase positive and negative human cancer cells. Sci Rep 2021; 11:17052. [PMID: 34426596 PMCID: PMC8382839 DOI: 10.1038/s41598-021-96096-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
Activation of a telomere length maintenance mechanism (TMM), including telomerase and alternative lengthening of telomeres (ALT), is essential for replicative immortality of tumor cells, although its regulatory mechanisms are incompletely understood. We conducted a microRNA (miRNA) microarray analysis on isogenic telomerase positive (TEP) and ALT cancer cell lines. Amongst nine miRNAs that showed difference in their expression in TEP and ALT cancer cells in array analysis, miR-708 was selected for further analysis since it was consistently highly expressed in a large panel of ALT cells. miR-708 in TEP and ALT cancer cells was not correlated with C-circle levels, an established feature of ALT cells. Its overexpression induced suppression of cell migration, invasion, and angiogenesis in both TEP and ALT cells, although cell proliferation was inhibited only in TEP cells suggesting that ALT cells may have acquired the ability to escape inhibition of cell proliferation by sustained miR-708 overexpression. Further, cell proliferation regulation in TEP cells by miR708 appears to be through the CARF-p53 pathway. We demonstrate here that miR-708 (i) is the first miRNA shown to be differentially regulated in TEP and ALT cancer cells, (ii) possesses tumor suppressor function, and (iii) deregulates CARF and p21WAF1-mediated signaling to limit proliferation in TEP cells.
Collapse
|
18
|
Akter J, Kamijo T. How Do Telomere Abnormalities Regulate the Biology of Neuroblastoma? Biomolecules 2021; 11:1112. [PMID: 34439779 PMCID: PMC8392161 DOI: 10.3390/biom11081112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 12/25/2022] Open
Abstract
Telomere maintenance plays important roles in genome stability and cell proliferation. Tumor cells acquire replicative immortality by activating a telomere-maintenance mechanism (TMM), either telomerase, a reverse transcriptase, or the alternative lengthening of telomeres (ALT) mechanism. Recent advances in the genetic and molecular characterization of TMM revealed that telomerase activation and ALT define distinct neuroblastoma (NB) subgroups with adverse outcomes, and represent promising therapeutic targets in high-risk neuroblastoma (HRNB), an aggressive childhood solid tumor that accounts for 15% of all pediatric-cancer deaths. Patients with HRNB frequently present with widely metastatic disease, with tumors harboring recurrent genetic aberrations (MYCN amplification, TERT rearrangements, and ATRX mutations), which are mutually exclusive and capable of promoting TMM. This review provides recent insights into our understanding of TMM in NB tumors, and highlights emerging therapeutic strategies as potential treatments for telomerase- and ALT-positive tumors.
Collapse
Affiliation(s)
- Jesmin Akter
- Saitama Cancer Center, Research Institute for Clinical Oncology, Saitama 362-0806, Japan;
| | - Takehiko Kamijo
- Saitama Cancer Center, Research Institute for Clinical Oncology, Saitama 362-0806, Japan;
- Laboratory of Tumor Molecular Biology, Department of Graduate School of Science and Engineering, Saitama University, Saitama 362-0806, Japan
| |
Collapse
|
19
|
Tong Y, Sun P, Yong J, Zhang H, Huang Y, Guo Y, Yu J, Zhou S, Wang Y, Wang Y, Ji Q, Wang Y, Chang C. Radiogenomic Analysis of Papillary Thyroid Carcinoma for Prediction of Cervical Lymph Node Metastasis: A Preliminary Study. Front Oncol 2021; 11:682998. [PMID: 34268116 PMCID: PMC8276635 DOI: 10.3389/fonc.2021.682998] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/09/2021] [Indexed: 12/19/2022] Open
Abstract
Background Papillary thyroid carcinoma (PTC) is characterized by frequent metastases to cervical lymph nodes (CLNs), and the presence of lymph node metastasis at diagnosis has a significant impact on the surgical approach. Therefore, we established a radiomic signature to predict the CLN status of PTC patients using preoperative thyroid ultrasound, and investigated the association between the radiomic features and underlying molecular characteristics of PTC tumors. Methods In total, 270 patients were enrolled in this prospective study, and radiomic features were extracted according to multiple guidelines. A radiomic signature was built with selected features in the training cohort and validated in the validation cohort. The total protein extracted from tumor samples was analyzed with LC/MS and iTRAQ technology. Gene modules acquired by clustering were chosen for their diagnostic significance. A radiogenomic map linking radiomic features to gene modules was constructed with the Spearman correlation matrix. Genes in modules related to metastasis were extracted for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses, and a protein-protein interaction (PPI) network was built to identify the hub genes in the modules. Finally, the screened hub genes were validated by immunohistochemistry analysis. Results The radiomic signature showed good performance for predicting CLN status in training and validation cohorts, with area under curve of 0.873 and 0.831 respectively. A radiogenomic map was created with nine significant correlations between radiomic features and gene modules, and two of them had higher correlation coefficient. Among these, MEmeganta representing the upregulation of telomere maintenance via telomerase and cell-cell adhesion was correlated with ‘Rectlike’ and ‘deviation ratio of tumor tissue and normal thyroid gland’ which reflect the margin and the internal echogenicity of the tumor, respectively. MEblue capturing cell-cell adhesion and glycolysis was associated with feature ‘minimum calcification area’ which measures the punctate calcification. The hub genes of the two modules were identified by protein-protein interaction network. Immunohistochemistry validated that LAMC1 and THBS1 were differently expressed in metastatic and non-metastatic tissues (p=0.003; p=0.002). And LAMC1 was associated with feature ‘Rectlike’ and ‘deviation ratio of tumor and normal thyroid gland’ (p<0.001; p<0.001); THBS1 was correlated with ‘minimum calcification area’ (p<0.001). Conclusions The radiomic signature proposed here has the potential to noninvasively predict the CLN status in PTC patients. Merging imaging phenotypes with genomic data could allow noninvasive identification of the molecular properties of PTC tumors, which might support clinical decision making and personalized management.
Collapse
Affiliation(s)
- Yuyang Tong
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Surgical Oncology, The Ohio State University, Columbus, OH, United States
| | - Peixuan Sun
- Diagnostic Imaging Center, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Yong
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, Finland.,Turku Biosciences Center, University of Turku and Åbo Akademi University, Turku, Finland
| | - Yunxia Huang
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Guo
- Department of Electronic Engineering, Fudan University and Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, China
| | - Jinhua Yu
- Department of Electronic Engineering, Fudan University and Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, China
| | - Shichong Zhou
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yulong Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qinghai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yuanyuan Wang
- Department of Electronic Engineering, Fudan University and Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, China
| | - Cai Chang
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Blattner-Johnson M, Jones DTW, Pfaff E. Precision medicine in pediatric solid cancers. Semin Cancer Biol 2021; 84:214-227. [PMID: 34116162 DOI: 10.1016/j.semcancer.2021.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/03/2021] [Accepted: 06/04/2021] [Indexed: 12/18/2022]
Abstract
Despite huge advances in the diagnosis and treatment of pediatric cancers over the past several decades, it remains one of the leading causes of death during childhood in developed countries. The development of new targeted treatments for these diseases has been hampered by two major factors. First, the extremely heterogeneous nature of the types of tumors encountered in this age group, and their fundamental differences from common adult carcinomas, has made it hard to truly get a handle on the complexities of the underlying biology driving tumor growth. Second, a reluctance of the pharmaceutical industry to develop products or trials for this population due to the relatively small size of the 'market', and a too-easy mechanism of obtaining waivers for pediatric development of adult oncology drugs based on disease type rather than mechanism of action, led to significant difficulties in getting access to new drugs. Thankfully, the field has now started to change, both scientifically and from a regulatory perspective, in order to address some of these challenges. In this review, we will examine some of the recent insights into molecular features which make pediatric tumors so unique and how these might represent therapeutic targets; highlight ongoing international initiatives for providing comprehensive, personalized genomic profiling of childhood tumors in a clinically-relevant timeframe, and look briefly at where the field of pediatric precision oncology may be heading in future.
Collapse
Affiliation(s)
- Mirjam Blattner-Johnson
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Elke Pfaff
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
21
|
Kockler ZW, Osia B, Lee R, Musmaker K, Malkova A. Repair of DNA Breaks by Break-Induced Replication. Annu Rev Biochem 2021; 90:165-191. [PMID: 33792375 DOI: 10.1146/annurev-biochem-081420-095551] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Double-strand DNA breaks (DSBs) are the most lethal type of DNA damage, making DSB repair critical for cell survival. However, some DSB repair pathways are mutagenic and promote genome rearrangements, leading to genome destabilization. One such pathway is break-induced replication (BIR), which repairs primarily one-ended DSBs, similar to those formed by collapsed replication forks or telomere erosion. BIR is initiated by the invasion of a broken DNA end into a homologous template, synthesizes new DNA within the context of a migrating bubble, and is associated with conservative inheritance of new genetic material. This mode of synthesis is responsible for a high level of genetic instability associated with BIR. Eukaryotic BIR was initially investigated in yeast, but now it is also actively studied in mammalian systems. Additionally, a significant breakthrough has been made regarding the role of microhomology-mediated BIR in the formation of complex genomic rearrangements that underly various human pathologies.
Collapse
Affiliation(s)
- Z W Kockler
- Department of Biology, University of Iowa, Iowa City, Iowa 52242, USA;
| | - B Osia
- Department of Biology, University of Iowa, Iowa City, Iowa 52242, USA;
| | - R Lee
- Department of Biology, University of Iowa, Iowa City, Iowa 52242, USA;
| | - K Musmaker
- Department of Biology, University of Iowa, Iowa City, Iowa 52242, USA;
| | - A Malkova
- Department of Biology, University of Iowa, Iowa City, Iowa 52242, USA;
| |
Collapse
|
22
|
Abstract
Epithelial ovarian cancer (EOC) is a heterogeneous group of diseases with distinct biological and clinical behaviour. Despite the differences between them, the capability of tumour cells to continuously proliferate and avoid death is maintained among histotypes. This ability is the result of alterations at different levels, causing the deregulation of cell cycle and proliferative-related pathways. Even if the leading role is played by RB and TP53, changes in other molecular pathways are involved in the development of EOC. This ability can be exploited to generate in vitro and in vivo models resembling the conditions of tumour development in a patient. In vivo models, such as patient-derived xenografts (PDX) or genetically engineered mouse models (GEMM), represent a fundamental tool in the study of the molecular mechanisms implicated in each EOC biotype for testing new therapeutic approaches. Herein we describe the major proliferation-related pathways and its disruption found in EOC and how these features can be used to establish in vivo models for translational research. Epithelial ovarian cancer (EOC) molecular biotypes are defined by distinct biology and clinical behaviour. Sustained proliferation and resistance to cell death mechanisms characterised tumour cells. RB and TP53 tumour-suppressor genes are highly implicated in EOC pathogenesis. In vitro and in vivo models have a key role in the study of molecular mechanisms involved in EOC pathogenesis. Development of animal models that mimic disease features constitute essential tools for new drugs testing.
Collapse
|
23
|
Goncalves T, Zoumpoulidou G, Alvarez-Mendoza C, Mancusi C, Collopy LC, Strauss SJ, Mittnacht S, Tomita K. Selective Elimination of Osteosarcoma Cell Lines with Short Telomeres by Ataxia Telangiectasia and Rad3-Related Inhibitors. ACS Pharmacol Transl Sci 2020; 3:1253-1264. [PMID: 33344901 PMCID: PMC7737214 DOI: 10.1021/acsptsci.0c00125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Indexed: 12/12/2022]
Abstract
![]()
To
avoid replicative senescence or telomere-induced apoptosis,
cancers employ telomere maintenance mechanisms (TMMs) involving either
the upregulation of telomerase or the acquisition of recombination-based
alternative telomere lengthening (ALT). The choice of TMM may differentially
influence cancer evolution and be exploitable in targeted therapies.
Here, we examine TMMs in a panel of 17 osteosarcoma-derived cell lines,
defining three separate groups according to TMM and the length of
telomeres maintained. Eight were ALT-positive, including the previously
uncharacterized lines, KPD and LM7. While ALT-positive lines all showed
excessive telomere length, ALT-negative cell lines fell into two groups
according to their telomere length: HOS-MNNG, OHSN, SJSA-1, HAL, 143b,
and HOS displayed subnormally short telomere length, while MG-63,
MHM, and HuO-3N1 displayed long telomeres. Hence, we further subcategorized
ALT-negative TMM into long-telomere (LT) and short-telomere (ST) maintenance groups.
Importantly, subnormally short telomeres were significantly associated
with hypersensitivity to three different therapeutics targeting the
protein kinase ataxia telangiectasia and Rad3-related (ATR) (AZD-6738/Ceralasertib,
VE-822/Berzoserib, and BAY-1895344) compared to long telomeres maintained
via ALT or telomerase. Within 24 h of ATR inhibition, cells with short
but not long telomeres displayed chromosome bridges and underwent
cell death, indicating a selective dependency on ATR for chromosome
stability. Collectively, our work provides a resource to identify
links between the mode of telomere maintenance and drug sensitivity
in osteosarcoma and indicates that telomere length predicts ATR inhibitor
sensitivity in cancer.
Collapse
Affiliation(s)
- Tomas Goncalves
- Centre for Genome Engineering and Maintenance, College of Health, Medicine and Life Sciences, Brunel University London, London UB8 3PH, United Kingdom.,Chromosome Maintenance Group, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Georgia Zoumpoulidou
- Cancer Cell Signalling, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Carlos Alvarez-Mendoza
- Cancer Cell Signalling, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Caterina Mancusi
- Cancer Cell Signalling, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Laura C Collopy
- Chromosome Maintenance Group, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Sandra J Strauss
- Department of Oncology, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom.,London Sarcoma Service, University College London Hospitals Foundation Trust, London WC1E 6DD, United Kingdom
| | - Sibylle Mittnacht
- Cancer Cell Signalling, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Kazunori Tomita
- Centre for Genome Engineering and Maintenance, College of Health, Medicine and Life Sciences, Brunel University London, London UB8 3PH, United Kingdom.,Chromosome Maintenance Group, UCL Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| |
Collapse
|
24
|
Wu Y, Poulos RC, Reddel RR. Role of POT1 in Human Cancer. Cancers (Basel) 2020; 12:cancers12102739. [PMID: 32987645 PMCID: PMC7598640 DOI: 10.3390/cancers12102739] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 09/20/2020] [Accepted: 09/22/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary The segmentation of eukaryotic genomes into discrete linear chromosomes requires processes to solve several major biological problems, including prevention of the chromosome ends being recognized as DNA breaks and compensation for the shortening that occurs when linear DNA is replicated. A specialized set of six proteins, collectively referred to as shelterin, is involved in both of these processes, and mutations in several of these are now known to be involved in cancer. Here, we focus on Protection of Telomeres 1 (POT1), the shelterin protein that appears to be most commonly involved in cancer, and consider the clinical significance of findings about its biological functions and the prevalence of inherited and acquired mutations in the POT1 gene. Abstract Telomere abnormalities facilitate cancer development by contributing to genomic instability and cellular immortalization. The Protection of Telomeres 1 (POT1) protein is an essential subunit of the shelterin telomere binding complex. It directly binds to single-stranded telomeric DNA, protecting chromosomal ends from an inappropriate DNA damage response, and plays a role in telomere length regulation. Alterations of POT1 have been detected in a range of cancers. Here, we review the biological functions of POT1, the prevalence of POT1 germline and somatic mutations across cancer predisposition syndromes and tumor types, and the dysregulation of POT1 expression in cancers. We propose a framework for understanding how POT1 abnormalities may contribute to oncogenesis in different cell types. Finally, we summarize the clinical implications of POT1 alterations in the germline and in cancer, and possible approaches for the development of targeted cancer therapies.
Collapse
Affiliation(s)
- Yangxiu Wu
- Cancer Research Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead NSW 2145, Australia;
- ProCan® Cancer Data Science Group, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead NSW 2145, Australia;
| | - Rebecca C. Poulos
- ProCan® Cancer Data Science Group, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead NSW 2145, Australia;
| | - Roger R. Reddel
- Cancer Research Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead NSW 2145, Australia;
- Correspondence: ; Tel.: +61-2-8865-2901
| |
Collapse
|
25
|
Bekmurzinova FK, Ospanov OB, Akilzhanova AR, Kozhamkulov UA, Rakhimova SE. The evaluation of chromosome telomere length change as a criterion of life expectancy in bariatric practice. OBESITY AND METABOLISM 2020; 17:125-129. [DOI: 10.14341/omet10331] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
In recent decades, the prevalence of obesity has been steadily increasing in most countries of the world. Overweight is a risk factor for a wide range of endocrine, cardiovascular, gastrointestinal, metabolic, neoplastic and musculoskeletal disorders and diseases. As you know, obesity is a state of chronic inflammation and severe oxidative stress, which will certainly affect the length of the chromosome telomeres. The dynamics of telomere length changes plays a decisive role in the regulation of cellular processes and cellular changes. Damage to telomeres, chromatin structures that help maintain the stability of the genome, leads to cell death or aging. However, information on how telomere length changes after weight loss through bariatric surgery remains limited to date. There are several types of bariatric surgery, each of which has its advantages and disadvantages. Based on this, it is possible that the restoration of the telomere length will differ depending on the technique used. This review describes the mechanisms for shortening leukocyte telomeres, and how bariatric surgery can affect this. The review also includes an analysis of evidence linking obesity and accelerated aging processes, as they are regulated by telomeres.
Collapse
|
26
|
Aramburu T, Plucinsky S, Skordalakes E. POT1-TPP1 telomere length regulation and disease. Comput Struct Biotechnol J 2020; 18:1939-1946. [PMID: 32774788 PMCID: PMC7385035 DOI: 10.1016/j.csbj.2020.06.040] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/24/2020] [Accepted: 06/27/2020] [Indexed: 12/27/2022] Open
Abstract
Telomeres are DNA repeats at the ends of linear chromosomes and are replicated by telomerase, a ribonucleoprotein reverse transcriptase. Telomere length regulation and chromosome end capping are essential for genome stability and are mediated primarily by the shelterin and CST complexes. POT1-TPP1, a subunit of shelterin, binds the telomeric overhang, suppresses ATR-dependent DNA damage response, and recruits telomerase to telomeres for DNA replication. POT1 localization to telomeres and chromosome end protection requires its interaction with TPP1. Therefore, the POT1-TPP1 complex is critical to telomere maintenance and full telomerase processivity. The aim of this mini-review is to summarize recent POT1-TPP1 structural studies and discuss how the complex contributes to telomere length regulation. In addition, we review how disruption of POT1-TPP1 function leads to human disease.
Collapse
Key Words
- ATM, Ataxia Telangiectasia Mutated protein
- ATR, Ataxia Telangiectasia and Rad3-related Protein
- CST, CTC1, Stn1 and Ten1
- CTC1, Conserved Telomere Capping Protein 1
- POT1
- POT1, Protection of telomere 1
- RAP1, Repressor/Activator Protein 1
- RPA, Replication Protein A
- SMCHD1, Structural Maintenance Of Chromosomes Flexible Hinge Domain Containing 1
- Shelterin
- Stn1, Suppressor of Cdc Thirteen
- TERC, Telomerase RNA
- TERT, Telomerase Reverse Transcriptase
- TIN2, TRF1- and TRF2-Interacting Nuclear Protein 2
- TPP1
- TPP1 also known as ACD, Adrenocortical Dysplasia Protein Homolog
- TRF1, Telomere Repeat binding Factor 1
- TRF2, Telomere Repeat binding Factor 2
- TSPYL5, Testis-specific Y-encoded-like protein 5
- Telomerase
- Telomeres
- Ten1, Telomere Length Regulation Protein
- USP7, ubiquitin-specific-processing protease 7
Collapse
|
27
|
Kahl VFS, Allen JAM, Nelson CB, Sobinoff AP, Lee M, Kilo T, Vasireddy RS, Pickett HA. Telomere Length Measurement by Molecular Combing. Front Cell Dev Biol 2020; 8:493. [PMID: 32612998 PMCID: PMC7308456 DOI: 10.3389/fcell.2020.00493] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 05/25/2020] [Indexed: 12/14/2022] Open
Abstract
Telomeres are repetitive regions of DNA bound by specialized proteins at the termini of linear chromosomes that prevent the natural chromosome ends from being recognized as DNA double strand breaks. Telomeric DNA is gradually eroded with each round of cell division, resulting in the accumulation of critically short or dysfunctional telomeres that eventually trigger cellular senescence. Consequently, telomere length is indicative of the proliferative capacity of a cell. Multiple methods exist to measure telomere length and telomere content, but a simple and reliable technique to accurately measure individual telomere lengths is currently lacking. We have developed the Telomere length Combing Assay (TCA) to measure telomere length on stretched DNA fibers. We used TCA to measure telomere erosion in primary human fibroblasts, and to detect telomere lengthening in response to activation of telomere maintenance pathways. TCA was also used to accurately measure telomere length in healthy individuals, and to identify critically short telomeres in patients with telomere biology disorders. TCA is performed on isolated DNA, negating the need for cycling cells. TCA is amenable to semi-automated image analysis, and can be fully automated using the Genomic Vision molecular combing platform. This not only precludes sampling bias, but also provides the potential for high-throughput applications and clinical development. TCA is a simple and versatile technique to measure the distribution of individual telomere lengths in a cell population, offering improved accuracy, and more detailed biological insight for telomere length measurement applications.
Collapse
Affiliation(s)
- Vivian F S Kahl
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Joshua A M Allen
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Christopher B Nelson
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Alexander P Sobinoff
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Michael Lee
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Tatjana Kilo
- Department of Hematology, Children's Hospital at Westmead, Sydney Children's Hospitals Network, Sydney, NSW, Australia
| | - Raja S Vasireddy
- Department of Hematology, Children's Hospital at Westmead, Sydney Children's Hospitals Network, Sydney, NSW, Australia
| | - Hilda A Pickett
- Telomere Length Regulation Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
28
|
M’kacher R, Colicchio B, Borie C, Junker S, Marquet V, Heidingsfelder L, Soehnlen K, Najar W, Hempel WM, Oudrhiri N, Wilhelm-Murer N, Miguet M, Arnoux M, Ferrapie C, Kerbrat W, Plesch A, Dieterlen A, Girinsky T, Voisin P, Deschenes G, Tabet AC, Yardin C, Bennaceur-Griscelli A, Fenech M, Carde P, Jeandidier E. Telomere and Centromere Staining Followed by M-FISH Improves Diagnosis of Chromosomal Instability and Its Clinical Utility. Genes (Basel) 2020; 11:E475. [PMID: 32349350 PMCID: PMC7291161 DOI: 10.3390/genes11050475] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022] Open
Abstract
Dicentric chromosomes are a relevant marker of chromosomal instability. Their appearance is associated with telomere dysfunction, leading to cancer progression and a poor clinical outcome. Here, we present Telomere and Centromere staining followed by M-FISH (TC+M-FISH) for improved detection of telomere dysfunction and the identification of dicentric chromosomes in cancer patients and various genetic syndromes. Significant telomere length shortening and significantly higher frequencies of telomere loss and deletion were found in the peripheral lymphocytes of patients with cancer and genetic syndromes relative to similar age-matched healthy donors. We assessed our technique against conventional cytogenetics for the detection of dicentric chromosomes by subjecting metaphase preparations to both approaches. We identified dicentric chromosomes in 28/50 cancer patients and 21/44 genetic syndrome patients using our approach, but only 7/50 and 12/44, respectively, using standard cytogenetics. We ascribe this discrepancy to the identification of the unique configuration of dicentric chromosomes. We observed significantly higher frequencies of telomere loss and deletion in patients with dicentric chromosomes (p < 10-4). TC+M-FISH analysis is superior to classical cytogenetics for the detection of chromosomal instability. Our approach is a relatively simple but useful tool for documenting telomere dysfunction and chromosomal instability with the potential to become a standard additional diagnostic tool in medical genetics and the clinic.
Collapse
Affiliation(s)
- Radhia M’kacher
- Cell Environment, DNA Damage R&D, 75020 Paris, France; (K.S.); (W.N.); (W.M.H.); (P.V.)
| | - Bruno Colicchio
- IRIMAS, Institut de Recherche en Informatique, Mathématiques, Automatique et Signal, Université de Haute-Alsace, 68093 Mulhouse, France; (B.C.); (A.D.)
| | - Claire Borie
- APHP-Service D’hématologie Oncohématologie Moléculaire et Cytogénétique Hôpital Paul Brousse Université Paris Saclay/ Inserm UMR 935, 94800 Villejuif, France; (C.B.); (N.O.); (M.A.); (C.F.); (W.K.); (A.B.-G.)
| | - Steffen Junker
- Institute of Biomedicine, University of Aarhus, DK-8000 Aarhus, Denmark;
| | - Valentine Marquet
- Service de Cytogénétique, Génétique Médicale, et Biologie de la Reproduction Hôpital de la Mère et de l’Enfant, CHU Dupuytren, 87042 Limoges, France; (V.M.); (C.Y.)
| | | | - Kevin Soehnlen
- Cell Environment, DNA Damage R&D, 75020 Paris, France; (K.S.); (W.N.); (W.M.H.); (P.V.)
| | - Wala Najar
- Cell Environment, DNA Damage R&D, 75020 Paris, France; (K.S.); (W.N.); (W.M.H.); (P.V.)
- IRIMAS, Institut de Recherche en Informatique, Mathématiques, Automatique et Signal, Université de Haute-Alsace, 68093 Mulhouse, France; (B.C.); (A.D.)
- APHP-Service D’hématologie Oncohématologie Moléculaire et Cytogénétique Hôpital Paul Brousse Université Paris Saclay/ Inserm UMR 935, 94800 Villejuif, France; (C.B.); (N.O.); (M.A.); (C.F.); (W.K.); (A.B.-G.)
- Institute of Biomedicine, University of Aarhus, DK-8000 Aarhus, Denmark;
- Service de Cytogénétique, Génétique Médicale, et Biologie de la Reproduction Hôpital de la Mère et de l’Enfant, CHU Dupuytren, 87042 Limoges, France; (V.M.); (C.Y.)
- MetaSystems GmbH, Robert-Bosch-Str., 6 D-68804 Altlussheim, Germany; (L.H.); (A.P.)
- Faculté de Médicine, Université Paris Descartes, 75005 Paris, France
| | - William M. Hempel
- Cell Environment, DNA Damage R&D, 75020 Paris, France; (K.S.); (W.N.); (W.M.H.); (P.V.)
| | - Noufissa Oudrhiri
- APHP-Service D’hématologie Oncohématologie Moléculaire et Cytogénétique Hôpital Paul Brousse Université Paris Saclay/ Inserm UMR 935, 94800 Villejuif, France; (C.B.); (N.O.); (M.A.); (C.F.); (W.K.); (A.B.-G.)
| | - Nadège Wilhelm-Murer
- Service de Génétique Groupe Hospitalier de la Région de Mulhouse et Sud Alsace Mulhouse, 68070 Mulhouse, France; (N.W.-M.); (M.M.); (E.J.)
| | - Marguerite Miguet
- Service de Génétique Groupe Hospitalier de la Région de Mulhouse et Sud Alsace Mulhouse, 68070 Mulhouse, France; (N.W.-M.); (M.M.); (E.J.)
| | - Micheline Arnoux
- APHP-Service D’hématologie Oncohématologie Moléculaire et Cytogénétique Hôpital Paul Brousse Université Paris Saclay/ Inserm UMR 935, 94800 Villejuif, France; (C.B.); (N.O.); (M.A.); (C.F.); (W.K.); (A.B.-G.)
| | - Catherine Ferrapie
- APHP-Service D’hématologie Oncohématologie Moléculaire et Cytogénétique Hôpital Paul Brousse Université Paris Saclay/ Inserm UMR 935, 94800 Villejuif, France; (C.B.); (N.O.); (M.A.); (C.F.); (W.K.); (A.B.-G.)
| | - Wendy Kerbrat
- APHP-Service D’hématologie Oncohématologie Moléculaire et Cytogénétique Hôpital Paul Brousse Université Paris Saclay/ Inserm UMR 935, 94800 Villejuif, France; (C.B.); (N.O.); (M.A.); (C.F.); (W.K.); (A.B.-G.)
| | - Andreas Plesch
- MetaSystems GmbH, Robert-Bosch-Str., 6 D-68804 Altlussheim, Germany; (L.H.); (A.P.)
| | - Alain Dieterlen
- IRIMAS, Institut de Recherche en Informatique, Mathématiques, Automatique et Signal, Université de Haute-Alsace, 68093 Mulhouse, France; (B.C.); (A.D.)
| | - Theodore Girinsky
- Department of Radiation Oncology, Gustave Roussy Cancer Campus, 94800 Villejuif, France;
| | - Philippe Voisin
- Cell Environment, DNA Damage R&D, 75020 Paris, France; (K.S.); (W.N.); (W.M.H.); (P.V.)
| | - Georges Deschenes
- Nephrology Department, APHP-Hopital Robert Debré, 75019 Paris, France;
| | - Anne-Claude Tabet
- Cytogenetic Laboratory, APHP-Hopital Robert Debré, 75019 Paris, France;
| | - Catherine Yardin
- Service de Cytogénétique, Génétique Médicale, et Biologie de la Reproduction Hôpital de la Mère et de l’Enfant, CHU Dupuytren, 87042 Limoges, France; (V.M.); (C.Y.)
| | - Annelise Bennaceur-Griscelli
- APHP-Service D’hématologie Oncohématologie Moléculaire et Cytogénétique Hôpital Paul Brousse Université Paris Saclay/ Inserm UMR 935, 94800 Villejuif, France; (C.B.); (N.O.); (M.A.); (C.F.); (W.K.); (A.B.-G.)
| | - Michael Fenech
- School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5000, Australia;
- Genome Health Foundation, North Brighton, SA 5048, Australia
| | - Patrice Carde
- Department of Hematology, Gustave Roussy Cancer Campus, 94800 Villejuif, France;
| | - Eric Jeandidier
- Service de Génétique Groupe Hospitalier de la Région de Mulhouse et Sud Alsace Mulhouse, 68070 Mulhouse, France; (N.W.-M.); (M.M.); (E.J.)
| |
Collapse
|
29
|
The Role of Alternative Lengthening of Telomeres Mechanism in Cancer: Translational and Therapeutic Implications. Cancers (Basel) 2020; 12:cancers12040949. [PMID: 32290440 PMCID: PMC7226354 DOI: 10.3390/cancers12040949] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/17/2022] Open
Abstract
Telomere maintenance mechanisms (i.e., telomerase activity (TA) and the alternative lengthening of telomere (ALT) mechanism) contribute to tumorigenesis by providing unlimited proliferative capacity to cancer cells. Although the role of either telomere maintenance mechanisms seems to be equivalent in providing a limitless proliferative ability to tumor cells, the contribution of TA and ALT to the clinical outcome of patients may differ prominently. In addition, several strategies have been developed to interfere with TA in cancer, including Imetelstat that has been the first telomerase inhibitor tested in clinical trials. Conversely, the limited information available on the molecular underpinnings of ALT has hindered thus far the development of genuine ALT-targeting agents. Moreover, whether anti-telomerase therapies may be hampered or not by possible adaptive responses is still debatable. Nonetheless, it is plausible hypothesizing that treatment with telomerase inhibitors may exert selective pressure for the emergence of cancer cells that become resistant to treatment by activating the ALT mechanism. This notion, together with the evidence that both telomere maintenance mechanisms may coexist within the same tumor and may distinctly impinge on patients' outcomes, suggests that ALT may exert an unexpected role in tumor biology that still needs to be fully elucidated.
Collapse
|
30
|
Zhang JM, Yadav T, Ouyang J, Lan L, Zou L. Alternative Lengthening of Telomeres through Two Distinct Break-Induced Replication Pathways. Cell Rep 2020; 26:955-968.e3. [PMID: 30673617 PMCID: PMC6366628 DOI: 10.1016/j.celrep.2018.12.102] [Citation(s) in RCA: 185] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 11/09/2018] [Accepted: 12/26/2018] [Indexed: 12/11/2022] Open
Abstract
Alternative lengthening of telomeres (ALT) is a telomerase-independent but recombination-dependent pathway that maintains telomeres. Here, we describe an assay to visualize ALT-mediated telomeric DNA synthesis in ALT-associated PML bodies (APBs) without DNA-damaging agents or replication inhibitors. Using this assay, we find that ALT occurs through two distinct mechanisms. One of the ALT mechanisms requires RAD52, a protein implicated in break-induced DNA replication (BIR). We demonstrate that RAD52 directly promotes telomeric D-loop formation in vitro and is required for maintaining telomeres in ALT-positive cells. Unexpectedly, however, RAD52 is dispensable for C-circle formation, a hallmark of ALT. In RAD52-knockout ALT cells, C-circle formation and RAD52-independent ALT DNA synthesis gradually increase as telomeres are shortened, and these activities are dependent on BLM and BIR proteins POLD3 and POLD4. These results suggest that ALT occurs through a RAD52-dependent and a RAD52-independent BIR pathway, revealing the bifurcated framework and dynamic nature of this process.
Collapse
Affiliation(s)
- Jia-Min Zhang
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Tribhuwan Yadav
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Jian Ouyang
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA
| | - Li Lan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Lee Zou
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA 02129, USA; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
31
|
Sui Y, Zhang W, Tang T, Gao L, Cao T, Zhu H, You Q, Yu B, Yang T. Insulin-like growth factor-II overexpression accelerates parthenogenetic stem cell differentiation into cardiomyocytes and improves cardiac function after acute myocardial infarction in mice. Stem Cell Res Ther 2020; 11:86. [PMID: 32102690 PMCID: PMC7045450 DOI: 10.1186/s13287-020-1575-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/01/2020] [Accepted: 01/28/2020] [Indexed: 12/11/2022] Open
Abstract
Background Parthenogenetic stem cells (PSCs) are a promising source of regenerated cardiomyocytes; however, their application may be limited without a paternal genome. Insulin-like growth factor-II (IGF-II), a paternally expressed growth hormone, is critical in embryonic differentiation. This study investigated whether forced expression of IGF-II in PSCs can accelerate their differentiation. Methods Overexpression and re-knockdown of IGF-II in PSCs were performed to investigate the role of IGF-II in PSC differentiation. The derivatives of PSCs with different IGF-II manipulations were transplanted into infarcted murine hearts to investigate the role of IGF-II in cardiomyocyte differentiation in vivo. Results Data showed that the expression of cardiac troponin T and troponin I in IGF-II-PSC outgrowths preceded that of parental PSC outgrowths, suggesting that IGF-II can accelerate PSC differentiation into cardiac lineage. Overexpression of IGF-II accelerated PSC differentiation towards cardiomyocytes while inhibiting PSC proliferation via the IGF-II/IGF1R signaling. Similar to that observed in cardiac marker expression, on differentiation day 24, IGF-II-PSCs showed PCNA and cyclin D2 expression comparable to juvenile mouse cardiomyocytes, showing that IGF-II-PSCs at this stage possess differential and proliferative properties similar to those of juvenile cardiomyocytes. Moreover, the expression pattern of cardiac markers in IGF-II-overexpressing PSC derivatives resembled that of juvenile mouse cardiomyocytes. After transplantation into the infarcted mouse hearts, IGF-II-PSC-derived cardiomyocytes displayed significant characteristics of mature cardiomyocytes, and IGF-II-depletion by shRNA significantly reversed these effects, suggesting the critical role of IGF-II in promoting cardiomyocyte maturation in vivo. Furthermore, IGF-II-overexpressing PSC derivatives reduced collagen deposition and mitochondrial damage in the infarcted areas and improved cardiac function. The re-knockdown of IGF-II could counteract these favorable effects of IGF-II. Conclusions These findings suggest that the ectopic expression of IGF-II accelerates PSC differentiation into the cardiac lineage and promotes cardiomyocyte maturation. The underlying process includes the IGF-II/IGF1R signaling, which is involved in the suppressive effect of IGF-II on PSC proliferation. Moreover, transplanting IGF-II-overexpressing PSC derivatives into the infarcted heart could reduce collagen deposition and improve mitochondria biogenesis and measurements of cardiac function, highlighting the importance of IGF-II in the application of PSCs in cardiac regeneration.
Collapse
Affiliation(s)
- Yi Sui
- Department of Nutrition, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Wei Zhang
- Department of Pharmacology, School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050200, China
| | - Tao Tang
- Department of Obstetrics and Gynaecology, Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Lili Gao
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Ting Cao
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Hongbo Zhu
- Department of Pathology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Qinghua You
- Department of Pathology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Bo Yu
- Department of General Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| | - Tao Yang
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| |
Collapse
|
32
|
Sieverling L, Hong C, Koser SD, Ginsbach P, Kleinheinz K, Hutter B, Braun DM, Cortés-Ciriano I, Xi R, Kabbe R, Park PJ, Eils R, Schlesner M, Brors B, Rippe K, Jones DTW, Feuerbach L. Genomic footprints of activated telomere maintenance mechanisms in cancer. Nat Commun 2020; 11:733. [PMID: 32024817 PMCID: PMC7002710 DOI: 10.1038/s41467-019-13824-9] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 11/26/2019] [Indexed: 12/14/2022] Open
Abstract
Cancers require telomere maintenance mechanisms for unlimited replicative potential. They achieve this through TERT activation or alternative telomere lengthening associated with ATRX or DAXX loss. Here, as part of the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium, we dissect whole-genome sequencing data of over 2500 matched tumor-control samples from 36 different tumor types aggregated within the ICGC/TCGA Pan-Cancer Analysis of Whole Genomes (PCAWG) Consortium to characterize the genomic footprints of these mechanisms. While the telomere content of tumors with ATRX or DAXX mutations (ATRX/DAXXtrunc) is increased, tumors with TERT modifications show a moderate decrease of telomere content. One quarter of all tumor samples contain somatic integrations of telomeric sequences into non-telomeric DNA. This fraction is increased to 80% prevalence in ATRX/DAXXtrunc tumors, which carry an aberrant telomere variant repeat (TVR) distribution as another genomic marker. The latter feature includes enrichment or depletion of the previously undescribed singleton TVRs TTCGGG and TTTGGG, respectively. Our systematic analysis provides new insight into the recurrent genomic alterations associated with telomere maintenance mechanisms in cancer.
Collapse
Affiliation(s)
- Lina Sieverling
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Chen Hong
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Sandra D Koser
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Philip Ginsbach
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Kortine Kleinheinz
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- Department for Bioinformatics and Functional Genomics, Institute for Pharmacy and Molecular Biotechnology (IPMB) and BioQuant, 69120, Heidelberg, Germany
| | - Barbara Hutter
- German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
- Heidelberg Center for Personalized Oncology (DKFZ-HIPO), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Delia M Braun
- Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
- Division of Chromatin Networks, German Cancer Research Center (DKFZ) and BioQuant, 69120, Heidelberg, Germany
| | - Isidro Cortés-Ciriano
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, 02115, USA
- Department of Chemistry, Centre for Molecular Science Informatics, University of Cambridge, Cambridge, CB2 1EW, UK
- Ludwig Center at Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Ruibin Xi
- School of Mathematical Sciences and Center for Statistical Science, Peking University, Beijing, 100871, China
| | - Rolf Kabbe
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Peter J Park
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts, 02115, USA
- Ludwig Center at Harvard Medical School, Boston, Massachusetts, 02115, USA
| | - Roland Eils
- Division of Theoretical Bioinformatics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- Department for Bioinformatics and Functional Genomics, Institute for Pharmacy and Molecular Biotechnology (IPMB) and BioQuant, 69120, Heidelberg, Germany
| | - Matthias Schlesner
- Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
| | - Benedikt Brors
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Karsten Rippe
- Division of Chromatin Networks, German Cancer Research Center (DKFZ) and BioQuant, 69120, Heidelberg, Germany
| | - David T W Jones
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany
- Pediatric Glioma Research Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lars Feuerbach
- Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
| |
Collapse
|
33
|
Sommer A, Royle NJ. ALT: A Multi-Faceted Phenomenon. Genes (Basel) 2020; 11:E133. [PMID: 32012790 PMCID: PMC7073516 DOI: 10.3390/genes11020133] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/17/2020] [Accepted: 01/22/2020] [Indexed: 01/13/2023] Open
Abstract
One of the hallmarks of cancer cells is their indefinite replicative potential, made possible by the activation of a telomere maintenance mechanism (TMM). The majority of cancers reactivate the reverse transcriptase, telomerase, to maintain their telomere length but a minority (10% to 15%) utilize an alternative lengthening of telomeres (ALT) pathway. Here, we review the phenotypes and molecular markers specific to ALT, and investigate the significance of telomere mutations and sequence variation in ALT cell lines. We also look at the recent advancements in understanding the different mechanisms behind ALT telomere elongation and finally, the progress made in identifying potential ALT-targeted therapies, including those already in use for the treatment of both hematological and solid tumors.
Collapse
Affiliation(s)
| | - Nicola J. Royle
- Department of Genetics and Genome Biology, University of Leicester, Leicester LE1 7RH, UK;
| |
Collapse
|
34
|
Panneerpandian P, Devanandan HJ, Marimuthu A, Karthikeyan C, Ganesan K. Abacavir induces the transcriptional activity of YY1 and other oncogenic transcription factors in gastric cancer cells. Antiviral Res 2019; 174:104695. [PMID: 31846633 DOI: 10.1016/j.antiviral.2019.104695] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 12/11/2019] [Accepted: 12/13/2019] [Indexed: 12/21/2022]
Abstract
Yin Yang 1 (YY1) is a ubiquitous transcription factor with both transcriptional activating and repressing functions. Targeting YY1 is considered as a potential therapeutic strategy for several malignancies. Telomerase Reverse Transcriptase (TERT) is also considered as a potential target for cancer therapeutics. To enable the large-scale screening and identification of potential YY1 targeting drugs, a gastric cancer cell line-based drug screening assay was developed. In a YY1 targeted drug repurpose screen, abacavir sulfate, a nucleoside analog reverse transcriptase inhibitor, known to target TERT was identified to show the feature of activating YY1 mediated transcription. We further explored i) the molecular targets of abacavir, ii) activation pattern of pathways regulated by abacavir in gastric tumors, and iii) therapeutic potential of abacavir for gastric cancer cells. Oncogenic signaling pathways like MYC, HIF1-α, ERK, WNT, E2F, NFκB and NRF1/2 were also found to be highly activated by abacavir. Abacavir was found to have less impact on the viability of gastric cancer cells. Across gastric tumors, we observed the co-activation of TERT, alternative lengthening of telomere (ALT), DNA repair, and the oncogenic pathways MYC, E2F/DP1, ERK, YY1, HIF1α, and NFκB specific gene-sets, in a subset of gastric tumors. The observed connectivity among TERT, DNA repair, and multiple oncogenic pathways indicate the need for the development of combinatorial therapeutics for the gastric tumors with the activated TERT.
Collapse
Affiliation(s)
- Ponmathi Panneerpandian
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India.
| | - Helen Jemimah Devanandan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | - Anantharaj Marimuthu
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India
| | | | - Kumaresan Ganesan
- Unit of Excellence in Cancer Genetics, Department of Genetics, Centre for Excellence in Genomic Sciences, School of Biological Sciences, Madurai Kamaraj University, Madurai, India.
| |
Collapse
|
35
|
Ko E, Kim JS, Bae JW, Kim J, Park SG, Jung G. SERPINA3 is a key modulator of HNRNP-K transcriptional activity against oxidative stress in HCC. Redox Biol 2019; 24:101217. [PMID: 31121493 PMCID: PMC6529774 DOI: 10.1016/j.redox.2019.101217] [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: 01/29/2019] [Revised: 04/26/2019] [Accepted: 05/10/2019] [Indexed: 02/07/2023] Open
Abstract
Most studies about serpin peptidase inhibitor, clade A member 3 (SERPINA3) has been limited to its inhibitory functions and mechanisms. Herein, we report a novel role of SERPINA3 in transcriptional regulation of HCC progression-related genes. Among 19 selected genes through HCC cell isolation system based on telomere length, microarray analyses, and cell-based studies, SERPINA3 was the strongest determinant of increases in telomere length, HCC cell proliferation, survival, migration, and invasion. We also found that SERPINA3 strongly interacted with heterogeneous nuclear ribonucleoprotein K (HNRNP-K) under H2O2 exposure, and the oxidation-elicited SERPINA3-HNRNP-K complex enhanced the promoter activities and transcript levels of a telomere-relating gene (POT1) and HCC-promoting genes (UHRF1 and HIST2H2BE). Intriguingly, the inhibition of SERPINA3 oxidation rendered the transcriptional activity of the SERPINA3-HNRNP-K complex suppressed. Moreover, the co-immunoprecipitated HNRNP-K with SERPINA3 quantitatively correlated with not only the level of SERPINA3 oxidation but also the level of POT1, UHRF1, and HIST2H2BE transcripts and telomere length in HCC tissues. Therefore, the upregulated transcriptional activity of HNRNP-K mediated by SERPINA3 promotes HCC cell survival and proliferation and could be an indicator of poor prognosis for HCC patients. SERPINA3-HNRNP-K complex promotes HCC survival and proliferation. Oxidation of SERPINA3 accentuated the role of complex on target regulatory DNA. Blockade of the SERPINA3-HNRNP-K complex could be valuable in HCC therapy.
Collapse
Affiliation(s)
- Eunkyong Ko
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - Jong-Seo Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea; Center for RNA Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Jong Woo Bae
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea; Center for RNA Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Jeesoo Kim
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea; Center for RNA Research, Institute of Basic Science (IBS), Seoul, 08826, Republic of Korea
| | - Sung-Gyoo Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Guhung Jung
- Department of Biological Sciences, College of Natural Sciences, Seoul National University, 599 Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
36
|
Goncalves J, Lussey-Lepoutre C, Favier J, Gimenez-Roqueplo AP, Castro-Vega LJ. Emerging molecular markers of metastatic pheochromocytomas and paragangliomas. ANNALES D'ENDOCRINOLOGIE 2019; 80:159-162. [PMID: 31053249 DOI: 10.1016/j.ando.2019.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Metastatic pheochromocytoma/paraganglioma (PPGL) represents a major clinical challenge due to limitations in accurate diagnostic tools and effective treatments. Currently, patients classified at high-risk by means of clinical, biochemical and genetic criteria, require a lifelong monitoring, while it remains difficult to determine the metastatic potential of PPGL only on the basis of histopathological features. Thus, tumor molecular markers that improve the risk stratification of these patients are needed. In the past few years, we have witnessed an unprecedented molecular characterization of PPGL, which led to the emergence of promising candidate biomarkers predictive of metastatic behavior. Here, we briefly discuss these breakthroughs and provide some insights for the prospective implementation of molecular markers of metastatic PPGL in the clinical setting in years to come.
Collapse
Affiliation(s)
- Judith Goncalves
- Inserm, UMR970, équipe labellisée Ligue Contre le Cancer, Paris-Cardiovascular Research Center, 75015 Paris, France; Faculté de médecine, PRES Sorbonne Paris-Cité, Paris-Descartes University, 75006 Paris, France
| | - Charlotte Lussey-Lepoutre
- Inserm, UMR970, équipe labellisée Ligue Contre le Cancer, Paris-Cardiovascular Research Center, 75015 Paris, France; Department of Nuclear Medicine, Pitié-Salpêtrière Hospital, Sorbonne University, 75013 Paris, France
| | - Judith Favier
- Inserm, UMR970, équipe labellisée Ligue Contre le Cancer, Paris-Cardiovascular Research Center, 75015 Paris, France; Faculté de médecine, PRES Sorbonne Paris-Cité, Paris-Descartes University, 75006 Paris, France
| | - Anne-Paule Gimenez-Roqueplo
- Inserm, UMR970, équipe labellisée Ligue Contre le Cancer, Paris-Cardiovascular Research Center, 75015 Paris, France; Faculté de médecine, PRES Sorbonne Paris-Cité, Paris-Descartes University, 75006 Paris, France; Genetics Department, hôpital européen Georges-Pompidou, AP-HP, 75015, Paris, France
| | - Luis Jaime Castro-Vega
- Inserm, UMR970, équipe labellisée Ligue Contre le Cancer, Paris-Cardiovascular Research Center, 75015 Paris, France; Faculté de médecine, PRES Sorbonne Paris-Cité, Paris-Descartes University, 75006 Paris, France.
| |
Collapse
|
37
|
Zhang X, Liu Z, Liu X, Wang S, Zhang Y, He X, Sun S, Ma S, Shyh-Chang N, Liu F, Wang Q, Wang X, Liu L, Zhang W, Song M, Liu GH, Qu J. Telomere-dependent and telomere-independent roles of RAP1 in regulating human stem cell homeostasis. Protein Cell 2019; 10:649-667. [PMID: 30796637 PMCID: PMC6711945 DOI: 10.1007/s13238-019-0610-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Accepted: 01/03/2019] [Indexed: 01/19/2023] Open
Abstract
RAP1 is a well-known telomere-binding protein, but its functions in human stem cells have remained unclear. Here we generated RAP1-deficient human embryonic stem cells (hESCs) by using CRISPR/Cas9 technique and obtained RAP1-deficient human mesenchymal stem cells (hMSCs) and neural stem cells (hNSCs) via directed differentiation. In both hMSCs and hNSCs, RAP1 not only negatively regulated telomere length but also acted as a transcriptional regulator of RELN by tuning the methylation status of its gene promoter. RAP1 deficiency enhanced self-renewal and delayed senescence in hMSCs, but not in hNSCs, suggesting complicated lineage-specific effects of RAP1 in adult stem cells. Altogether, these results demonstrate for the first time that RAP1 plays both telomeric and nontelomeric roles in regulating human stem cell homeostasis.
Collapse
Affiliation(s)
- Xing Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zunpeng Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoqian Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yiyuan Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaojuan He
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Shuhui Sun
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuai Ma
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ng Shyh-Chang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Feng Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qiang Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiaoqun Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.,University of Chinese Academy of Sciences, Beijing, 100049, China.,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Weiqi Zhang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China. .,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Guang-Hui Liu
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China. .,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China. .,Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, 510632, China. .,Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China. .,University of Chinese Academy of Sciences, Beijing, 100049, China. .,Institute for Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
38
|
Cayuela ML, Claes KBM, Ferreira MG, Henriques CM, van Eeden F, Varga M, Vierstraete J, Mione MC. The Zebrafish as an Emerging Model to Study DNA Damage in Aging, Cancer and Other Diseases. Front Cell Dev Biol 2019; 6:178. [PMID: 30687705 PMCID: PMC6335974 DOI: 10.3389/fcell.2018.00178] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 12/22/2018] [Indexed: 12/17/2022] Open
Abstract
Cancer is a disease of the elderly, and old age is its largest risk factor. With age, DNA damage accumulates continuously, increasing the chance of malignant transformation. The zebrafish has emerged as an important vertebrate model to study these processes. Key mechanisms such as DNA damage responses and cellular senescence can be studied in zebrafish throughout its life course. In addition, the zebrafish is becoming an important resource to study telomere biology in aging, regeneration and cancer. Here we review some of the tools and resources that zebrafish researchers have developed and discuss their potential use in the study of DNA damage, cancer and aging related diseases.
Collapse
Affiliation(s)
- Maria Luisa Cayuela
- Telomerase, Cancer and Aging Group, Surgery Unit, Instituto Murciano de Investigación Biosanitaria-Arrixaca, Murcia, Spain
| | | | | | - Catarina Martins Henriques
- Department of Oncology and Metabolism, Bateson Centre, University of Sheffield, Sheffield, United Kingdom
| | | | - Máté Varga
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
- MTA-SE Lendület Nephrogenetic Laboratory, Budapest, Hungary
| | | | | |
Collapse
|
39
|
Epigenetic Alterations: The Relation Between Occupational Exposure and Biological Effects in Humans. RNA TECHNOLOGIES 2019. [DOI: 10.1007/978-3-030-14792-1_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
40
|
Job S, Draskovic I, Burnichon N, Buffet A, Cros J, Lépine C, Venisse A, Robidel E, Verkarre V, Meatchi T, Sibony M, Amar L, Bertherat J, de Reyniès A, Londoño-Vallejo A, Favier J, Castro-Vega LJ, Gimenez-Roqueplo AP. Telomerase Activation and ATRX Mutations Are Independent Risk Factors for Metastatic Pheochromocytoma and Paraganglioma. Clin Cancer Res 2018; 25:760-770. [PMID: 30301828 DOI: 10.1158/1078-0432.ccr-18-0139] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/30/2018] [Accepted: 10/03/2018] [Indexed: 11/16/2022]
Abstract
PURPOSE Pheochromocytomas and paragangliomas (PPGLs) are rare neuroendocrine tumors. Whereas most PPGLs are benign, up to 20% may become metastatic with SDHB- and FH-mutated tumors showing the higher risk. We aimed at determining the contribution of immortalization mechanisms to metastatic progression.Experimental Design: Immortalization mechanisms were investigated in 200 tumors. To identify telomerase (+) tumors, we analyzed genomic alterations leading to transcriptional activation of TERT comprising promoter mutations, hypermethylation and gain copy number. To identify tumors that activated the alternative lengthening of telomere (ALT) mechanism, we combined analyses of telomere length by slot blot, telomere heterogeneity by telomere FISH, and ATRX mutations by next-generation sequencing. Univariate/multivariate and metastasis-free survival (MFS) and overall survival (OS) analyses were carried out for assessment of risk factors and clinical outcomes. RESULTS Only 37 of 200 (18.5%) tumors achieved immortalization. Telomerase activation occurred in 12 metastatic tumors and was prevalent in SDHB-mutated paragangliomas (P = 2.42e-09). ALT features were present in 25 tumors, mostly pheochromocytomas, regardless of metastatic status or molecular group (P = 0.169), yet ATRX mutations were found preferentially in SDHB/FH-mutated metastatic tumors (P = 0.0014). Telomerase activation and ATRX mutations were independent factors of poor prognosis: MFS (hazard ratio, 48.2 and 33.1; P = 6.50E-07 and 1.90E-07, respectively); OS (hazard ratio, 97.4 and 44.1; P = 4.30E-03 and 2.00E-03, respectively) and were associated with worse MFS and OS (log-rank tests P < 0.0001). CONCLUSIONS Assessment of telomerase activation and ATRX mutations could be used to identify metastatic PPGLs, particularly in tumors at high risk of progression.
Collapse
Affiliation(s)
- Sylvie Job
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Irena Draskovic
- CNRS, UMR3244, Institut Curie, PSL Research University, Paris, France.,Sorbonne Universités, UPMC, Univ Paris 06, Paris, France
| | - Nelly Burnichon
- INSERM, UMR970, Paris-Cardiovascular Research Center, Equipe Labellisée par la Ligue contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Genetics, Paris, France
| | - Alexandre Buffet
- INSERM, UMR970, Paris-Cardiovascular Research Center, Equipe Labellisée par la Ligue contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Jérôme Cros
- INSERM, UMR1149, Hôpital Beaujon, Department of Pathology, Clichy, France
| | - Charles Lépine
- Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Genetics, Paris, France
| | - Annabelle Venisse
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Genetics, Paris, France
| | - Estelle Robidel
- INSERM, UMR970, Paris-Cardiovascular Research Center, Equipe Labellisée par la Ligue contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Virginie Verkarre
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Pathology, Paris, France
| | - Tchao Meatchi
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Pathology, Paris, France
| | - Mathilde Sibony
- INSERM, U1016, Institut Cochin, Paris, France. 10 CNRS UMR8104, Paris, France.,Assistance Publique Hôpitaux de Paris, Hôpital Cochin, Department of Pathology, Paris, France
| | - Laurence Amar
- INSERM, UMR970, Paris-Cardiovascular Research Center, Equipe Labellisée par la Ligue contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Hypertension Unit, Paris, France
| | - Jérôme Bertherat
- INSERM, U1016, Institut Cochin, Paris, France. 10 CNRS UMR8104, Paris, France.,Rare Adrenal Cancer Network COMETE, Paris, France
| | - Aurélien de Reyniès
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France
| | - Arturo Londoño-Vallejo
- CNRS, UMR3244, Institut Curie, PSL Research University, Paris, France.,Sorbonne Universités, UPMC, Univ Paris 06, Paris, France
| | - Judith Favier
- INSERM, UMR970, Paris-Cardiovascular Research Center, Equipe Labellisée par la Ligue contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Luis Jaime Castro-Vega
- INSERM, UMR970, Paris-Cardiovascular Research Center, Equipe Labellisée par la Ligue contre le Cancer, Paris, France. .,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France
| | - Anne-Paule Gimenez-Roqueplo
- INSERM, UMR970, Paris-Cardiovascular Research Center, Equipe Labellisée par la Ligue contre le Cancer, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Faculté de Médecine, Paris, France.,Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Genetics, Paris, France.,Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre Le Cancer, Paris, France.,CNRS, UMR3244, Institut Curie, PSL Research University, Paris, France
| |
Collapse
|
41
|
Abstract
Telomere length measurement is increasingly recognized as a clinical gauge for age-related disease risk. There are several methods for studying blood telomere length (BTL) as a clinical biomarker. The first is an observational study approach, which directly measures telomere lengths using either cross-sectional or longitudinal patient cohorts and compares them to a population of age- and sex-matched individuals. These direct traceable measurements can be considered reflective of an individual's current health or disease state. Escalating interest in personalized medicine, access to high-throughput genotyping and resulting acquisition of large volumes of genetic data corroborates the second method, Mendelian randomization (MR). MR employs telomere length-associated genetic variants to indicate predisposition to disease risk based on the genomic composition of the individual. When assessed from cells in the bloodstream, telomeres can show variation from their genetically predisposed lengths due to environmental-induced changes. These alterations in telomere length act as an indicator of cellular health, which, in turn, can provide disease risk status. Overall, BTL measurement is a dynamic marker of biological health and well-being that together with genetically defined telomere lengths can provide insights into improved healthcare for the individual.
Collapse
|
42
|
M'kacher R, Cuceu C, Al Jawhari M, Morat L, Frenzel M, Shim G, Lenain A, Hempel WM, Junker S, Girinsky T, Colicchio B, Dieterlen A, Heidingsfelder L, Borie C, Oudrhiri N, Bennaceur-Griscelli A, Moralès O, Renaud S, Van de Wyngaert Z, Jeandidier E, Delhem N, Carde P. The Transition between Telomerase and ALT Mechanisms in Hodgkin Lymphoma and Its Predictive Value in Clinical Outcomes. Cancers (Basel) 2018; 10:E169. [PMID: 29848986 PMCID: PMC6025489 DOI: 10.3390/cancers10060169] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/18/2018] [Accepted: 05/25/2018] [Indexed: 11/16/2022] Open
Abstract
Background: We analyzed telomere maintenance mechanisms (TMMs) in lymph node samples from HL patients treated with standard therapy. The TMMs correlated with clinical outcomes of patients. Materials and Methods: Lymph node biopsies obtained from 38 HL patients and 24 patients with lymphadenitis were included in this study. Seven HL cell lines were used as in vitro models. Telomerase activity (TA) was assessed by TRAP assay and verified through hTERT immunofluorescence expression; alternative telomere lengthening (ALT) was also assessed, along with EBV status. Results: Both TA and ALT mechanisms were present in HL lymph nodes. Our findings were reproduced in HL cell lines. The highest levels of TA were expressed in CD30-/CD15- cells. Small cells were identified with ALT and TA. Hodgkin and Reed Sternberg cells contained high levels of PML bodies, but had very low hTERT expression. There was a significant correlation between overall survival (p < 10-3), event-free survival (p < 10-4), and freedom from progression (p < 10-3) and the presence of an ALT profile in lymph nodes of EBV+ patients. Conclusion: The presence of both types of TMMs in HL lymph nodes and in HL cell lines has not previously been reported. TMMs correlate with the treatment outcome of EBV+ HL patients.
Collapse
Affiliation(s)
- Radhia M'kacher
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
- Cell Environment, DNA Damages R&D, Oncology Section, 75020 Paris, France.
| | - Corina Cuceu
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - Mustafa Al Jawhari
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - Luc Morat
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - Monika Frenzel
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - Grace Shim
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - Aude Lenain
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - William M Hempel
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - Steffen Junker
- Institute of Biomedicine, University of Aarhus, DK-8000 Aarhus C, Denmark.
| | - Theodore Girinsky
- Department of Radiation Therapy, Gustave Roussy Cancer Campus, 94808 Villejuif, France.
| | - Bruno Colicchio
- IRIMAS, Institut de Recherche en Informatique, Mathématiques, Automatique et Signal, Université de Haute-Alsace, 68093 Mulhouse, France.
| | - Alain Dieterlen
- IRIMAS, Institut de Recherche en Informatique, Mathématiques, Automatique et Signal, Université de Haute-Alsace, 68093 Mulhouse, France.
| | | | - Claire Borie
- Université Paris Sud, Service d'hématologie moléculaire et cytogénétique Paul brousse CHU paris Sud, Inserm UMRS935, 94800 Villejuif, France.
| | - Noufissa Oudrhiri
- Université Paris Sud, Service d'hématologie moléculaire et cytogénétique Paul brousse CHU paris Sud, Inserm UMRS935, 94800 Villejuif, France.
| | - Annelise Bennaceur-Griscelli
- Université Paris Sud, Service d'hématologie moléculaire et cytogénétique Paul brousse CHU paris Sud, Inserm UMRS935, 94800 Villejuif, France.
| | - Olivier Moralès
- CNRS, Institut Pasteur de Lille, UMR 8161-Immunoregulation of Virus-induced Cancers Team, F-59000 Lille, France.
| | - Sarah Renaud
- CNRS, Institut Pasteur de Lille, UMR 8161-Immunoregulation of Virus-induced Cancers Team, F-59000 Lille, France.
| | - Zoé Van de Wyngaert
- CHRU Lille Service des Maladies du Sang, Hopital Huriez, 59000 Lille, France.
| | - Eric Jeandidier
- Service de génétique, Groupe hospitalier de la région de Mulhouse Sud-Alsace, 68093 Mulhouse, France.
| | - Nadira Delhem
- CNRS, Institut Pasteur de Lille, UMR 8161-Immunoregulation of Virus-induced Cancers Team, F-59000 Lille, France.
| | - Patrice Carde
- Department of Medicine, Gustave Roussy Cancer Campus, 94808 Villejuif, France.
| |
Collapse
|
43
|
Abstract
Studies of rare and common illnesses have led to remarkable progress in the understanding of the role of telomeres (nucleoprotein complexes at chromosome ends essential for chromosomal integrity) in human disease. Telomere biology disorders encompass a growing spectrum of conditions caused by rare pathogenic germline variants in genes encoding essential aspects of telomere function. Dyskeratosis congenita, a disorder at the severe end of this spectrum, typically presents in childhood with the classic triad of abnormal skin pigmentation, nail dystrophy, and oral leukoplakia, accompanied by a very high risk of bone marrow failure, cancer, pulmonary fibrosis, and other medical problems. In contrast, the less severe end of the telomere biology disorder spectrum consists of middle-age or older adults with just one feature typically seen in dyskeratosis congenita, such as pulmonary fibrosis or bone marrow failure. In the common disease realm, large-scale molecular epidemiology studies have discovered novel associations between illnesses, such as cancer, heart disease, and mental health, and both telomere length and common genetic variants in telomere biology genes. This review highlights recent findings of telomere biology in human disease from both the rare and common disease perspectives. Multi-disciplinary collaborations between clinicians, basic scientists, and epidemiologist are essential as we seek to incorporate new telomere biology discoveries to improve health outcomes.
Collapse
Affiliation(s)
- Sharon A. Savage
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
44
|
Bonfigli AR, Spazzafumo L, Prattichizzo F, Bonafè M, Mensà E, Micolucci L, Giuliani A, Fabbietti P, Testa R, Boemi M, Lattanzio F, Olivieri F. Leukocyte telomere length and mortality risk in patients with type 2 diabetes. Oncotarget 2018; 7:50835-50844. [PMID: 27437767 PMCID: PMC5239440 DOI: 10.18632/oncotarget.10615] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 07/01/2016] [Indexed: 12/22/2022] Open
Abstract
Leukocyte telomere length (LTL) shortening is found in a number of age-related diseases, including type 2 diabetes (T2DM). In this study its possible association with mortality was analyzed in a sample of 568 T2DM patients (mean age 65.9 ± 9 years), who were followed for a median of 10.2 years (interquartile range 2.2). A number of demographic, laboratory and clinical parameters determined at baseline were evaluated as mortality risk factors. LTL was measured by quantitative real-time PCR and reported as T/S (telomere-to-single copy gene ratio). Age, gender, creatinine, diabetes duration at baseline, and LTL were significantly different between T2DM patients who were dead and alive at follow-up. In the Cox regression analysis adjusted for the confounding variables, shorter LTL, older age, and longer disease duration significantly increased the risk of all-cause mortality (HR = 3.45, 95%CI 1.02-12.5, p = 0.004). Kaplan-Maier analysis also found a different cumulative mortality risk for patients having an LTL shorter than the median (T/S ≤0.04) and disease duration longer than the median (>10 years) (log-rank = 11.02, p = 0.011). Time-dependent mortality risk stratification showed that T2DM duration and LTL combined was a fairly good predictor of mortality over the first 76 months of follow-up. In conclusion, LTL combined with clinical parameters can provide additive prognostic information on mortality risk in T2DM patients.
Collapse
Affiliation(s)
| | - Liana Spazzafumo
- Center of Biostatistics, INRCA-IRCCS National Institute, Ancona, Italy
| | | | - Massimiliano Bonafè
- Department of Experimental, Diagnostic and Specialty Medicine, DIMES, University of Bologna, Bologna, Italy
| | - Emanuela Mensà
- Center of Clinical Pathology and Innovative Therapy, National Institute INRCA-IRCCS, Ancona, Italy
| | - Luigina Micolucci
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Angelica Giuliani
- Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| | - Paolo Fabbietti
- Center of Biostatistics, INRCA-IRCCS National Institute, Ancona, Italy
| | - Roberto Testa
- Experimental Models in Clinical Pathology, INRCA-IRCCS National Institute, Ancona, Italy
| | - Massimo Boemi
- Metabolic Diseases and Diabetology Unit, INRCA-IRCCS National Institute, Ancona, Italy
| | | | - Fabiola Olivieri
- Center of Clinical Pathology and Innovative Therapy, National Institute INRCA-IRCCS, Ancona, Italy.,Department of Clinical and Molecular Sciences, DISCLIMO, Università Politecnica delle Marche, Ancona, Italy
| |
Collapse
|
45
|
Dogan F, Biray Avci C. Correlation between telomerase and mTOR pathway in cancer stem cells. Gene 2018; 641:235-239. [DOI: 10.1016/j.gene.2017.09.072] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 09/14/2017] [Accepted: 09/26/2017] [Indexed: 12/17/2022]
|
46
|
Yu P, Shen X, Yang W, Zhang Y, Liu C, Huang T. ZEB1 stimulates breast cancer growth by up-regulating hTERT expression. Biochem Biophys Res Commun 2017; 495:2505-2511. [PMID: 29288666 DOI: 10.1016/j.bbrc.2017.12.139] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 12/22/2017] [Indexed: 02/06/2023]
Abstract
Dysfunctional cell proliferation and death are the foundation of the malignant biological characteristics of cancers. In this study, we discovered that ZEB1 was positively correlated with hTERT in breast invasive ductal carcinoma samples at both the mRNA and protein levels. Further, our in vitro study in breast cancer cell lines confirmed that ZEB1 regulates hTERT expression at the mRNA and protein levels; thus, hTERT promotes or inhibits telomerase activity, and telomere length is either protected or reduced. Finally, we verified that ZEB1, which mostly functions as a transcriptional repressor, can recruit the co-activator YAP to enhance the transcriptional activation of hTERT. Fascinatingly, instead of acting on E-boxes, the ZEB1/YAP complex tends to function as a transcriptional activator by binding with sequences potentially located in the hTERT promoter. Consequently, our research revealed a new ZEB1-hTERT signaling pathway involved in cell proliferation regulation that has never before been illuminated in breast cancer.
Collapse
Affiliation(s)
- Pan Yu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Xi Shen
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Wen Yang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yunke Zhang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunping Liu
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Tao Huang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
47
|
Faleiro I, Apolónio JD, Price AJ, De Mello RA, Roberto VP, Tabori U, Castelo-Branco P. The TERT hypermethylated oncologic region predicts recurrence and survival in pancreatic cancer. Future Oncol 2017; 13:2045-2051. [DOI: 10.2217/fon-2017-0167] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: We explore the biomarker potential of the TERT hypermethylated oncologic region (THOR) in pancreatic cancer. Materials & methods: We assessed the methylation status of THOR using the cancer genome atlas data on the cohort of pancreatic cancer (n = 193 patients). Results: THOR was significantly hypermethylated in pancreatic tumor tissue when compared with the normal tissue used as control (p < 0.0001). Also, THOR hypermethylation could distinguish early stage I disease from normal tissue and was associated with worse prognosis. Discussion: We found that THOR is hypermethylated in pancreatic tumor tissue when compared with normal tissue and that THOR methylation correlates with TERT expression in tumor samples. Conclusion: Our preliminary findings support the diagnostic and prognostic values of THOR in pancreatic cancer.
Collapse
Affiliation(s)
- Inês Faleiro
- Department of Biomedical Sciences & Medicine, University of Algarve, Campus de Gambelas, Edifício 2, 8005–139 Faro, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, 8005–139 Faro, Portugal
- Algarve Biomedical Center, University of Algarve, Campus de Gambelas, 8005–139, Faro, Portugal
| | - Joana Dias Apolónio
- Department of Biomedical Sciences & Medicine, University of Algarve, Campus de Gambelas, Edifício 2, 8005–139 Faro, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, 8005–139 Faro, Portugal
- Algarve Biomedical Center, University of Algarve, Campus de Gambelas, 8005–139, Faro, Portugal
| | - Aryeh J Price
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Ramon Andrade De Mello
- Department of Biomedical Sciences & Medicine, University of Algarve, Campus de Gambelas, Edifício 2, 8005–139 Faro, Portugal
- Algarve Biomedical Center, University of Algarve, Campus de Gambelas, 8005–139, Faro, Portugal
| | - Vânia Palma Roberto
- Department of Biomedical Sciences & Medicine, University of Algarve, Campus de Gambelas, Edifício 2, 8005–139 Faro, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, 8005–139 Faro, Portugal
- Algarve Biomedical Center, University of Algarve, Campus de Gambelas, 8005–139, Faro, Portugal
| | - Uri Tabori
- Arthur & Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Pedro Castelo-Branco
- Department of Biomedical Sciences & Medicine, University of Algarve, Campus de Gambelas, Edifício 2, 8005–139 Faro, Portugal
- Centre for Biomedical Research (CBMR), University of Algarve, 8005–139 Faro, Portugal
- Algarve Biomedical Center, University of Algarve, Campus de Gambelas, 8005–139, Faro, Portugal
| |
Collapse
|
48
|
Daniloski Z, Smith S. Loss of Tumor Suppressor STAG2 Promotes Telomere Recombination and Extends the Replicative Lifespan of Normal Human Cells. Cancer Res 2017; 77:5530-5542. [PMID: 28819029 DOI: 10.1158/0008-5472.can-17-1260] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/11/2017] [Accepted: 08/08/2017] [Indexed: 01/19/2023]
Abstract
Sister chromatids are held together by cohesin, a tripartite ring with a peripheral SA1/2 subunit, where SA1 is required for telomere cohesion and SA2 for centromere cohesion. The STAG2 gene encoding SA2 is often inactivated in human cancer, but not in in a manner associated with aneuploidy. Thus, how these tumors maintain chromosomal cohesion and how STAG2 loss contributes to tumorigenesis remain open questions. Here we show that, despite a loss in centromere cohesion, sister chromatids in STAG2 mutant tumor cells maintain cohesion in mitosis at chromosome arms and telomeres. Telomere maintenance in STAG2 mutant tumor cells occurred by either telomere recombination or telomerase activation mechanisms. Notably, these cells were refractory to telomerase inhibitors, indicating recombination can provide an alternative means of telomere maintenance. STAG2 silencing in normal human cells that lack telomerase led to increased recombination at telomeres, delayed telomere shortening, and postponed senescence onset. Insofar as telomere shortening and replicative senescence prevent genomic instability and cancer by limiting the number of cell divisions, our findings suggest that extending the lifespan of normal human cells due to inactivation of STAG2 could promote tumorigenesis by extending the period during which tumor-driving mutations occur. Cancer Res; 77(20); 5530-42. ©2017 AACR.
Collapse
Affiliation(s)
- Zharko Daniloski
- Department of Pathology, Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, New York
| | - Susan Smith
- Department of Pathology, Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, New York.
| |
Collapse
|
49
|
Pelizzo G, Avanzini MA, Folini M, Bussani R, Mantelli M, Croce S, Acquafredda G, Travaglino P, Cimino-Reale G, Boni M, Dambruoso I, Calcaterra V. CPAM type 2-derived mesenchymal stem cells: Malignancy risk study in a 14-month-old boy. Pediatr Pulmonol 2017; 52:990-999. [PMID: 28493304 DOI: 10.1002/ppul.23734] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/20/2017] [Indexed: 12/19/2022]
Abstract
INTRODUCTION The association between congenital pulmonary airway malformations (CPAM) and malignancy is reported in the literature. Interactions between the tumor, immune, and mesenchymal stromal/stem cells (MSCs) have been recognized as crucial for understanding tumorigenesis. We characterized MSCs isolated from CPAM lesions in order to define potential malignancy risks. METHODS CPAM II pulmonary tissue was used for MSC expansion; a "healthy" lung section from the same child was used as a comparator. Morphology, immunophenotype, differentiation and immunological capacity, proliferative growth, gene signature telomerase activity, and in vivo tumorigenicity in nude mice were evaluated. RESULTS MSCs were successfully isolated and propagated from CPAM tissue. CPAM-MSCs presented the typical MSC morphology and phenotype, while exhibiting high proliferative capacity, reaching confluence at a median time of 5 days as well as differentiation capabilities. CPAM-MSCs at early passages were not neoplastic and chromosomally normal, even though unbalanced chromosomal rearrangements were noted by molecular karyotype. CONCLUSIONS CPAM-MSCs exhibited specific features similar to tumor derived MSCs. Whilst there was no evidence of malignant transformation in the cystic tissue, our results provide evidence that this abnormal tissue has malignant potential. MSCs are considered important players in the tumor microenvironment and they have been closely linked to regulation of tumor survival, growth, and progression. Thus, early lesion resection also in asymptomatic patients might be indicated to exclude that the microenvironment may be potentially permissive to cancer development.
Collapse
Affiliation(s)
- Gloria Pelizzo
- Pediatric Surgery Unit, Children's Hospital, Istituto Mediterraneo di Eccellenza Pediatrica, Palermo, Italy
| | - Maria A Avanzini
- Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Marco Folini
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Rossana Bussani
- Institute of Pathologic Anatomy, University of Trieste, Trieste, Italy
| | - Melissa Mantelli
- Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Stefania Croce
- Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Gloria Acquafredda
- Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Paola Travaglino
- Immunology and Transplantation Laboratory/Cell Factory/Pediatric Hematology/Oncology Department, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Graziella Cimino-Reale
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Marina Boni
- Hematology Department, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Irene Dambruoso
- Hematology Department, Fondazione IRCCS Policlinico S. Matteo, Pavia, Italy
| | - Valeria Calcaterra
- Pediatric Unit, Department of Internal Medicine, University of Pavia and Department of Maternal and Children's Health, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
50
|
Naderlinger E, Holzmann K. Epigenetic Regulation of Telomere Maintenance for Therapeutic Interventions in Gliomas. Genes (Basel) 2017; 8:E145. [PMID: 28513547 PMCID: PMC5448019 DOI: 10.3390/genes8050145] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/08/2017] [Accepted: 05/12/2017] [Indexed: 02/07/2023] Open
Abstract
High-grade astrocytoma of WHO grade 4 termed glioblastoma multiforme (GBM) is a common human brain tumor with poor patient outcome. Astrocytoma demonstrates two known telomere maintenance mechanisms (TMMs) based on telomerase activity (TA) and on alternative lengthening of telomeres (ALT). ALT is associated with lower tumor grades and better outcome. In contrast to ALT, regulation of TA in tumors by direct mutation and epigenetic activation of the hTERT promoter is well established. Here, we summarize the genetic background of TMMs in non-malignant cells and in cancer, in addition to clinical and pathological features of gliomas. Furthermore, we present new evidence for epigenetic mechanisms (EMs) involved in regulation of ALT and TA with special emphasis on human diffuse gliomas as potential therapeutic drug targets. We discuss the role of TMM associated telomeric chromatin factors such as DNA and histone modifying enzymes and non-coding RNAs including microRNAs and long telomeric TERRA transcripts.
Collapse
Affiliation(s)
- Elisabeth Naderlinger
- Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, Vienna 1090, Austria.
| | - Klaus Holzmann
- Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Borschkegasse 8a, Vienna 1090, Austria.
| |
Collapse
|