1
|
Baylie T, Jemal M, Baye G, Getinet M, Amare GA, Adugna A, Abebaw D, Hibstu Z, Tegegne BA, Gugsa E, Adane T, Getie G, Ashenef B, Sinamaw D. The role of telomere and telomerase in cancer and novel therapeutic target: narrative review. Front Oncol 2025; 15:1542930. [PMID: 40151802 PMCID: PMC11947687 DOI: 10.3389/fonc.2025.1542930] [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: 12/10/2024] [Accepted: 01/27/2025] [Indexed: 03/29/2025] Open
Abstract
Telomeres are dynamic complexes at the ends of chromosomes that are made up of protective proteins and tandem repeating DNA sequences. In the large majority of cancer cells, telomere length is maintained by telomerase, an enzyme that elongates telomeres. Telomerase activation is seen in the majority of cancer, which permits uncontrol cell proliferation. About 90% of human malignancies show telomere dysfunction and telomerase reactivation; as a result, telomerase activation plays a special role as a practically universal stage on the way to malignancy. This review understands the structural and functional of telomere and telomerase, mechanisms of telomerase activation in oncogenesis, biomarkers and therapeutic targets. Therapeutic strategies targeting telomerase, including antisense oligonucleotides, G-quadruplex stabilizers, immunotherapy, small-molecule inhibitors, gene therapy, Telomerase-Responsive Drug Release System, have shown promise in preclinical and clinical settings. Advances in telomere biology not only illuminate the complex interplay between telomeres, telomerase, and cancer progression but also open avenues for innovative, targeted cancer therapies.
Collapse
Affiliation(s)
- Temesgen Baylie
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Mohammed Jemal
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Gelagay Baye
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Mamaru Getinet
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Gashaw Azanaw Amare
- Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Adane Adugna
- Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Desalegn Abebaw
- Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Zigale Hibstu
- Medical Laboratory Science, College of Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Bantayehu Addis Tegegne
- Department of Pharmacy, College of Medicine and Health Sciences, Debre Markos University, Debre Markos, Ethiopia
| | - Endalkachew Gugsa
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Tadegew Adane
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Gedefaw Getie
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Baye Ashenef
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| | - Deresse Sinamaw
- Department of Biomedical Sciences, School of Medicine, Debre Markos University, Debre Markos, Ethiopia
| |
Collapse
|
2
|
Yakovenko I, Mihai IS, Selinger M, Rosenbaum W, Dernstedt A, Gröning R, Trygg J, Carroll L, Forsell M, Henriksson J. Telomemore enables single-cell analysis of cell cycle and chromatin condensation. Nucleic Acids Res 2025; 53:gkaf031. [PMID: 39878215 PMCID: PMC11775621 DOI: 10.1093/nar/gkaf031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/15/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
Single-cell RNA-seq methods can be used to delineate cell types and states at unprecedented resolution but do little to explain why certain genes are expressed. Single-cell ATAC-seq and multiome (ATAC + RNA) have emerged to give a complementary view of the cell state. It is however unclear what additional information can be extracted from ATAC-seq data besides transcription factor binding sites. Here, we show that ATAC-seq telomere-like reads counter-inituively cannot be used to infer telomere length, as they mostly originate from the subtelomere, but can be used as a biomarker for chromatin condensation. Using long-read sequencing, we further show that modern hyperactive Tn5 does not duplicate 9 bp of its target sequence, contrary to common belief. We provide a new tool, Telomemore, which can quantify nonaligning subtelomeric reads. By analyzing several public datasets and generating new multiome fibroblast and B-cell atlases, we show how this new readout can aid single-cell data interpretation. We show how drivers of condensation processes can be inferred, and how it complements common RNA-seq-based cell cycle inference, which fails for monocytes. Telomemore-based analysis of the condensation state is thus a valuable complement to the single-cell analysis toolbox.
Collapse
Affiliation(s)
- Iryna Yakovenko
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Universitetstorget 4, 901 87, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
| | - Ionut Sebastian Mihai
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Universitetstorget 4, 901 87, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Industrial Doctoral School, Umeå University, Umeå, Sweden
| | - Martin Selinger
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Universitetstorget 4, 901 87, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Department of Chemistry, Faculty of Science, University of South Bohemia, Ceske Budejovice 37005, Czech Republic
| | - William Rosenbaum
- Department of Molecular Biology, Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
| | - Andy Dernstedt
- Department of Clinical Microbiology, Umeå University, Biomedicinbyggnaden 6M, Umeå universitetssjukhus, 901 87, Umeå, Sweden
| | - Remigius Gröning
- Department of Clinical Microbiology, Umeå University, Biomedicinbyggnaden 6M, Umeå universitetssjukhus, 901 87, Umeå, Sweden
| | - Johan Trygg
- Department of Chemistry, Umeå University, Linnaeus väg 10, Umeå universitet, 901 87, Umeå, Sweden
- Sartorius Corporate Research, Östra Strandgatan 24, 903 33, Umeå, Sweden
| | - Laura Carroll
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Universitetstorget 4, 901 87, Umeå, Sweden
- Department of Clinical Microbiology, Umeå University, Biomedicinbyggnaden 6M, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Integrated Science Lab (IceLab), Umeå University, Naturvetarhuset, Universitetsvägen, 901 87, Umeå, Sweden
| | - Mattias Forsell
- Department of Clinical Microbiology, Umeå University, Biomedicinbyggnaden 6M, Umeå universitetssjukhus, 901 87, Umeå, Sweden
| | - Johan Henriksson
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Umeå Centre for Microbial Research (UCMR), Universitetstorget 4, 901 87, Umeå, Sweden
- Department of Molecular Biology, Umeå University, Biomedicinbyggnaden 6K och 6L, Umeå universitetssjukhus, 901 87, Umeå, Sweden
- Integrated Science Lab (IceLab), Umeå University, Naturvetarhuset, Universitetsvägen, 901 87, Umeå, Sweden
| |
Collapse
|
3
|
Loukopoulou C, Nikolouzakis T, Koliarakis I, Vakonaki E, Tsiaoussis J. Telomere Length and Telomerase Activity as Potential Biomarkers for Gastrointestinal Cancer. Cancers (Basel) 2024; 16:3370. [PMID: 39409990 PMCID: PMC11482595 DOI: 10.3390/cancers16193370] [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: 08/13/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Gastrointestinal (GI) cancers, such as colorectal and gastric cancers, pose significant global health challenges due to their high rates of incidence and mortality. Even with advancements in treatment and early detection, many patients still face poor outcomes, highlighting the critical need for new biomarkers and therapeutic targets. Telomere length (TL) and telomerase activity (TA) have gained attention in this context. Telomeres, protective nucleotide sequences at chromosome ends, shorten with each cell division, leading to cellular aging. Telomerase, a ribonucleoprotein enzyme, counteracts this shortening by adding telomeric repeats, a process tightly regulated in normal cells but often dysregulated in cancer. This review critically evaluates the role of TL and TA in the pathogenesis of GI cancers, examining their potential as diagnostic, prognostic, and predictive biomarkers. It explores how alterations in telomere biology contribute to the initiation and progression of GI tumors and assesses the therapeutic implications of targeting telomerase. By integrating findings from diverse studies, this review aims to elucidate the intricate relationship between telomere dynamics and gastrointestinal carcinogenesis, offering insights into how TL and TA could be leveraged to enhance the early detection, treatment, and prognosis of GI cancers.
Collapse
Affiliation(s)
- Christina Loukopoulou
- Department of Anatomy, School of Medicine, University of Crete, 71003 Heraklion, Greece; (C.L.); (T.N.); (I.K.)
| | - Taxiarchis Nikolouzakis
- Department of Anatomy, School of Medicine, University of Crete, 71003 Heraklion, Greece; (C.L.); (T.N.); (I.K.)
| | - Ioannis Koliarakis
- Department of Anatomy, School of Medicine, University of Crete, 71003 Heraklion, Greece; (C.L.); (T.N.); (I.K.)
| | - Elena Vakonaki
- Department of Forensic Sciences and Toxicology, School of Medicine, University of Crete, 71003 Heraklion, Greece;
| | - John Tsiaoussis
- Department of Anatomy, School of Medicine, University of Crete, 71003 Heraklion, Greece; (C.L.); (T.N.); (I.K.)
| |
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
|
Atroosh F, Al-Habori M, Al-Eryani E, Saif-Ali R. Impact of khat (Catha edulis) and oral contraceptive use on telomerase levels and tumor suppressor genes p53 and p21 in normal subjects and breast cancer patients. Sci Rep 2024; 14:16365. [PMID: 39013992 PMCID: PMC11252306 DOI: 10.1038/s41598-024-67355-5] [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: 04/20/2024] [Accepted: 07/10/2024] [Indexed: 07/18/2024] Open
Abstract
This study aimed to evaluate the effects of oral contraceptive (OC) use, khat chewing, and their combined effect on telomerase level and tumor suppressor genes, p53 and p21 in breast cancer (BC) patients and normal volunteers. 140 Yemeni women aged 25-40 years old enrolled, 60 newly diagnosed pretreated BC patients, and 80 control subjects. Venous blood (5 ml) was collected and the results showed BC patients to have significantly raised levels of telomerase, p53, and p21 compared to the control group. The use of OCs significantly raised telomerase in control group with no effect in BC patients; whereas p53 and p21 were significantly increased in BC patients. On the other hand, khat chewing significantly increased p53 in controls and BC patients, whereas p21 was significantly raised in BC patients. The combined use of OCs and khat chewing significantly increased telomerase and p53 in control group, and significantly increased p53 and p21 in BC patients. Telomerase was shown to be a risk factor (OR 4.4) for BC, and the use of OCs was a high-risk factor for increasing telomerase (OR 27.8) in normal subjects. In contrast, khat chewing was shown to be protective (OR 0.142), and the combined use of OCs and khat chewing decreased the risk factor of telomerase from OR 27.8 to 2.1.
Collapse
Affiliation(s)
- Fairooz Atroosh
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, Sana'a University, Sana'a, Republic of Yemen
| | - Molham Al-Habori
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, Sana'a University, Sana'a, Republic of Yemen.
| | - Ekram Al-Eryani
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, Sana'a University, Sana'a, Republic of Yemen
| | - Riyadh Saif-Ali
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, Sana'a University, Sana'a, Republic of Yemen
| |
Collapse
|
6
|
Hakobyan M, Binder H, Arakelyan A. Pan-cancer analysis of telomere maintenance mechanisms. J Biol Chem 2024; 300:107392. [PMID: 38763334 PMCID: PMC11225560 DOI: 10.1016/j.jbc.2024.107392] [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: 01/19/2024] [Revised: 04/28/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024] Open
Abstract
Telomeres, protective caps at chromosome ends, maintain genomic stability and control cell lifespan. Dysregulated telomere maintenance mechanisms (TMMs) are cancer hallmarks, enabling unchecked cell proliferation. We conducted a pan-cancer evaluation of TMM using RNA sequencing data from The Cancer Genome Atlas for 33 different cancer types and analyzed the activities of telomerase-dependent (TEL) and alternative lengthening of telomeres (ALT) TMM pathways in detail. To further characterize the TMM profiles, we categorized the tumors based on their ALT and TEL TMM pathway activities into five major phenotypes: ALT high TEL low, ALT low TEL low, ALT middle TEL middle, ALT high TEL high, and ALT low TEL high. These phenotypes refer to variations in telomere maintenance strategies, shedding light on the heterogeneous nature of telomere regulation in cancer. Moreover, we investigated the clinical implications of TMM phenotypes by examining their associations with clinical characteristics and patient outcomes. Specific TMM profiles were linked to specific survival patterns, emphasizing the potential of TMM profiling as a prognostic indicator and aiding in personalized cancer treatment strategies. Gene ontology analysis of the TMM phenotypes unveiled enriched biological processes associated with cell cycle regulation (both TEL and ALT), DNA replication (TEL), and chromosome dynamics (ALT) showing that telomere maintenance is tightly intertwined with cellular processes governing proliferation and genomic stability. Overall, our study provides an overview of the complexity of transcriptional regulation of telomere maintenance mechanisms in cancer.
Collapse
Affiliation(s)
- Meline Hakobyan
- Bioinformatics Group, Institute of Molecular Biology NAS RA, Yerevan, Armenia.
| | - Hans Binder
- Interdisciplinary Centre for Bioinformatics, University of Leipzig, Leipzig, Germany; Armenian Bioinformatics Institute, Yerevan, Armenia
| | - Arsen Arakelyan
- Bioinformatics Group, Institute of Molecular Biology NAS RA, Yerevan, Armenia
| |
Collapse
|
7
|
Córdoba-Lanús E, Montuenga LM, Domínguez-de-Barros A, Oliva A, Mayato D, Remírez-Sanz A, Gonzalvo F, Celli B, Zulueta JJ, Casanova C. Oxidative Damage and Telomere Length as Markers of Lung Cancer Development among Chronic Obstructive Pulmonary Disease (COPD) Smokers. Antioxidants (Basel) 2024; 13:156. [PMID: 38397754 PMCID: PMC10886051 DOI: 10.3390/antiox13020156] [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: 12/29/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Lung cancer (LC) constitutes an important cause of death among patients with Chronic Obstructive Pulmonary Disease (COPD). Both diseases may share pathobiological mechanisms related to oxidative damage and cellular senescence. In this study, the potential value of leucocyte telomere length, a hallmark of aging, and 8-OHdG concentrations, indicative of oxidative DNA damage, as risk biomarkers of LC was evaluated in COPD patients three years prior to LC diagnosis. Relative telomere length measured using qPCR and serum levels of 8-OHdG were determined at the baseline in 99 COPD smokers (33 with LC and 66 age-matched COPD without LC as controls). Of these, 21 COPD with LC and 42 controls had the biomarkers measured 3 years before. Single nucleotide variants (SNVs) in TERT, RTEL, and NAF1 genes were also determined. COPD cases were evaluated, which showed greater telomere length (p < 0.001) and increased serum 8-OHdG levels (p = 0.004) three years prior to LC diagnosis compared to the controls. This relationship was confirmed at the time of LC diagnosis. No significant association was found between the studied SNVs in cases vs. controls. In conclusion, this preliminary study shows that longer leucocyte telomere length and increased 8-OHdG serum levels can be useful as early biomarkers of the risk for future lung cancer development among COPD patients.
Collapse
Affiliation(s)
- Elizabeth Córdoba-Lanús
- Department of Internal Medicine, Dermatology and Psychiatry, University of La Laguna, 38296 San Cristóbal de La Laguna, Spain;
- Instituto de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), 38029 San Cristóbal de La Laguna, Spain; (A.D.-d.-B.); (A.O.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Luis M. Montuenga
- Department of Pathology, Anatomy and Physiology, School of Medicine and Sciences, University of Navarra, 31008 Pamplona, Spain;
- CIMA—Centro de Investigación Médica Aplicada, University of Navarra, 31008 Pamplona, Spain;
- Navarra Institute for Health Research (IdISNA), 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Angélica Domínguez-de-Barros
- Instituto de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), 38029 San Cristóbal de La Laguna, Spain; (A.D.-d.-B.); (A.O.)
| | - Alexis Oliva
- Instituto de Enfermedades Tropicales y Salud Pública de Canarias (IUETSPC), 38029 San Cristóbal de La Laguna, Spain; (A.D.-d.-B.); (A.O.)
- Department of Pharmaceutical Technology, University of La Laguna, 38296 San Cristóbal de La Laguna, Spain
| | - Delia Mayato
- Pulmonary Division, Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain; (D.M.); (F.G.)
| | - Ana Remírez-Sanz
- CIMA—Centro de Investigación Médica Aplicada, University of Navarra, 31008 Pamplona, Spain;
| | - Francisca Gonzalvo
- Pulmonary Division, Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain; (D.M.); (F.G.)
| | - Bartolomé Celli
- Pulmonary Critical Care Medicine Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Javier J. Zulueta
- Pulmonary, Critical Care and Sleep Medicine Division, Mount Sinai Morningside Hospital, Icahn School of Medicine, New York, NY 10025, USA;
| | - Ciro Casanova
- Department of Internal Medicine, Dermatology and Psychiatry, University of La Laguna, 38296 San Cristóbal de La Laguna, Spain;
- Pulmonary Division, Research Unit, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain; (D.M.); (F.G.)
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
8
|
Belić M, Sopić M, Roksandić-Milenković M, Ćeriman V, Guzonijić A, Vukašinović A, Ostanek B, Dimić N, Jovanović D, Kotur-Stevuljević J. Correlation of Short Leukocyte Telomeres and Oxidative Stress with the Presence and Severity of Lung Cancer Explored by Principal Component Analysis. Folia Biol (Praha) 2023; 69:59-68. [PMID: 38063002 DOI: 10.14712/fb2023069020059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Lung cancer (LC) is the second most common malignancy and leading cause of cancer death. The potential "culprit" for local and systemic telomere shortening in LC patients is oxidative stress. We investigated the correlation between the peripheral blood leukocyte (PBL) telomere length (TL) and the presence/severity of LC and oxidative stress, and its usefulness as LC diagnostic marker. PBL TL was measured in 89 LC patients and 83 healthy subjects using the modified Cawthon RTq-PCR method. The relative PBL TL, found to be a potential diagnostic marker for LC with very good accuracy (P < 0.001), was significantly shorter in patients compared to the control group (CG) (P < 0.001). Significantly shorter telomeres were found in patients with LC TNM stage IV than in patients with stages I-III (P = 0.014), in patients without therapy compared to those on therapy (P = 0.008), and in patients with partial response and stable/progressive disease compared to those with complete response (P = 0.039). The total oxidant status (TOS), advanced oxidation protein products (AOPP), prooxidant-antioxidant balance (PAB) and C-reactive protein (CRP) were significantly higher in patients compared to CG (P < 0.001) and correlated negatively with TL in both patients and CG (P < 0.001). PCA showed a relation between PAB and TL, and between the EGFR status and TL. Oxidative stress and PBL telomere shortening are probably associated with LC development and progression.
Collapse
Affiliation(s)
| | - Miron Sopić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Serbia.
| | | | - Vesna Ćeriman
- Institute for Lung Diseases, Thoracic Surgery and Tuberculosis, Clinical Center of Serbia, Belgrade, Serbia
| | - Azra Guzonijić
- Department of Medical Biochemistry, Faculty of Pharmacy, University of Belgrade, Serbia
| | - Aleksandra Vukašinović
- Department of Experimental Medicine and Biochemical Sciences, University of Perugia, Italy
| | - Barbara Ostanek
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Slovenia
| | - Nemanja Dimić
- University Clinical-Hospital Center Dr. Dragisa Misovic, Belgrade, Serbia
| | | | | |
Collapse
|
9
|
Mishra R, Haldar S, Biondi S, Bhari VK, Singh G, Bhowmick NA. TGF-β controls stromal telomere length through epigenetic modifications. 3 Biotech 2022; 12:290. [PMID: 36276465 PMCID: PMC9512944 DOI: 10.1007/s13205-022-03346-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/01/2022] [Indexed: 11/01/2022] Open
Abstract
Telomere length is primarily controlled by the enzyme telomerase, but being chromatin structures, telomeres undergo epigenetic regulation for their maintenance and function. Altered telomere length among cancer cells combined with shorter telomere length in cancer-associated stromal cells, strongly implicated with progression to prostate cancer metastasis and cancer death and providing a novel target for therapeutics. Transforming growth factor-β (TGF-β) signaling pathways are well-recognized for their role in stromal-epithelial interactions responsible for prostate androgen responsiveness, promoting tumorigenesis. However, the underlying mechanism remains unclear. We sought to establish a role for TGF-β in the regulation of telomere length in mouse and human prostate fibroblast. Polymerase chain reaction (PCR)-based telomere length measuring methods are widely used due to their repeatability and reproducibility. Using real-time RT-PCR-based telomere length measuring method, we identified that TGF-beta regulates telomere length via increased expression of histone methyltransferase, Suv39h1, which in turn affected histone methylation levels at the telomeric ends. Moreover, treatment of DAPT and non-steroidal antiandrogen bicalutamide demonstrated that notch and androgen signaling co-operated with TGF-ß in regulating stromal telomere length. Telomere variation in tumor cells and non-tumor cells within the tumor microenvironment greatly facilitates the clinical assessment of prostate cancer; therefore, understanding stromal telomere length regulation mechanism will hold significant prospects for cancer treatment, diagnosis, and prognosis. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03346-5.
Collapse
Affiliation(s)
- Rajeev Mishra
- Department of Life Sciences and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kalyanpur, Kanpur, UP 208024 India
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Subhash Haldar
- Department of Food and Nutrition, University of Gour Banga, Mokdumpur, West Bengal 732101 India
| | - Shea Biondi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Vikash Kumar Bhari
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan 303007 India
| | - Gyanendra Singh
- Toxicology Department, ICMR-National Institute of Occupational Health, Ahmedabad, 380016 India
| | - Neil A Bhowmick
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
- Department of Research, Greater Los Angeles Veterans Administration, Los Angeles, CA 90073 USA
| |
Collapse
|
10
|
Khodadadi E, Mir SM, Memar MY, Sadeghi H, Kashiri M, Faeghiniya M, Jamalpoor Z, Sheikh Arabi M. Shelterin complex at telomeres: Roles in cancers. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
11
|
Pańczyszyn A, Boniewska-Bernacka E, Głąb G. Telomere length in leukocytes and cervical smears of women with high-risk human papillomavirus (HR HPV) infection. Taiwan J Obstet Gynecol 2020; 59:51-55. [PMID: 32039800 DOI: 10.1016/j.tjog.2019.11.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2019] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE Persistent high-risk HPV (HR HPV) infection leads to the development of squamous intraepithelial lesions, which in turn may progress to cervical cancer. Telomere elongation or shortening may indicate a carcinogenesis process. In the present study, we analyzed telomere length from blood and cervical smears of women without and with high-risk HPV infection. MATERIALS AND METHODS Telomere length was quantified by real-time PCR in blood and cervical smears from 48 women with high-risk HPV infection and HGSIL or LGSIL, 29 women HR-HPV positive without SIL, and 11 HPV-negative women. RESULTS No correlation was found between age and telomere length in blood and cervical smears. Women with high-risk HPV infection had shorter telomeres in cervical smears, but not in blood compared to the control group. CONCLUSION These findings suggest that telomere shortening occurs in cervical cells of women with HR HPV infection both with LGSIL and HGSIL and may indicate the onset of carcinogenesis. In turn, there is no correlation between leukocyte telomere length and cervical cancer risk in women with HR HPV infection.
Collapse
Affiliation(s)
| | | | - Grzegorz Głąb
- Public Higher Medical Professional School in Opole, Poland
| |
Collapse
|
12
|
Zhu T, Zhu Y, Xuan Y, Gao H, Cai X, Piersma SR, Pham TV, Schelfhorst T, Haas RRGD, Bijnsdorp IV, Sun R, Yue L, Ruan G, Zhang Q, Hu M, Zhou Y, Van Houdt WJ, Le Large TYS, Cloos J, Wojtuszkiewicz A, Koppers-Lalic D, Böttger F, Scheepbouwer C, Brakenhoff RH, van Leenders GJLH, Ijzermans JNM, Martens JWM, Steenbergen RDM, Grieken NC, Selvarajan S, Mantoo S, Lee SS, Yeow SJY, Alkaff SMF, Xiang N, Sun Y, Yi X, Dai S, Liu W, Lu T, Wu Z, Liang X, Wang M, Shao Y, Zheng X, Xu K, Yang Q, Meng Y, Lu C, Zhu J, Zheng J, Wang B, Lou S, Dai Y, Xu C, Yu C, Ying H, Lim TK, Wu J, Gao X, Luan Z, Teng X, Wu P, Huang S, Tao Z, Iyer NG, Zhou S, Shao W, Lam H, Ma D, Ji J, Kon OL, Zheng S, Aebersold R, Jimenez CR, Guo T. DPHL: A DIA Pan-human Protein Mass Spectrometry Library for Robust Biomarker Discovery. GENOMICS PROTEOMICS & BIOINFORMATICS 2020; 18:104-119. [PMID: 32795611 PMCID: PMC7646093 DOI: 10.1016/j.gpb.2019.11.008] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 09/03/2019] [Accepted: 11/08/2019] [Indexed: 12/21/2022]
Abstract
To address the increasing need for detecting and validating protein biomarkers in clinical specimens, mass spectrometry (MS)-based targeted proteomic techniques, including the selected reaction monitoring (SRM), parallel reaction monitoring (PRM), and massively parallel data-independent acquisition (DIA), have been developed. For optimal performance, they require the fragment ion spectra of targeted peptides as prior knowledge. In this report, we describe a MS pipeline and spectral resource to support targeted proteomics studies for human tissue samples. To build the spectral resource, we integrated common open-source MS computational tools to assemble a freely accessible computational workflow based on Docker. We then applied the workflow to generate DPHL, a comprehensive DIA pan-human library, from 1096 data-dependent acquisition (DDA) MS raw files for 16 types of cancer samples. This extensive spectral resource was then applied to a proteomic study of 17 prostate cancer (PCa) patients. Thereafter, PRM validation was applied to a larger study of 57 PCa patients and the differential expression of three proteins in prostate tumor was validated. As a second application, the DPHL spectral resource was applied to a study consisting of plasma samples from 19 diffuse large B cell lymphoma (DLBCL) patients and 18 healthy control subjects. Differentially expressed proteins between DLBCL patients and healthy control subjects were detected by DIA-MS and confirmed by PRM. These data demonstrate that the DPHL supports DIA and PRM MS pipelines for robust protein biomarker discovery. DPHL is freely accessible at https://www.iprox.org/page/project.html?id=IPX0001400000.
Collapse
Affiliation(s)
- Tiansheng Zhu
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China; School of Computer Science, Shanghai Key Laboratory of Intelligent Information Processing, Fudan University, Shanghai 200433, China
| | - Yi Zhu
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China.
| | - Yue Xuan
- Thermo Fisher Scientific (BREMEN) GmbH, Bremen 28195, Germany
| | - Huanhuan Gao
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Xue Cai
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Sander R Piersma
- OncoProteomics Laboratory, Department of Medical Oncology, VU University Medical Center, VU University, Amsterdam 1011, The Netherlands
| | - Thang V Pham
- OncoProteomics Laboratory, Department of Medical Oncology, VU University Medical Center, VU University, Amsterdam 1011, The Netherlands
| | - Tim Schelfhorst
- OncoProteomics Laboratory, Department of Medical Oncology, VU University Medical Center, VU University, Amsterdam 1011, The Netherlands
| | - Richard R G D Haas
- OncoProteomics Laboratory, Department of Medical Oncology, VU University Medical Center, VU University, Amsterdam 1011, The Netherlands
| | - Irene V Bijnsdorp
- OncoProteomics Laboratory, Department of Medical Oncology, VU University Medical Center, VU University, Amsterdam 1011, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Urology, Cancer Center Amsterdam, Amsterdam 1011, The Netherlands
| | - Rui Sun
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Liang Yue
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Guan Ruan
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Qiushi Zhang
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Mo Hu
- Thermo Fisher Scientific, Shanghai 201206, China
| | - Yue Zhou
- Thermo Fisher Scientific, Shanghai 201206, China
| | - Winan J Van Houdt
- The Netherlands Cancer Institute, Surgical Oncology, Amsterdam 1011, The Netherlands
| | - Tessa Y S Le Large
- Amsterdam UMC, Vrije Universiteit Amsterdam, Surgery, Cancer Center Amsterdam, Amsterdam 1011, The Netherlands
| | - Jacqueline Cloos
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam 1011, The Netherlands
| | - Anna Wojtuszkiewicz
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam 1011, The Netherlands
| | - Danijela Koppers-Lalic
- Amsterdam UMC, Vrije Universiteit Amsterdam, Hematology, Cancer Center Amsterdam, Amsterdam 1011, The Netherlands
| | - Franziska Böttger
- Amsterdam UMC, Vrije Universiteit Amsterdam, Medical Oncology, Cancer Center Amsterdam, Amsterdam 1011, The Netherlands
| | - Chantal Scheepbouwer
- Amsterdam UMC, Vrije Universiteit Amsterdam, Neurosurgery, Cancer Center Amsterdam, Amsterdam 1011, The Netherlands; Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, Cancer Center Amsterdam, Amsterdam 1011, The Netherlands
| | - Ruud H Brakenhoff
- Amsterdam UMC, Vrije Universiteit Amsterdam, Otolaryngology/Head and Neck Surgery, Cancer Center Amsterdam, Amsterdam 1011, The Netherlands
| | | | - Jan N M Ijzermans
- Erasmus MC University Medical Center, Surgery, Rotterdam 1016LV, The Netherlands
| | - John W M Martens
- Erasmus MC University Medical Center, Medical Oncology, Rotterdam 1016LV, The Netherlands
| | - Renske D M Steenbergen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, Cancer Center Amsterdam, Amsterdam 1011, The Netherlands
| | - Nicole C Grieken
- Amsterdam UMC, Vrije Universiteit Amsterdam, Pathology, Cancer Center Amsterdam, Amsterdam 1011, The Netherlands
| | | | - Sangeeta Mantoo
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169608, Singapore
| | - Sze S Lee
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169608, Singapore
| | - Serene J Y Yeow
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169608, Singapore
| | - Syed M F Alkaff
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169608, Singapore
| | - Nan Xiang
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Yaoting Sun
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Xiao Yi
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Shaozheng Dai
- School of Computer Science and Engineering, Beihang University, Beijing 100191, China
| | - Wei Liu
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Tian Lu
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Zhicheng Wu
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China; School of Computer Science, Shanghai Key Laboratory of Intelligent Information Processing, Fudan University, Shanghai 200433, China
| | - Xiao Liang
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Man Wang
- MOE Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Translational Research, Peking University Cancer Hospital, Beijing 100142, China
| | - Yingkuan Shao
- Cancer Institute (MOE Key Laboratory of Cancer Prevention and Intervention, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xi Zheng
- Cancer Institute (MOE Key Laboratory of Cancer Prevention and Intervention, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Kailun Xu
- Cancer Institute (MOE Key Laboratory of Cancer Prevention and Intervention, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Qin Yang
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yifan Meng
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Cong Lu
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiang Zhu
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jin'e Zheng
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Bo Wang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Sai Lou
- Phase I Clinical Research Center, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Yibei Dai
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Chao Xu
- College of Mathematics and Informatics, Digital Fujian Institute of Big Data Security Technology, Fujian Normal University, Fuzhou 350108, China
| | - Chenhuan Yu
- Zhejiang Provincial Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310015, China
| | - Huazhong Ying
- Zhejiang Provincial Key Laboratory of Experimental Animal and Safety Evaluation, Zhejiang Academy of Medical Sciences, Hangzhou 310015, China
| | - Tony K Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169608, Singapore
| | - Jianmin Wu
- MOE Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Translational Research, Peking University Cancer Hospital, Beijing 100142, China
| | - Xiaofei Gao
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
| | - Zhongzhi Luan
- School of Computer Science and Engineering, Beihang University, Beijing 100191, China
| | - Xiaodong Teng
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Peng Wu
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shi'ang Huang
- Center for Stem Cell Research and Application, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Narayanan G Iyer
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169608, Singapore
| | - Shuigeng Zhou
- School of Computer Science, Shanghai Key Laboratory of Intelligent Information Processing, Fudan University, Shanghai 200433, China
| | - Wenguang Shao
- Department of Biology, Institute for Molecular Systems Biology, ETH Zurich, Zurich 8092, Switzerland
| | - Henry Lam
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong Special Administrative Region, China
| | - Ding Ma
- Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jiafu Ji
- MOE Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Translational Research, Peking University Cancer Hospital, Beijing 100142, China
| | - Oi L Kon
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169608, Singapore
| | - Shu Zheng
- Cancer Institute (MOE Key Laboratory of Cancer Prevention and Intervention, Zhejiang Provincial Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Ruedi Aebersold
- Department of Biology, Institute for Molecular Systems Biology, ETH Zurich, Zurich 8092, Switzerland; Faculty of Science, University of Zurich, Zurich 8092, Switzerland
| | - Connie R Jimenez
- OncoProteomics Laboratory, Department of Medical Oncology, VU University Medical Center, VU University, Amsterdam 1011, The Netherlands
| | - Tiannan Guo
- Zhejiang Provincial Laboratory of Life Sciences and Biomedicine, Westlake University, Hangzhou 310024, China; Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China.
| |
Collapse
|
13
|
Li Z, Liu H, Qian Y, Li X, Guo C, Wang Z, Wei Y. Influence of metals from e-waste dismantling on telomerelength and mitochondrial DNA copy number in people living near recycling sites. ENVIRONMENT INTERNATIONAL 2020; 140:105769. [PMID: 32387852 DOI: 10.1016/j.envint.2020.105769] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 06/11/2023]
Abstract
Metals are the primary toxicants released by electronic waste (e-waste) recycling, but their adverse effects on people working in e-waste recycling or living near e-waste sites have not been studied well. Taizhou is one of the three largest e-waste recycling locations in China. Atpresent, to prevent the environmental problems stem from e-waste dismantling, the local government has shut down all the industries in 2015. In this study, we collected blood samples of residents living near e-waste dismantling factories, and in matched reference areas in Taizhou, in December 2017, after the factories have been shut down for two years. Seventeen metals were quantified in all blood samples. Among them, the concentrations of arsenic (As), nickel (Ni), silver (Ag), lanthanum (La), and Cerium (Ce) were statistically significant higher in individuals in e-waste recycling locations than those in reference location. Length of telomere (LOT) and mitochondrialDNA copy number (MCN) were measured in blood as a marker of human health. In the e-waste dismantling location, the level LOT and MCN were elevated in resident living near e-waste sites (RE) and former working in e-waste recycling (OE) than residents living in the reference area (RF). Furthermore, Pearson correlation and multiple linear regression analysis between the changed metals and LOT, MCN in blood were performed. In RE and OE, the concentration of Ni significantly positively correlated with MCN; in OE, the Ni level significantly positively correlated with MCN and LOT. Considering that the high level of Ni, TL and mtDNA were correlated with the risk of cancer, we speculated that e-waste exposure elevate the risk of cancer, and Ni that has long been present in the body was the potential hazardous element causing cancer.
Collapse
Affiliation(s)
- Zhigang Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Huijie Liu
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China; Institute of Basic Medical Sciences, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Yan Qian
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Xiaoqian Li
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Chen Guo
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Zhanshan Wang
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China
| | - Yongjie Wei
- State Key Laboratory of Environmental Criteria and Risk Assessment, Chinese Research Academy of Environmental Sciences, Beijing 100012, China; Center for Global Health, School of Public Health, Nanjing Medical University, China.
| |
Collapse
|
14
|
Pepper C, Norris K, Fegan C. Clinical utility of telomere length measurements in cancer. Curr Opin Genet Dev 2020; 60:107-111. [PMID: 32220800 DOI: 10.1016/j.gde.2020.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 10/24/2022]
Abstract
Cancer remains one of the leading causes of death in the developed world and despite impressive advances in therapeutic modalities, only a small subset of patients are currently cured. The underlying genetic heterogeneity of cancers clearly plays a crucial role in determining both the clinical course of individual pathologies and their responses to standard treatments. Although every tumour is to some extent distinct, there are recurrent features of cancers that can be exploited as therapeutic targets and as prognostic and predictive biomarkers; one such attribute is telomere length. Here we discuss the utility of telomere length evaluation in cancer and describe some of the promise and challenges of bringing this into clinical practice.
Collapse
Affiliation(s)
- Chris Pepper
- Brighton and Sussex Medical School, University of Sussex, Brighton, BN1 9PX, United Kingdom.
| | - Kevin Norris
- Division of Cancer & Genetics, Cardiff University Medical School, Cardiff, CF14 4XN, United Kingdom
| | - Christopher Fegan
- Division of Cancer & Genetics, Cardiff University Medical School, Cardiff, CF14 4XN, United Kingdom
| |
Collapse
|
15
|
Morais M, Dias F, Teixeira AL, Medeiros R. Telomere Length in Renal Cell Carcinoma: The Jekyll and Hyde Biomarker of Ageing of the Kidney. Cancer Manag Res 2020; 12:1669-1679. [PMID: 32184670 PMCID: PMC7064280 DOI: 10.2147/cmar.s211225] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 11/02/2019] [Indexed: 12/17/2022] Open
Abstract
Renal cell carcinoma (RCC) is a heterogeneous group of cancers where the clear cell (ccRCC) is the most common and the most lethal. The absence of accurate diagnostic and follow-up biomarkers along with the time-limited response to therapies may explain the lethality and shows the necessity of new sensitive and specific biomarkers. One of the most studied molecules are the telomeres: specialized ribonucleoprotein structures that keep the structural integrity of the genome. Among other features, telomere length (TL) has been widely studied in several tumor models regarding its biomarker potential, due to the easy detection and quantification. The scope of this review was to analyze all the information about this parameter in RCC. There was some disparity in the results of the studies, since some pointed to an association between short TL and risk or poor outcome of RCC; others between long TL and RCC outcome and some did not find any association. We propose some epidemiological and biological explanations to these differences. The telomeres may play a dual role during RCC carcinogenesis in the early stages, short telomeres may increase RCC risk and in late carcinogenesis, long telomeres seem to be associated with tumor prognosis. However, the controversy of the results along with the lack of specificity are some problems that need to be clarified for the usage of TL as a prognostic biomarker.
Collapse
Affiliation(s)
- Mariana Morais
- Molecular Oncology and Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto4200-072, Portugal
- Research Department, LPCC- Portuguese League Against Cancer (NRNorte), Porto4200-172, Portugal
| | - Francisca Dias
- Molecular Oncology and Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto4200-072, Portugal
- ICBAS, Abel Salazar Institute for the Biomedical Sciences, University of Porto, Porto, Portugal
| | - Ana Luísa Teixeira
- Molecular Oncology and Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto4200-072, Portugal
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, IPO-Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO-Porto), Porto4200-072, Portugal
- Research Department, LPCC- Portuguese League Against Cancer (NRNorte), Porto4200-172, Portugal
- ICBAS, Abel Salazar Institute for the Biomedical Sciences, University of Porto, Porto, Portugal
- FMUP, Faculty of Medicine, University of Porto, Porto4200-319, Portugal
- CEBIMED, Faculty of Health Sciences, Fernando Pessoa University, Porto4249-004, Portugal
| |
Collapse
|
16
|
Díaz-Cartagena D, Hernández-Cancel G, Bracho-Rincón DP, González-Feliciano JA, Cunci L, González CI, Cabrera CR. Label-Free Telomerase Activity Detection via Electrochemical Impedance Spectroscopy. ACS OMEGA 2019; 4:16724-16732. [PMID: 31646217 PMCID: PMC6796945 DOI: 10.1021/acsomega.9b00783] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/16/2019] [Indexed: 05/14/2023]
Abstract
In the last decade, researchers have been searching for innovative platforms, methods, and techniques able to address recurring problems with the current cancer detection methods. Early disease detection, fast results, point-of-care sensing, and cost are among the most prevalent issues that need further exploration in this field. Herein, studies are focused on overcoming these problems by developing an electrochemical device able to detect telomerase as a cancer biomarker. Electrochemical platforms and techniques are more appealing for cancer detection, offering lower costs than the established cancer detection methods, high sensitivity inherent to the technique, rapid signal processing, and their capacity of being miniaturized. Therefore, Au interdigital electrodes and electrochemical impedance spectroscopy were used to detect telomerase activity in acute T cell leukemia. Different cancer cell concentrations were evaluated, and a detection limit of 1.9 × 105 cells/mL was obtained. X-ray photoelectron spectroscopy was used to characterize the telomerase substrate (TS) DNA probe self-assembled monolayer on gold electrode surfaces. Atomic force microscopy displayed three-dimensional images of the surface to establish a height difference of 9.0 nm between the bare electrode and TS-modified Au electrodes. The TS probe is rich in guanines, thus forming secondary structures known as G-quadruplex that can be triggered with a fluorescence probe. Confocal microscopy fluorescence images showed the formation of DNA G-quadruplex because of TS elongation by telomerase on the Au electrode surface. Moreover, electrodes exposed to telomerase containing 2',3'-dideoxyguanosine-5'-triphosphate (ddGTP) did not exhibit high fluorescence, as ddGTP is a telomerase inhibitor, thus making this device suitable for telomerase inhibitors capacity studies. The electrochemical method and Au microchip device may be developed as a biosensor for a point-of-care medical device.
Collapse
Affiliation(s)
- Diana
C. Díaz-Cartagena
- Department
of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, Puerto Rico 00925-2537, United States
- Molecular
Sciences Research Center, University of
Puerto Rico, San Juan, Puerto Rico 00926, United States
| | - Griselle Hernández-Cancel
- Molecular
Sciences Research Center, University of
Puerto Rico, San Juan, Puerto Rico 00926, United States
| | - Dina P. Bracho-Rincón
- Molecular
Sciences Research Center, University of
Puerto Rico, San Juan, Puerto Rico 00926, United States
- Department
of Biology, University of Puerto Rico, Río Piedras Campus, San Juan, Puerto Rico 00931, United States
| | - José A. González-Feliciano
- Molecular
Sciences Research Center, University of
Puerto Rico, San Juan, Puerto Rico 00926, United States
| | - Lisandro Cunci
- School
of Natural Sciences and Technology, Universidad
Ana G. Méndez, Gurabo Campus, Gurabo, Puerto
Rico 00778, United
States
| | - Carlos I. González
- Molecular
Sciences Research Center, University of
Puerto Rico, San Juan, Puerto Rico 00926, United States
- Department
of Biology, University of Puerto Rico, Río Piedras Campus, San Juan, Puerto Rico 00931, United States
| | - Carlos R. Cabrera
- Department
of Chemistry, University of Puerto Rico, Río Piedras Campus, San Juan, Puerto Rico 00925-2537, United States
- Molecular
Sciences Research Center, University of
Puerto Rico, San Juan, Puerto Rico 00926, United States
- E-mail:
| |
Collapse
|
17
|
Hu R, Hua X, Jiang Q. Associations of telomere length in risk and recurrence of prostate cancer: A meta‐analysis. Andrologia 2019; 51:e13304. [DOI: 10.1111/and.13304] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 03/28/2019] [Accepted: 04/03/2019] [Indexed: 01/01/2023] Open
Affiliation(s)
- Rui Hu
- Department of Medical Insurance, School of Health Services Management Anhui Medical University Hefei China
| | - Xiao‐Guo Hua
- Department of Epidemiology and Biostatistics, School of Public Health Anhui Medical University Hefei China
| | - Qi‐Cheng Jiang
- Department of Medical Insurance, School of Health Services Management Anhui Medical University Hefei China
| |
Collapse
|
18
|
Tanpaisankit M, Hongsaprabhas C, Chareonlap C, Honsawek S. Relative telomere length and oxidative stress in musculoskeletal tumors. Mol Biol Rep 2019; 46:4009-4016. [PMID: 31069615 DOI: 10.1007/s11033-019-04847-y] [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: 01/16/2019] [Accepted: 04/30/2019] [Indexed: 11/29/2022]
Abstract
Telomeres are capped at the end of the chromosome and gradually shorten when the cell divides. When there is an oxidative stress, it can cause the DNA to be damaged. Hence, 8-hydroxy-2'-deoxyguanosine (8-OHdG) has been shown to be an indicator for oxidative DNA damage. This study aimed to determine the relative telomere length (RTL) and 8-OHdG levels in neoplastic tissues, adjacent non-neoplastic tissues, and blood leukocytes of musculoskeletal (MS) tumor patients. Neoplastic tissues were compared to adjacent non-neoplastic tissues in MS tumor patients (n = 46). Peripheral blood leukocytes (PBLs) of MS tumor subjects were compared to those of age-matched healthy controls (n = 107). RTL was evaluated by quantitative real-time polymerase chain reaction and 8-OHdG levels were quantified by enzyme-linked immunosorbent assay. The RTL in neoplastic tissues was significantly shorter than that in non-neoplastic tissues [1.12 (0.86-1.46) vs 1.45 (1.25-1.65), P = 0.001]. PBLs had lower RTL than non-neoplastic tissues in MS tumor patients [1.04 (0.85-1.13) vs 1.45 (1.25-1.65), P < 0.001]. However, there was no significant difference between RTL in PBLs and in neoplastic tissues. In addition, PBLs of MS tumor patients had higher RTL than those of the controls [1.04 (0.85-1.13) versus 0.78 (0.68-0.90), P < 0.001]. The 8-OHdG levels in neoplastic tissues were remarkably higher than those in non-neoplastic tissues [8.14 (6.81-11.37) nM/μg/μl vs. 3.79 (2.53-6.17) nM/μg/μl, P < 0.001]. Furthermore, plasma 8-OHdG levels in MS tumor patients were markedly greater than those in the controls [102.50 (73.16-133.50) nM vs. 41.09 (6.81-11.37) nM, P < 0.001]. Area under the curve (AUC) was 0.7536 (95% confident interval (CI) 0.6602-0.8469) when the cut-off value of RTL in PBLs was 0.97. Also, plasma 8-OHdG levels depicted that when the cut-off value was 38.67 nM, the AUC was 0.7723 (95% CI 0.6920-0.8527). Moreover, ROC curve analysis showed that both RTL and 8-OHdG appeared to improve the sensitivity (85.68%) and specificity (70.91%) with the AUC 0.8639 (95% CI 0.7500-0.9500). This study suggested that blood leukocyte RTL and plasma 8-OHdG could serve as promising non-invasive biomarkers to differentiate between MS tumor patients and healthy controls. Additionally, telomere attrition and increased oxidative DNA damage might play contributory roles in the pathogenesis of MS tumors.
Collapse
Affiliation(s)
- Montira Tanpaisankit
- Department of Biochemistry, Osteoarthritis and Musculoskeleton Research Unit, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chindanai Hongsaprabhas
- Department of Orthopaedics, Faculty of Medicine, Vinai Parkpian Orthopaedic Research Center, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chris Chareonlap
- Department of Orthopaedics, Faculty of Medicine, Vinai Parkpian Orthopaedic Research Center, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sittisak Honsawek
- Department of Biochemistry, Osteoarthritis and Musculoskeleton Research Unit, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand. .,Department of Orthopaedics, Faculty of Medicine, Vinai Parkpian Orthopaedic Research Center, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Chulalongkorn University, Bangkok, 10330, Thailand. .,Department of Biochemistry, Osteoarthritis and Musculoskeleton Research Unit, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University, 1873 Rama IV Road Patumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
19
|
Establishment of two data mining models of lung cancer screening based on three gene promoter methylations combined with telomere damage. Int J Biol Markers 2017; 32:e141-e146. [PMID: 27716889 DOI: 10.5301/jbm.5000232] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2016] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To identify the significance of a support vector machine (SVM) model and a decision tree (DT) model for the diagnosis of lung cancer combined with the detection of fragile histidine triad (FHIT), RAS association domain family 1 (RASSF1A) and cyclin-dependent kinase inhibitor 2A (p16) promoter methylation levels and relative telomere length (RTL) of white blood cells from peripheral blood DNA. METHODS The levels of p16, RASSF1A and FHIT promoter methylation and the RTL of white blood cells in peripheral blood DNA of 200 healthy individuals and 200 lung cancer patients were analyzed by SYBR Green-based quantitative methylation-specific PCR and quantitative PCR. Based on the 4 biomarkers, SVM and DT models were developed. RESULTS The levels of FHIT, RASSF1A and p16 promoter methylation were 3.33 (1.86-6.40) and 2.85 (1.39-5.44) (p = 0.002); 27.62 (9.09-52.86) and 17.17 (3.86-50.87) (p = 0.038); and 0.59 (0.16-4.50) and 0.36 (0.06-4.00) (p = 0.008) in cases and controls, respectively. RTL was 0.93 ± 0.32 and 1.16 ± 0.57 (p<0.001). The areas under the receiver operating characteristic (ROC) curves of the Fisher discriminant analysis, SVM and DT models were 0.670 (0.569-0.761), 0.810 (0.719-0.882) and 0.810 (0.719-0.882), respectively. CONCLUSIONS The SVM and DT models for diagnosing lung cancer were successfully developed through the combined detection of p16, RASSF1A and FHIT promoter methylation and RTL, which provided useful tools for screening lung cancer.
Collapse
|
20
|
Influence of In Utero Maternal and Neonate Factors on Cord Blood Leukocyte Telomere Length: Clues to the Racial Disparity in Prostate Cancer? Prostate Cancer 2016; 2016:3691650. [PMID: 28070423 PMCID: PMC5192337 DOI: 10.1155/2016/3691650] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 07/23/2016] [Indexed: 02/06/2023] Open
Abstract
Background. Modifiable factors in adulthood that explain the racial disparity in prostate cancer have not been identified. Because racial differences in utero that may account for this disparity are understudied, we investigated the association of maternal and neonate factors with cord blood telomere length, as a cumulative marker of cell proliferation and oxidative damage, by race. Further, we evaluated whether cord blood telomere length differs by race. Methods. We measured venous umbilical cord blood leukocyte relative telomere length by qPCR in 38 black and 38 white full-term male neonates. Using linear regression, we estimated geometric mean relative telomere length and tested for differences by race. Results. Black mothers were younger and had higher parity and black neonates had lower birth and placental weights. These factors were not associated with relative telomere length, even after adjusting for or stratifying by race. Relative telomere length in black (2.72) and white (2.73) neonates did not differ, even after adjusting for maternal or neonate factors (all p > 0.9). Conclusions. Maternal and neonate factors were not associated with cord blood telomere length, and telomere length did not differ by race. These findings suggest that telomere length at birth does not explain the prostate cancer racial disparity.
Collapse
|
21
|
Duan X, Yang Y, Tan S, Wang S, Feng X, Cui L, Feng F, Yu S, Wang W, Wu Y. Application of artificial neural network model combined with four biomarkers in auxiliary diagnosis of lung cancer. Med Biol Eng Comput 2016; 55:1239-1248. [PMID: 27766520 DOI: 10.1007/s11517-016-1585-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 10/10/2016] [Indexed: 12/11/2022]
Abstract
The purpose of the study was to explore the application of artificial neural network model in the auxiliary diagnosis of lung cancer and compare the effects of back-propagation (BP) neural network with Fisher discrimination model for lung cancer screening by the combined detections of four biomarkers of p16, RASSF1A and FHIT gene promoter methylation levels and the relative telomere length. Real-time quantitative methylation-specific PCR was used to detect the levels of three-gene promoter methylation, and real-time PCR method was applied to determine the relative telomere length. BP neural network and Fisher discrimination analysis were used to establish the discrimination diagnosis model. The levels of three-gene promoter methylation in patients with lung cancer were significantly higher than those of the normal controls. The values of Z(P) in two groups were 2.641 (0.008), 2.075 (0.038) and 3.044 (0.002), respectively. The relative telomere lengths of patients with lung cancer (0.93 ± 0.32) were significantly lower than those of the normal controls (1.16 ± 0.57), t = 4.072, P < 0.001. The areas under the ROC curve (AUC) and 95 % CI of prediction set from Fisher discrimination analysis and BP neural network were 0.670 (0.569-0.761) and 0.760 (0.664-0.840). The AUC of BP neural network was higher than that of Fisher discrimination analysis, and Z(P) was 0.76. Four biomarkers are associated with lung cancer. BP neural network model for the prediction of lung cancer is better than Fisher discrimination analysis, and it can provide an excellent and intelligent diagnosis tool for lung cancer.
Collapse
Affiliation(s)
- Xiaoran Duan
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Yongli Yang
- Department of Epidemiology and Biostatistics, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Shanjuan Tan
- Department of Hospital Infection Management, Qingdao Municipal Hospital, Qingdao, China
| | - Sihua Wang
- Department of Occupational Health, Henan Institute of Occupational Health, Zhengzhou, China
| | - Xiaolei Feng
- Department of Occupational Health and Occupational Medicine, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Liuxin Cui
- Department of Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Feifei Feng
- Department of Health Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Songcheng Yu
- Department of Sanitary Chemistry, College of Public Health, Zhengzhou University, Zhengzhou, China
| | - Wei Wang
- Department of Occupational Health and Occupational Medicine, College of Public Health, Zhengzhou University, Zhengzhou, China.
| | - Yongjun Wu
- Department of Health Toxicology, College of Public Health, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
22
|
Gunkel M, Chung I, Wörz S, Deeg KI, Simon R, Sauter G, Jones DTW, Korshunov A, Rohr K, Erfle H, Rippe K. Quantification of telomere features in tumor tissue sections by an automated 3D imaging-based workflow. Methods 2016; 114:60-73. [PMID: 27725304 DOI: 10.1016/j.ymeth.2016.09.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 09/26/2016] [Accepted: 09/27/2016] [Indexed: 01/08/2023] Open
Abstract
The microscopic analysis of telomere features provides a wealth of information on the mechanism by which tumor cells maintain their unlimited proliferative potential. Accordingly, the analysis of telomeres in tissue sections of patient tumor samples can be exploited to obtain diagnostic information and to define tumor subgroups. In many instances, however, analysis of the image data is conducted by manual inspection of 2D images at relatively low resolution for only a small part of the sample. As the telomere feature signal distribution is frequently heterogeneous, this approach is prone to a biased selection of the information present in the image and lacks subcellular details. Here we address these issues by using an automated high-resolution imaging and analysis workflow that quantifies individual telomere features on tissue sections for a large number of cells. The approach is particularly suited to assess telomere heterogeneity and low abundant cellular subpopulations with distinct telomere characteristics in a reproducible manner. It comprises the integration of multi-color fluorescence in situ hybridization, immunofluorescence and DNA staining with targeted automated 3D fluorescence microscopy and image analysis. We apply our method to telomeres in glioblastoma and prostate cancer samples, and describe how the imaging data can be used to derive statistically reliable information on telomere length distribution or colocalization with PML nuclear bodies. We anticipate that relating this approach to clinical outcome data will prove to be valuable for pretherapeutic patient stratification.
Collapse
Affiliation(s)
- Manuel Gunkel
- VIROQUANT CellNetworks RNAi Screening Facility and Research Group High-Content Analysis of the Cell (HiCell), Bioquant Center, University of Heidelberg, Germany
| | - Inn Chung
- Research Group Genome Organization & Function, German Cancer Research Center (DKFZ) and Bioquant Center, Germany.
| | - Stefan Wörz
- Department of Bioinformatics and Functional Genomics, Biomedical Computer Vision Group, Bioquant Center and IPMB, University of Heidelberg and German Cancer Research Center (DKFZ), Germany
| | - Katharina I Deeg
- Research Group Genome Organization & Function, German Cancer Research Center (DKFZ) and Bioquant Center, Germany
| | - Ronald Simon
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - Guido Sauter
- Department of Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | - David T W Jones
- Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), Germany
| | - Andrey Korshunov
- Department of Neuropathology, Heidelberg University Hospital, Germany
| | - Karl Rohr
- Department of Bioinformatics and Functional Genomics, Biomedical Computer Vision Group, Bioquant Center and IPMB, University of Heidelberg and German Cancer Research Center (DKFZ), Germany.
| | - Holger Erfle
- VIROQUANT CellNetworks RNAi Screening Facility and Research Group High-Content Analysis of the Cell (HiCell), Bioquant Center, University of Heidelberg, Germany.
| | - Karsten Rippe
- Research Group Genome Organization & Function, German Cancer Research Center (DKFZ) and Bioquant Center, Germany.
| |
Collapse
|
23
|
Baljevic M, Dumitriu B, Lee JW, Paietta EM, Wiernik PH, Racevskis J, Chen C, Stein EM, Gallagher RE, Rowe JM, Appelbaum FR, Powell BL, Larson RA, Coutré SE, Lancet J, Litzow MR, Luger SM, Young NS, Tallman MS. Telomere Length Recovery: A Strong Predictor of Overall Survival in Acute Promyelocytic Leukemia. Acta Haematol 2016; 136:210-218. [PMID: 27632567 DOI: 10.1159/000448160] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 07/01/2016] [Indexed: 11/19/2022]
Abstract
Telomeres are the capping ends of chromosomes that protect the loss of genetic material and prevent chromosomal instability. In human tissue-specific stem/progenitor cells, telomere length (TL) is maintained by the telomerase complex, which consists of a reverse transcriptase catalytic subunit (TERT) and an RNA template (TERC). Very short telomeres and loss-of-function mutations in the TERT and TERC genes have been reported in acute myeloid leukemia, but the role of telomeres in acute promyelocytic leukemia (APL) has not been well established. We report the results for a large cohort of 187 PML/RARα-positive APL patients. No germline mutations in the TERT or TERC genes were identified. Codon 279 and 1062 TERT polymorphisms were present at a frequency similar to that in the general population. TL measured in blood or marrow mononuclear cells at diagnosis was significantly shorter in the APL patients than in healthy volunteers, and shorter telomeres at diagnosis were significantly associated with high-risk disease. For patients who achieved complete remission, the median increase in TL from diagnosis to remission (delta TL) was 2.0 kilobase (kb), and we found delta TL to be the most powerful predictor of overall survival when compared with well-established risk factors for poor outcomes in APL.
Collapse
Affiliation(s)
- Muhamed Baljevic
- Oncology/Hematology Division, The University of Nebraska Medical Center, Omaha, Nebr., USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kong PL, Looi LM, Lau TP, Cheah PL. Assessment of Telomere Length in Archived Formalin-Fixed, Paraffinized Human Tissue Is Confounded by Chronological Age and Storage Duration. PLoS One 2016; 11:e0161720. [PMID: 27598341 PMCID: PMC5012687 DOI: 10.1371/journal.pone.0161720] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 08/10/2016] [Indexed: 01/13/2023] Open
Abstract
Telomeres shorten with physiological aging but undergo substantial restoration during cancer immortalization. Increasingly, cancer studies utilize the archive of formalin-fixed, paraffin-embedded (FFPE) tissues in diagnostic pathology departments. Conceptually, such studies would be confounded by physiological telomere attrition and loss of DNA integrity from prolonged tissue storage. Our study aimed to investigate these two confounding factors. 145 FFPE tissues of surgically-resected, non-diseased appendixes were retrieved from our pathology archive, from years 2008 to 2014. Cases from 2013 to 2014 were categorized by patient chronological age (0–20 years, 21–40 years, 41–60 years, > 60 years). Telomere lengths of age categories were depicted by telomere/chromosome 2 centromere intensity ratio (TCR) revealed by quantitative fluorescence in situ hybridization. Material from individuals aged 0–20 years from years 2013/2014, 2011/2012, 2009/2010, and 2008 were compared for storage effect. Telomere integrity was assessed by telomere fluorescence intensity (TFI). Epithelial TCRs (mean ± SD) for the respective age groups were 4.84 ± 2.08, 3.64 ± 1.21, 2.03 ± 0.37, and 1.93 ± 0.45, whereas corresponding stromal TCRs were 5.16 ± 2.55, 3.84 ± 1.36, 2.49 ± 1.20, and 2.93 ± 1.24. A trend of inverse correlation with age in both epithelial and stromal tissues is supported by r = -0.69, p < 0.001 and r = -0.42, p < 0.001 respectively. Epithelial TFIs (mean ± SD) of years 2013/2014, 2011/2012, 2009/2010 and 2008 were 852.60 ± 432.46, 353.04 ± 127.12, 209.24 ± 55.57 and 429.22 ± 188.75 respectively. Generally, TFIs reduced with storage duration (r = -0.42, p < 0.001). Our findings agree that age-related telomere attrition occurs in normal somatic tissues, and suggest that an age-based reference can be established for telomere studies on FFPE tissues. We also showed that FFPE tissues archived beyond 2 years are suboptimal for telomere analysis.
Collapse
Affiliation(s)
- Po-Lian Kong
- Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Federal Territory, Malaysia
| | - Lai-Meng Looi
- Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Federal Territory, Malaysia
- * E-mail:
| | - Tze-Pheng Lau
- Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Federal Territory, Malaysia
| | - Phaik-Leng Cheah
- Department of Pathology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Federal Territory, Malaysia
| |
Collapse
|
25
|
Danforth DN. Genomic Changes in Normal Breast Tissue in Women at Normal Risk or at High Risk for Breast Cancer. BREAST CANCER-BASIC AND CLINICAL RESEARCH 2016; 10:109-46. [PMID: 27559297 PMCID: PMC4990153 DOI: 10.4137/bcbcr.s39384] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 04/17/2016] [Accepted: 04/19/2016] [Indexed: 12/12/2022]
Abstract
Sporadic breast cancer develops through the accumulation of molecular abnormalities in normal breast tissue, resulting from exposure to estrogens and other carcinogens beginning at adolescence and continuing throughout life. These molecular changes may take a variety of forms, including numerical and structural chromosomal abnormalities, epigenetic changes, and gene expression alterations. To characterize these abnormalities, a review of the literature has been conducted to define the molecular changes in each of the above major genomic categories in normal breast tissue considered to be either at normal risk or at high risk for sporadic breast cancer. This review indicates that normal risk breast tissues (such as reduction mammoplasty) contain evidence of early breast carcinogenesis including loss of heterozygosity, DNA methylation of tumor suppressor and other genes, and telomere shortening. In normal tissues at high risk for breast cancer (such as normal breast tissue adjacent to breast cancer or the contralateral breast), these changes persist, and are increased and accompanied by aneuploidy, increased genomic instability, a wide range of gene expression differences, development of large cancerized fields, and increased proliferation. These changes are consistent with early and long-standing exposure to carcinogens, especially estrogens. A model for the breast carcinogenic pathway in normal risk and high-risk breast tissues is proposed. These findings should clarify our understanding of breast carcinogenesis in normal breast tissue and promote development of improved methods for risk assessment and breast cancer prevention in women.
Collapse
Affiliation(s)
- David N Danforth
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
26
|
Kulić A, Plavetić ND, Gamulin S, Jakić-Razumović J, Vrbanec D, Sirotković-Skerlev M. Telomerase activity in breast cancer patients: association with poor prognosis and more aggressive phenotype. Med Oncol 2016; 33:23. [PMID: 26833480 DOI: 10.1007/s12032-016-0736-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Accepted: 01/18/2016] [Indexed: 11/28/2022]
Abstract
Telomerase expression is an important mechanism of tumor unlimited replicative potential. The aim of this study was to evaluate prognostic impact of telomerase activity in breast cancer patients and to correlate telomerase activity with established prognostic factors. We analyzed tissue of 102 malignant breast lesions and 20 healthy breast tissues. Telomerase activity was determined by telomeric repeat amplification protocol assay. Telomerase activity was present in 77 (75.49 %) of 102 breast cancers. Telomerase activity in breast cancers was statistically significantly higher in comparison with the activity in normal breast tissue. The levels of telomerase activity were significantly positively correlated with tumor size, axillary nodal status, histological grade, HER-2/neu protein expression in tumor tissue and expression of the nuclear antigen Ki-67. A statistically significant negative correlation was found between the presence of ER and telomerase activity. There was no correlation between telomerase activity and concentration of PR or the age of patients. Kaplan-Meier analysis showed that patients with higher telomerase activity had significantly shorter 10-year disease-free survival (p < 0.0001) and 10-year overall survival (p < 0.0001) than those with lower telomerase activity. These results were confirmed by logistic regression analysis. Our results support the prognostic role of telomerase activity and its relationship with the more aggressive phenotype of breast cancer.
Collapse
Affiliation(s)
- Ana Kulić
- Department of Oncology, Division of Pathophysiology and Experimental Oncology, University Hospital Center Zagreb, Kišpatićeva 12, 10000, Zagreb, Croatia
| | - Natalija Dedić Plavetić
- Department of Oncology, Division of Medical Oncology, University Hospital Center Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | | | - Jasminka Jakić-Razumović
- Department of Pathology, University Hospital Center Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Damir Vrbanec
- Department of Oncology, Division of Medical Oncology, University Hospital Center Zagreb, Zagreb, Croatia.,School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Maja Sirotković-Skerlev
- Department of Oncology, Division of Pathophysiology and Experimental Oncology, University Hospital Center Zagreb, Kišpatićeva 12, 10000, Zagreb, Croatia. .,School of Medicine, University of Zagreb, Zagreb, Croatia.
| |
Collapse
|
27
|
Ferguson LR. Fish oils in parenteral nutrition: Why could these be important for gastrointestinal oncology? World J Gastrointest Oncol 2015; 7:128-131. [PMID: 26380055 PMCID: PMC4569589 DOI: 10.4251/wjgo.v7.i9.128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 08/07/2015] [Indexed: 02/05/2023] Open
Abstract
By the time a gastroenterology patient is moved to parenteral nutrition, he or she is usually in poor health. All parenteral nutrition formulae contain essential nutrients, avoiding components that could cause an adverse reaction. The lipid component is often provided by a soy extract, containing all the fatty acids considered to be essential in the diet. Several trials have considered parenteral nutrition formulas with added fish oils, high in the long chain omega-3 polyunsaturated fatty acids, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA). Given the range of biological functions associated with such compounds, especially in reducing inflammatory symptoms, this move would appear rational. However, while data from such trials are often positive, there has been variability among results. Some of this variability could be caused by environmental contaminants in the fish, and/or oxidation of the lipids because of poor storage. The situation is complicated by a recent report that fish oils may counter the effects of platinum chemotherapy. However, this effect associated with a minor component, hexadeca-4,7,10,13-tetraenoic acid. It is suggested that pure DHA and EPA would be beneficial additions to parenteral nutrition, reducing the probability of carcinogenesis and enhancing rational disease management. However, the jury is still out on fish oils more generally.
Collapse
|
28
|
Fernández-Marcelo T, Gómez A, Pascua I, de Juan C, Head J, Hernando F, Jarabo JR, Calatayud J, Torres-García AJ, Iniesta P. Telomere length and telomerase activity in non-small cell lung cancer prognosis: clinical usefulness of a specific telomere status. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:78. [PMID: 26250468 PMCID: PMC4528384 DOI: 10.1186/s13046-015-0195-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/21/2015] [Indexed: 01/14/2023]
Abstract
Background Considering previous data and the need to incorporate new biomarkers for the prognosis of solid tumours into the clinic, our aim in this work consists of evaluating the potential clinical use of telomeres and telomerase in non-small cell lung cancer (NSCLC). Methods Telomere status was established by determination of telomere length using the Terminal Restriction Fragment length method, and telomerase activity by the Telomeric Repeat Amplification Protocol in 142 NSCLCs and their corresponding control samples, obtained from patients submitted to surgery. Group-oriented curves for disease-free survival were calculated according to the Kaplan-Meier method considering telomere length, T/N ratio (telomere length in tumour to control tissue) and telomerase activity. Results Overall, tumours had significantly shorter telomeres compared with telomeres in control tissues (P = 0.027). More than 80 % of NSCLCs displayed telomerase activity. Regarding prognosis studies, patients whose tumours showed a mean telomere length (MTL) <7.29 Kb or T/N ratio <0.97 showed a significantly poor clinical evolution (P = 0.034 and P = 0.040, respectively). As result of a Cox multivariate analysis including pathologic state and lymph node dissemination, the MTL and T/N ratio emerged as independent significant prognostic factors. Conclusions Telomerase activity was identified as a marker of poor prognosis. The novel finding of this study is the independent prognosis role of a specific telomere status in NSCLC patients. According to our results, telomere function may emerge as a useful molecular tool that allow to select groups of NSCLC patients with different clinical evolution, in order to establish personalized therapy protocols.
Collapse
Affiliation(s)
- Tamara Fernández-Marcelo
- Department of Biochemistry and Molecular Biology II. Faculty of Pharmacy, Complutense University, Sq. Ramón y Cajal s/n (Ciudad Universitaria), Madrid, 28040, Spain. .,Sanitary Research Institute of San Carlos Hospital (IdISSC), Madrid, 28040, Spain.
| | - Ana Gómez
- Service of Thoracic Surgery, San Carlos Hospital, Madrid, 28040, Spain. .,Sanitary Research Institute of San Carlos Hospital (IdISSC), Madrid, 28040, Spain.
| | - Irene Pascua
- Department of Biochemistry and Molecular Biology II. Faculty of Pharmacy, Complutense University, Sq. Ramón y Cajal s/n (Ciudad Universitaria), Madrid, 28040, Spain. .,Sanitary Research Institute of San Carlos Hospital (IdISSC), Madrid, 28040, Spain.
| | - Carmen de Juan
- Department of Biochemistry and Molecular Biology II. Faculty of Pharmacy, Complutense University, Sq. Ramón y Cajal s/n (Ciudad Universitaria), Madrid, 28040, Spain. .,Sanitary Research Institute of San Carlos Hospital (IdISSC), Madrid, 28040, Spain.
| | - Jacqueline Head
- Department of Biochemistry and Molecular Biology II. Faculty of Pharmacy, Complutense University, Sq. Ramón y Cajal s/n (Ciudad Universitaria), Madrid, 28040, Spain. .,Sanitary Research Institute of San Carlos Hospital (IdISSC), Madrid, 28040, Spain.
| | - Florentino Hernando
- Service of Thoracic Surgery, San Carlos Hospital, Madrid, 28040, Spain. .,Sanitary Research Institute of San Carlos Hospital (IdISSC), Madrid, 28040, Spain.
| | - Jose-Ramón Jarabo
- Service of Thoracic Surgery, San Carlos Hospital, Madrid, 28040, Spain. .,Sanitary Research Institute of San Carlos Hospital (IdISSC), Madrid, 28040, Spain.
| | - Joaquín Calatayud
- Service of Thoracic Surgery, San Carlos Hospital, Madrid, 28040, Spain. .,Sanitary Research Institute of San Carlos Hospital (IdISSC), Madrid, 28040, Spain.
| | - Antonio-José Torres-García
- Service of General Surgery and Digestive Tract, San Carlos Hospital, Madrid, 28040, Spain. .,Sanitary Research Institute of San Carlos Hospital (IdISSC), Madrid, 28040, Spain.
| | - Pilar Iniesta
- Department of Biochemistry and Molecular Biology II. Faculty of Pharmacy, Complutense University, Sq. Ramón y Cajal s/n (Ciudad Universitaria), Madrid, 28040, Spain. .,Sanitary Research Institute of San Carlos Hospital (IdISSC), Madrid, 28040, Spain.
| |
Collapse
|
29
|
Heaphy CM, Gaonkar G, Peskoe SB, Joshu CE, De Marzo AM, Lucia MS, Goodman PJ, Lippman SM, Thompson IM, Platz EA, Meeker AK. Prostate stromal cell telomere shortening is associated with risk of prostate cancer in the placebo arm of the Prostate Cancer Prevention Trial. Prostate 2015; 75:1160-6. [PMID: 25893825 PMCID: PMC4475463 DOI: 10.1002/pros.22997] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/09/2015] [Indexed: 11/09/2022]
Abstract
BACKGROUND Telomeres are repetitive nucleoproteins that help maintain chromosomal stability by inhibiting exonucleolytic degradation, prohibiting inappropriate homologous recombination, and preventing chromosomal fusions by suppressing double-strand break signals. We recently observed that men treated for clinically localized prostate cancer with shorter telomeres in their cancer-associated stromal cells, in combination with greater variation in cancer cell telomere lengths, were significantly more likely to progress to distant metastases, and die from their disease. Here, we hypothesized that shorter stromal cell telomere length would be associated with prostate cancer risk at time of biopsy. METHODS Telomere-specific fluorescence in situ hybridization (FISH) analysis was performed in normal-appearing stromal, basal epithelial, and luminal epithelial cells in biopsies from men randomized to the placebo arm of the Prostate Cancer Prevention Trial. Prostate cancer cases (N = 32) were either detected on a biopsy performed for cause or at the end of the study per trial protocol, and controls (N = 50), defined as negative for cancer on an end-of-study biopsy performed per trial protocol (e.g., irrespective of indication), were sampled. Logistic regression was used to estimate the association between mean telomere length of the particular cell populations, cell-to-cell telomere length variability, and risk of prostate cancer. RESULTS Men with short stromal cell telomere lengths (below median) had 2.66 (95% CI 1.04-3.06; P = 0.04) times the odds of prostate cancer compared with men who had longer lengths (at or above median). Conversely, we did not observe statistically significant associations for short telomere lengths in normal-appearing basal (OR = 2.15, 95% CI 0.86-5.39; P= 0 .10) or luminal (OR = 1.15, 95% CI 0.47-2.80; P = 0.77) cells. CONCLUSIONS These findings suggest that telomere shortening in normal stromal cells is associated with prostate cancer risk. It is essential to extend and validate these findings, while also identifying the cellular milieu that comprises the subset of cells with short telomeres within the prostate tumor microenvironment.
Collapse
Affiliation(s)
- Christopher M. Heaphy
- Department of Pathology, Johns Hopkins University School of Medicine
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | - Gaurav Gaonkar
- Department of Pathology, Johns Hopkins University School of Medicine
| | - Sarah B. Peskoe
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health
| | - Corinne E. Joshu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | - Angelo M. De Marzo
- Department of Pathology, Johns Hopkins University School of Medicine
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | | | - Phyllis J. Goodman
- SWOG Statistical Center, and the Division of Public Health Sciences, Fred Hutchinson Cancer Research Center
| | | | | | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| | - Alan K. Meeker
- Department of Pathology, Johns Hopkins University School of Medicine
- Department of Urology and the James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins
| |
Collapse
|
30
|
Bull C, Christensen H, Fenech M. Cortisol is not associated with telomere shortening or chromosomal instability in human lymphocytes cultured under low and high folate conditions. PLoS One 2015; 10:e0119367. [PMID: 25748629 PMCID: PMC4352017 DOI: 10.1371/journal.pone.0119367] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 01/13/2015] [Indexed: 01/13/2023] Open
Abstract
Chronic psychological stress and nutritional deficiencies are factors that impact negatively on human health and disease risk. Chronic stress has been associated with accelerated leukocyte telomere shortening in numerous cohorts, however, a mechanistic link has proven elusive. This study tested the hypotheses that chronic exposure to the stress hormone, cortisol, causes telomere shortening and chromosome instability (CIN) in vitro, and that these effects would be further exacerbated by folate (vitamin B9) deficiency. Primary human lymphocytes were maintained in vitro for 12 days in medium containing either 25 nM folic acid (FA(low)) or 100 nM FA (FA(high)), together with either 0, 400, 1000 or 3500 nM cortisol. The interactive effects of cortisol and FA were examined by comparing telomere length (TL), biomarkers of DNA damage, and cytostasis. At day 12 TL was 5-17% longer in lymphocytes cultured in FA(low) conditions (mean ± SD;10.2% ± 1.6), compared with those in FA(high) medium (9.1% ± 1, p = 0.02). Refuting the hypothesis, TL was consistently greater in the presence of cortisol. The effect of FA deficiency on the frequency of DNA damage was significant for nucleoplasmic bridges, circular nuclei, micronuclei and nuclear buds, (p < 0.0001-0.001). The effect of cortisol, however, was negligible, only reaching statistical significance for the frequency of fused nuclei (p = 0.04). Cortisol was significantly associated with reduced cell division and growth and had an apparent protective effect on cell viability in the FA(low) conditions. Conclusions: Both chronic cortisol exposure, and folate deficiency, resulted in telomere elongation, however, the effect of cortisol was marginal relative to that of folate. Cortisol was not associated with increased chromosomal instability, but caused a significant reduction in cell division and growth. Together these results indicate that cortisol is not directly genotoxic and that the telomere shortening associated with increased psychological stress in vivo may not be explained by a direct effect of cortisol.
Collapse
Affiliation(s)
- Caroline Bull
- Nutritional Genomics and DNA Damage Diagnostics Laboratory, CSIRO Animal, Food and Health Sciences, Adelaide, South Australia, Australia
- Department of Microbiology & Immunology, School of Molecular & Biomedical Science, University of Adelaide, Adelaide, South Australia, Australia
- * E-mail:
| | - Helen Christensen
- Black Dog Institute, Prince of Wales Hospital, Randwick, New South Wales, Australia
| | - Michael Fenech
- Nutritional Genomics and DNA Damage Diagnostics Laboratory, CSIRO Animal, Food and Health Sciences, Adelaide, South Australia, Australia
| |
Collapse
|
31
|
Prognostic value of telomere function in gastric cancers with and without microsatellite instability. Eur J Gastroenterol Hepatol 2015; 27:162-9. [PMID: 25486025 DOI: 10.1097/meg.0000000000000250] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To identify molecular markers that may be useful in the selection of gastric cancer patients with different prognoses, we investigated telomere function in gastric cancers with and without microsatellite instability (MSI). MATERIALS AND METHODS We analyzed 83 gastric cancers and its paired-normal tissues to investigate MSI and telomere function. MSI was established using five polymorphic human repeat DNA markers. Telomere function was evaluated by determining telomerase activity, telomere length, and telomere-repeat factors 1 and 2 (TRF1 and TRF2) expression. RESULTS Patients with high microsatellite instability (MSI-H) gastric cancers showed a significantly better prognosis than those affected by microsatellite stable or low microsatellite instability (MSS/MSI-L) tumors (P = 0.03). The lowest expression levels of TRF1 and TRF2 were associated with MSI-H gastric cancers (P = 0.008 and 0.006, respectively). Moreover, a clear trend toward a worse prognosis was found in the group of patients who had tumors with the shortest telomeres (P = 0.01). Cox multivariate analysis showed that MSI emerged as a protective prognostic factor; MSS/MSI-L tumors conferred a significantly poor prognosis in patients (relative risk = 4.862-fold greater than the MSI-H group) (P = 0.033). Telomere length of gastric tumors less than 2.86 kbp was a factor that led to a poor prognosis (relative risk = 4.420, with respect to tumors showing telomere length ≥ 2.86 kbp) (P = 0.002). CONCLUSION We propose telomere status as a potential molecular marker with usefulness in the establishment of the prognosis of gastric cancers both for the mutator phenotype and for the suppressor pathway.
Collapse
|
32
|
Tanaka H, Beam MJ, Caruana K. The presence of telomere fusion in sporadic colon cancer independently of disease stage, TP53/KRAS mutation status, mean telomere length, and telomerase activity. Neoplasia 2014; 16:814-23. [PMID: 25379018 PMCID: PMC4212252 DOI: 10.1016/j.neo.2014.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 08/19/2014] [Accepted: 08/20/2014] [Indexed: 11/25/2022] Open
Abstract
Defects in telomere maintenance can result in telomere fusions that likely play a causative role in carcinogenesis by promoting genomic instability. However, this proposition remains to be fully understood in human colon carcinogenesis. In the present study, the temporal sequence of telomere dysfunction dynamics was delineated by analyzing telomere fusion, telomere length, telomerase activity, hotspot mutations in KRAS or BRAF, and TP53 of tissue samples obtained from 18 colon cancer patients. Our results revealed that both the deficiency of p53 and the shortening of mean telomere length were not necessary for producing telomere fusions in colon tissue. In five cases, telomere fusion was observed even in tissue adjacent to cancerous lesions, suggesting that genomic instability is initiated in pathologically non-cancerous lesions. The extent of mean telomere attrition increased with lymph node invasiveness of tumors, implying that mean telomere shortening correlates with colon cancer progression. Telomerase activity was relatively higher in most cancer tissues containing mutation(s) in KRAS or BRAF and/or TP53 compared to those without these hotspot mutations, suggesting that telomerase could become fully active at the late stage of colon cancer development. Interestingly, the majority of telomere fusion junctions in colon cancer appeared to be a chromatid-type containing chromosome 7q or 12q. In sum, this meticulous correlative study not only highlights the concept that telomere fusion is present in the early stages of cancer regardless of TP53/KRAS mutation status, mean telomere length, and telomerase activity, but also provides additional insights targeting key telomere fusion junctions which may have significant implications for colon cancer diagnoses.
Collapse
Affiliation(s)
- Hiromi Tanaka
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, IN, USA
| | - Matthew J Beam
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, IN, USA
| | - Kevin Caruana
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, IN, USA
| |
Collapse
|
33
|
Spiegel D. Minding the body: Psychotherapy and cancer survival. Br J Health Psychol 2013; 19:465-85. [DOI: 10.1111/bjhp.12061] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Revised: 06/24/2013] [Indexed: 01/02/2023]
Affiliation(s)
- David Spiegel
- Department of Psychiatry & Behavioral Sciences; Stanford University School of Medicine; Stanford California USA
| |
Collapse
|
34
|
A comprehensive model for the recognition of human telomeres by TRF1. J Mol Biol 2013; 425:2910-21. [PMID: 23702294 PMCID: PMC3776228 DOI: 10.1016/j.jmb.2013.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2013] [Revised: 05/10/2013] [Accepted: 05/13/2013] [Indexed: 01/18/2023]
Abstract
Eukaryotic chromosomes are capped by telomeres, nucleoprotein complexes that prevent chromosome end-to-end fusions and control cell ageing. Two proteins in this complex, telomere repeat binding factors (TRF1 and TRF2), specifically recognise the double-stranded TTAGGG tandem repeat sequence. TRF1 is a homodimer with roles governing DNA architecture and negatively regulating telomere length. We explore the conformational space of this protein-DNA complex using molecular dynamics and, for the first time, generate a complete model of TRF1-DNA recognition that has not been possible on the basis of crystallographic and NMR data alone. The results reconcile previous conflicting experimental models for the sequence selectivity of the recognition process, by confirming many of the findings while identifying important new interactions and behaviour. This improved characterisation also reveals extensive indirect readout, which suggests that recognition will be affected by changes to DNA helical parameters such as bending.
Collapse
|
35
|
Savage SA, Gadalla SM, Chanock SJ. The long and short of telomeres and cancer association studies. J Natl Cancer Inst 2013; 105:448-9. [PMID: 23468461 PMCID: PMC3614507 DOI: 10.1093/jnci/djt041] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2013] [Indexed: 01/02/2023] Open
|
36
|
Williamson SR, Zhang S, Lopez-Beltran A, Montironi R, Wang M, Cheng L. Telomere shortening distinguishes inverted urothelial neoplasms. Histopathology 2013; 62:595-601. [DOI: 10.1111/his.12030] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Accepted: 09/14/2012] [Indexed: 01/18/2023]
Affiliation(s)
- Sean R Williamson
- Department of Pathology and Laboratory Medicine; Indiana University School of Medicine; Indianapolis; IN; USA
| | - Shaobo Zhang
- Department of Pathology and Laboratory Medicine; Indiana University School of Medicine; Indianapolis; IN; USA
| | | | - Rodolfo Montironi
- Institute of Pathological Anatomy and Histopathology; School of Medicine, Polytechnic University of the Marche Region (Ancona); United Hospitals; Ancona; Italy
| | - Mingsheng Wang
- Department of Pathology and Laboratory Medicine; Indiana University School of Medicine; Indianapolis; IN; USA
| | | |
Collapse
|
37
|
Svenson U, Grönlund E, Söderström I, Sitaram RT, Ljungberg B, Roos G. Telomere length in relation to immunological parameters in patients with renal cell carcinoma. PLoS One 2013; 8:e55543. [PMID: 23383336 PMCID: PMC3562315 DOI: 10.1371/journal.pone.0055543] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 12/28/2012] [Indexed: 12/13/2022] Open
Abstract
Over the last decade, telomere length (TL) has gained attention as a potential biomarker in cancer disease. We previously reported that long blood TL was associated with a poorer outcome in patients with breast cancer and renal cell carcinoma. Based on these findings, we hypothesized that certain immunological components may have an impact on TL dynamics in cancer patients. One aim of the present study was to investigate a possible association between serum cytokines and TL of peripheral blood cells, tumors and corresponding kidney cortex, in patients with clear cell renal cell carcinoma. For this purpose, a multiplex cytokine assay was used. Correlation analysis revealed significant positive correlations between tumor TL and peripheral levels of three cytokines (IL-7, IL-8 and IL-10). In a parallel patient group with various kidney tumors, TL was investigated in whole blood and in immune cell subsets in relation to peripheral levels of regulatory T cells (Tregs). A significant positive association was found between whole blood TL and Treg levels. However, the strongest correlation was found between Tregs and TL of the T lymphocyte fraction. Thus, patients with higher Treg levels displayed longer T cell telomeres, which might reflect a suppressed immune system with fewer cell divisions and hence less telomere shortening. These results are in line with our earlier observation that long blood TL is an unfavorable prognostic factor for cancer-specific survival. In summary, we here show that immunological components are associated with TL in patients with renal cell carcinoma, providing further insight into the field of telomere biology in cancer.
Collapse
Affiliation(s)
- Ulrika Svenson
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | | | - Ingegerd Söderström
- Department of Public Health and Clinical Medicine, Medicine, Umeå University, Umeå, Sweden
| | - Raviprakash T. Sitaram
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden
| | - Börje Ljungberg
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden
| | - Göran Roos
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
- * E-mail:
| |
Collapse
|
38
|
Shen YC, Chen ZN, Yang T, Chen L, Wang T, Wen FQ, Yi Q. Telomerase activity assay for the diagnosis of malignant pleural effusion: A meta-analysis. Exp Ther Med 2012. [PMID: 23181123 PMCID: PMC3503797 DOI: 10.3892/etm.2012.623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The telomerase activity assay has been established for the detection of malignant pleural effusion (MPE), however, the overall diagnostic accuracy of the telomerase activity assay for MPE remains unclear. We performed a systematic search in the Pubmed, Embase and Cochrane databases to identify published studies that have evaluated the diagnostic role of the telomerase activity assay for MPE. Sensitivity, specificity and other measures of accuracy of the telomerase activity assay in the diagnosis of MPE were pooled using the random effects models. A summary receiver operating characteristic (SROC) curve was used to summarize overall test performance. A total of eight studies met the inclusion criteria for the meta-analysis. The pooled sensitivity and specificity for diagnosing MPE were 0.76 [95% confidence intervals (CI), 0.72–0.80] and 0.87 (95% CI, 0.83–0.91), respectively. The positive likelihood ratio was 5.19 (95% CI, 2.36–11.42), the negative likelihood ratio was 0.25 (95% CI, 0.11–0.53) and the diagnostic odds ratio was 23.18 (95% CI, 6.11–87.83). The area under the SROC curve was 0.92. The telomerase activity assay plays a role in the diagnosis of MPE with a relatively high specificity. The results of a telomerase activity assay should be interpreted together with the combination of other test results and clinical findings.
Collapse
Affiliation(s)
- Yong-Chun Shen
- Division of Pulmonary Diseases, State Key Laboratory of Biotherapy of China, Department of Respiratory Medicine, West China Hospital of Sichuan University
| | | | | | | | | | | | | |
Collapse
|
39
|
Expression of telomeres in astrocytoma WHO grade 2 to 4: TERRA level correlates with telomere length, telomerase activity, and advanced clinical grade. Transl Oncol 2012; 5:56-65. [PMID: 22348177 DOI: 10.1593/tlo.11202] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 10/20/2011] [Accepted: 10/25/2011] [Indexed: 12/19/2022] Open
Abstract
Cancer cells bypass replicative senescence, the major barrier to tumor progression, by using telomerase or alternative lengthening of telomeres (ALT) as telomere maintenance mechanisms (TMMs). Correlation between ALT and patient survival was demonstrated for high-grade astrocytomas. Transcription from subtelomeres produces telomeric repeat-containing RNA (TERRA), a natural inhibitor of telomerase activity (TA). This led us to evaluate correlations of TERRA and TMM with tumor grade and outcome in astrocytoma patients. SYBR Green real-time reverse transcription-polymerase chain reaction assays for quantitation of total and chromosome 2p and 18p specific TERRA levels were developed. Tumor samples from 46 patients with astrocytoma grade 2 to 4, tissue controls, and cell lines were assessed. TMMs were evaluated by measuring TA and by detecting long telomeres due to ALT. In glioblastoma multiforme (GBM) grade 4, total TERRA levels were similar to cell lines but 14-, 31-, and 313-fold lower compared with grade 3, grade 2, and nonmalignant tissue, respectively. Total TERRA levels differed from chromosomal levels. Low 2p TERRA levels correlated with dense promoter methylation of subtelomeric CpG islands, indicating that TERRA expression in gliomas may be chromosome specific and epigenetically regulated. Total TERRA levels correlated with diagnosis, with low or absent TA and the presence of ALT, and were tentatively associated with favorable patient prognosis in our cohort (P = .06). TA and short telomeres identified a subset of GBM with a median survival of only 14.8 months. TERRA and TA may be prognostic in astrocytic tumors.
Collapse
|