1
|
Saad FA. Gene Therapy for Skin Aging. Curr Gene Ther 2024; 25:2-9. [PMID: 38529607 DOI: 10.2174/0115665232286489240320051925] [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: 10/09/2023] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 03/27/2024]
Abstract
Extrinsic and intrinsic factors contribute to skin aging; nonetheless, they are intertwined. Moreover, intrinsic skin aging mirrors age-related declines in the entire human body's internal organs. There is evidence that skin appearance is an indicator of the general health of somebody or a visual certificate of health. Earlier, it was apparent that the intrinsic factors are unalterable, but the sparkling of skin aging gene therapy on the horizon is changing this narrative. Skin aging gene therapy offers tools for skin rejuvenation, natural beauty restoration, and therapy for diseases affecting the entire skin. However, skin aging gene therapy is an arduous and sophisticated task relying on precise interim stimulation of telomerase to extend telomeres and wend back the biological clock in the hopes to find the fountain of youth, while preserving cells innate biological features. Finding the hidden fountain of youth will be a remarkable discovery for promoting aesthetics medicine, genecosmetics, and healthy aging. Caloric restriction offers ultimate health benefits and a reproducible way to promote longevity in mammals, while delaying age-related diseases. Moreover, exercise further enhances these health benefits. This article highlights the potential of skin aging gene therapy and foretells the emerging dawn of the genecosmetics era.
Collapse
Affiliation(s)
- Fawzy A Saad
- Department of Gene Therapy, Saad Pharmaceuticals, Juhkentali 8, Tallinn, 10132, Estonia
| |
Collapse
|
2
|
Sun H, Chen G, Guo B, Lv S, Yuan G. Potential clinical treatment prospects behind the molecular mechanism of alternative lengthening of telomeres (ALT). J Cancer 2023; 14:417-433. [PMID: 36860927 PMCID: PMC9969575 DOI: 10.7150/jca.80097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 12/25/2022] [Indexed: 02/04/2023] Open
Abstract
Normal somatic cells inevitably experience replicative stress and senescence during proliferation. Somatic cell carcinogenesis can be prevented in part by limiting the reproduction of damaged or old cells and removing them from the cell cycle [1, 2]. However, Cancer cells must overcome the issues of replication pressure and senescence as well as preserve telomere length in order to achieve immortality, in contrast to normal somatic cells [1, 2]. Although telomerase accounts for the bulk of telomere lengthening methods in human cancer cells, there is a non-negligible portion of telomere lengthening pathways that depend on alternative lengthening of telomeres (ALT) [3]. For the selection of novel possible therapeutic targets for ALT-related disorders, a thorough understanding of the molecular biology of these diseases is crucial [4]. The roles of ALT, typical ALT tumor cell traits, the pathophysiology and molecular mechanisms of ALT tumor disorders, such as adrenocortical carcinoma (ACC), are all summarized in this work. Additionally, this research compiles as many of its hypothetically viable but unproven treatment targets as it can (ALT-associated PML bodies (APB), etc.). This review is intended to contribute as much as possible to the development of research, while also trying to provide a partial information for prospective investigations on ALT pathways and associated diseases.
Collapse
Affiliation(s)
- Haolu Sun
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230011, China
| | - Guijuan Chen
- School of Environment and Chemical Engineering, Anhui Vocational and Technical College, Hefei, 230011, China
| | - Baochang Guo
- Rehabilitation Department of Traditional Chinese Medicine, 969 Hospital of the Joint Support Force of the Chinese People's Liberation Army, Hohhot, 010000, China
| | - Shushu Lv
- Department of Pathology, The First Affiliated Hospital of Huzhou University, Huzhou 313000, China
| | - Guojun Yuan
- School of Environment and Chemical Engineering, Anhui Vocational and Technical College, Hefei, 230011, China
| |
Collapse
|
3
|
Denham J, Sellami M. Exercise training increases telomerase reverse transcriptase gene expression and telomerase activity: A systematic review and meta-analysis. Ageing Res Rev 2021; 70:101411. [PMID: 34284150 DOI: 10.1016/j.arr.2021.101411] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/03/2021] [Accepted: 07/14/2021] [Indexed: 01/12/2023]
Abstract
Telomeres protect genomic stability and shortening is one of the hallmarks of ageing. Telomerase reverse transcriptase (TERT) is the major protein component of telomerase, which elongates telomeres. Given that short telomeres are linked to a host of chronic diseases and the therapeutic potential of telomerase-based therapies as treatments and a strategy to extend lifespan, lifestyle factors that increase TERT gene expression and telomerase activity could attenuate telomere attrition and contribute to healthy biological ageing. Physical activity and maximal aerobic fitness are associated with telomere maintenance, yet the molecular mechanisms remain unclear. Therefore, the purpose of this systematic review and meta-analysis was to identify the influence of a single bout of exercise and long-term exercise training on TERT expression and telomerase activity. A search of human and rodent trials using the PubMed, Scopus, Science Direct and Embase databases was performed. Based on findings from the identified and eligible trials, both a single bout of exercise (n; standardised mean difference [95%CI]: 5; SMD: 1.19 [0.41-1.97], p = 0.003) and long-term exercise training (10; 0.31 [0.03-0.60], p = 0.03) up-regulates TERT and telomerase activity in non-cancerous somatic cells. As human and rodent studies were included in the meta-analyses both exhibited heterogeneity (I2 = 55-87%, p < 0.05). Endurance athletes also exhibited increased leukocyte TERT and telomerase activity compared to their inactive counterparts. These findings suggest exercise training as an inexpensive lifestyle factor that increases TERT expression and telomerase activity. Regular exercise training could attenuate telomere attrition through a telomerase-dependent mechanism and ultimately extend health-span and longevity.
Collapse
Affiliation(s)
- Joshua Denham
- RMIT University, School of Health and Biomedical Sciences, Melbourne, Victoria, Australia.
| | - Maha Sellami
- Physical Education Department (PE), College of Education, Qatar University, Doha, Qatar
| |
Collapse
|
4
|
Yang J, Liu M, Hong D, Zeng M, Zhang X. The Paradoxical Role of Cellular Senescence in Cancer. Front Cell Dev Biol 2021; 9:722205. [PMID: 34458273 PMCID: PMC8388842 DOI: 10.3389/fcell.2021.722205] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/20/2021] [Indexed: 12/12/2022] Open
Abstract
Cellular senescence occurs in proliferating cells as a consequence of various triggers including telomere shortening, DNA damage, and inappropriate expression of oncogenes. The senescent state is accompanied by failure to reenter the cell cycle under mitotic stimulation, resistance to cell death and enhanced secretory phenotype. A growing number of studies have convincingly demonstrated a paradoxical role for spontaneous senescence and therapy-induced senescence (TIS), that senescence may involve both cancer prevention and cancer aggressiveness. Cellular senescence was initially described as a physiological suppressor mechanism of tumor cells, because cancer development requires cell proliferation. However, there is growing evidence that senescent cells may contribute to oncogenesis, partly in a senescence-associated secretory phenotype (SASP)-dependent manner. On the one hand, SASP prevents cell division and promotes immune clearance of damaged cells, thereby avoiding tumor development. On the other hand, SASP contributes to tumor progression and relapse through creating an immunosuppressive environment. In this review, we performed a review to summarize both bright and dark sides of senescence in cancer, and the strategies to handle senescence in cancer therapy were also discussed.
Collapse
Affiliation(s)
- Jing Yang
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mengmeng Liu
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Dongchun Hong
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Musheng Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xing Zhang
- Melanoma and Sarcoma Medical Oncology Unit, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
5
|
Zhu X, Xu T, Zhu J. The regulatory function of tandem repeat VNTR2-1 in hTERT gene involves basic Helix-loop-helix family transcription factors. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2021; 62:338-349. [PMID: 34115413 PMCID: PMC9648534 DOI: 10.1002/em.22447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/06/2021] [Accepted: 06/09/2021] [Indexed: 06/12/2023]
Abstract
Telomerase is a ribonucleoprotein enzyme that synthesizes telomere end sequence. The expression of hTERT gene, encoding the catalytic subunit of human telomerase, is restricted to highly proliferative tissues and is undetectable in most somatic cells. Abnormal activation of hTERT gene is found in 90% of human tumors. Previously, we identified tandem repeat of 42-bp/unit, VNTR2-1, in intron 2 of the hTERT gene, as a novel regulatory element important for hTERT transcription in cancer cells. In the current study, we found that multiple 42-bp repeats of VNTR2-1 activated luciferase gene in reporter plasmids. Mutation of the predicted cis-regulatory elements within the 42-bp repeats, including a E-box motif, resulted in a partial or complete loss of its enhancer activity. Moreover, MYC family proteins, c-MYC, MAX, and MNT, regulated hTERT gene transcription through both VNTR2-1 and E-boxes at the proximal hTERT promoter. Chromatin segmentation analysis of published ChIP-sequencing data from K562 cells indicated that VNTR2-1 was a bivalent enhancer. In telomerase-expressing human melanoma cell line MelJuSo, deletion of VNTR2-1 caused the hTERT promoter chromatin status to change from an active state to a repressed state, accompanied by increases of H3K27me3 and H3K9me3 marks. Therefore, we provided additional evidence for VNTR2-1 as a functional regulatory element that regulated hTERT expression by MYC family transcription factors. These results have improved our knowledge on the functions of repetitive genomic DNAs and the regulatory mechanisms of human telomerase gene.
Collapse
Affiliation(s)
| | | | - Jiyue Zhu
- To whom correspondence should be addressed. Tel: 1-509-368-6565; Fax: 1-509-368-6561; ; 412E Spokane Falls Blvd, PBS323, Spokane, WA, 99202, USA
| |
Collapse
|
6
|
Xu T, Cheng D, Zhao Y, Zhang J, Zhu X, Zhang F, Chen G, Wang Y, Yan X, Robertson GP, Gaddameedhi S, Lazarus P, Wang S, Zhu J. Polymorphic tandem DNA repeats activate the human telomerase reverse transcriptase gene. Proc Natl Acad Sci U S A 2021; 118:e2019043118. [PMID: 34155099 PMCID: PMC8256013 DOI: 10.1073/pnas.2019043118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Multiple independent sequence variants of the hTERT locus have been associated with telomere length and cancer risks in genome-wide association studies. Here, we identified an intronic variable number tandem repeat, VNTR2-1, as an enhancer-like element, which activated hTERT transcription in a cell in a chromatin-dependent manner. VNTR2-1, consisting of 42-bp repeats with an array of enhancer boxes, cooperated with the proximal promoter in the regulation of hTERT transcription by basic helix-loop-helix transcription factors and maintained hTERT expression during embryonic stem-cell differentiation. Genomic deletion of VNTR2-1 in MelJuSo melanoma cells markedly reduced hTERT transcription, leading to telomere shortening, cellular senescence, and impairment of xenograft tumor growth. Interestingly, VNTR2-1 lengths varied widely in human populations; hTERT alleles with shorter VNTR2-1 were underrepresented in African American centenarians, indicating its role in human aging. Therefore, this polymorphic element is likely a missing link in the telomerase regulatory network and a molecular basis for genetic diversities of telomere homeostasis and age-related disease susceptibilities.
Collapse
Affiliation(s)
- Tao Xu
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99210
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - De Cheng
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99210
| | - Yuanjun Zhao
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Jinglong Zhang
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99210
| | - Xiaolu Zhu
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99210
| | - Fan Zhang
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99210
| | - Gang Chen
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99210
| | - Yang Wang
- College of Life Science, Northeast Forestry University, Harbin 150040, China
| | - Xiufeng Yan
- College of Life Science, Northeast Forestry University, Harbin 150040, China
- College of Life and Environmental Science, Wenzhou University, Chashan University Town, Wenzhou 325035, China
| | - Gavin P Robertson
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Dermatology, Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Surgery, Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Shobhan Gaddameedhi
- Center for Human Health and the Environment, Department of Biological Sciences, North Carolina State University, Raleigh, NC 27606
| | - Philip Lazarus
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99210
| | - Shuwen Wang
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99210
| | - Jiyue Zhu
- College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, WA 99210;
| |
Collapse
|
7
|
Zarei M, Najafi M, Movahedi E, Javanbakht MH, Choi YH, Yaseri M, Shirvani A, Sellke FW, Stranges S. The predictive role of circulating telomerase and vitamin D for long-term survival in patients undergoing coronary artery bypass grafting surgery (CABG). PLoS One 2020; 15:e0237477. [PMID: 32790742 PMCID: PMC7425905 DOI: 10.1371/journal.pone.0237477] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/27/2020] [Indexed: 11/21/2022] Open
Abstract
BACKGROUNDS Cardiovascular disease (CVD) is the leading cause of mortality all over the globe. Inflammation is believed to play a pivotal role in the pathophysiology of CVD. While there are studies on the interrelationship of telomerase and vitamin D and their involvement in CVD, their independent contributions to long-term outcomes in patients with CVD are not well-defined. This study aimed to investigate the association of both telomerase and vitamin D concentrations with 10-year survival among candidates of coronary artery bypass grafting (CABG) surgery. METHODS Participants were 404 patients from Tehran Heart Center-Coronary Outcome Measurement (THC-COM) cohort who were recruited from CABG surgery candidates in 2006. In addition to demographic and clinical data including risk factors for coronary artery disease, laboratory parameters such as markers of inflammation as well as baseline serum 25-hydroxy vitamin D [25(OH)D] and telomerase concentrations were measured. Cardiac function indexes alongside outcome measures such as mortality and survival days were recorded for every patient up to 10 years after CABG. Cox-proportional hazard model was used to study the association between all-cause mortality and research parameters. RESULTS The mean serum telomerase enzyme level was 24.92 ±21.4 nmol/L and the mean serum 25(OH)D was 27.27±10.3 ng/mL. 10-year mortality was reported in 64 (15.8%) patients. 25(OH)D was categorized into three groups (<20, 20-30, and >30) and the cut-point for telomerase was set at 25.0 nmol/L. In Cox regression analysis, higher levels of telomerase (>25 nmol/L) were significantly associated with longer survival (p = 0.041), whereas 25(OH)D concentrations were not associated with survival time. Further analysis showed that telomerase concentrations significantly predicted survival only in the presence of insufficient levels of 25(OH)D (20-30 ng/mL) (p = 0.037). CONCLUSIONS Telomerase can be regarded as a potential predictor of long-term outcomes in patients who underwent CABG. However, the association of telomerase with the mortality may be modified by vitamin D concentrations.
Collapse
Affiliation(s)
- Mahtab Zarei
- Department of Cellular and Molecular Nutrition, Faculty of Nutritional Sciences & Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Najafi
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anesthesiology, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elnaz Movahedi
- Department of Cellular and Molecular Nutrition, Faculty of Nutritional Sciences & Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamad Hassan Javanbakht
- Department of Cellular and Molecular Nutrition, Faculty of Nutritional Sciences & Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Yun-Hee Choi
- Department of Epidemiology and Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Mehdi Yaseri
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Shirvani
- Department of Medicine, Section of Endocrinology, Nutrition, and Diabetes, Vitamin D, Skin and Bone Research Laboratory, Boston University Medical Campus, Boston, Massachusetts, United States of America
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Saverio Stranges
- Department of Epidemiology and Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Family Medicine, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| |
Collapse
|
8
|
Bahrami A, Bo S, Jamialahmadi T, Sahebkar A. Effects of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors on ageing: Molecular mechanisms. Ageing Res Rev 2020; 58:101024. [PMID: 32006687 DOI: 10.1016/j.arr.2020.101024] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 12/11/2019] [Accepted: 01/27/2020] [Indexed: 12/15/2022]
Abstract
Human ageing is determined by degenerative alterations and processes with different manifestations such as gradual organ dysfunction, tissue function loss, increased population of aged (senescent) cells, incapability of maintaining homeostasis and reduced repair capacity, which collectively lead to an increased risk of diseases and death. The inhibitors of HMG-CoA reductase (statins) are the most widely used lipid-lowering agents, which can reduce cardiovascular morbidity and mortality. Accumulating evidence has documented several pleiotropic effects of statins in addition to their lipid-lowering properties. Recently, several studies have highlighted that statins may have the potential to delay the ageing process and inhibit the onset of senescence. In this review, we focused on the anti-ageing mechanisms of statin drugs and their effects on cardiovascular and non-cardiovascular diseases.
Collapse
|
9
|
Perera ON, Sobinoff AP, Teber ET, Harman A, Maritz MF, Yang SF, Pickett HA, Cesare AJ, Arthur JW, MacKenzie KL, Bryan TM. Telomerase promotes formation of a telomere protective complex in cancer cells. SCIENCE ADVANCES 2019; 5:eaav4409. [PMID: 31616780 PMCID: PMC6774720 DOI: 10.1126/sciadv.aav4409] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 09/09/2019] [Indexed: 05/04/2023]
Abstract
Telomerase is a ribonucleoprotein complex that catalyzes addition of telomeric DNA repeats to maintain telomeres in replicating cells. Here, we demonstrate that the telomerase protein hTERT performs an additional role at telomeres that is independent of telomerase catalytic activity yet essential for telomere integrity and cell proliferation. Short-term depletion of endogenous hTERT reduced the levels of heat shock protein 70 (Hsp70-1) and the telomere protective protein Apollo at telomeres, and induced telomere deprotection and cell cycle arrest, in the absence of telomere shortening. Short-term expression of hTERT promoted colocalization of Hsp70-1 with telomeres and Apollo and reduced numbers of deprotected telomeres, in a manner independent of telomerase catalytic activity. These data reveal a previously unidentified noncanonical function of hTERT that promotes formation of a telomere protective complex containing Hsp70-1 and Apollo and is essential for sustained proliferation of telomerase-positive cancer cells, likely contributing to the known cancer-promoting effects of both hTERT and Hsp70-1.
Collapse
Affiliation(s)
- Omesha N. Perera
- Cell Biology Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Alexander P. Sobinoff
- Telomere Length Regulation Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Erdahl T. Teber
- Bioinformatics Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Ashley Harman
- Cell Biology Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Michelle F. Maritz
- Children’s Cancer Institute, School of Women’s and Children’s Health, University of NSW, NSW 2052, Australia
| | - Sile F. Yang
- Telomere Length Regulation Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Hilda A. Pickett
- Telomere Length Regulation Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Anthony J. Cesare
- Genome Integrity Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Jonathan W. Arthur
- Bioinformatics Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
| | - Karen L. MacKenzie
- Children’s Cancer Institute, School of Women’s and Children’s Health, University of NSW, NSW 2052, Australia
| | - Tracy M. Bryan
- Cell Biology Unit, Children’s Medical Research Institute, Faculty of Medicine and Health, University of Sydney, Westmead, NSW 2145, Australia
- Corresponding author.
| |
Collapse
|
10
|
Engineering a humanized telomerase reverse transcriptase gene in mouse embryonic stem cells. Sci Rep 2019; 9:9683. [PMID: 31273310 PMCID: PMC6609615 DOI: 10.1038/s41598-019-46160-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 06/24/2019] [Indexed: 12/18/2022] Open
Abstract
Telomerase is expressed in adult mouse, but not in most human, tissues and mouse telomeres are much longer than those in humans. This interspecies difference of telomere homeostasis poses a challenge in modeling human diseases using laboratory mice. Using chromatinized bacterial artificial chromosome reporters, we discovered that the 5′ intergenic region, introns 2 and 6 of human telomerase gene (hTERT) were critical for regulating its promoter in somatic cells. Accordingly, we engineered a humanized gene, hmTert, by knocking-in a 47-kilobase hybrid fragment containing these human non-coding sequences into the mTert locus in mouse embryonic stem cells (mESCs). The hmTert gene, encoding the wildtype mTert protein, was fully functional, as a mESC line with homozygous hmTert alleles proliferated for over 400 population doublings without exhibiting chromosomal abnormalities. Like human ESCs, the engineered mESCs contained high telomerase activity, which was repressed upon their differentiation into fibroblast-like cells in a histone deacetylase-dependent manner. Fibroblast-like cells differentiated from these mESCs contained little telomerase activity. Thus, telomerase in mESCs with the hmTert alleles was subjected to human-like regulation. Our study revealed a novel approach to engineer a humanized telomerase gene in mice, achieving a milestone in creating a mouse model with humanized telomere homeostasis.
Collapse
|
11
|
Wazir U, Orakzai MMAW, Martin TA, Jiang WG, Mokbel K. Correlation of TERT and Stem Cell Markers in the Context of Human Breast Cancer. Cancer Genomics Proteomics 2019; 16:121-127. [PMID: 30850363 DOI: 10.21873/cgp.20117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/24/2019] [Accepted: 02/26/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Telomerase reverse transcriptase (TERT) has a well-known role in carcinogenesis due to its functions in inducing cell immortality and preventing senescence. In this study, the relationships between TERT and a panel of known stem cell markers was examined in order to direct future enquiries into the role of 'stem-ness' in human breast cancer. MATERIALS AND METHODS Breast cancer tissues (n=124) and adjacent normal tissues (n=30) underwent reverse transcription and quantitative polymerase chain reaction. Transcript levels were analyzed for the correlation with that of TERT. RESULTS A significant direct correlation was found in cancerous tissue between TERT and BMI1 proto-oncogene polycomb ring finger 4 (BMI1; n=88, p<0.001), nestin (NES; n=88, p<0.001), POU domain, class 5, transcription factor 1 (POU5F1; n=88, p<0.001), aldehyde dehydrogenase 1 family member A2 (ALDH1A2; n=87, p=0.0298), cyclin-dependent kinase inhibitor 1A (CDKN1A; n=88, p<0.001), integrin subunit beta 1 (ITGNB1; n=88, p<0.001), integrin subunit alpha 6 (ITGA6; n=88, p<0.001), cluster of differentiation antigen 24 (CD24; n=88, p=0.0114), MET proto-oncogene (MET; n=78, p<0.001) and noggin (NOG; n=88, p<0.001). CONCLUSION The evidence presented in this article of possible interactions between TERT and a discrete subset of known stem cell markers would significantly contribute to further enquiries regarding clonal dynamics in the context of human breast cancer.
Collapse
Affiliation(s)
- Umar Wazir
- The London Breast Institute, Princess Grace Hospital, London, U.K.,Department of General Surgery, Khyber Teaching Hospital, Peshawar, Pakistan
| | | | - Tracey Amanda Martin
- Cardiff-China Cancer Research Collaboration, Cardiff University School of Medicine, Cardiff University, Cardiff, U.K
| | - Wen G Jiang
- Cardiff-China Cancer Research Collaboration, Cardiff University School of Medicine, Cardiff University, Cardiff, U.K
| | - Kefah Mokbel
- The London Breast Institute, Princess Grace Hospital, London, U.K. .,Department of General Surgery, Khyber Teaching Hospital, Peshawar, Pakistan
| |
Collapse
|
12
|
Sato M, Saitoh I, Inada E, Nakamura S, Watanabe S. Potential for Isolation of Immortalized Hepatocyte Cell Lines by Liver-Directed In Vivo Gene Delivery of Transposons in Mice. Stem Cells Int 2019; 2019:5129526. [PMID: 31281376 PMCID: PMC6589260 DOI: 10.1155/2019/5129526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Accepted: 05/06/2019] [Indexed: 12/14/2022] Open
Abstract
Isolation of hepatocytes and their culture in vitro represent important avenues to explore the function of such cells. However, these studies are often difficult to perform because of the inability of hepatocytes to proliferate in vitro. Immortalization of isolated hepatocytes is thus an important step toward continuous in vitro culture. For cellular immortalization, integration of relevant genes into the host chromosomes is a prerequisite. Transposons, which are mobile genetic elements, are known to facilitate integration of genes of interest (GOI) into chromosomes in vitro and in vivo. Here, we proposed that a combination of transposon- and liver-directed introduction of nucleic acids may confer acquisition of unlimited cellular proliferative potential on hepatocytes, enabling the possible isolation of immortalized hepatocyte cell lines, which has often failed using more traditional immortalization methods.
Collapse
Affiliation(s)
- Masahiro Sato
- Section of Gene Expression Regulation, Frontier Science Research Center, Kagoshima University, Kagoshima 890-8544, Japan
| | - Issei Saitoh
- Division of Pediatric Dentistry, Graduate School of Medical and Dental Science, Niigata University, Niigata 951-8514, Japan
| | - Emi Inada
- Department of Pediatric Dentistry, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
| | - Shingo Nakamura
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Saitama 359-8513, Japan
| | - Satoshi Watanabe
- Animal Genome Unit, Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization (NARO), Tsukuba, Ibaraki 305-0901, Japan
| |
Collapse
|
13
|
Lin S, Wei J, Wunderlich M, Chou FS, Mulloy JC. Immortalization of human AE pre-leukemia cells by hTERT allows leukemic transformation. Oncotarget 2018; 7:55939-55950. [PMID: 27509060 PMCID: PMC5302887 DOI: 10.18632/oncotarget.11093] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 06/13/2016] [Indexed: 01/21/2023] Open
Abstract
Human CD34+ hematopoietic stem and progenitor cells (HSPC) expressing fusion protein AML1-ETO (AE), generated by the t(8;21)(q22;q22) rearrangement, manifest enhanced self-renewal and dysregulated differentiation without leukemic transformation, representing a pre-leukemia stage. Enabling replicative immortalization via telomerase reactivation is a crucial step in cancer development. However, AE expression alone is not sufficient to maintain high telomerase activity to immortalize human HSPC cells, which may hamper transformation. Here, we investigated the cooperativity of telomerase reverse transcriptase (hTERT), the catalytic subunit of telomerase, and AE in disease progression. Enforced expression of hTERT immortalized human AE pre-leukemia cells in a telomere-lengthening independent manner, and improved the pre-leukemia stem cell function by enhancing cell proliferation and survival. AE-hTERT cells retained cytokine dependency and multi-lineage differentiation potential similar to parental AE clones. Over the short-term, AE-hTERT cells did not show features of stepwise transformation, with no leukemogenecity evident upon initial injection into immunodeficient mice. Strikingly, after extended culture, we observed full transformation of one AE-hTERT clone, which recapitulated the disease evolution process in patients and emphasizes the importance of acquiring cooperating mutations in t(8;21) AML leukemogenesis. In summary, achieving unlimited proliferative potential via hTERT activation, and thereby allowing for acquisition of additional mutations, is a critical link for transition from pre-leukemia to overt disease in human cells. AE-hTERT cells represent a tractable model to study cooperating genetic lesions important for t(8;21) AML disease progression.
Collapse
Affiliation(s)
- Shan Lin
- Cancer and Blood Disease Institute, Cincinnati Children's Hospital Research Center, Cincinnati, OH, USA
| | - Junping Wei
- Cancer and Blood Disease Institute, Cincinnati Children's Hospital Research Center, Cincinnati, OH, USA
| | - Mark Wunderlich
- Cancer and Blood Disease Institute, Cincinnati Children's Hospital Research Center, Cincinnati, OH, USA
| | - Fu-Sheng Chou
- Cancer and Blood Disease Institute, Cincinnati Children's Hospital Research Center, Cincinnati, OH, USA
| | - James C Mulloy
- Cancer and Blood Disease Institute, Cincinnati Children's Hospital Research Center, Cincinnati, OH, USA
| |
Collapse
|
14
|
Regulation of human and mouse telomerase genes by genomic contexts and transcription factors during embryonic stem cell differentiation. Sci Rep 2017; 7:16444. [PMID: 29180668 PMCID: PMC5703907 DOI: 10.1038/s41598-017-16764-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/16/2017] [Indexed: 12/14/2022] Open
Abstract
Differential regulation of telomerase reverse transcriptase (TERT) genes contribute to distinct aging and tumorigenic processes in humans and mice. To study TERT regulation, we generated mouse embryonic stem cell (ESC) lines containing single-copy bacterial artificial chromosome (BAC) reporters, covering hTERT and mTERT genes and their neighboring loci, via recombinase-mediated BAC targeting. ESC lines with chimeric BACs, in which two TERT promoters were swapped, were also generated. Using these chromatinized BACs, we showed that hTERT silencing during differentiation to embryoid bodies (EBs) and to fibroblast-like cells was driven by the human-specific genomic context and accompanied by increases of repressive epigenetic marks, H3K9me3 and H3K27me3, near its promoter. Conversely, the mouse genomic context did not repress TERT transcription until late during differentiation. The hTERT promoter was more active than its mouse counterpart when compared in the same genomic contexts. Mutations of E-box and E2F consensus sites at the promoter had little effect on hTERT transcription in ESCs. However, the mutant promoters were rapidly silenced upon EB differentiation, indicating that transcription factors (TFs) bound to these sites were critical in maintaining hTERT transcription during differentiation. Together, our study revealed a dynamic hTERT regulation by chromatin environment and promoter-bound TFs during ESC differentiation.
Collapse
|
15
|
Dolivo D, Hernandez S, Dominko T. Cellular lifespan and senescence: a complex balance between multiple cellular pathways. Bioessays 2017; 38 Suppl 1:S33-44. [PMID: 27417120 DOI: 10.1002/bies.201670906] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2015] [Revised: 09/14/2015] [Accepted: 09/17/2015] [Indexed: 11/09/2022]
Abstract
The study of cellular senescence and proliferative lifespan is becoming increasingly important because of the promises of autologous cell therapy, the need for model systems for tissue disease and the implication of senescent cell phenotypes in organismal disease states such as sarcopenia, diabetes and various cancers, among others. Here, we explain the concepts of proliferative cellular lifespan and cellular senescence, and we present factors that have been shown to mediate cellular lifespan positively or negatively. We review much recent literature and present potential molecular mechanisms by which lifespan mediation occurs, drawing from the fields of telomere biology, metabolism, NAD(+) and sirtuin biology, growth factor signaling and oxygen and antioxidants. We conclude that cellular lifespan and senescence are complex concepts that are governed by multiple independent and interdependent pathways, and that greater understanding of these pathways, their interactions and their convergence upon specific cellular phenotypes may lead to viable therapies for tissue regeneration and treatment of age-related pathologies, which are caused by or exacerbated by senescent cells in vivo.
Collapse
Affiliation(s)
- David Dolivo
- Biology and Biotechnology Department, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Sarah Hernandez
- Biology and Biotechnology Department, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Tanja Dominko
- Biology and Biotechnology Department, Worcester Polytechnic Institute, Worcester, MA, USA
| |
Collapse
|
16
|
Abstract
The number of diabetic patients in the world is increasing in recent years and the prevention of diabetes mellitus is therefore one of the urgent medical issues. Exogenous insulin is used for the control of blood glucose in diabetic patients; however, hypoglycemic episodes are unavoidable. Over the last several decades, islet transplantation has been developed as a promising method to achieve strict control of blood glucose and a potential cure for type 1 diabetes. However, due to the shortage of donor pancreata, alternative sources of islets have been sought through the generation of beta cells from stem cells, use of porcine islets, and beta cell expansion with growth factors. However, differentiation and expansion of embryonic and pancreatic stem cells and expansion of differentiated beta cells in vitro is limited. Expansion of primary beta cells by growth factors is also hampered by the senescence of the cells. Thus, we focused on establishing a human pancreatic beta cell line that is functionally equivalent to primary beta cells and can yield large amounts of cells for transplantation. Using Cre/loxP-based reversible immortalization, we constructed a reversibly immortalized pancreatic beta cell clone (NAKT-15). The cells may overcome the limitation of primary pancreatic beta cells for transplantation to control type 1 diabetes. In order to avoid the use of immunosuppressive agents, we are currently engaged in developing an implantable bag-type bioartificial pancreas. In this article, I discuss the hurdles of the current therapy for diabetes and introduce the possible future treatment of diabetes.
Collapse
Affiliation(s)
- Naoya Kobayashi
- Department of Surgery, Okayama University Graduate School of Medicine and Dentistry, Okayama 700-8558, Japan.
| |
Collapse
|
17
|
Cheng D, Zhao Y, Wang S, Zhang F, Russo M, McMahon SB, Zhu J. Repression of telomerase gene promoter requires human-specific genomic context and is mediated by multiple HDAC1-containing corepressor complexes. FASEB J 2016; 31:1165-1178. [PMID: 27940549 DOI: 10.1096/fj.201601111r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 11/28/2016] [Indexed: 12/16/2022]
Abstract
The human telomerase reverse transcriptase (hTERT) gene is repressed in most somatic cells, whereas the expression of the mouse mTert gene is widely detected. To understand the mechanisms of this human-specific repression, we constructed bacterial artificial chromosome (BAC) reporters using human and mouse genomic DNAs encompassing the TERT genes and neighboring loci. Upon chromosomal integration, the hTERT, but not the mTert, reporter was stringently repressed in telomerase-negative human cells in a histone deacetylase (HDAC)-dependent manner, replicating the expression of their respective endogenous genes. In chimeric BACs, the mTert promoter became strongly repressed in the human genomic context, but the hTERT promoter was highly active in the mouse genomic context. Furthermore, an unrelated herpes simplex virus-thymidine kinase (HSV-TK) promoter was strongly repressed in the human, but not in the mouse, genomic context. These results demonstrated that the repression of hTERT gene was dictated by distal elements and its chromatin environment. This repression depended on class I HDACs and involved multiple corepressor complexes, including HDAC1/2-containing Sin3B, nucleosome remodeling and histone deacetylase (NuRD), and corepressor of RE1 silencing transcription factor (CoREST) complexes. Together, our data indicate that the lack of telomerase expression in most human somatic cells results from its repressive genomic environment, providing new insight into the mechanism of long-recognized differential telomerase regulation in mammalian species.-Cheng, D., Zhao, Y., Wang, S., Zhang, F., Russo, M., McMahon, S. B., Zhu, J. Repression of telomerase gene promoter requires human-specific genomic context and is mediated by multiple HDAC1-containing corepressor complexes.
Collapse
Affiliation(s)
- De Cheng
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington, USA
| | - Yuanjun Zhao
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA; and
| | - Shuwen Wang
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington, USA
| | - Fan Zhang
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington, USA
| | - Mariano Russo
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA; and
| | - Steven B McMahon
- Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jiyue Zhu
- Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington, USA;
| |
Collapse
|
18
|
Abstract
Telomerase activity is responsible for the maintenance of chromosome end structures (telomeres) and cancer cell immortality in most human malignancies, making telomerase an attractive therapeutic target. The rationale for targeting components of the telomerase holoenzyme has been strengthened by accumulating evidence indicating that these molecules have extra-telomeric functions in tumour cell survival and proliferation. This Review discusses current knowledge of the biogenesis, structure and multiple functions of telomerase-associated molecules intertwined with recent advances in drug discovery approaches. We also describe the fertile ground available for the pursuit of next-generation small-molecule inhibitors of telomerase.
Collapse
Affiliation(s)
- Greg M Arndt
- Australian Cancer Research Foundation (ACRF) Drug Discovery Centre for Childhood Cancer, Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Randwick, New South Wales 2031, Australia
| | - Karen L MacKenzie
- Personalised Medicine Program, Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales, Randwick, New South Wales 2031, Australia
| |
Collapse
|
19
|
Yaswen P, MacKenzie KL, Keith WN, Hentosh P, Rodier F, Zhu J, Firestone GL, Matheu A, Carnero A, Bilsland A, Sundin T, Honoki K, Fujii H, Georgakilas AG, Amedei A, Amin A, Helferich B, Boosani CS, Guha G, Ciriolo MR, Chen S, Mohammed SI, Azmi AS, Bhakta D, Halicka D, Niccolai E, Aquilano K, Ashraf SS, Nowsheen S, Yang X. Therapeutic targeting of replicative immortality. Semin Cancer Biol 2015; 35 Suppl:S104-S128. [PMID: 25869441 PMCID: PMC4600408 DOI: 10.1016/j.semcancer.2015.03.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 03/06/2015] [Accepted: 03/13/2015] [Indexed: 12/15/2022]
Abstract
One of the hallmarks of malignant cell populations is the ability to undergo continuous proliferation. This property allows clonal lineages to acquire sequential aberrations that can fuel increasingly autonomous growth, invasiveness, and therapeutic resistance. Innate cellular mechanisms have evolved to regulate replicative potential as a hedge against malignant progression. When activated in the absence of normal terminal differentiation cues, these mechanisms can result in a state of persistent cytostasis. This state, termed "senescence," can be triggered by intrinsic cellular processes such as telomere dysfunction and oncogene expression, and by exogenous factors such as DNA damaging agents or oxidative environments. Despite differences in upstream signaling, senescence often involves convergent interdependent activation of tumor suppressors p53 and p16/pRB, but can be induced, albeit with reduced sensitivity, when these suppressors are compromised. Doses of conventional genotoxic drugs required to achieve cancer cell senescence are often much lower than doses required to achieve outright cell death. Additional therapies, such as those targeting cyclin dependent kinases or components of the PI3K signaling pathway, may induce senescence specifically in cancer cells by circumventing defects in tumor suppressor pathways or exploiting cancer cells' heightened requirements for telomerase. Such treatments sufficient to induce cancer cell senescence could provide increased patient survival with fewer and less severe side effects than conventional cytotoxic regimens. This positive aspect is countered by important caveats regarding senescence reversibility, genomic instability, and paracrine effects that may increase heterogeneity and adaptive resistance of surviving cancer cells. Nevertheless, agents that effectively disrupt replicative immortality will likely be valuable components of new combinatorial approaches to cancer therapy.
Collapse
Affiliation(s)
- Paul Yaswen
- Life Sciences Division, Lawrence Berkeley National Lab, Berkeley, CA, United States.
| | - Karen L MacKenzie
- Children's Cancer Institute Australia, Kensington, New South Wales, Australia.
| | | | | | | | - Jiyue Zhu
- Washington State University College of Pharmacy, Pullman, WA, United States.
| | | | | | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, HUVR, Consejo Superior de Investigaciones Cientificas, Universdad de Sevilla, Seville, Spain.
| | | | | | | | | | | | | | - Amr Amin
- United Arab Emirates University, Al Ain, United Arab Emirates; Cairo University, Cairo, Egypt
| | - Bill Helferich
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| | | | - Gunjan Guha
- SASTRA University, Thanjavur, Tamil Nadu, India
| | | | - Sophie Chen
- Ovarian and Prostate Cancer Research Trust, Guildford, Surrey, United Kingdom
| | | | - Asfar S Azmi
- Karmanos Cancer Institute, Wayne State University, Detroit, MI, United States
| | | | | | | | | | - S Salman Ashraf
- United Arab Emirates University, Al Ain, United Arab Emirates; Cairo University, Cairo, Egypt
| | | | - Xujuan Yang
- University of Illinois at Urbana Champaign, Champaign, IL, United States
| |
Collapse
|
20
|
Telomere homeostasis in mammalian germ cells: a review. Chromosoma 2015; 125:337-51. [DOI: 10.1007/s00412-015-0555-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 10/19/2015] [Accepted: 10/20/2015] [Indexed: 02/03/2023]
|
21
|
Cheng D, Zhao Y, Wang S, Jia W, Kang J, Zhu J. Human Telomerase Reverse Transcriptase (hTERT) Transcription Requires Sp1/Sp3 Binding to the Promoter and a Permissive Chromatin Environment. J Biol Chem 2015; 290:30193-203. [PMID: 26487723 DOI: 10.1074/jbc.m115.662221] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Indexed: 12/14/2022] Open
Abstract
The transcription of human telomerase gene hTERT is regulated by transcription factors (TFs), including Sp1 family proteins, and its chromatin environment. To understand its regulation in a relevant chromatin context, we employed bacterial artificial chromosome reporters containing 160 kb of human genomic sequence containing the hTERT gene. Upon chromosomal integration, the bacterial artificial chromosomes recapitulated endogenous hTERT expression, contrary to transient reporters. Sp1/Sp3 expression did not correlate with hTERT promoter activity, and these TFs bound to the hTERT promoters in both telomerase-positive and telomerase-negative cells. Mutation of the proximal GC-box resulted in a dramatic decrease of hTERT promoter activity, and mutations of all five GC-boxes eliminated its transcriptional activity. Neither mutations of GC-boxes nor knockdown of endogenous Sp1 impacted promoter binding by other TFs, including E-box-binding proteins, and histone acetylation and trimethylation of histone H3K9 at the hTERT promoter in telomerase-positive and -negative cells. The result indicated that promoter binding by Sp1/Sp3 was essential, but not a limiting step, for hTERT transcription. hTERT transcription required a permissive chromatin environment. Importantly, our data also revealed different functions of GC-boxes and E-boxes in hTERT regulation; although GC-boxes were essential for promoter activity, factors bound to the E-boxes functioned to de-repress hTERT promoter.
Collapse
Affiliation(s)
- De Cheng
- From the Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington 99210
| | - Yuanjun Zhao
- the Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, and
| | - Shuwen Wang
- From the Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington 99210
| | - Wenwen Jia
- the School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jiuhong Kang
- the School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jiyue Zhu
- From the Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington 99210, the Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033, and
| |
Collapse
|
22
|
MU XIYAN, SANG YAXIONG, FANG CHUNJU, SHAO BIN, YANG LU, YAO KUI, ZHAO XITONG, GOU JINHAI, WEI YUQUAN, YI TAO, WU YANG, ZHAO XIA. Immunotherapy of tumors with human telomerase reverse transcriptase immortalized human umbilical vein endothelial cells. Int J Oncol 2015; 47:1901-11. [DOI: 10.3892/ijo.2015.3175] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/19/2015] [Indexed: 11/06/2022] Open
|
23
|
Trochet D, Mergui X, Ivkovic I, Porreca RM, Gerbault-Seureau M, Sidibe A, Richard F, Londono-Vallejo A, Perret M, Aujard F, Riou JF. Telomere regulation during ageing and tumorigenesis of the grey mouse lemur. Biochimie 2015; 113:100-10. [PMID: 25882681 DOI: 10.1016/j.biochi.2015.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/03/2015] [Indexed: 01/01/2023]
Abstract
Telomere erosion leading to replicative senescence has been well documented in human and anthropoid primates, and provides a clue against tumorigenesis. In contrast, other mammals, such as laboratory mice, with short lifespan and low body weight mass have different telomere biology without replicative senescence. We analyzed telomere biology in the grey mouse lemur, a small prosimian model with a relative long lifespan currently used in ageing research. We report an average telomere length by telomere restriction fragment (TRF) among the longest reported so far for a primate species (25-30 kb), but without detectable overall telomere shortening with ageing on blood samples. However, we demonstrate using universal STELA (Single Telomere Length Amplification) the existence of short telomeres, the increase of which, while correlating with ageing might be related to another mechanism than replicative senescence. We also found a low stringency of telomerase restriction in tissues and an ease to immortalize fibroblasts in vitro upon spontaneous telomerase activation. Finally, we describe the first grey mouse lemur cancer cell line showing a dramatic telomere shortening and high telomerase activity associated with polyploidy. Our overall results suggest that telomere biology in grey mouse lemur is an exception among primates, with at best a physiologically limited replicative telomere ageing and closest to that observed in small rodents.
Collapse
Affiliation(s)
- Delphine Trochet
- Structure et Instabilité des Génomes, Sorbonne Universités, Muséum National d'Histoire Naturelle, Inserm U 1154, CNRS UMR 7196, CP26, 57 rue Cuvier, 75005 Paris, France
| | - Xénia Mergui
- Structure et Instabilité des Génomes, Sorbonne Universités, Muséum National d'Histoire Naturelle, Inserm U 1154, CNRS UMR 7196, CP26, 57 rue Cuvier, 75005 Paris, France
| | - Ivana Ivkovic
- Structure et Instabilité des Génomes, Sorbonne Universités, Muséum National d'Histoire Naturelle, Inserm U 1154, CNRS UMR 7196, CP26, 57 rue Cuvier, 75005 Paris, France
| | - Rosa Maria Porreca
- Telomeres and Cancer Laboratory, CNRS UMR 3244, Institut Curie, 26 rue d'Ulm, 75248 Paris, France; UPMC Univ. Paris 06, 75005 Paris, France
| | - Michèle Gerbault-Seureau
- Institut de Systématique, Evolution, Biodiversité, Sorbonne Universités, Muséum National d'Histoire Naturelle, UMR 7205 CNRS, UPMC Univ. Paris 06, EPHE, 57 rue Cuvier, 75005 Paris, France
| | - Assitan Sidibe
- Structure et Instabilité des Génomes, Sorbonne Universités, Muséum National d'Histoire Naturelle, Inserm U 1154, CNRS UMR 7196, CP26, 57 rue Cuvier, 75005 Paris, France
| | - Florence Richard
- Institut de Systématique, Evolution, Biodiversité, Sorbonne Universités, Muséum National d'Histoire Naturelle, UMR 7205 CNRS, UPMC Univ. Paris 06, EPHE, 57 rue Cuvier, 75005 Paris, France
| | - Arturo Londono-Vallejo
- Telomeres and Cancer Laboratory, CNRS UMR 3244, Institut Curie, 26 rue d'Ulm, 75248 Paris, France; UPMC Univ. Paris 06, 75005 Paris, France
| | - Martine Perret
- Mécanismes Adaptatifs et Evolution, Muséum National d'Histoire Naturelle, Sorbonne Universités, UMR 7179 CNRS, 1 Avenue du Petit Château, 91800 Brunoy, France
| | - Fabienne Aujard
- Mécanismes Adaptatifs et Evolution, Muséum National d'Histoire Naturelle, Sorbonne Universités, UMR 7179 CNRS, 1 Avenue du Petit Château, 91800 Brunoy, France
| | - Jean-François Riou
- Structure et Instabilité des Génomes, Sorbonne Universités, Muséum National d'Histoire Naturelle, Inserm U 1154, CNRS UMR 7196, CP26, 57 rue Cuvier, 75005 Paris, France.
| |
Collapse
|
24
|
The role of telomeres in predicting individual radiosensitivity of patients with cancer in the era of personalized radiotherapy. Cancer Treat Rev 2015; 41:354-60. [PMID: 25704912 DOI: 10.1016/j.ctrv.2015.02.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 02/02/2015] [Accepted: 02/04/2015] [Indexed: 02/06/2023]
Abstract
Radiotherapy plays a key role in cancer treatments, but tumor cell death differs from one tumor to another. The response of patients to radiotherapy varies considerably and adverse side effects are difficult to prevent. The mechanisms involved in the heterogeneity of this response are not well understood. In order to enhance the efficacy and safety of radiotherapy, it is important to identify subpopulations most at risk of developing a late adverse response to radiotherapy. Telomeres are composed of multiple repeats of a unique sequence of nucleotides forming a TTAGGG pattern. They protect chromosomes from end-to-end fusion and maintain genomic stability. Telomeres have been shown to be extremely sensitive to radiotherapy especially because of their atypical DNA damage repair response, which includes partial inhibition of the non-homologous end joining repair pathway. Ionizing Radiation (IR)-induced damage to telomere DNA could lead to chromosome instability and the initiation or progression of tumor processes. Telomeres could thus be a reliable marker of IR exposure and as such become a new parameter for predicting radiosensitivity. Furthermore, short telomeres are more sensitive to radiotherapy, which could partially explain differences in tumor cell death and in inter-individual sensitivity to radiotherapy. Telomere length could be used to identify subpopulations of patients who could benefit from higher or lower doses per fraction. Finally, pharmacological interference with tumor-cell telomere biology to reduce telomere length and/or telomere stability could also enhance the effectiveness and safety of radiotherapy. Telomeres could play a key role in radiotherapy in the era of personalized medicine.
Collapse
|
25
|
Ma Y, Hao S, Wang S, Zhao Y, Lim B, Lei M, Spector DJ, El-Deiry WS, Zheng SY, Zhu J. A Combinatory Strategy for Detection of Live CTCs Using Microfiltration and a New Telomerase-Selective Adenovirus. Mol Cancer Ther 2015; 14:835-43. [PMID: 25589497 DOI: 10.1158/1535-7163.mct-14-0693] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 12/29/2014] [Indexed: 01/12/2023]
Abstract
Circulating tumor cells (CTC) have become an important biomarker for early cancer diagnosis, prognosis, and treatment monitoring. Recently, a replication-competent recombinant adenovirus driven by a human telomerase gene (hTERT) promoter was shown to detect live CTCs in blood samples of patients with cancer. Here, we report a new class of adenoviruses containing regulatory elements that repress the hTERT gene in normal cells. Compared with the virus with only the hTERT core promoter, the new viruses showed better selectivity for replication in cancer cells than in normal cells. In particular, Ad5GTSe, containing three extra copies of a repressor element, displayed a superior tropism for cancer cells among leukocytes and was thus selected for CTC detection in blood samples. To further improve the efficiency and specificity of CTC identification, we tested a combinatory strategy of microfiltration enrichment using flexible micro spring arrays and Ad5GTSe imaging. Our experiments showed that this method efficiently detected both cancer cells spiked into healthy blood and potential CTCs in blood samples of patients with breast and pancreatic cancer, demonstrating its potential as a highly sensitive and reliable system for detection and capture of CTCs of different tumor types.
Collapse
Affiliation(s)
- Yanchun Ma
- College of Life Science, Northwest A&F University, Taicheng Road, Yangling, Shaanxi, China. Department of C&M Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Sijie Hao
- Department of C&M Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Shuwen Wang
- Department of C&M Physiology, Penn State College of Medicine, Hershey, Pennsylvania. Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington
| | - Yuanjun Zhao
- Department of C&M Physiology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Bora Lim
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington
| | - Ming Lei
- College of Life Science, Northwest A&F University, Taicheng Road, Yangling, Shaanxi, China
| | - David J Spector
- Department of Microbiology and Immunology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Wafik S El-Deiry
- Division of Hematology-Oncology, Penn State Hershey Cancer Institute, Hershey, Pennsylvania
| | - Si-Yang Zheng
- Micro & Nano Integrated Biosystem Laboratory, Department of Biomedical Engineering and Materials Research Institute, Pennsylvania State University, University Park, Pennsylvania
| | - Jiyue Zhu
- Department of C&M Physiology, Penn State College of Medicine, Hershey, Pennsylvania. Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington.
| |
Collapse
|
26
|
Métifiot M, Amrane S, Litvak S, Andreola ML. G-quadruplexes in viruses: function and potential therapeutic applications. Nucleic Acids Res 2014; 42:12352-66. [PMID: 25332402 PMCID: PMC4227801 DOI: 10.1093/nar/gku999] [Citation(s) in RCA: 187] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 09/26/2014] [Accepted: 10/06/2014] [Indexed: 12/15/2022] Open
Abstract
G-rich nucleic acids can form non-canonical G-quadruplex structures (G4s) in which four guanines fold in a planar arrangement through Hoogsteen hydrogen bonds. Although many biochemical and structural studies have focused on DNA sequences containing successive, adjacent guanines that spontaneously fold into G4s, evidence for their in vivo relevance has recently begun to accumulate. Complete sequencing of the human genome highlighted the presence of ∼300,000 sequences that can potentially form G4s. Likewise, the presence of putative G4-sequences has been reported in various viruses genomes [e.g., Human immunodeficiency virus (HIV-1), Epstein-Barr virus (EBV), papillomavirus (HPV)]. Many studies have focused on telomeric G4s and how their dynamics are regulated to enable telomere synthesis. Moreover, a role for G4s has been proposed in cellular and viral replication, recombination and gene expression control. In parallel, DNA aptamers that form G4s have been described as inhibitors and diagnostic tools to detect viruses [e.g., hepatitis A virus (HAV), EBV, cauliflower mosaic virus (CaMV), severe acute respiratory syndrome virus (SARS), simian virus 40 (SV40)]. Here, special emphasis will be given to the possible role of these structures in a virus life cycle as well as the use of G4-forming oligonucleotides as potential antiviral agents and innovative tools.
Collapse
Affiliation(s)
- Mathieu Métifiot
- CNRS UMR-5234, Université de Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France
| | - Samir Amrane
- INSERM, U869, IECB, ARNA laboratory, Université de Bordeaux, 2 Rue Robert Escarpit 33600 Pessac, France
| | - Simon Litvak
- CNRS UMR-5234, Université de Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France
| | - Marie-Line Andreola
- CNRS UMR-5234, Université de Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France
| |
Collapse
|
27
|
Eva R, Bram DC, Joery DK, Tamara V, Geert B, Vera R, Mathieu V. Strategies for immortalization of primary hepatocytes. J Hepatol 2014; 61:925-43. [PMID: 24911463 PMCID: PMC4169710 DOI: 10.1016/j.jhep.2014.05.046] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Revised: 04/17/2014] [Accepted: 05/30/2014] [Indexed: 02/06/2023]
Abstract
The liver has the unique capacity to regenerate in response to a damaging event. Liver regeneration is hereby largely driven by hepatocyte proliferation, which in turn relies on cell cycling. The hepatocyte cell cycle is a complex process that is tightly regulated by several well-established mechanisms. In vitro, isolated hepatocytes do not longer retain this proliferative capacity. However, in vitro cell growth can be boosted by immortalization of hepatocytes. Well-defined immortalization genes can be artificially overexpressed in hepatocytes or the cells can be conditionally immortalized leading to controlled cell proliferation. This paper discusses the current immortalization techniques and provides a state-of-the-art overview of the actually available immortalized hepatocyte-derived cell lines and their applications.
Collapse
Affiliation(s)
- Ramboer Eva
- Department of Toxicology, Center for Pharmaceutical Research, Vrije Universiteit Brussel Laarbeeklaan 103, 1090 Brussel, Belgium
| | - De Craene Bram
- Unit of Molecular and Cellular Oncology, Inflammation Research Center, VIB, Technologiepark 927, 9052 Zwijnaarde, Belgium
,Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - De Kock Joery
- Department of Toxicology, Center for Pharmaceutical Research, Vrije Universiteit Brussel Laarbeeklaan 103, 1090 Brussel, Belgium
| | - Vanhaecke Tamara
- Department of Toxicology, Center for Pharmaceutical Research, Vrije Universiteit Brussel Laarbeeklaan 103, 1090 Brussel, Belgium
| | - Berx Geert
- Unit of Molecular and Cellular Oncology, Inflammation Research Center, VIB, Technologiepark 927, 9052 Zwijnaarde, Belgium
,Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Rogiers Vera
- Department of Toxicology, Center for Pharmaceutical Research, Vrije Universiteit Brussel Laarbeeklaan 103, 1090 Brussel, Belgium
| | - Vinken Mathieu
- Department of Toxicology, Center for Pharmaceutical Research, Vrije Universiteit Brussel Laarbeeklaan 103, 1090 Brussel, Belgium
| |
Collapse
|
28
|
Reig-Viader R, Capilla L, Vila-Cejudo M, Garcia F, Anguita B, Garcia-Caldés M, Ruiz-Herrera A. Telomere homeostasis is compromised in spermatocytes from patients with idiopathic infertility. Fertil Steril 2014; 102:728-738.e1. [DOI: 10.1016/j.fertnstert.2014.06.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 06/04/2014] [Accepted: 06/04/2014] [Indexed: 01/06/2023]
|
29
|
Zhao Y, Cheng D, Wang S, Zhu J. Dual roles of c-Myc in the regulation of hTERT gene. Nucleic Acids Res 2014; 42:10385-98. [PMID: 25170084 PMCID: PMC4176324 DOI: 10.1093/nar/gku721] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Revised: 07/24/2014] [Accepted: 07/25/2014] [Indexed: 12/04/2022] Open
Abstract
Human telomerase gene hTERT is important for cancer and aging. hTERT promoter is regulated by multiple transcription factors (TFs) and its activity is dependent on the chromatin environment. However, it remains unsolved how the interplay between TFs and chromatin environment controls hTERT transcription. In this study, we employed the recombinase-mediated BAC targeting and BAC recombineering techniques to dissect the functions of two proximal E-box sites at -165 and +44 nt in regulating the hTERT promoter in the native genomic contexts. Our data showed that mutations of these sites abolished promoter binding by c-Myc/Max, USF1 and USF2, decreased hTERT promoter activity, and prevented its activation by overexpressed c-Myc. Upon inhibition of histone deacetylases, mutant and wildtype promoters were induced to the same level, indicating that the E-boxes functioned to de-repress the hTERT promoter and allowed its transcription in a repressive chromatin environment. Unexpectedly, knockdown of endogenous c-Myc/Max proteins activated hTERT promoter. This activation did not require the proximal E-boxes but was accompanied by increased promoter accessibility, as indicated by augmented active histone marks and binding of multiple TFs at the promoter. Our studies demonstrated that c-Myc/Max functioned in maintaining chromatin-dependent repression of the hTERT gene in addition to activating its promoter.
Collapse
Affiliation(s)
- Yuanjun Zhao
- Department of C & M Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA
| | - De Cheng
- Department of C & M Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington, USA
| | - Shuwen Wang
- Department of C & M Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington, USA
| | - Jiyue Zhu
- Department of C & M Physiology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania, USA Department of Pharmaceutical Sciences, Washington State University College of Pharmacy, Spokane, Washington, USA
| |
Collapse
|
30
|
Falandry C, Bonnefoy M, Freyer G, Gilson E. Biology of Cancer and Aging: A Complex Association With Cellular Senescence. J Clin Oncol 2014; 32:2604-10. [DOI: 10.1200/jco.2014.55.1432] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Over the last 50 years, major improvements have been made in our understanding of the driving forces, both parallel and opposing, that lead to aging and cancer. Many theories on aging first proposed in the 1950s, including those associated with telomere biology, senescence, and adult stem-cell regulation, have since gained support from cumulative experimental evidence. These views suggest that the accumulation of mutations might be a common driver of both aging and cancer. Moreover, some tumor suppressor pathways lead to aging in line with the theory of antagonist pleiotropy. According to the evolutionary-selected disposable soma theory, aging should affect primarily somatic cells. At the cellular level, both intrinsic and extrinsic pathways regulate aging and senescence. However, increasing lines of evidence support the hypothesis that these driving forces might be regulated by evolutionary-conserved pathways that modulate energy balance. According to the hyperfunction theory, aging is a quasi-program favoring both age-related diseases and cancer that could be inhibited by the regulation of longevity pathways. This review summarizes these hypotheses, as well as the experimental data that have accumulated over the last 60 years linking aging and cancer.
Collapse
Affiliation(s)
- Claire Falandry
- Claire Falandry, Marc Bonnefoy, and Gilles Freyer, Hospices Civils de Lyon and Lyon University, Lyon; and Eric Gilson, Centre Hospitalier Universitaire of Nice and Nice University Sophia Antipolis, Nice, France
| | - Marc Bonnefoy
- Claire Falandry, Marc Bonnefoy, and Gilles Freyer, Hospices Civils de Lyon and Lyon University, Lyon; and Eric Gilson, Centre Hospitalier Universitaire of Nice and Nice University Sophia Antipolis, Nice, France
| | - Gilles Freyer
- Claire Falandry, Marc Bonnefoy, and Gilles Freyer, Hospices Civils de Lyon and Lyon University, Lyon; and Eric Gilson, Centre Hospitalier Universitaire of Nice and Nice University Sophia Antipolis, Nice, France
| | - Eric Gilson
- Claire Falandry, Marc Bonnefoy, and Gilles Freyer, Hospices Civils de Lyon and Lyon University, Lyon; and Eric Gilson, Centre Hospitalier Universitaire of Nice and Nice University Sophia Antipolis, Nice, France
| |
Collapse
|
31
|
Reig-Viader R, Vila-Cejudo M, Vitelli V, Buscà R, Sabaté M, Giulotto E, Caldés MG, Ruiz-Herrera A. Telomeric Repeat-Containing RNA (TERRA) and Telomerase Are Components of Telomeres During Mammalian Gametogenesis1. Biol Reprod 2014; 90:103. [DOI: 10.1095/biolreprod.113.116954] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
32
|
Involvement of telomerase reverse transcriptase in heterochromatin maintenance. Mol Cell Biol 2014; 34:1576-93. [PMID: 24550003 DOI: 10.1128/mcb.00093-14] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
In the fission yeast Schizosaccharomyces pombe, centromeric heterochromatin is maintained by an RNA-directed RNA polymerase complex (RDRC) and the RNA-induced transcriptional silencing (RITS) complex in a manner that depends on the generation of short interfering RNA. In association with the telomerase RNA component (TERC), the telomerase reverse transcriptase (TERT) forms telomerase and counteracts telomere attrition, and without TERC, TERT has been implicated in the regulation of heterochromatin at locations distinct from telomeres. Here, we describe a complex composed of human TERT (hTERT), Brahma-related gene 1 (BRG1), and nucleostemin (NS) that contributes to heterochromatin maintenance at centromeres and transposons. This complex produced double-stranded RNAs homologous to centromeric alpha-satellite (alphoid) repeat elements and transposons that were processed into small interfering RNAs targeted to these heterochromatic regions. These small interfering RNAs promoted heterochromatin assembly and mitotic progression in a manner dependent on the RNA interference machinery. These observations implicate the hTERT/BRG1/NS (TBN) complex in heterochromatin assembly at particular sites in the mammalian genome.
Collapse
|
33
|
Essential role for mammalian apurinic/apyrimidinic (AP) endonuclease Ape1/Ref-1 in telomere maintenance. Proc Natl Acad Sci U S A 2013; 110:17844-9. [PMID: 24127576 DOI: 10.1073/pnas.1304784110] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The major mammalian apurinic/apyrimidinic endonuclease Ape1 is a multifunctional protein operating in protection of cells from oxidative stress via its DNA repair, redox, and transcription regulatory activities. The importance of Ape1 has been marked by previous work demonstrating its requirement for viability in mammalian cells. However, beyond a requirement for Ape1-dependent DNA repair activity, deeper molecular mechanisms of the fundamental role of Ape1 in cell survival have not been defined. Here, we report that Ape1 is an essential factor stabilizing telomeric DNA, and its deficiency is associated with telomere dysfunction and segregation defects in immortalized cells maintaining telomeres by either the alternative lengthening of telomeres pathway (U2OS) or telomerase expression (BJ-hTERT), or in normal human fibroblasts (IMR90). Through the expression of Ape1 derivatives with site-specific changes, we found that the DNA repair and N-terminal acetylation domains are required for the Ape1 function at telomeres. Ape1 associates with telomere proteins in U2OS cells, and Ape1 depletion causes dissociation of TRF2 protein from telomeres. Consistent with this effect, we also observed that Ape1 depletion caused telomere shortening in both BJ-hTERT and in HeLa cells. Thus, our study describes a unique and unpredicted role for Ape1 in telomere protection, providing a direct link between base excision DNA repair activities and telomere metabolism.
Collapse
|
34
|
Telomerase and its extracurricular activities. Cell Mol Biol Lett 2013; 18:538-54. [PMID: 24048710 PMCID: PMC6275585 DOI: 10.2478/s11658-013-0105-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 09/11/2013] [Indexed: 11/20/2022] Open
Abstract
The classical activity of telomerase is to synthesize telomeric repeats and thus maintain telomere length, which in turn ensures chromosome stability and cellular proliferation. However, there is growing evidence that implicates telomerase in many other functions that are independent of TERC being used as its template. Telomerase has an RNA-dependent RNA polymerase (RdRP) activity in the mitochondria. Other than viral RdRPs, it is the only RNA-dependent RNA polymerase that has been identified in mammals. It also plays a role in the Wnt signaling pathway by acting as a transcriptional modulator. Telomerase acts as a reverse transcriptase independent of its core subunit, TERC. Studies indicate that telomerase is also involved in apoptosis and DNA repair.
Collapse
|
35
|
Boccardi V, Barbieri M, Rizzo MR, Marfella R, Esposito A, Marano L, Paolisso G. A new pleiotropic effect of statins in elderly: modulation of telomerase activity. FASEB J 2013; 27:3879-3885. [PMID: 23748973 DOI: 10.1096/fj.13-232066] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Recent evidence suggests a link between statins and telomere biology. Whether statin treatment may modulate telomerase activity and affect telomere erosion rate is unknown. We aimed at investigating the potential impact of statin therapy on peripheral blood mononuclear cells telomerase activity, its implication on LTL variability, and its association with telomere shortening rates along with aging. The cross-sectional study was conducted in 230 subjects (age range: 30-86 y) stratified according to statins treatment. LTL was measured by quantitative polymerase chain reaction and telomerase activity by a PCR-ELISA protocol. Subjects on statin treatment showed higher telomerase activity (P<0.0001) and longer LTL (P=0.028) levels compared to the nonstatin group. Statin therapy was associated with higher telomerase activity independently of multiple covariates, including age, gender, smoking habits, lipid, systemic inflammation, glucose, and blood pressure levels (P=0.019). Indeed, subjects on statin treatment showed significant lower telomere erosion along with aging. Every 1 y increment in age, LTL decreases by 0.058 Kb in no statin and 0.033 Kb in statin groups, respectively, as well as the major difference in telomere attrition between groups was found after the age of 65 yr (P<0.0001). In summary, statins, modulating telomerase activity, affect telomere erosion along with aging.
Collapse
Affiliation(s)
- Virginia Boccardi
- Department of Internal Medicine, Surgical, Neurological, Metabolic Disease and Geriatric Medicine, Second University of Naples, P.zza Miraglia 2, 80138 Naples, Italy
| | | | | | | | | | | | | |
Collapse
|
36
|
Lanna A, Coutavas E, Levati L, Seidel J, Rustin MHA, Henson SM, Akbar AN, Franzese O. IFN-α inhibits telomerase in human CD8⁺ T cells by both hTERT downregulation and induction of p38 MAPK signaling. THE JOURNAL OF IMMUNOLOGY 2013; 191:3744-52. [PMID: 23997212 DOI: 10.4049/jimmunol.1301409] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The cytokine IFN-α is secreted during viral infections and has been shown to inhibit telomerase activity and accelerate T cell differentiation in vivo. However, the mechanism for this inhibition is not clear. In this study, we show that IFN-α inhibits both the transcription and translation of human telomerase reverse transcriptase (hTERT), the catalytic component of telomerase, in activated CD8(+) T cells. This was associated with increased activity of the repressor of hTERT transcription E2 transcription factor and decreased activation of NF-κB that promotes hTERT transcription. However IFN-α did not affect the translocation of hTERT from the cytoplasm to the nucleus. IFN-α also inhibits AKT kinase activation but increases p38 MAPK activity, and both of these events have been shown previously to inhibit telomerase activity. Addition of BIRB796, an inhibitor of p38 activity, to IFN-α-treated cells reversed, in part, the inhibition of telomerase by this cytokine. Therefore, IFN-α can inhibit the enzyme telomerase in CD8(+) T cells by transcriptional and posttranslational mechanisms. Furthermore, the addition of IFN-α to CD8(+)CD27(+)CD28(+) T cells accelerates the loss of both these costimulatory molecules. This suggests that persistent viral infections may contribute to the accumulation of highly differentiated/senescent CD8(+)CD27(-)CD28(-) T cells during aging by promoting IFN-α secretion during repeated episodes of viral reactivation.
Collapse
Affiliation(s)
- Alessio Lanna
- Pharmacology Section, Department of System Medicine, University of Rome Tor Vergata, 00133, Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Olyslaegers DAJ, Desmarets LMB, Dedeurwaerder A, Dewerchin HL, Nauwynck HJ. Generation and characterization of feline arterial and venous endothelial cell lines for the study of the vascular endothelium. BMC Vet Res 2013; 9:170. [PMID: 23987139 PMCID: PMC3847150 DOI: 10.1186/1746-6148-9-170] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 08/16/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The in vitro culture of endothelial cells (ECs) is an indispensable tool for studying the role of the endothelium in physical and pathological conditions. Primary ECs, however, have a restricted proliferative lifespan which hampers their use in long-term studies. The need for standardized experimental conditions to obtain relevant and reproducible results has increased the demand for well-characterized, continuous EC lines that retain the phenotypic and functional characteristics of their non-transformed counterparts. RESULTS Primary feline ECs from aorta and vena cava were successfully immortalized through the successive introduction of simian virus 40 large T (SV40LT) antigen and the catalytic subunit of human telomerase (hTERT). In contrast to the parental ECs, the transformed cells were able to proliferate continuously in culture. Established cell lines exhibited several inherent endothelial properties, including typical cobblestone morphology, binding of endothelial cell-specific lectins and internalization of acetylated low-density lipoprotein. In addition, the immortalization did not affect the functional phenotype as demonstrated by their capacity to rapidly form cord-like structures on matrigel and to express cell adhesion molecules following cytokine stimulation. CONCLUSION The ability to immortalize feline ECs, and the fact that these cells maintain the EC phenotype will enable a greater understanding of fundamental mechanisms of EC biology and endothelial-related diseases. Furthermore, the use of cell lines is an effective implementation of the 3-R principles formulated by Russel and Burch.
Collapse
Affiliation(s)
- Dominique A J Olyslaegers
- Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B-9820 Merelbeke, Belgium.
| | | | | | | | | |
Collapse
|
38
|
Boccardi V, Esposito A, Rizzo MR, Marfella R, Barbieri M, Paolisso G. Mediterranean diet, telomere maintenance and health status among elderly. PLoS One 2013; 8:e62781. [PMID: 23646142 PMCID: PMC3640022 DOI: 10.1371/journal.pone.0062781] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 03/24/2013] [Indexed: 11/18/2022] Open
Abstract
Leukocyte telomere length (LTL) and rate of telomere shortening are known biomarkers of aging while, numerous studies showed that Mediterranean diet (MD) may boost longevity. We studied association between telomere length, telomerase activity and different adherence to MD and its effects on healthy status. The study was conducted in 217 elderly subjects stratified according Mediterranean diet score (MDS) in low adherence (MDS≤3), medium adherence (MDS 4–5) and high adherence (MDS≥6) groups. LTL was measured by quantitative polymerase chain reaction and telomerase activity by a PCR-ELISA protocol. High adherence group showed longer LTL (p = 0.003) and higher telomerase activity (p = 0.013) compared to others. Linear regression analysis including age, gender, smoking habit and MDS showed that MDS was independently associated with LTL (p = 0.024) and telomerase activity levels (p = 0.006). Telomerase activity was independently associated with LTL (p = 0.007) and negatively modulated by inflammation and oxidative stress. Indeed, telomerase levels were associated with healthy status independently of multiple covariates (p = 0.048). These results support a novel role of MD in promoting health-span suggesting that telomere maintenance, rather than LTL variability is the major determinant of healthy status among elderly.
Collapse
Affiliation(s)
- Virginia Boccardi
- Department of Internal Medicine, Surgical, Neurological Metabolic Disease and Geriatric Medicine, Second University of Naples, Naples, Italy
| | - Antonietta Esposito
- Department of Internal Medicine, Surgical, Neurological Metabolic Disease and Geriatric Medicine, Second University of Naples, Naples, Italy
| | - Maria Rosaria Rizzo
- Department of Internal Medicine, Surgical, Neurological Metabolic Disease and Geriatric Medicine, Second University of Naples, Naples, Italy
| | - Raffaele Marfella
- Department of Internal Medicine, Surgical, Neurological Metabolic Disease and Geriatric Medicine, Second University of Naples, Naples, Italy
| | - Michelangela Barbieri
- Department of Internal Medicine, Surgical, Neurological Metabolic Disease and Geriatric Medicine, Second University of Naples, Naples, Italy
| | - Giuseppe Paolisso
- Department of Internal Medicine, Surgical, Neurological Metabolic Disease and Geriatric Medicine, Second University of Naples, Naples, Italy
- * E-mail:
| |
Collapse
|
39
|
Miller J, Dakic A, Chen R, Palechor-Ceron N, Dai Y, Kallakury B, Schlegel R, Liu X. HPV16 E7 protein and hTERT proteins defective for telomere maintenance cooperate to immortalize human keratinocytes. PLoS Pathog 2013; 9:e1003284. [PMID: 23592995 PMCID: PMC3617164 DOI: 10.1371/journal.ppat.1003284] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2012] [Accepted: 02/15/2013] [Indexed: 12/13/2022] Open
Abstract
Previous studies have shown that wild-type human telomerase reverse transcriptase (hTERT) protein can functionally replace the human papillomavirus type 16 (HPV-16) E6 protein, which cooperates with the viral E7 protein in the immortalization of primary keratinocytes. In the current study, we made the surprising finding that catalytically inactive hTERT (hTERT-D868A), elongation-defective hTERT (hTERT-HA), and telomere recruitment-defective hTERT (hTERT N+T) also cooperate with E7 in mediating bypass of the senescence blockade and effecting cell immortalization. This suggests that hTERT has activities independent of its telomere maintenance functions that mediate transit across this restriction point. Since hTERT has been shown to have a role in gene activation, we performed microarray studies and discovered that E6, hTERT and mutant hTERT proteins altered the expression of highly overlapping sets of cellular genes. Most important, the E6 and hTERT proteins induced mRNA and protein levels of Bmi1, the core subunit of the Polycomb Group (PcG) complex 1. We show further that Bmi1 substitutes for E6 or hTERT in cell immortalization. Finally, tissue array studies demonstrated that expression of Bmi1 increased with the severity of cervical dysplasia, suggesting a potential role in the progression of cervical cancer. Together, these data demonstrate that hTERT has extra-telomeric activities that facilitate cell immortalization and that its induction of Bmi1 is one potential mechanism for mediating this activity. The human papillomaviruses (HPVs) are critical elements in the etiology of cervical cancer, as well as several other human cancers. The E6 protein, in combination with the E7 protein of these viruses, immortalizes epithelial cells and increases the expression of the hTERT protein. In the current study we show that the enzymatic activity of hTERT is not required for cooperating in cell immortalization. We further demonstrate that hTERT proteins increase the expression of the Bmi1 protein, which is also capable of cooperating in cell immortalization. We anticipate that these findings will stimulate new studies of telomerase in HPV biology, cancer etiology, and stem cell reprogramming.
Collapse
Affiliation(s)
- Jonathan Miller
- Department of Pathology, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Aleksandra Dakic
- Department of Pathology, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Renxiang Chen
- Department of Pathology, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Nancy Palechor-Ceron
- Department of Pathology, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Yuhai Dai
- Department of Pathology, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Bhaskar Kallakury
- Department of Pathology, Georgetown University Medical Center, Washington, D.C., United States of America
| | - Richard Schlegel
- Department of Pathology, Georgetown University Medical Center, Washington, D.C., United States of America
- * E-mail: (RS); (XL)
| | - Xuefeng Liu
- Department of Pathology, Georgetown University Medical Center, Washington, D.C., United States of America
- * E-mail: (RS); (XL)
| |
Collapse
|
40
|
Jullien L, Mestre M, Roux P, Gire V. Eroded human telomeres are more prone to remain uncapped and to trigger a G2 checkpoint response. Nucleic Acids Res 2012. [PMID: 23193277 PMCID: PMC3553962 DOI: 10.1093/nar/gks1121] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Telomeres cap the ends of chromosomes and regulate the replicative life span of human somatic cells. Telomere function is lost upon critical shortening and a p53-dependent checkpoint that detects altered telomere states at the G1/S transition was proposed to act as a regulator of the telomere damage response. We show that telomerase-negative human fibroblasts spend more time in G2 phase as they approach senescence and this delay is associated with manifestations of telomere dysfunction and the triggering of an ATM/ATR-dependent DNA damage signal. This correlates with a partial release of telomeric proteins TRF1 and TRF2. Analysis of the consequences of TRF1 and TRF2 depletion or over-expression of mutated versions revealed that telomere uncapping or telomere replication stress also led to DNA damage signalling in G2. Progression through mitosis of these cells was associated with signs of incomplete telomere terminal processing. We also observed an increase in sister chromatid-type telomere aberrations in senescing fibroblasts indicating that defects of telomere post-replicative events increased as cells age. Our results link a post-replicative damage response at eroded telomeres to G2 arrest signalling and challenge the current paradigm that the checkpoint response to short telomeres occurs primarily at the G1/S transition in human cells.
Collapse
Affiliation(s)
- Laurent Jullien
- Universités Montpellier 2 et 1, CRBM-CNRS UMR 5237, 1919, route de Mende, 34293 Montpellier, France
| | | | | | | |
Collapse
|
41
|
An alternative telomerase RNA in Arabidopsis modulates enzyme activity in response to DNA damage. Genes Dev 2012; 26:2512-23. [PMID: 23109676 DOI: 10.1101/gad.202960.112] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Telomerase replenishes telomere tracts by reiteratively copying its RNA template, TER. Unlike other model organisms, Arabidopsis thaliana harbors two divergent TER genes. However, only TER1 is required for telomere maintenance. Here we examine the function of TER2. We show that TER2 is spliced and its 3' end is truncated in vivo to generate a third TER isoform, TER2(S). TERT preferentially associates with TER2 > TER1 > TER2(S). Moreover, TER2 and TER2(S) assemble with Ku and POT1b (protection of telomeres), forming RNP (ribonucleoprotein) complexes distinct from TER1 RNP. Plants null for TER2 display increased telomerase enzyme activity, while TER2 overexpression inhibits telomere synthesis from TER1 and leads to telomere shortening. These findings argue that TER2 negatively regulates telomerase by sequestering TERT in a nonproductive RNP complex. Introduction of DNA double-strand breaks by zeocin leads to an immediate and specific spike in TER2 and a concomitant decrease in telomerase enzyme activity. This response is not triggered by replication stress or telomere dysfunction and is abrogated in ter2 mutants. We conclude that Arabidopsis telomerase is modulated by TER2, a novel DNA damage-induced noncoding RNA that works in concert with the canonical TER to promote genome integrity.
Collapse
|
42
|
Chiodi I, Mondello C. Telomere-independent functions of telomerase in nuclei, cytoplasm, and mitochondria. Front Oncol 2012; 2:133. [PMID: 23061047 PMCID: PMC3460319 DOI: 10.3389/fonc.2012.00133] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 09/18/2012] [Indexed: 12/25/2022] Open
Abstract
Telomerase canonical activity at telomeres prevents telomere shortening, allowing chromosome stability and cellular proliferation. To perform this task, the catalytic subunit (telomerase reverse transcriptase, TERT) of the enzyme works as a reverse transcriptase together with the telomerase RNA component (TERC), adding telomeric repeats to DNA molecule ends. Growing evidence indicates that, besides the telomeric-DNA synthesis activity, TERT has additional functions in tumor development and is involved in many different biological processes, among which cellular proliferation, gene expression regulation, and mitochondrial functionality. TERT has been shown to act independently of TERC in the Wnt-β-catenin signaling pathway, regulating the expression of Wnt target genes, which play a role in development and tumorigenesis. Moreover, TERT RNA-dependent RNA polymerase activity has been found, leading to the genesis of double-stranded RNAs that act as precursor of silencing RNAs. In mitochondria, a TERT TERC-independent reverse transcriptase activity has been described that could play a role in the protection of mitochondrial integrity. In this review, we will discuss some of the extra-telomeric functions of telomerase.
Collapse
Affiliation(s)
- Ilaria Chiodi
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche Pavia, Italy
| | | |
Collapse
|
43
|
Abstract
Human telomerase reverse transcriptase (hTERT) is the catalytic subunit of the human telomerase complex. Growing evidence suggests that hTERT also contributes to the cell physiology independently of telomere elongation. However, its role in bacterial infection is unknown. Here we show that hTERT is critical for Listeria monocytogenes infection, as the depletion of hTERT impaired bacterial intracellular replication. In addition, we observed that L. monocytogenes caused a decrease in hTERT levels at early time points of the infectious process. This effect was mediated by the pore-forming toxin listeriolysin O (LLO) and did not require bacterial entry into host cells. Calcium influx through the LLO pores contributed to a proteasome-independent decrease in hTERT protein levels. Together, our data provide evidence that these bacteria trigger hTERT degradation, an event that is detrimental to bacterial replication.
Collapse
|
44
|
Hostility and cellular aging in men from the Whitehall II cohort. Biol Psychiatry 2012; 71:767-73. [PMID: 21974787 PMCID: PMC3657139 DOI: 10.1016/j.biopsych.2011.08.020] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 08/28/2011] [Accepted: 08/30/2011] [Indexed: 01/01/2023]
Abstract
BACKGROUND Hostility is associated with a significantly increased risk of age-related disease and mortality, yet the pathophysiological mechanisms involved remain unclear. Here we investigated the hypothesis that hostility might impact health by promoting cellular aging. METHODS We tested the relationship between cynical hostility and two known markers of cellular aging, leukocyte telomere length (TL) and leukocyte telomerase activity (TA), in 434 men and women from the Whitehall II cohort. RESULTS High-hostile men had significantly shorter leukocyte TL than their low-hostile counterparts. They also had elevated leukocyte TA, with a significantly increased likelihood of having both short TL and high TA, compared with low-hostile individuals. CONCLUSIONS Because telomerase is known to counteract telomere shortening by synthesizing telomeric DNA repeats, particularly in the context of shortened telomeres, heightened TA might represent a compensatory response in high-hostile individuals. The relationship between hostility and disease is stronger in men than in women, and men generally have a shorter life expectancy than women. Our findings suggest that telomere attrition might represent a novel mechanism mediating the detrimental effects of hostility on men's health.
Collapse
|
45
|
Hoffmann J, Spyridopoulos I. Telomere length in cardiovascular disease: new challenges in measuring this marker of cardiovascular aging. Future Cardiol 2012; 7:789-803. [PMID: 22050065 DOI: 10.2217/fca.11.55] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Atherosclerosis is an age-related systemic disease characterized by systemic oxidative stress and low grade chronic inflammation. Various types of leukocytes play an important role within this process. Telomeres, the ends of chromosomes, shorten during each and every cell division and have therefore been regarded as a cellular clock. Telomere dysfunction has been implicated in aging and senescence, and shorter leukocyte telomere length (LTL) has been demonstrated to predict cardiovascular disease and mortality. However, although LTL can predict cardiovascular events in population studies, a number of factors have prevented its broad use as a surrogate end point, such as serum levels of LDL cholesterol. In this article we will provide an overview of telomere biology and telomere dynamics of different leukocyte populations, and we will also discuss pitfalls in the methodology of LTL quantification, in context with landmark studies, which measured LTL in cardiovascular disease. Finally, we will attempt to critically assess and explain the shortcomings of LTL as a biomarker and identify further research avenues that require further investigation before telomere length can be implemented as an individual biomarker for cardiovascular aging. From this it becomes evident that LTL can be susceptible to methodological errors affecting longitudinal reproducibility. LTL is generally confounded at least by genetic factors, population variation and leukocyte composition.
Collapse
Affiliation(s)
- Jedrzej Hoffmann
- Newcastle University, Institute of Genetic Medicine, Central Parkway, Newcastle Upon Tyne, NE1 3BZ, UK
| | | |
Collapse
|
46
|
Zhu H, Guo D, Li K, Pedersen-White J, Stallmann-Jorgensen IS, Huang Y, Parikh S, Liu K, Dong Y. Increased telomerase activity and vitamin D supplementation in overweight African Americans. Int J Obes (Lond) 2011; 36:805-9. [PMID: 21986705 DOI: 10.1038/ijo.2011.197] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE We aimed to investigate whether vitamin D supplementation modulates peripheral blood mononuclear cell (PBMC) telomerase activity in overweight African Americans. DESIGN A double blind, randomized and placebo-controlled clinical trial (#NCT01141192) was recently conducted. SUBJECTS AND METHODS African-American adults were randomly assigned to either the placebo, or the vitamin D group (60,000 IU per month (equivalent to ~2000 IU per day) oral vitamin D3 supplementation). Fresh PBMCs were collected from 37 subjects (18 in the placebo group and 19 in the vitamin D group), both at baseline and 16 weeks. PBMC telomerase activity was measured by the telomeric repeat amplification protocol. RESULTS Serum 25 hydroxyvitamin D levels increased from 40.7±15.7 at baseline to 48.1±17.5 nmol l(-1) at posttest (P=0.004) in the placebo group, and from 35.4±11.3 at baseline to 103.7±31.5 nmol l(-1) at posttests (P<0.0001) in the vitamin D group. In the vitamin D group, PBMC telomerase activity increased by 19.2% from baseline (1.56±0.29 absorbance reading unit (AU)) to posttest (1.86±0.42 AU, P<0.0001). The significance persisted after controlling for age, sex and body mass index (P=0.039). PBMC telomerase activity in the placebo group did not change from baseline (1.43±0.26 AU) to posttest (1.46±0.27 AU, P=0.157). CONCLUSION Vitamin D supplementation significantly increased PBMC telomerase activity in overweight African Americans. Our data suggest that vitamin D may improve telomere maintenance and prevent cell senescence and counteract obesity-induced acceleration of cellular aging.
Collapse
Affiliation(s)
- H Zhu
- Georgia Prevention Institute, Department of Pediatrics, Georgia Health Sciences University, Augusta, GA 30912, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Educational attainment but not measures of current socioeconomic circumstances are associated with leukocyte telomere length in healthy older men and women. Brain Behav Immun 2011; 25:1292-8. [PMID: 21536122 DOI: 10.1016/j.bbi.2011.04.010] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2011] [Revised: 04/01/2011] [Accepted: 04/15/2011] [Indexed: 11/23/2022] Open
Abstract
Low socioeconomic status (SES) may be associated with accelerated biological aging, but findings relating SES with telomere length have been inconsistent. We tested the hypotheses that shorter telomere length and telomerase activity would be related more robustly to education, an early life indicator of socioeconomic position, than to current indicators of socioeconomic circumstances. Healthy men and women aged 53-76 years from the Whitehall II epidemiological cohort provided blood samples from which telomere length was assessed in 448 and telomerase activity in 416. Educational attainment was classified into four levels, while household income and grade of employment were measured as indicators of current socioeconomic circumstances. Age, gender, blood pressure, glycated hemoglobin, high density lipoprotein cholesterol, smoking, body mass index and physical activity were included as covariates. We found that lower educational attainment was associated with shorter telomere length after controlling statistically for biological and behavioral covariates. Neither household income nor employment grade was related to telomere length. The association between telomere length and education remained significant after adjusting for current socioeconomic circumstances. In men, highest levels of telomerase activity were found in the lowest education group. We conclude that low SES defined in terms of education but not current socioeconomic circumstances is associated with shortened telomeres. Low educational attainment may be an indicator of long-term SES trajectories, and be associated with accumulated allostatic load resulting in telomere shortening. Education may also promote problem-solving skills leading to reduced biological stress responsivity, with favorable consequences for biological aging.
Collapse
|
48
|
Separation of telomerase functions by reverse genetics. Proc Natl Acad Sci U S A 2011; 108:E1363-71. [PMID: 21949400 DOI: 10.1073/pnas.1112414108] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The canonical function of the human telomerase protein (hTERT) is to synthesize telomeric DNA, but it has other biological activities, including enhancing cell proliferation, decreasing apoptosis, regulating DNA damage responses, and increasing cellular proliferative lifespan. The mechanistic relationships among these activities are not understood. We previously demonstrated that ectopic hTERT expression in primary human mammary epithelial cells diminishes their requirement for exogenous mitogens, thus giving them a proliferative advantage in a mitogen-depleted environment. Here, we show that this phenotype is caused by a combination of increased cell division and decreased apoptosis. In addition, we use a panel of hTERT mutants to demonstrate that this enhanced cell proliferation can be uncoupled not only from telomere elongation, but also from other telomerase activities, including cellular lifespan extension and regulation of DNA damage responses. We also find that the proliferative function of hTERT, which requires hTERT catalytic activity, is not caused by increased Wnt signaling, but is accompanied by alterations in key cell cycle regulators and is linked to an hTERT-catalyzed decrease in the levels of the RNA component of mitochondrial RNA processing endoribonuclease. Thus, enhanced cell proliferation is an independent function of hTERT that could provide a new target for the development of anti-telomerase cancer therapeutic agents.
Collapse
|
49
|
Zvereva MI, Shcherbakova DM, Dontsova OA. Telomerase: structure, functions, and activity regulation. BIOCHEMISTRY (MOSCOW) 2011; 75:1563-83. [PMID: 21417995 DOI: 10.1134/s0006297910130055] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Telomerase is the enzyme responsible for maintenance of the length of telomeres by addition of guanine-rich repetitive sequences. Telomerase activity is exhibited in gametes and stem and tumor cells. In human somatic cells proliferation potential is strictly limited and senescence follows approximately 50-70 cell divisions. In most tumor cells, on the contrary, replication potential is unlimited. The key role in this process of the system of the telomere length maintenance with involvement of telomerase is still poorly studied. No doubt, DNA polymerase is not capable to completely copy DNA at the very ends of chromosomes; therefore, approximately 50 nucleotides are lost during each cell cycle, which results in gradual telomere length shortening. Critically short telomeres cause senescence, following crisis, and cell death. However, in tumor cells the system of telomere length maintenance is activated. Besides catalytic telomere elongation, independent telomerase functions can be also involved in cell cycle regulation. Inhibition of the telomerase catalytic function and resulting cessation of telomere length maintenance will help in restriction of tumor cell replication potential. On the other hand, formation of temporarily active enzyme via its intracellular activation or due to stimulation of expression of telomerase components will result in telomerase activation and telomere elongation that can be used for correction of degenerative changes. Data on telomerase structure and function are summarized in this review, and they are compared for evolutionarily remote organisms. Problems of telomerase activity measurement and modulation by enzyme inhibitors or activators are considered as well.
Collapse
Affiliation(s)
- M I Zvereva
- Faculty of Chemistry, Lomonosov Moscow State University, Russia.
| | | | | |
Collapse
|
50
|
Gamble CM, Barton PA. Baculoviral expression of telomerase in primary human fibroblasts to rejuvenate cells for tissue engineering. J Tissue Eng Regen Med 2011; 6:414-20. [PMID: 21751423 DOI: 10.1002/term.447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Accepted: 05/12/2011] [Indexed: 11/09/2022]
Abstract
Tissue engineering involves the use of synthetic or natural materials as a scaffold to support the growth of replacement tissue or organs. The use of autologous cells to populate the scaffold avoids problems associated with rejection; however, a major limitation of this approach is the finite lifespan of primary cells in culture. This finite lifespan is due to the shortening of telomeres, short repetitive sequences of DNA located at the ends of eukaryotic chromosomes. Ectopic expression of telomerase reverse transcriptase (hTERT) is able to reconstitute telomerase activity and maintain the length of telomeres. This study investigated an alternative gene delivery vector, baculovirus, for the expression of hTERT in primary human cells. A recombinant baculovirus was used to efficiently deliver the hTERT gene to primary fibroblasts and the telomerase enzyme was found to be active. Although no increase in telomere length was detected, expression of hTERT in primary fibroblasts resulted in a significant extension of replicative lifespan. To our knowledge this is a novel attempt to use a recombinant baculovirus for the extension of cellular lifespan by exogenous expression of telomerase.
Collapse
Affiliation(s)
- C M Gamble
- Faculty of Life and Social Sciences, Swinburne University of Technology, Melbourne, Australia
| | | |
Collapse
|