1
|
Zhang L, Pang B, Wang R, Yang B, Jia X. Nesfatin-1 attenuated lipopolysaccharide-induced inflammatory response and senescence in human dental pulp cells. Heliyon 2024; 10:e32108. [PMID: 38975143 PMCID: PMC11226773 DOI: 10.1016/j.heliyon.2024.e32108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/19/2024] [Accepted: 05/28/2024] [Indexed: 07/09/2024] Open
Abstract
Lipopolysaccharide (LPS)-triggered damage in human dental pulp cells (hDPCs) is associated with the progression of gingivitis, which is inflammation of the gingival tissue. Nesfatin-1 is a peptide secreted by neurons and peripheral tissues. Here, we report a novel property of Nesfatin-1 in ameliorating LPS-induced inflammatory response and senescence in hDPCs. First, we demonstrate that Nesfatin-1 repressed LPS-triggered expression of inflammatory factors. Secondly, Nesfatin-1 restored telomerase activity and the expression of human telomerase reverse transcriptase (hTERT) and telomeric repeat binding factor 2 (TERF2) against LPS. Senescence-associated β-galactosidase (SA-β-gal) staining assay revealed that Nesfatin-1 attenuated LPS-induced cellular senescence in hDPCs. We also found that Nesfatin-1 increased telomerase activity in LPS-challenged hDPCs. It is also shown that Nesfatin-1 reduced the expression of plasminogen activator inhibitor-1 (PAI-1) and p16. Additionally, LPS stimulation reduced the expression of SIRT1, which was rescued by Nesfatin-1. However, the silencing of sirtuin1 (SIRT1) abrogated the protective property of Nesfatin-1 in preventing cellular senescence, implying that the function of Nesfatin-1 is regulated by SIRT1. Taken together, our findings suggest that Nesfatin-1 might possess a protective effect against gingivitis.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Stomatology, Shijingshan Teaching Hospital of Capital Medical University, Beijing Shijingshan Hospital, Beijing, 100043, China
| | - Bo Pang
- Department of Big Data, Computer Network Information Center, Chinese Academy of Sciences, Beijing, 100190, China
| | - Rong Wang
- Department of Stomatology, Shijingshan Teaching Hospital of Capital Medical University, Beijing Shijingshan Hospital, Beijing, 100043, China
| | - Bin Yang
- Department of Stomatology, Shijingshan Teaching Hospital of Capital Medical University, Beijing Shijingshan Hospital, Beijing, 100043, China
| | - Xubei Jia
- Department of Stomatology, Shijingshan Teaching Hospital of Capital Medical University, Beijing Shijingshan Hospital, Beijing, 100043, China
| |
Collapse
|
2
|
El Maï M, Bird M, Allouche A, Targen S, Şerifoğlu N, Lopes-Bastos B, Guigonis JM, Kang D, Pourcher T, Yue JX, Ferreira MG. Gut-specific telomerase expression counteracts systemic aging in telomerase-deficient zebrafish. NATURE AGING 2023; 3:567-584. [PMID: 37142828 DOI: 10.1038/s43587-023-00401-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/21/2023] [Indexed: 05/06/2023]
Abstract
Telomere shortening is a hallmark of aging and is counteracted by telomerase. As in humans, the zebrafish gut is one of the organs with the fastest rate of telomere decline, triggering early tissue dysfunction during normal zebrafish aging and in prematurely aged telomerase mutants. However, whether telomere-dependent aging of an individual organ, the gut, causes systemic aging is unknown. Here we show that tissue-specific telomerase expression in the gut can prevent telomere shortening and rescues premature aging of tert-/-. Induction of telomerase rescues gut senescence and low cell proliferation, while restoring tissue integrity, inflammation and age-dependent microbiota dysbiosis. Averting gut aging causes systemic beneficial impacts, rescuing aging of distant organs such as reproductive and hematopoietic systems. Conclusively, we show that gut-specific telomerase expression extends the lifespan of tert-/- by 40%, while ameliorating natural aging. Our work demonstrates that gut-specific rescue of telomerase expression leading to telomere elongation is sufficient to systemically counteract aging in zebrafish.
Collapse
Affiliation(s)
- Mounir El Maï
- Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR7284, INSERM U1081, Université Côte d'Azur, Nice, France
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Malia Bird
- Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR7284, INSERM U1081, Université Côte d'Azur, Nice, France
| | - Asma Allouche
- Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR7284, INSERM U1081, Université Côte d'Azur, Nice, France
| | - Seniye Targen
- Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR7284, INSERM U1081, Université Côte d'Azur, Nice, France
| | - Naz Şerifoğlu
- Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR7284, INSERM U1081, Université Côte d'Azur, Nice, France
| | - Bruno Lopes-Bastos
- Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR7284, INSERM U1081, Université Côte d'Azur, Nice, France
| | - Jean-Marie Guigonis
- Laboratory Transporter in Imaging and Radiotherapy in Oncology, Institut des Sciences du Vivant Frederic Joliot, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Côte d'Azur, Nice, France
| | - Da Kang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Thierry Pourcher
- Laboratory Transporter in Imaging and Radiotherapy in Oncology, Institut des Sciences du Vivant Frederic Joliot, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Côte d'Azur, Nice, France
| | - Jia-Xing Yue
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Miguel Godinho Ferreira
- Institute for Research on Cancer and Aging of Nice (IRCAN), CNRS UMR7284, INSERM U1081, Université Côte d'Azur, Nice, France.
- Instituto Gulbenkian de Ciência, Oeiras, Portugal.
| |
Collapse
|
3
|
Lansdorp PM. Telomeres, aging, and cancer: the big picture. Blood 2022; 139:813-821. [PMID: 35142846 PMCID: PMC8832478 DOI: 10.1182/blood.2021014299] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 12/16/2021] [Indexed: 12/13/2022] Open
Abstract
The role of telomeres in human health and disease is yet to be fully understood. The limitations of mouse models for the study of human telomere biology and difficulties in accurately measuring the length of telomere repeats in chromosomes and cells have diverted attention from many important and relevant observations. The goal of this perspective is to summarize some of these observations and to discuss the antagonistic role of telomere loss in aging and cancer in the context of developmental biology, cell turnover, and evolution. It is proposed that both damage to DNA and replicative loss of telomeric DNA contribute to aging in humans, with the differences in leukocyte telomere length between humans being linked to the risk of developing specific diseases. These ideas are captured in the Telomere Erosion in Disposable Soma theory of aging proposed herein.
Collapse
Affiliation(s)
- Peter M Lansdorp
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, BC, Canada; and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
4
|
Galati A, Scatolini L, Micheli E, Bavasso F, Cicconi A, Maccallini P, Chen L, Roake CM, Schoeftner S, Artandi SE, Gatti M, Cacchione S, Raffa GD. The S-adenosylmethionine analog sinefungin inhibits the trimethylguanosine synthase TGS1 to promote telomerase activity and telomere lengthening. FEBS Lett 2022; 596:42-52. [PMID: 34817067 DOI: 10.1002/1873-3468.14240] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 10/16/2021] [Accepted: 11/09/2021] [Indexed: 12/11/2022]
Abstract
Mutations in many genes that control the expression, the function, or the stability of telomerase cause telomere biology disorders (TBDs), such as dyskeratosis congenita, pulmonary fibrosis, and aplastic anemia. Mutations in a subset of the genes associated with TBDs cause reductions of the telomerase RNA moiety hTR, thus limiting telomerase activity. We have recently found that loss of the trimethylguanosine synthase TGS1 increases both hTR abundance and telomerase activity and leads to telomere elongation. Here, we show that treatment with the S-adenosylmethionine analog sinefungin inhibits TGS1 activity, increases the hTR levels, and promotes telomere lengthening in different cell types. Our results hold promise for restoring telomere length in stem and progenitor cells from TBD patients with reduced hTR levels.
Collapse
Affiliation(s)
- Alessandra Galati
- Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Italy
| | - Livia Scatolini
- Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Italy
| | - Emanuela Micheli
- Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Italy
| | - Francesca Bavasso
- Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Italy
| | - Alessandro Cicconi
- Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Italy
| | - Paolo Maccallini
- Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Italy
| | - Lu Chen
- Cancer Signaling and Epigenetics Program-Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Caitlin M Roake
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Stefan Schoeftner
- Dipartimento di Scienze della Vita, Università degli studi di Trieste, Italy
| | - Steven E Artandi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Maurizio Gatti
- Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Italy
- Istituto di Biologia e Patologia Molecolari del CNR, Roma, Italy
| | - Stefano Cacchione
- Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Italy
| | - Grazia D Raffa
- Dipartimento di Biologia e Biotecnologie, Sapienza Università di Roma, Italy
| |
Collapse
|
5
|
Vonderheide RH, Kraynyak KA, Shields AF, McRee AJ, Johnson JM, Sun W, Chintakuntlawar AV, Pawlicki J, Sylvester AJ, McMullan T, Samuels R, Kim JJ, Weiner D, Boyer JD, Morrow MP, Humeau L, Skolnik JM. Phase 1 study of safety, tolerability and immunogenicity of the human telomerase (hTERT)-encoded DNA plasmids INO-1400 and INO-1401 with or without IL-12 DNA plasmid INO-9012 in adult patients with solid tumors. J Immunother Cancer 2021; 9:jitc-2021-003019. [PMID: 34230114 PMCID: PMC8261871 DOI: 10.1136/jitc-2021-003019] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2021] [Indexed: 01/09/2023] Open
Abstract
Background Human telomerase reverse transcriptase (hTERT) is frequently classified as a ‘universal’ tumor associated antigen due to its expression in a vast number of cancers. We evaluated plasmid DNA-encoded hTERT as an immunotherapy across nine cancer types. Methods A phase 1 clinical trial was conducted in adult patients with no evidence of disease following definitive surgery and standard therapy, who were at high risk of relapse. Plasmid DNA encoding one of two hTERT variants (INO-1400 or INO-1401) with or without plasmid DNA encoding interleukin 12 (IL-12) (INO-9012) was delivered intramuscularly concurrent with the application of the CELLECTRA constant-current electroporation device 4 times across 12 weeks. Safety assessments and immune monitoring against native (germline, non-mutated, non-plasmid matched) hTERT antigen were performed. The largest cohort of patients enrolled had pancreatic cancer, allowing for additional targeted assessments for this tumor type. Results Of the 93 enrolled patients who received at least one dose, 88 had at least one adverse event; the majority were grade 1 or 2, related to injection site. At 18 months, 54.8% (51/93) patients were disease-free, with median disease-free survival (DFS) not reached by end of study. For patients with pancreatic cancer, the median DFS was 9 months, with 41.4% of these patients remaining disease-free at 18 months. hTERT immunotherapy induced a de novo cellular immune response or enhanced pre-existing cellular responses to native hTERT in 96% (88/92) of patients with various cancer types. Treatment with INO-1400/INO-1401±INO-9012 drove hTERT-specific IFN-γ production, generated hTERT-specific CD4+ and CD8+ T cells expressing the activation marker CD38, and induced hTERT-specific activated CD8 +CTLs as defined by cells expressing perforin and granzymes. The addition of plasmid IL-12 adjuvant elicited higher magnitudes of cellular responses including IFN-γ production, activated CD4+ and CD8+ T cells, and activated CD8+CTLs. In a subset analysis of pancreatic cancer patients, the presence of immunotherapy-induced activated CD8+ T cells expressing PD-1, granzymes and perforin correlated with survival. Conclusions Plasmid DNA-encoded hTERT/IL-12 DNA immunotherapy was well-tolerated, immune responses were noted across all tumor types, and a specific CD8+ phenotype increased by the immunotherapy was significantly correlated with survival in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Robert H Vonderheide
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | - Anthony F Shields
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, USA
| | - Autumn J McRee
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Jennifer M Johnson
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Weijing Sun
- University of Kansas Medical Center, Department of Medicine, Division of Medical Oncology, Kansas City, Kansas, USA
| | | | - Jan Pawlicki
- Inovio Pharmaceuticals, Plymouth Meeting, Pennsylvania, USA
| | | | | | - Robert Samuels
- Inovio Pharmaceuticals, Plymouth Meeting, Pennsylvania, USA
| | - Joseph J Kim
- Inovio Pharmaceuticals, Plymouth Meeting, Pennsylvania, USA
| | - David Weiner
- Wistar Institute, Philadelphia, Pennsylvania, USA
| | - Jean D Boyer
- Inovio Pharmaceuticals, Plymouth Meeting, Pennsylvania, USA
| | | | - Laurent Humeau
- Inovio Pharmaceuticals, Plymouth Meeting, Pennsylvania, USA
| | | |
Collapse
|
6
|
Jacczak B, Rubiś B, Totoń E. Potential of Naturally Derived Compounds in Telomerase and Telomere Modulation in Skin Senescence and Aging. Int J Mol Sci 2021; 22:6381. [PMID: 34203694 PMCID: PMC8232155 DOI: 10.3390/ijms22126381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/26/2021] [Accepted: 06/07/2021] [Indexed: 12/14/2022] Open
Abstract
Proper functioning of cells-their ability to divide, differentiate, and regenerate-is dictated by genomic stability. The main factors contributing to this stability are the telomeric ends that cap chromosomes. Telomere biology and telomerase activity have been of interest to scientists in various medical science fields for years, including the study of both cancer and of senescence and aging. All these processes are accompanied by telomere-length modulation. Maintaining the key levels of telomerase component (hTERT) expression and telomerase activity that provide optimal telomere length as well as some nontelomeric functions represents a promising step in advanced anti-aging strategies, especially in dermocosmetics. Some known naturally derived compounds contribute significantly to telomere and telomerase metabolism. However, before they can be safely used, it is necessary to assess their mechanisms of action and potential side effects. This paper focuses on the metabolic potential of natural compounds to modulate telomerase and telomere biology and thus prevent senescence and skin aging.
Collapse
Affiliation(s)
| | | | - Ewa Totoń
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, 49 Przybyszewskiego St., 60-355 Poznań, Poland; (B.J.); (B.R.)
| |
Collapse
|
7
|
Herrmann W, Herrmann M. The Importance of Telomere Shortening for Atherosclerosis and Mortality. J Cardiovasc Dev Dis 2020; 7:jcdd7030029. [PMID: 32781553 PMCID: PMC7570376 DOI: 10.3390/jcdd7030029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/11/2022] Open
Abstract
Telomeres are the protective end caps of chromosomes and shorten with every cell division. Short telomeres are associated with older age and adverse lifestyle factors. Leucocyte telomere length (LTL) has been proposed as a biomarker of biological age. The shortening of LTL with age is the result of the end-replication problem, environmental, and lifestyle-related factors. Epidemiologic studies have shown that LTL predicts cardiovascular disease, all-cause mortality, and death from vascular causes. Age appears to be an important co-variate that explains a substantial fraction of this effect. Although it has been proposed that short telomeres promote atherosclerosis and impair the repair of vascular lesions, existing results are inconsistent. Oxidative stress and chronic inflammation can both accelerate telomere shortening. Multiple factors, including homocysteine (HCY), vitamin B6, and vitamin B12 modulate oxidative stress and inflammation through direct and indirect mechanisms. This review provides a compact overview of telomere physiology and the utility of LTL measurements in atherosclerosis and cardiovascular disease. In addition, it summarizes existing knowledge regarding the impact of oxidative stress, inflammation, HCY, and B-vitamins on telomere function.
Collapse
Affiliation(s)
- Wolfgang Herrmann
- Department of Clinical Chemistry, Medical School of the Saarland University, 66421 Homburg, Saar, Germany;
| | - Markus Herrmann
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, 8036 Graz, Austria
- Correspondence: or ; Tel.: +43-316-385-13145; Fax: +43-316-385-13430
| |
Collapse
|
8
|
Roake CM, Artandi SE. Regulation of human telomerase in homeostasis and disease. Nat Rev Mol Cell Biol 2020; 21:384-397. [PMID: 32242127 DOI: 10.1038/s41580-020-0234-z] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2020] [Indexed: 12/14/2022]
Abstract
Telomerase is a ribonucleoprotein complex, the catalytic core of which includes the telomerase reverse transcriptase (TERT) and the non-coding human telomerase RNA (hTR), which serves as a template for the addition of telomeric repeats to chromosome ends. Telomerase expression is restricted in humans to certain cell types, and telomerase levels are tightly controlled in normal conditions. Increased levels of telomerase are found in the vast majority of human cancers, and we have recently begun to understand the mechanisms by which cancer cells increase telomerase activity. Conversely, germline mutations in telomerase-relevant genes that decrease telomerase function cause a range of genetic disorders, including dyskeratosis congenita, idiopathic pulmonary fibrosis and bone marrow failure. In this Review, we discuss the transcriptional regulation of human TERT, hTR processing, assembly of the telomerase complex, the cellular localization of telomerase and its recruitment to telomeres, and the regulation of telomerase activity. We also discuss the disease relevance of each of these steps of telomerase biogenesis.
Collapse
Affiliation(s)
- Caitlin M Roake
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven E Artandi
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
9
|
de Pedro N, Díez M, García I, García J, Otero L, Fernández L, García B, González R, Rincón S, Pérez D, Rodríguez E, Segovia E, Najarro P. Analytical Validation of Telomere Analysis Technology® for the High-Throughput Analysis of Multiple Telomere-Associated Variables. Biol Proced Online 2020; 22:2. [PMID: 31956299 PMCID: PMC6961256 DOI: 10.1186/s12575-019-0115-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 12/23/2019] [Indexed: 02/07/2023] Open
Abstract
Background A large number of studies have suggested a correlation between the status of telomeres and disease risk. High-throughput quantitative fluorescence in situ hybridization (HT Q-FISH) is a highly accurate telomere measurement technique that can be applied to the study of large cell populations. Here we describe the analytical performance testing and validation of Telomere Analysis Technology (TAT®), a laboratory-developed HT Q-FISH-based methodology that includes HT imaging and software workflows that provide a highly detailed view of telomere populations. Methods TAT was developed for the analysis of telomeres in peripheral blood mononuclear cells (PBMCs). TAT was compared with Terminal Restriction Fragment (TRF) length analysis, and tested for accuracy, precision, limits of detection (LOD) and specificity, reportable range and reference range. Results Using 6 different lymphocyte cell lines, we found a high correlation between TAT and TRF for telomere length (R2 ≥ 0.99). The standard variation (assay error) of TAT was 454 base pairs, and the limit of detection of 800 base pairs. A standard curve was constructed to cover human median reportable range values and defined its lower limit at 4700 bp and upper limits at 14,400 bp. Using TAT, up to 223 telomere associated variables (TAVs) can be obtained from a single sample. A pilot, population study, of telomere analysis using TAT revealed high accuracy and reliability of the methodology. Conclusions Analytical validation of TAT shows that is a robust and reliable technique for the characterization of a detailed telomere profile in large cell populations. The combination of high-throughput imaging and software workflows allows for the collection of a large number of telomere-associated variables from each sample, which can then be used in epidemiological and clinical studies.
Collapse
Affiliation(s)
| | - María Díez
- Life Length SL, Miguel Ángel 11, 28010 Madrid, Spain
| | - Irene García
- Life Length SL, Miguel Ángel 11, 28010 Madrid, Spain
| | - Jorge García
- Life Length SL, Miguel Ángel 11, 28010 Madrid, Spain
| | | | | | | | - Rut González
- Life Length SL, Miguel Ángel 11, 28010 Madrid, Spain
| | - Sara Rincón
- Life Length SL, Miguel Ángel 11, 28010 Madrid, Spain
| | - Diego Pérez
- Life Length SL, Miguel Ángel 11, 28010 Madrid, Spain
| | | | | | - Pilar Najarro
- Life Length SL, Miguel Ángel 11, 28010 Madrid, Spain
| |
Collapse
|
10
|
Inflammation and Vascular Ageing: From Telomeres to Novel Emerging Mechanisms. High Blood Press Cardiovasc Prev 2019; 26:321-329. [DOI: 10.1007/s40292-019-00331-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/12/2019] [Indexed: 12/20/2022] Open
|
11
|
Corrêa T, Feltes BC, Riegel M. Integrated analysis of the critical region 5p15.3-p15.2 associated with cri-du-chat syndrome. Genet Mol Biol 2019; 42:186-196. [PMID: 30985858 PMCID: PMC6687350 DOI: 10.1590/1678-4685-gmb-2018-0173] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 07/29/2018] [Indexed: 11/21/2022] Open
Abstract
Cri-du-chat syndrome (CdCs) is one of the most common contiguous gene syndromes, with an incidence of 1:15,000 to 1:50,000 live births. To better understand the etiology of CdCs at the molecular level, we investigated theprotein-protein interaction (PPI) network within the critical chromosomal region 5p15.3-p15.2 associated with CdCs using systemsbiology. Data were extracted from cytogenomic findings from patients with CdCs. Based on clinical findings, molecular characterization of chromosomal rearrangements, and systems biology data, we explored possible genotype-phenotype correlations involving biological processes connected with CdCs candidate genes. We identified biological processes involving genes previously found to be associated with CdCs, such as TERT, SLC6A3, and CTDNND2, as well as novel candidate proteins with potential contributions to CdCs phenotypes, including CCT5, TPPP, MED10, ADCY2, MTRR, CEP72, NDUFS6, and MRPL36. Although further functional analyses of these proteins are required, we identified candidate proteins for the development of new multi-target genetic editing tools to study CdCs. Further research may confirm those that are directly involved in the development of CdCs phenotypes and improve our understanding of CdCs-associated molecular mechanisms.
Collapse
Affiliation(s)
- Thiago Corrêa
- Post-Graduate Program in Genetics and Molecular Biology,
Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bruno César Feltes
- Institute of Informatics, Universidade Federal do Rio Grande
do Sul, Porto Alegre, RS, Brazil
| | - Mariluce Riegel
- Post-Graduate Program in Genetics and Molecular Biology,
Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Medical Genetics Service, Hospital de Clínicas de Porto
Alegre, Porto Alegre, RS, Brazil
| |
Collapse
|
12
|
Chahine MN, Toupance S, El-Hakim S, Labat C, Gautier S, Moussallem T, Yared P, Asmar R, Benetos A. Telomere length and age-dependent telomere attrition: the blood-and-muscle model. Can J Physiol Pharmacol 2019; 97:328-334. [DOI: 10.1139/cjpp-2018-0582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Short telomere length (TL) is associated with atherosclerotic cardiovascular disease (ACVD) and other age-related diseases. It is unclear whether these associations originate from having inherently short TL or a faster TL attrition before or during disease development. We proposed the blood-and-muscle model to assess TL dynamics throughout life course. Our objective was to measure TL in leukocytes (LTL) and in skeletal muscle (MTL), which served as a proxy of TL at birth. The delta (MTL–LTL) represented life-long telomere attrition. Blood draws and skeletal muscle biopsies were performed on 35 Lebanese individuals undergoing surgery. Following DNA extraction, LTL and MTL were measured by Southern blot. In every individual aged between 30 and 85 years, MTL was longer than LTL. With age, MTL and LTL decreased, but the delta (MTL–LTL) increased by 14 bp/year. We validated the blood-and-muscle model that allowed us to identify TL, TL at birth, and lifelong TL attrition in a cross-sectional study. This model can be used in larger cross-sectional studies to evaluate the association of telomere dynamics with age-related diseases onset and progression.
Collapse
Affiliation(s)
- Mirna N. Chahine
- Foundation-Medical Research Institutes, Beirut, Lebanon
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
| | - Simon Toupance
- Université de Lorraine, Inserm, DCAC, F-54000 Nancy, France
- Université de Lorraine, CHRU-Nancy, Pôle “Maladies du Vieillissement, Gérontologie et Soins Palliatifs”, F-54000, France
- Nancyclotep-GIE, F-54000 Nancy, France
| | - Sandy El-Hakim
- Faculty of Public Health II, Lebanese University, Fanar, Lebanon
| | - Carlos Labat
- Université de Lorraine, Inserm, DCAC, F-54000 Nancy, France
| | - Sylvie Gautier
- Université de Lorraine, CHRU-Nancy, Pôle “Maladies du Vieillissement, Gérontologie et Soins Palliatifs”, F-54000, France
| | | | - Pierre Yared
- Faculty of Medical Sciences, Lebanese University, Hadath, Lebanon
| | - Roland Asmar
- Foundation-Medical Research Institutes, Beirut, Lebanon
| | - Athanase Benetos
- Université de Lorraine, Inserm, DCAC, F-54000 Nancy, France
- Université de Lorraine, CHRU-Nancy, Pôle “Maladies du Vieillissement, Gérontologie et Soins Palliatifs”, F-54000, France
| |
Collapse
|
13
|
Should we consider telomere length and telomerase activity in male factor infertility? Curr Opin Obstet Gynecol 2019; 30:197-202. [PMID: 29664790 DOI: 10.1097/gco.0000000000000451] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to analyze what is known to date about the relation between telomeres and male fertility, and if it is possible for telomeres, or elements related to them, to be used as new prognostic biomarkers in fertility treatment. RECENT FINDINGS Cells in germ series, including spermatozoids, have longer telomeres (10-20 kb), and do not seem to undergo the shortening that takes place in somatic cells with age as they present telomerase activity. Longer telomere length found in the sperm of older fathers, influences their offspring possessing cells with longer telomere length. Infertile patients have spermatozoids with shorter telomere length than fertile people, but telomere length does neither correlate with the sperm concentration, mobility or morphology, nor with the DNA fragmentation indices (DFI) of spermatozoids. Embryo quality rate and transplantable embryo rate are related with the telomere length of spermatozoids (STL), but pregnancy rates are not affected. SUMMARY Telomere length and telomerase levels can be used as biomarkers of male fertility. Higher STL can have beneficial effects on fertility, thus the use of spermatozoids with longer telomere length in an assisted reproduction technique (ART) could be one way of solving some infertility cases.
Collapse
|
14
|
Telomeres in Plants and Humans: Not So Different, Not So Similar. Cells 2019; 8:cells8010058. [PMID: 30654521 PMCID: PMC6356271 DOI: 10.3390/cells8010058] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/07/2019] [Accepted: 01/07/2019] [Indexed: 01/01/2023] Open
Abstract
Parallel research on multiple model organisms shows that while some principles of telomere biology are conserved among all eukaryotic kingdoms, we also find some deviations that reflect different evolutionary paths and life strategies, which may have diversified after the establishment of telomerase as a primary mechanism for telomere maintenance. Much more than animals, plants have to cope with environmental stressors, including genotoxic factors, due to their sessile lifestyle. This is, in principle, made possible by an increased capacity and efficiency of the molecular systems ensuring maintenance of genome stability, as well as a higher tolerance to genome instability. Furthermore, plant ontogenesis differs from that of animals in which tissue differentiation and telomerase silencing occur during early embryonic development, and the “telomere clock” in somatic cells may act as a preventive measure against carcinogenesis. This does not happen in plants, where growth and ontogenesis occur through the serial division of apical meristems consisting of a small group of stem cells that generate a linear series of cells, which differentiate into an array of cell types that make a shoot and root. Flowers, as generative plant organs, initiate from the shoot apical meristem in mature plants which is incompatible with the human-like developmental telomere shortening. In this review, we discuss differences between human and plant telomere biology and the implications for aging, genome stability, and cell and organism survival. In particular, we provide a comprehensive comparative overview of telomere proteins acting in humans and in Arabidopsis thaliana model plant, and discuss distinct epigenetic features of telomeric chromatin in these species.
Collapse
|
15
|
M'kacher R, Cuceu C, Al Jawhari M, Morat L, Frenzel M, Shim G, Lenain A, Hempel WM, Junker S, Girinsky T, Colicchio B, Dieterlen A, Heidingsfelder L, Borie C, Oudrhiri N, Bennaceur-Griscelli A, Moralès O, Renaud S, Van de Wyngaert Z, Jeandidier E, Delhem N, Carde P. The Transition between Telomerase and ALT Mechanisms in Hodgkin Lymphoma and Its Predictive Value in Clinical Outcomes. Cancers (Basel) 2018; 10:E169. [PMID: 29848986 PMCID: PMC6025489 DOI: 10.3390/cancers10060169] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 05/18/2018] [Accepted: 05/25/2018] [Indexed: 11/16/2022] Open
Abstract
Background: We analyzed telomere maintenance mechanisms (TMMs) in lymph node samples from HL patients treated with standard therapy. The TMMs correlated with clinical outcomes of patients. Materials and Methods: Lymph node biopsies obtained from 38 HL patients and 24 patients with lymphadenitis were included in this study. Seven HL cell lines were used as in vitro models. Telomerase activity (TA) was assessed by TRAP assay and verified through hTERT immunofluorescence expression; alternative telomere lengthening (ALT) was also assessed, along with EBV status. Results: Both TA and ALT mechanisms were present in HL lymph nodes. Our findings were reproduced in HL cell lines. The highest levels of TA were expressed in CD30-/CD15- cells. Small cells were identified with ALT and TA. Hodgkin and Reed Sternberg cells contained high levels of PML bodies, but had very low hTERT expression. There was a significant correlation between overall survival (p < 10-3), event-free survival (p < 10-4), and freedom from progression (p < 10-3) and the presence of an ALT profile in lymph nodes of EBV+ patients. Conclusion: The presence of both types of TMMs in HL lymph nodes and in HL cell lines has not previously been reported. TMMs correlate with the treatment outcome of EBV+ HL patients.
Collapse
Affiliation(s)
- Radhia M'kacher
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
- Cell Environment, DNA Damages R&D, Oncology Section, 75020 Paris, France.
| | - Corina Cuceu
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - Mustafa Al Jawhari
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - Luc Morat
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - Monika Frenzel
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - Grace Shim
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - Aude Lenain
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - William M Hempel
- Laboratoire de Radiobiologie et d'Oncologie, IRCM/DSV/CEA, 92265 Fontenay aux Roses, France.
| | - Steffen Junker
- Institute of Biomedicine, University of Aarhus, DK-8000 Aarhus C, Denmark.
| | - Theodore Girinsky
- Department of Radiation Therapy, Gustave Roussy Cancer Campus, 94808 Villejuif, France.
| | - Bruno Colicchio
- IRIMAS, Institut de Recherche en Informatique, Mathématiques, Automatique et Signal, Université de Haute-Alsace, 68093 Mulhouse, France.
| | - Alain Dieterlen
- IRIMAS, Institut de Recherche en Informatique, Mathématiques, Automatique et Signal, Université de Haute-Alsace, 68093 Mulhouse, France.
| | | | - Claire Borie
- Université Paris Sud, Service d'hématologie moléculaire et cytogénétique Paul brousse CHU paris Sud, Inserm UMRS935, 94800 Villejuif, France.
| | - Noufissa Oudrhiri
- Université Paris Sud, Service d'hématologie moléculaire et cytogénétique Paul brousse CHU paris Sud, Inserm UMRS935, 94800 Villejuif, France.
| | - Annelise Bennaceur-Griscelli
- Université Paris Sud, Service d'hématologie moléculaire et cytogénétique Paul brousse CHU paris Sud, Inserm UMRS935, 94800 Villejuif, France.
| | - Olivier Moralès
- CNRS, Institut Pasteur de Lille, UMR 8161-Immunoregulation of Virus-induced Cancers Team, F-59000 Lille, France.
| | - Sarah Renaud
- CNRS, Institut Pasteur de Lille, UMR 8161-Immunoregulation of Virus-induced Cancers Team, F-59000 Lille, France.
| | - Zoé Van de Wyngaert
- CHRU Lille Service des Maladies du Sang, Hopital Huriez, 59000 Lille, France.
| | - Eric Jeandidier
- Service de génétique, Groupe hospitalier de la région de Mulhouse Sud-Alsace, 68093 Mulhouse, France.
| | - Nadira Delhem
- CNRS, Institut Pasteur de Lille, UMR 8161-Immunoregulation of Virus-induced Cancers Team, F-59000 Lille, France.
| | - Patrice Carde
- Department of Medicine, Gustave Roussy Cancer Campus, 94808 Villejuif, France.
| |
Collapse
|
16
|
Eitsuka T, Nakagawa K, Kato S, Ito J, Otoki Y, Takasu S, Shimizu N, Takahashi T, Miyazawa T. Modulation of Telomerase Activity in Cancer Cells by Dietary Compounds: A Review. Int J Mol Sci 2018; 19:E478. [PMID: 29415465 PMCID: PMC5855700 DOI: 10.3390/ijms19020478] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 01/25/2018] [Accepted: 02/01/2018] [Indexed: 12/26/2022] Open
Abstract
Telomerase is expressed in ~90% of human cancer cell lines and tumor specimens, whereas its enzymatic activity is not detectable in most human somatic cells, suggesting that telomerase represents a highly attractive target for selective cancer treatment. Accordingly, various classes of telomerase inhibitors have been screened and developed in recent years. We and other researchers have successfully found that some dietary compounds can modulate telomerase activity in cancer cells. Telomerase inhibitors derived from food are subdivided into two groups: one group directly blocks the enzymatic activity of telomerase (e.g., catechin and sulfoquinovosyldiacylglycerol), and the other downregulates the expression of human telomerase reverse transcriptase (hTERT), the catalytic subunit of human telomerase, via signal transduction pathways (e.g., retinoic acid and tocotrienol). In contrast, a few dietary components, including genistein and glycated lipid, induce cellular telomerase activity in several types of cancer cells, suggesting that they may be involved in tumor progression. This review summarizes the current knowledge about the effects of dietary factors on telomerase regulation in cancer cells and discusses their molecular mechanisms of action.
Collapse
Affiliation(s)
- Takahiro Eitsuka
- Food & Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai 980-0845, Japan.
| | - Kiyotaka Nakagawa
- Food & Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai 980-0845, Japan.
| | - Shunji Kato
- Food & Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai 980-0845, Japan.
| | - Junya Ito
- Food & Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai 980-0845, Japan.
| | - Yurika Otoki
- Food & Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai 980-0845, Japan.
| | - Soo Takasu
- Food & Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai 980-0845, Japan.
| | - Naoki Shimizu
- Food & Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai 980-0845, Japan.
| | - Takumi Takahashi
- Food & Biodynamic Chemistry Laboratory, Graduate School of Agricultural Science, Tohoku University, Sendai 980-0845, Japan.
| | - Teruo Miyazawa
- Food and Biotechnology Innovation Project, New Industry Creation Hatchery Center (NICHe), Tohoku University, Sendai 980-8579, Japan.
- Food and Health Science Research Unit, Graduate School of Agricultural Science, Tohoku University, Sendai 980-0845, Japan.
| |
Collapse
|
17
|
Lu AT, Xue L, Salfati EL, Chen BH, Ferrucci L, Levy D, Joehanes R, Murabito JM, Kiel DP, Tsai PC, Yet I, Bell JT, Mangino M, Tanaka T, McRae AF, Marioni RE, Visscher PM, Wray NR, Deary IJ, Levine ME, Quach A, Assimes T, Tsao PS, Absher D, Stewart JD, Li Y, Reiner AP, Hou L, Baccarelli AA, Whitsel EA, Aviv A, Cardona A, Day FR, Wareham NJ, Perry JRB, Ong KK, Raj K, Lunetta KL, Horvath S. GWAS of epigenetic aging rates in blood reveals a critical role for TERT. Nat Commun 2018; 9:387. [PMID: 29374233 PMCID: PMC5786029 DOI: 10.1038/s41467-017-02697-5] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/19/2017] [Indexed: 02/02/2023] Open
Abstract
DNA methylation age is an accurate biomarker of chronological age and predicts lifespan, but its underlying molecular mechanisms are unknown. In this genome-wide association study of 9907 individuals, we find gene variants mapping to five loci associated with intrinsic epigenetic age acceleration (IEAA) and gene variants in three loci associated with extrinsic epigenetic age acceleration (EEAA). Mendelian randomization analysis suggests causal influences of menarche and menopause on IEAA and lipoproteins on IEAA and EEAA. Variants associated with longer leukocyte telomere length (LTL) in the telomerase reverse transcriptase gene (TERT) paradoxically confer higher IEAA (P < 2.7 × 10-11). Causal modeling indicates TERT-specific and independent effects on LTL and IEAA. Experimental hTERT-expression in primary human fibroblasts engenders a linear increase in DNA methylation age with cell population doubling number. Together, these findings indicate a critical role for hTERT in regulating the epigenetic clock, in addition to its established role of compensating for cell replication-dependent telomere shortening.
Collapse
Affiliation(s)
- Ake T Lu
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Luting Xue
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Elias L Salfati
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Brian H Chen
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
- National Heart, Lung and Blood Institute, Bethesda, MD, 20824-0105, USA
| | - Luigi Ferrucci
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Daniel Levy
- National Heart, Lung and Blood Institute, Bethesda, MD, 20824-0105, USA
| | - Roby Joehanes
- National Heart, Lung and Blood Institute, Bethesda, MD, 20824-0105, USA
| | - Joanne M Murabito
- Department of Medicine, Section of General Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Douglas P Kiel
- Institute for Aging Research, Hebrew SeniorLife, Beth Israel Deaconess Medical Centre, Harvard Medical School, Boston, MA, 02215, USA
| | - Pei-Chien Tsai
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, SE1 7EH, UK
| | - Idil Yet
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, SE1 7EH, UK
| | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, SE1 7EH, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, SE1 7EH, UK
| | - Toshiko Tanaka
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Allan F McRae
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, QLD, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Riccardo E Marioni
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, QLD, Australia
- Centre for Cognitive Aging and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Peter M Visscher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, QLD, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Naomi R Wray
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, QLD, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Ian J Deary
- Centre for Cognitive Aging and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
| | - Morgan E Levine
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Austin Quach
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Themistocles Assimes
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Philip S Tsao
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, 35806, USA
| | - James D Stewart
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Yun Li
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Alex P Reiner
- Fred Hutchinson Cancer Research Center Box 358080, WHI Clinical Coordinating Ctr/Public Health Sciences M3-A4, Seattle, WA, 98109, USA
| | - Lifang Hou
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University Chicago, Evanston, IL, 60611, USA
- Center for Population Epigenetics, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University Chicago, Evanston, IL, 60611, USA
| | - Andrea A Baccarelli
- Laboratory of Environmental Epigenetics, Departments of Environmental Health Sciences Epidemiology, Columbia University Mailman School of Public Health, New York, NY, 10032, USA
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, 27516, USA
| | - Abraham Aviv
- The Center for Human Development and Aging, University of Medicine and Dentistry, New Jersey Medical School, Rutgers, Newark, NJ, 07103, USA
| | - Alexia Cardona
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, CB2 0SL, UK
| | - Felix R Day
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, CB2 0SL, UK
| | - Nicholas J Wareham
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, CB2 0SL, UK
| | - John R B Perry
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, CB2 0SL, UK
| | - Ken K Ong
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, CB2 0SL, UK
- Department of Paediatrics, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 0SP, UK
| | - Kenneth Raj
- Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, Oxfordshire, OX11 0RQ, UK
| | - Kathryn L Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Steve Horvath
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Biostatistics, School of Public Health, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
18
|
Abstract
BACKGROUND Telomeres are protein DNA structures present at the ends of chromosomes and are essential for genetic stability and cell replication. Telomerase is the enzyme complex that maintains telomere integrity. Hematopoietic stem cells express telomerase and contain long telomeres, which become shorter as cells differentiate and mature. The extent of telomere shortening and the level of telomerase activity often correlate with the presence and severity of some hematopoietic diseases. METHODS The fundamentals of telomeres and telomerase are reviewed, and the telomere biology of human hematopoietic cells is discussed. RESULTS Telomere length and telomerase activity are important in the self-renewal of hematopoietic stem cells. Changes within these compartments affect both normal hematopoietic cells and the generation of hematopoietic disease. Telomere length provides information pertaining to the proliferative history and potential of a hematopoietic cell. CONCLUSIONS The role of telomerase and telomeres within the hematopoietic compartment needs further clarification. Advances in our knowledge in this field may improve clinical outcomes for the treatment of hematologic disease.
Collapse
Affiliation(s)
- Ngaire Elwood
- Leukaemia Research Fund Stem Cell Laboratory, Department of Clinical Haematology and Oncology, Murdoch Children's Research Institute, Melbourne, Australia.
| |
Collapse
|
19
|
p53 Mediates Failure of Human Definitive Hematopoiesis in Dyskeratosis Congenita. Stem Cell Reports 2017; 9:409-418. [PMID: 28757166 PMCID: PMC5550027 DOI: 10.1016/j.stemcr.2017.06.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 06/27/2017] [Accepted: 06/27/2017] [Indexed: 12/23/2022] Open
Abstract
Dyskeratosis congenita (DC) is a bone marrow failure syndrome associated with telomere dysfunction. The progression and molecular determinants of hematopoietic failure in DC remain poorly understood. Here, we use the directed differentiation of human embryonic stem cells harboring clinically relevant mutations in telomerase to understand the consequences of DC-associated mutations on the primitive and definitive hematopoietic programs. Interestingly, telomere shortening does not broadly impair hematopoiesis, as primitive hematopoiesis is not impaired in DC cells. In contrast, while phenotypic definitive hemogenic endothelium is specified, the endothelial-to-hematopoietic transition is impaired in cells with shortened telomeres. This failure is caused by DNA damage accrual and is mediated by p53 stabilization. These observations indicate that detrimental effects of telomere shortening in the hematopoietic system are specific to the definitive hematopoietic lineages. This work illustrates how telomere dysfunction impairs hematopoietic development and creates a robust platform for therapeutic discovery for treatment of DC patients. CRISPR/Cas9 was used to generate different DC-associated mutations in hESCs Telomere shortening specifically impairs definitive hematopoietic specification Genetic instability and p53 activation regulate hematopoietic specification in DC In vitro hematopoietic failure resembles phenotypes of aplastic anemia in DC
Collapse
|
20
|
Kang HJ, Cui Y, Yin H, Scheid A, Hendricks WPD, Schmidt J, Sekulic A, Kong D, Trent JM, Gokhale V, Mao H, Hurley LH. A Pharmacological Chaperone Molecule Induces Cancer Cell Death by Restoring Tertiary DNA Structures in Mutant hTERT Promoters. J Am Chem Soc 2016; 138:13673-13692. [PMID: 27643954 DOI: 10.1021/jacs.6b07598] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Activation of human telomerase reverse transcriptase (hTERT) is necessary for limitless replication in tumorigenesis. Whereas hTERT is transcriptionally silenced in normal cells, most tumor cells reactivate hTERT expression by alleviating transcriptional repression through diverse genetic and epigenetic mechanisms. Transcription-activating hTERT promoter mutations have been found to occur at high frequencies in multiple cancer types. These mutations have been shown to form new transcription factor binding sites that drive hTERT expression, but this model cannot fully account for differences in wild-type (WT) and mutant promoter activation and has not yet enabled a selective therapeutic strategy. Here, we demonstrate a novel mechanism by which promoter mutations activate hTERT transcription, which also sheds light on a unique therapeutic opportunity. Promoter mutations occur in a core promoter region that forms tertiary structures consisting of a pair of G-quadruplexes involved in transcriptional silencing. We show that promoter mutations exert a detrimental effect on the folding of one of these G-quadruplexes, resulting in a nonfunctional silencer element that alleviates transcriptional repression. We have also identified a small drug-like pharmacological chaperone (pharmacoperone) molecule, GTC365, that acts at an early step in the G-quadruplex folding pathway to redirect mutant promoter G-quadruplex misfolding, partially reinstate the correct folding pathway, and reduce hTERT activity through transcriptional repression. This transcription-mediated repression produces cancer cell death through multiple routes including both induction of apoptosis through inhibition of hTERT's role in regulating apoptosis-related proteins and induction of senescence by decreasing telomerase activity and telomere length. We demonstrate the selective therapeutic potential of this strategy in melanoma cells that overexpress hTERT.
Collapse
Affiliation(s)
- Hyun-Jin Kang
- University of Arizona , College of Pharmacy, 1703 East Mabel Street, Tucson, Arizona 85721, United States
| | - Yunxi Cui
- Department of Chemistry and Biochemistry and School of Biomedical Sciences, Kent State University , Kent, Ohio 44242, United States
| | - Holly Yin
- Translational Genomics Research Institute , 445 North Fifth Street, Phoenix, Arizona 85004, United States
| | - Amy Scheid
- College of Science, University of Arizona , 1040 East Fourth Street, Tucson, Arizona 85721, United States
| | - William P D Hendricks
- Translational Genomics Research Institute , 445 North Fifth Street, Phoenix, Arizona 85004, United States
| | - Jessica Schmidt
- Department of Dermatology, Mayo Clinic , 13400 East Shea Boulevard, Scottsdale, Arizona 85259, United States
| | - Aleksandar Sekulic
- Department of Dermatology, Mayo Clinic , 13400 East Shea Boulevard, Scottsdale, Arizona 85259, United States
| | - Deming Kong
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry, Nankai University , Tianjin 300071, People's Republic of China
| | - Jeffrey M Trent
- Translational Genomics Research Institute , 445 North Fifth Street, Phoenix, Arizona 85004, United States
| | - Vijay Gokhale
- BIO5 Institute , 1657 East Helen Street, Tucson, Arizona 85721, United States
| | - Hanbin Mao
- Department of Chemistry and Biochemistry and School of Biomedical Sciences, Kent State University , Kent, Ohio 44242, United States
| | - Laurence H Hurley
- University of Arizona , College of Pharmacy, 1703 East Mabel Street, Tucson, Arizona 85721, United States.,BIO5 Institute , 1657 East Helen Street, Tucson, Arizona 85721, United States.,Arizona Cancer Center , 1515 North Campbell Avenue, Tucson, Arizona 85724, United States
| |
Collapse
|
21
|
Zanetti M. A second chance for telomerase reverse transcriptase in anticancer immunotherapy. Nat Rev Clin Oncol 2016; 14:115-128. [DOI: 10.1038/nrclinonc.2016.67] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
22
|
Bell RJA, Rube HT, Xavier-Magalhães A, Costa BM, Mancini A, Song JS, Costello JF. Understanding TERT Promoter Mutations: A Common Path to Immortality. Mol Cancer Res 2016; 14:315-23. [PMID: 26941407 PMCID: PMC4852159 DOI: 10.1158/1541-7786.mcr-16-0003] [Citation(s) in RCA: 201] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 02/24/2016] [Indexed: 12/23/2022]
Abstract
Telomerase (TERT) activation is a fundamental step in tumorigenesis. By maintaining telomere length, telomerase relieves a main barrier on cellular lifespan, enabling limitless proliferation driven by oncogenes. The recently discovered, highly recurrent mutations in the promoter of TERT are found in over 50 cancer types, and are the most common mutation in many cancers. Transcriptional activation of TERT, via promoter mutation or other mechanisms, is the rate-limiting step in production of active telomerase. Although TERT is expressed in stem cells, it is naturally silenced upon differentiation. Thus, the presence of TERT promoter mutations may shed light on whether a particular tumor arose from a stem cell or more differentiated cell type. It is becoming clear that TERT mutations occur early during cellular transformation, and activate the TERT promoter by recruiting transcription factors that do not normally regulate TERT gene expression. This review highlights the fundamental and widespread role of TERT promoter mutations in tumorigenesis, including recent progress on their mechanism of transcriptional activation. These somatic promoter mutations, along with germline variation in the TERT locus also appear to have significant value as biomarkers of patient outcome. Understanding the precise molecular mechanism of TERT activation by promoter mutation and germline variation may inspire novel cancer cell-specific targeted therapies for a large number of cancer patients.
Collapse
Affiliation(s)
- Robert J A Bell
- Department of Neurological Surgery, University of California, San Francisco, California
| | - H Tomas Rube
- Department of Biological Sciences, Columbia University, New York, New York
| | - Ana Xavier-Magalhães
- Department of Neurological Surgery, University of California, San Francisco, California. Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal. ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Braga, Portugal
| | - Bruno M Costa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal. ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Braga, Portugal
| | - Andrew Mancini
- Department of Neurological Surgery, University of California, San Francisco, California
| | - Jun S Song
- Departments of Bioengineering and Physics, University of Illinois, Urbana-Champaign, Illinois
| | - Joseph F Costello
- Department of Neurological Surgery, University of California, San Francisco, California.
| |
Collapse
|
23
|
Barczak W, Rozwadowska N, Romaniuk A, Lipińska N, Lisiak N, Grodecka-Gazdecka S, Książek K, Rubiś B. Telomere length assessment in leukocytes presents potential diagnostic value in patients with breast cancer. Oncol Lett 2016; 11:2305-2309. [PMID: 26998167 DOI: 10.3892/ol.2016.4188] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 01/05/2016] [Indexed: 11/05/2022] Open
Abstract
Telomere shortening is associated with cancer development, primarily through the induction of genomic instability. The majority of studies have indicated that individuals with shorter blood telomeres may be at a higher risk of developing various types of cancer. There is increasing evidence that the study of the alterations in telomere length may improve cancer prognosis. The aim of the present study was to verify the use of telomere length parameters in the diagnostics of breast cancer stage. Telomere length was analyzed in the blood leukocytes of 52 patients with breast cancer relative to 47 control subjects using quantitative polymerase chain reaction. The effects of stage, grade, estrogen receptor, progesterone receptor and human epidermal growth factor 2 (HER2) status were assessed. The current study demonstrated that the average telomeric sequence length was significantly shorter in leukocytes from individuals diagnosed with a more severe stage of breast cancer (T2N1M0) than in leukocytes in the early stages of the disease (T1N0M0) (P=0.0207). Furthermore, the data indicated that telomeres in leukocytes derived from patients with HER2+ breast cancer were significantly longer compared with those with the HER2- type (P=0.0347). These results suggest that the assessment of telomeres in blood leukocytes may, at least partially, correspond with breast cancer staging and HER2 receptor status.
Collapse
Affiliation(s)
- Wojciech Barczak
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, The Greater Poland Cancer Centre, Poznan 61-866, Poland; Radiobiology Laboratory, Department of Medical Physics, The Greater Poland Cancer Centre, Poznan 61-866, Poland
| | - Natalia Rozwadowska
- Institute of Human Genetics, Polish Academy of Sciences, Poznan 60-479, Poland
| | - Aleksandra Romaniuk
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Poznan 60-355, Poland
| | - Natalia Lipińska
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Poznan 60-355, Poland
| | - Natalia Lisiak
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Poznan 60-355, Poland
| | | | - Krzysztof Książek
- Department of Pathophysiology, Laboratory of Gerontology, Poznan University of Medical Sciences, Poznan 60-781, Poland
| | - Błażej Rubiś
- Department of Clinical Chemistry and Molecular Diagnostics, Poznan University of Medical Sciences, Poznan 60-355, Poland
| |
Collapse
|
24
|
Liu T, Yu H, Ding L, Wu Z, Gonzalez De Los Santos F, Liu J, Ullenbruch M, Hu B, Martins V, Phan SH. Conditional Knockout of Telomerase Reverse Transcriptase in Mesenchymal Cells Impairs Mouse Pulmonary Fibrosis. PLoS One 2015; 10:e0142547. [PMID: 26555817 PMCID: PMC4640706 DOI: 10.1371/journal.pone.0142547] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/25/2015] [Indexed: 12/31/2022] Open
Abstract
Telomerase is typically expressed in cellular populations capable of extended replication, such as germ cells, tumor cells, and stem cells, but is also induced in tissue injury, repair and fibrosis. Its catalytic component, telomerase reverse transcriptase (TERT) is induced in lung fibroblasts from patients with fibrotic interstitial lung disease and in rodents with bleomycin-induced pulmonary fibrosis. To evaluate the fibroblast specific role of TERT in pulmonary fibrosis, transgenic mice bearing a floxed TERT allele were generated, and then crossed with an inducible collagen α2(I)-Cre mouse line to generate fibroblast specific TERT conditional knockout mice. TERT-specific deficiency in mesenchymal cells caused attenuation of pulmonary fibrosis as manifested by reduced lung hydroxyproline content, type I collagen and α-smooth muscle actin mRNA levels. The TERT-deficient mouse lung fibroblasts displayed decreased cell proliferative capacity and higher susceptibility to induced apoptosis compared with control cells. Additionally TERT deficiency was associated with heightened α-smooth muscle actin expression indicative of myofibroblast differentiation. However the impairment of cell proliferation and increased susceptibility to apoptosis would cause a reduction in the myofibroblast progenitor population necessary to mount a successful myofibroblast-dependent fibrotic response. These findings identified a key role for TERT in fibroblast proliferation and survival essential for pulmonary fibrosis.
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Hongfeng Yu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Lin Ding
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Zhe Wu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | | | - Jianhua Liu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Matthew Ullenbruch
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Biao Hu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Vanessa Martins
- Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Sem H. Phan
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
25
|
Hjelmborg JB, Dalgård C, Möller S, Steenstrup T, Kimura M, Christensen K, Kyvik KO, Aviv A. The heritability of leucocyte telomere length dynamics. J Med Genet 2015; 52:297-302. [PMID: 25770094 PMCID: PMC4413805 DOI: 10.1136/jmedgenet-2014-102736] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Accepted: 10/14/2014] [Indexed: 01/17/2023]
Abstract
Background Leucocyte telomere length (LTL) is a complex trait associated with ageing and longevity. LTL dynamics are defined by LTL and its age-dependent attrition. Strong, but indirect evidence suggests that LTL at birth and its attrition during childhood largely explains interindividual LTL variation among adults. A number of studies have estimated the heritability of LTL, but none has assessed the heritability of age-dependent LTL attrition. Methods We examined the heritability of LTL dynamics based on a longitudinal evaluation (an average follow-up of 12 years) in 355 monozygotic and 297 dizygotic same-sex twins (aged 19–64 years at baseline). Results Heritability of LTL at baseline was estimated at 64% (95% CI 39% to 83%) with 22% (95% CI 6% to 49%) of shared environmental effects. Heritability of age-dependent LTL attrition rate was estimated at 28% (95% CI 16% to 44%). Individually unique environmental factors, estimated at 72% (95% CI 56% to 84%) affected LTL attrition rate with no indication of shared environmental effects. Conclusions This is the first study that estimated heritability of LTL and also its age-dependent attrition. As LTL attrition is much slower in adults than in children and given that having a long or a short LTL is largely determined before adulthood, our findings suggest that heritability and early life environment are the main determinants of LTL throughout the human life course. Thus, insights into factors that influence LTL at birth and its dynamics during childhood are crucial for understanding the role of telomere genetics in human ageing and longevity.
Collapse
Affiliation(s)
- Jacob B Hjelmborg
- Department of Epidemiology, Biostatistics and Biodemography, Institute of Public Health, University of Southern Denmark, Odense, Denmark The Danish Twin Registry, University of Southern Denmark, Odense, Denmark
| | - Christine Dalgård
- Department of Environmental Medicine, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | - Soren Möller
- Department of Epidemiology, Biostatistics and Biodemography, Institute of Public Health, University of Southern Denmark, Odense, Denmark The Danish Twin Registry, University of Southern Denmark, Odense, Denmark
| | - Troels Steenstrup
- Department of Epidemiology, Biostatistics and Biodemography, Institute of Public Health, University of Southern Denmark, Odense, Denmark The Danish Twin Registry, University of Southern Denmark, Odense, Denmark
| | - Masayuki Kimura
- Center of Human Development and Aging, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| | - Kaare Christensen
- Department of Epidemiology, Biostatistics and Biodemography, Institute of Public Health, University of Southern Denmark, Odense, Denmark The Danish Twin Registry, University of Southern Denmark, Odense, Denmark Department of Clinical Genetics, Odense University Hospital, Odense, Denmark Department of Clinical Biochemistry and Pharmacology, Odense University Hospital, Odense, Denmark
| | - Kirsten O Kyvik
- Institute of Regional Health Services Research, University of Southern Denmark and Odense Patient data Explorative Network (OPEN), Odense University Hospital, Odense, Denmark
| | - Abraham Aviv
- Center of Human Development and Aging, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
26
|
Indumathi S, Mishra R, Harikrishnan R, Dhanasekaran M. Subcutaneous Adipose Tissue-Derived Stem Cells: Advancement and Applications in Regenerative Medicine. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
27
|
Solaimani Kartalaei P, Yamada-Inagawa T, Vink CS, de Pater E, van der Linden R, Marks-Bluth J, van der Sloot A, van den Hout M, Yokomizo T, van Schaick-Solernó ML, Delwel R, Pimanda JE, van IJcken WFJ, Dzierzak E. Whole-transcriptome analysis of endothelial to hematopoietic stem cell transition reveals a requirement for Gpr56 in HSC generation. ACTA ACUST UNITED AC 2014; 212:93-106. [PMID: 25547674 PMCID: PMC4291529 DOI: 10.1084/jem.20140767] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Using highly sensitive RNAseq to examine the whole transcriptome of enriched aortic hematopoietic stem cells and endothelial cells, the authors find G-protein–coupled receptor, Gpr56, is required to generate the first HSCs during endothelial to hematopoietic cell transition. Hematopoietic stem cells (HSCs) are generated via a natural transdifferentiation process known as endothelial to hematopoietic cell transition (EHT). Because of small numbers of embryonal arterial cells undergoing EHT and the paucity of markers to enrich for hemogenic endothelial cells (ECs [HECs]), the genetic program driving HSC emergence is largely unknown. Here, we use a highly sensitive RNAseq method to examine the whole transcriptome of small numbers of enriched aortic HSCs, HECs, and ECs. Gpr56, a G-coupled protein receptor, is one of the most highly up-regulated of the 530 differentially expressed genes. Also, highly up-regulated are hematopoietic transcription factors, including the “heptad” complex of factors. We show that Gpr56 (mouse and human) is a target of the heptad complex and is required for hematopoietic cluster formation during EHT. Our results identify the processes and regulators involved in EHT and reveal the surprising requirement for Gpr56 in generating the first HSCs.
Collapse
Affiliation(s)
- Parham Solaimani Kartalaei
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Center for Biomics, and Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Tomoko Yamada-Inagawa
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Center for Biomics, and Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Chris S Vink
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Center for Biomics, and Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Emma de Pater
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Center for Biomics, and Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Reinier van der Linden
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Center for Biomics, and Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Jonathon Marks-Bluth
- Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Anthon van der Sloot
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Center for Biomics, and Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Mirjam van den Hout
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Center for Biomics, and Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Tomomasa Yokomizo
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599
| | - M Lucila van Schaick-Solernó
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Center for Biomics, and Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Ruud Delwel
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Center for Biomics, and Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - John E Pimanda
- Lowy Cancer Research Centre and Prince of Wales Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Wilfred F J van IJcken
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Center for Biomics, and Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| | - Elaine Dzierzak
- Erasmus MC Stem Cell Institute, Department of Cell Biology, Center for Biomics, and Department of Hematology, Erasmus University Medical Center, 3000 CA Rotterdam, Netherlands
| |
Collapse
|
28
|
Won YS, Jeong JS, Kim SJ, Ju MH, Lee SW. Targeted anticancer effect through microRNA-181a regulated tumor-specific hTERT replacement. Cancer Lett 2014; 356:918-28. [PMID: 25444904 DOI: 10.1016/j.canlet.2014.11.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 11/01/2014] [Accepted: 11/04/2014] [Indexed: 01/11/2023]
Abstract
We previously generated a group I intron-based ribozyme that can reprogram human telomerase reverse transcriptase (hTERT) RNA to stimulate transgene activity in cancer cells expressing the target RNA via an accurate and specific trans-splicing reaction. One of the major concerns of the hTERT RNA targeting anti-cancer approach is the potential side effects to hTERT(+) hematopoietic stem cell-derived blood cells. Thus, here we modified the ribozyme by inserting target sites against microRNA-181a, which is a blood cell-specific microRNA, downstream of its 3' exon. The specificity of transgene induction and anticancer activity in hTERT(+) cancer cells improved significantly with the modified ribozyme, resulting in selective targeting of hTERT(+) cancer cells, but not hematopoietic cells even if they are hTERT-positive. Importantly, the trans-splicing reaction of the microRNA-regulated ribozyme worked equally well in a nude mouse model of hepatocarcinoma-derived intrasplenic carcinomatosis, inducing highly specific expression of a therapeutic transgene and efficiently regressing hTERT-positive liver tumors with minimal liver toxicity when systemically delivered with an adenoviral vector encoding the ribozyme. These results suggest that a combined approach of microRNA regulation with targeted RNA replacement is more useful for effective anti-cancer treatment.
Collapse
Affiliation(s)
- You-Sub Won
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, and Research Institute of Advanced Omics, Dankook University, Yongin, Republic of Korea
| | - Jin-Sook Jeong
- Department of Pathology and Medical Research Center for Cancer Molecular Therapy, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Sung Jin Kim
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, and Research Institute of Advanced Omics, Dankook University, Yongin, Republic of Korea
| | - Mi Ha Ju
- Department of Pathology and Medical Research Center for Cancer Molecular Therapy, Dong-A University College of Medicine, Busan, Republic of Korea
| | - Seong-Wook Lee
- Department of Molecular Biology, Institute of Nanosensor and Biotechnology, and Research Institute of Advanced Omics, Dankook University, Yongin, Republic of Korea.
| |
Collapse
|
29
|
Liu AH, Sun X, Wei XQ, Zhang YZ. Efficacy of multiple low-dose photodynamic TMPYP4 therapy on cervical cancer tumour growth in nude mice. Asian Pac J Cancer Prev 2014; 14:5371-4. [PMID: 24175828 DOI: 10.7314/apjcp.2013.14.9.5371] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE Photodynamic therapy (PDT) is an emerging therapeutic procedure suitable for the treatment of cervical cancer. However, the side effects of PDT are severe, including skin ulceration, so we designed an experiment to examine the effects of multiple low- dose photodynamic therapy of 5, 10, 15, 20-tetrakis(1- methylpyridinium-4-yl) porphyrin (Tmpyp4) on tumour growth by utilizing a model in nude mice implanted with Hela cervical cancer cells. MATERIALS AND METHODS Female BALB/c nude mice (aged 5-6 weeks, weighing 18-20 g) were used. Hela cervical cancer cells were injected subcutaneously (1 x 10(7) cells/200 μL). Ten days after injection, the mice were divided into three groups (n=6), the A group of controls without any treatment, the B group receiving a single-treatment with Tmpyp4 (10 mg/kg, intratumor injection) and irradiation (blue laser, 108 J/cm(2)), and the C group given three-treatments with Tmpyp4 (10 mg/ kg, intratumor injection) and irradiation at intervals of two days. After starting treatment, tumours were measured every two days, to assess growth. At 2 weeks after the last treatment of C group, tumour tissue and organs were collected from each mouse to evaluate tumor histology and organ damage. RESULTS Tumour growth in C group was significantly inhibited compared with A and B groups (P <0.05), without any injury to the skin and internal organs. CONCLUSION Our novel findings demonstrated that multiple low-dose photodynamic therapy of Tmpyp4 could inhibit cervical cancer growth significantly with no apparent side effects.
Collapse
Affiliation(s)
- Ai-Hong Liu
- Department of Gynecology and Obstetrics, Qilu Hospital of Shandong Univeristy, Jinan, China E-mail :
| | | | | | | |
Collapse
|
30
|
Marrone A, Dokal I. Dyskeratosis congenita: a disorder of telomerase deficiency and its relationship to other diseases. ACTA ACUST UNITED AC 2014. [DOI: 10.1586/17469872.1.3.463] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
31
|
Batista LFZ. Telomere biology in stem cells and reprogramming. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 125:67-88. [PMID: 24993698 DOI: 10.1016/b978-0-12-397898-1.00003-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Telomerase expression in humans is restricted to different populations of stem and progenitor cells, being silenced in most somatic tissues. Efficient telomere homeostasis is essential for embryonic and adult stem cell function and therefore essential for tissue homeostasis throughout organismal life. Accordingly, the mutations in telomerase culminate in reduced stem cell function both in vivo and in vitro and have been associated with tissue dysfunction in human patients. Despite the importance of telomerase for stem cell biology, the mechanisms behind telomerase regulation during development are still poorly understood, mostly due to difficulties in acquiring and maintaining pluripotent stem cell populations in culture. In this chapter, we will analyze recent developments in this field, including the importance of efficient telomere homeostasis in different stem cell types and the role of telomerase in different techniques used for cellular reprogramming.
Collapse
Affiliation(s)
- Luis F Z Batista
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA; Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
32
|
Daniali L, Benetos A, Susser E, Kark JD, Labat C, Kimura M, Desai K, Granick M, Aviv A. Telomeres shorten at equivalent rates in somatic tissues of adults. Nat Commun 2013; 4:1597. [PMID: 23511462 PMCID: PMC3615479 DOI: 10.1038/ncomms2602] [Citation(s) in RCA: 468] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 02/14/2013] [Indexed: 01/22/2023] Open
Abstract
Telomere shortening in somatic tissues largely reflects stem cell replication. Previous human studies of telomere attrition were predominantly conducted on leukocytes. However, findings in leukocytes cannot be generalized to other tissues. Here we measure telomere length in leukocytes, skeletal muscle, skin and subcutaneous fat of 87 adults (aged 19–77 years). Telomeres are longest in muscle and shortest in leukocytes, yet are strongly correlated between tissues. Notably, the rates of telomere shortening are similar in the four tissues. We infer from these findings that differences in telomere length between proliferative (blood and skin) and minimally proliferative tissues (muscle and fat) are established during early life, and that in adulthood, stem cells of the four tissues replicate at a similar rate. Telomere shortening as a result of cell proliferation has been implicated in human ageing. Here, Daniali and colleagues show that telomere length and the rate of age-dependent shortening vary between adults but are similar within tissues of the same individual.
Collapse
Affiliation(s)
- Lily Daniali
- Division of Plastic Surgery, Department of Surgery, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, New Jersey 07103, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Batista LF, Artandi SE. Understanding telomere diseases through analysis of patient-derived iPS cells. Curr Opin Genet Dev 2013; 23:526-33. [PMID: 23993228 PMCID: PMC3925063 DOI: 10.1016/j.gde.2013.07.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 07/25/2013] [Accepted: 07/26/2013] [Indexed: 12/30/2022]
Abstract
A unique characteristic of tissue stem cells is the ability to self-renew, a process that enables the life-long maintenance of many organs. Stem cell self-renewal is dependent in part on the synthesis of telomere repeats by the enzyme telomerase. Defects in telomerase and in genes in the telomere maintenance pathway result in diverse disease states, including dyskeratosis congenita, pulmonary fibrosis, aplastic anemia, liver cirrhosis and cancer. Many of these disease states share a tissue failure phenotype, such as loss of bone marrow cells or failure of pulmonary epithelium, suggesting that stem cell dysfunction is a common pathophysiological mechanism underlying these telomere diseases. Studies of telomere diseases in undifferentiated iPS cells have provided a quantitative relationship between the magnitude of biochemical defects in the telomerase pathway and disease severity in patients, thereby establishing a clear correlation between genotype and phenotype in telomere disease states. Modeling telomere diseases in iPS cells has also revealed diverse underlying disease mechanisms, including reduced telomerase catalytic activity, diminished assembly of the telomerase holoenzyme and impaired trafficking of the enzyme within the nucleus. These studies highlight the need for therapies tailored to the underlying biochemical defect in each class of patients.
Collapse
Affiliation(s)
- Luis F.Z. Batista
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Steven E. Artandi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
- Biochemistry Department, Stanford University School of Medicine, Stanford, CA 94305, USA
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA 94305, USA
- The Glenn Laboratories for the Biology of Aging, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
34
|
Ozturk S, Sozen B, Demir N. Telomere length and telomerase activity during oocyte maturation and early embryo development in mammalian species. Mol Hum Reprod 2013; 20:15-30. [DOI: 10.1093/molehr/gat055] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
35
|
Benetos A, Kark JD, Susser E, Kimura M, Sinnreich R, Chen W, Steenstrup T, Christensen K, Herbig U, von Bornemann Hjelmborg J, Srinivasan SR, Berenson GS, Labat C, Aviv A. Tracking and fixed ranking of leukocyte telomere length across the adult life course. Aging Cell 2013; 12:615-21. [PMID: 23601089 PMCID: PMC3798089 DOI: 10.1111/acel.12086] [Citation(s) in RCA: 170] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2013] [Indexed: 01/20/2023] Open
Abstract
Short leukocyte telomere length (LTL) is associated with atherosclerosis in adults and diminished survival in the elderly. LTL dynamics are defined by LTL at birth, which is highly variable, and its age-dependent attrition thereafter, which is rapid during the first 20 years of life. We examined whether age-dependent LTL attrition during adulthood can substantially affect individuals’ LTL ranking (e.g., longer or shorter LTL) in relation to their peers. We measured LTL in samples donated 12 years apart on average by 1156 participants in four longitudinal studies. We observed correlations of 0.91–0.96 between baseline and follow-up LTLs. Ranking individuals by deciles revealed that 94.1% (95% confidence interval of 92.6–95.4%) showed no rank change or a 1 decile change over time. We conclude that in adults, LTL is virtually anchored to a given rank with the passage of time. Accordingly, the links of LTL with atherosclerosis and longevity appear to be established early in life. It is unlikely that lifestyle and its modification during adulthood exert a major impact on LTL ranking.
Collapse
Affiliation(s)
- Athanase Benetos
- Geriatric Service Nancy University Hospital Nancy 54511France
- Inserm U1116 Faculty of Medicine, Université de Lorraine Nancy 54500France
| | - Jeremy D. Kark
- The Hebrew University–Hadassah School of Public Health and Community Medicine Ein Kerem Jerusalem Israel
| | - Ezra Susser
- Department of Epidemiology Mailman School of Public Health, Columbia University New York NY 10032USA
- New York State Psychiatric Institute New York NY 10032USA
| | - Masayuki Kimura
- The Center of Human Development and Aging University of Medicine and Dentistry of New Jersey, New Jersey Medical School Newark NJ 07103 USA
| | - Ronit Sinnreich
- The Hebrew University–Hadassah School of Public Health and Community Medicine Ein Kerem Jerusalem Israel
| | - Wei Chen
- Center for Cardiovascular Health Tulane University New Orleans LA 70118USA
| | - Troels Steenstrup
- Epidemiology and Statistics Units Institute of Public Health, Danish Twin Registry and Danish Aging Research Center, University of Southern Denmark Odense Denmark
| | - Kaare Christensen
- Epidemiology and Statistics Units Institute of Public Health, Danish Twin Registry and Danish Aging Research Center, University of Southern Denmark Odense Denmark
| | - Utz Herbig
- Department of Microbiology and Molecular Genetics University of Medicine and Dentistry of New Jersey, New Jersey Medical School Newark NJ 07103 USA
| | - Jacob von Bornemann Hjelmborg
- Epidemiology and Statistics Units Institute of Public Health, Danish Twin Registry and Danish Aging Research Center, University of Southern Denmark Odense Denmark
| | | | - Gerald S. Berenson
- Center for Cardiovascular Health Tulane University New Orleans LA 70118USA
| | - Carlos Labat
- Geriatric Service Nancy University Hospital Nancy 54511France
- Inserm U1116 Faculty of Medicine, Université de Lorraine Nancy 54500France
| | - Abraham Aviv
- Department of Epidemiology Mailman School of Public Health, Columbia University New York NY 10032USA
- The Center of Human Development and Aging University of Medicine and Dentistry of New Jersey, New Jersey Medical School Newark NJ 07103 USA
| |
Collapse
|
36
|
Aviv A, Susser E. Leukocyte telomere length and the father's age enigma: implications for population health and for life course. Int J Epidemiol 2013; 42:457-62. [PMID: 23382366 PMCID: PMC3619950 DOI: 10.1093/ije/dys236] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
What are the implications for population health of the demographic trend toward increasing paternal age at conception (PAC) in modern societies? We propose that the effects of older PAC are likely to be broad and harmful in some domains of health but beneficial in others. Harmful effects of older PAC have received the most attention. Thus, for example, older PAC is associated with an increased risk of offspring having rare conditions such as achondroplasia and Marfan syndrome, as well as with neurodevelopmental disorders such as autism. However, newly emerging evidence in the telomere field suggests potentially beneficial effects, since older PAC is associated with a longer leukocyte telomere length (LTL) in offspring, and a longer LTL is associated with a reduced risk of atherosclerosis and with increased survival in the elderly. Thus, older PAC may cumulatively increase resistance to atherosclerosis and lengthen lifespan in successive generations of modern humans. In this paper we: (i) introduce these novel findings; (ii) discuss potential explanations for the effect of older PAC on offspring LTL; (iii) draw implications for population health and for life course; (iv) put forth an evolutionary perspective as a context for the multigenerational effects of PAC; and (v) call for broad and intensive research to understand the mechanisms underlying the effects of PAC. We draw together work across a range of disciplines to offer an integrated perspective of this issue.
Collapse
Affiliation(s)
- Abraham Aviv
- The Center of Human Development and Aging, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, NJ 07103, USA.
| | | |
Collapse
|
37
|
Steenstrup T, Hjelmborg JVB, Mortensen LH, Kimura M, Christensen K, Aviv A. Leukocyte telomere dynamics in the elderly. Eur J Epidemiol 2013; 28:181-7. [PMID: 23430034 PMCID: PMC3604590 DOI: 10.1007/s10654-013-9780-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 02/06/2013] [Indexed: 11/24/2022]
Abstract
Limited data suggest that leukocytes of the elderly display ultra-short telomeres. It was reported that in some elderly persons leukocyte telomere length (LTL) shows age-dependent elongation. Using cross-sectional and longitudinal models, we characterized LTL dynamics in participants of the Longitudinal Study of Aging Danish Twins. We measured LTL by Southern blots of the terminal restriction fragment length (TRFL) in 476 individuals (73–94 years) in a cross-sectional evaluation and in a subset of this cohort comprising 80 individuals (73–81 years at baseline) who were followed–up for approximately 10 years. Based on the mean TRFL, we found that a) the average rate of LTL attrition was respectively, 27 bp/year (P < 0.001) and 31 bp/year (P < 0.001) for the cross-sectional and longitudinal evaluations, and b) mean TRFL was 180 bp (95 % CI 43, 320) longer in females than males (P < 0.010). For the TRFL distribution, which captures telomeres of all lengths in the DNA sample, we observed significant shifts with age toward shorter telomeres. Based on the measurement error of the TRFLs, we computed that in the longitudinal evaluation 10.6 % of individuals would manifest LTL elongation over 10 years, assuming a 340 bp attrition during this period. This was not significantly different from the empirical observation of 7.5 % of individuals showing LTL elongation. We conclude that accumulation of ultra-short telomeres in leukocytes of the elderly reflects a shift toward shorter telomeres in the entire telomere distribution. Measurement error is the probable explanation for LTL elongation in longitudinal studies.
Collapse
Affiliation(s)
- Troels Steenstrup
- Department of Biostatistics, Institute of Public Health, University of Southern Denmark, Odense, Denmark
| | | | | | | | | | | |
Collapse
|
38
|
Establishment of ultra long-lived cell lines by transfection of TERT into normal human fibroblast TIG-1 and their characterization. Cell Biol Int 2012; 36:519-27. [PMID: 22273270 DOI: 10.1042/cbi20110460] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
To establish useful human normal cell lines, TERT (telomerase reverse transcriptase) cDNA was transfected into normal female lung fibroblast, TIG-1. After long-term-sub-cultivation of 74 individual clones selected for resistance to G418, we obtained 55 cultures with normal range of life span [75 PDL (population doubling level)], 16 cultures with extended life span (75-140 PDL). In addition, 3 immortal cell strains and unexpectedly, one ultra long-lived cell line (ULT-1) with life span of 166 PDL were established. IMT-1, one of the immortal cell strains was confirmed to maintain long telomere length, high telomerase activity and an extremely low level of p16INK4A. They also showed moderate p53 and p21CIP1 expression, keeping vigorous growth rate even at 450 PDL. High level of fibronectin and collagen 1α expression confirmed IMT-1 as normal fibroblasts, although one X chromosome had been lost. ULT-1, however, kept a near normal karyotypes and had shortening of telomere length, high expression of p16INK4A, moderate levels of senescence associated-β-galactosidase positive cells and decreased growth rate only after 150 PDs (population doublings), and finally reached senescence at 166 PDL with morphology of normal senescent fibroblasts. As resources of standard normal human cell, abundant vials of early and middle passages of ULT-1 have been stocked. The use of the cell line is discussed, focusing on isograft of artificial skin and screening of anti-aging or safe chemical agents.
Collapse
|
39
|
Xu L, Li S, Stohr BA. The role of telomere biology in cancer. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2012; 8:49-78. [PMID: 22934675 DOI: 10.1146/annurev-pathol-020712-164030] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Telomere biology plays a critical and complex role in the initiation and progression of cancer. Although telomere dysfunction resulting from replicative attrition constrains tumor growth by engaging DNA-damage signaling pathways, it can also promote tumorigenesis by causing oncogenic chromosomal rearrangements. Expression of the telomerase enzyme enables telomere-length homeostasis and allows tumor cells to escape the antiproliferative barrier posed by short telomeres. Telomeres and telomerase also function independently of one another. Recent work has suggested that telomerase promotes cell growth through pathways unrelated to telomere maintenance, and a subset of tumors elongate telomeres through telomerase-independent mechanisms. In an effort to exploit the integral link between telomere biology and cancer growth, investigators have developed several telomerase-based therapeutic strategies, which are currently in clinical trials. Here, we broadly review the state of the field with a particular focus on recent developments of interest.
Collapse
Affiliation(s)
- Lifeng Xu
- Department of Microbiology, University of California-Davis, CA 95616, USA
| | | | | |
Collapse
|
40
|
Aston KI, Hunt SC, Susser E, Kimura M, Factor-Litvak P, Carrell D, Aviv A. Divergence of sperm and leukocyte age-dependent telomere dynamics: implications for male-driven evolution of telomere length in humans. Mol Hum Reprod 2012; 18:517-22. [PMID: 22782639 PMCID: PMC3480822 DOI: 10.1093/molehr/gas028] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Telomere length (TL) dynamics in vivo are defined by TL and its age-dependent change, brought about by cell replication. Leukocyte TL (LTL), which reflects TL in hematopoietic stem cells (HSCs), becomes shorter with age. In contrast, sperm TL, which reflects TL in the male germ cells, becomes longer with age. Moreover, offspring of older fathers display longer LTL. Thus far, no study has examined LTL and sperm TL relations with age in the same individuals, nor considered their implications for the paternal age at conception (PAC) effect on offspring LTL. We report that in 135 men (mean age: 34.4 years; range: 18–68 years) on average, LTL became shorter by 19 bp/year (r = −0.3; P = 0.0004), while sperm TL became longer by 57 bp/year (r = 0.32; P = 0.0002). Based on previously reported replication rates of HSCs and male germ cells, we estimate that HSCs lose 26 bp per replication. However, male germ cells gain only 2.48 bp per replication. As TL is inherited in an allele-specific manner, the magnitude of the PAC effect on the offspring's LTL should be approximately half of age-dependent sperm-TL elongation. When we compared the PAC effect data from previous studies with sperm-TL data from this study, the result was consistent with this prediction. As older paternal age is largely a feature of contemporary humans, we suggest that there may be progressive elongation of TL in future generations. In this sense, germ cell TL dynamics could be driving the evolution of TL in modern humans and perhaps telomere-related diseases in the general population.
Collapse
Affiliation(s)
- Kenneth I Aston
- Andrology and IVF Laboratories, Department of Surgery, University of Utah School of Medicine, Salt Lake City, UT 84108, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
Aviv A. Genetics of leukocyte telomere length and its role in atherosclerosis. Mutat Res 2012; 730:68-74. [PMID: 21600224 PMCID: PMC3202050 DOI: 10.1016/j.mrfmmm.2011.05.001] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 04/27/2011] [Accepted: 05/02/2011] [Indexed: 02/07/2023]
Abstract
Humans display a large inter-individual variation in leukocyte telomere length (LTL), which is influenced by heredity, sex, race/ethnicity, paternal age at conception and environmental exposures. LTL dynamics (birth LTL and its age-dependent attrition thereafter) mirror telomere dynamics in hematopoietic stem cells (HSCs). LTL at birth is evidently a major determinant of LTL throughout the human lifespan, such that individuals endowed with short (or long) LTL at birth probably have short (or long) LTL later in life. Therefore, the associations of short LTL with atherosclerosis and with diminished survival in the elderly may relate to short birth LTL, accelerated age-dependent LTL attrition, or both. The mechanisms underlying these associations are still not well understood, but they stem in part from genetic factors in control of telomere maintenance and the rate of HSC replication.
Collapse
Affiliation(s)
- Abraham Aviv
- The Center for Human Development and Aging, University of Medicine and Dentistry, New Jersey Medical School, Newark, NJ 07103, USA.
| |
Collapse
|
42
|
Slatter TL, Tan X, Yuen YC, Gunningham S, Ma SS, Daly E, Packer S, Devenish C, Royds JA, Hung NA. The alternative lengthening of telomeres pathway may operate in non-neoplastic human cells. J Pathol 2012; 226:509-18. [DOI: 10.1002/path.2981] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 07/27/2011] [Accepted: 08/04/2011] [Indexed: 01/20/2023]
|
43
|
Kong Y, Cui H, Ramkumar C, Zhang H. Regulation of senescence in cancer and aging. J Aging Res 2011; 2011:963172. [PMID: 21423549 PMCID: PMC3056284 DOI: 10.4061/2011/963172] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2010] [Accepted: 01/12/2011] [Indexed: 12/12/2022] Open
Abstract
Senescence is regarded as a physiological response of cells to stress, including telomere dysfunction, aberrant oncogenic activation, DNA damage, and oxidative stress. This stress response has an antagonistically pleiotropic effect to organisms: beneficial as a tumor suppressor, but detrimental by contributing to aging. The emergence of senescence as an effective tumor suppression mechanism is highlighted by recent demonstration that senescence prevents proliferation of cells at risk of neoplastic transformation. Consequently, induction of senescence is recognized as a potential treatment of cancer. Substantial evidence also suggests that senescence plays an important role in aging, particularly in aging of stem cells. In this paper, we will discuss the molecular regulation of senescence its role in cancer and aging. The potential utility of senescence in cancer therapeutics will also be discussed.
Collapse
Affiliation(s)
- Yahui Kong
- Department of Cell Biology, University of Massachusetts Medical School, 55 Lake Avenue North, S7-125, Worcester, MA 01655, USA
| | | | | | | |
Collapse
|
44
|
Kajstura J, Hosoda T, Bearzi C, Rota M, Maestroni S, Urbanek K, Leri A, Anversa P. The human heart: a self-renewing organ. Clin Transl Sci 2010; 1:80-6. [PMID: 20443822 DOI: 10.1111/j.1752-8062.2008.00030.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The dogma that the heart is a static organ which contains an irreplaceable population of cardiomyocytes prevailed in the cardiovascular field for the last several decades. However, the recent identification of progenitor cells that give rise to differentiated myocytes has prompted a re-interpretation of cardiac biology. The heart cannot be viewed any longer as a postmitotic organ characterized by a predetermined number of myocytes that is defined at birth and is preserved throughout life. The myocardium constitutes a dynamic entity in which new young parenchymal cells are formed to substitute old damaged dying myocytes. The regenerative ability of the heart was initially documented with a classic morphometric approach and more recently with the demonstration that DNA synthesis, mitosis, and cytokinesis take place in the newly formed myocytes of the normal and pathologic heart. Importantly, replicating myocytes correspond to the differentiated progeny of cardiac stem cells. These findings point to the possibility of novel therapeutic strategies for the diseased heart.
Collapse
Affiliation(s)
- Jan Kajstura
- Departments of Anesthesia and Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Brennan SK, Wang Q, Tressler R, Harley C, Go N, Bassett E, Huff CA, Jones RJ, Matsui W. Telomerase inhibition targets clonogenic multiple myeloma cells through telomere length-dependent and independent mechanisms. PLoS One 2010; 5. [PMID: 20824134 PMCID: PMC2931698 DOI: 10.1371/journal.pone.0012487] [Citation(s) in RCA: 58] [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: 06/12/2010] [Accepted: 08/09/2010] [Indexed: 12/28/2022] Open
Abstract
Background Plasma cells constitute the majority of tumor cells in multiple myeloma (MM) but lack the potential for sustained clonogenic growth. In contrast, clonotypic B cells can engraft and recapitulate disease in immunodeficient mice suggesting they serve as the MM cancer stem cell (CSC). These tumor initiating B cells also share functional features with normal stem cells such as drug resistance and self-renewal potential. Therefore, the cellular processes that regulate normal stem cells may serve as therapeutic targets in MM. Telomerase activity is required for the maintenance of normal adult stem cells, and we examined the activity of the telomerase inhibitor imetelstat against MM CSC. Moreover, we carried out both long and short-term inhibition studies to examine telomere length-dependent and independent activities. Methodology/Principal Findings Human MM CSC were isolated from cell lines and primary clinical specimens and treated with imetelstat, a specific inhibitor of the reverse transcriptase activity of telomerase. Two weeks of exposure to imetelstat resulted in a significant reduction in telomere length and the inhibition of clonogenic MM growth both in vitro and in vivo. In addition to these relatively long-term effects, 72 hours of imetelstat treatment inhibited clonogenic growth that was associated with MM CSC differentiation based on expression of the plasma cell antigen CD138 and the stem cell marker aldehyde dehydrogenase. Short-term treatment of MM CSC also decreased the expression of genes typically expressed by stem cells (OCT3/4, SOX2, NANOG, and BMI1) as revealed by quantitative real-time PCR. Conclusions Telomerase activity regulates the clonogenic growth of MM CSC. Moreover, reductions in MM growth following both long and short-term telomerase inhibition suggest that it impacts CSC through telomere length-dependent and independent mechanisms.
Collapse
Affiliation(s)
- Sarah K. Brennan
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Qiuju Wang
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Robert Tressler
- Geron Corporation, Menlo Park, California, United States of America
| | - Calvin Harley
- Geron Corporation, Menlo Park, California, United States of America
| | - Ning Go
- Geron Corporation, Menlo Park, California, United States of America
| | | | - Carol Ann Huff
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Richard J. Jones
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - William Matsui
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
46
|
Maritz MF, Napier CE, Wen VW, MacKenzie KL. Targeting telomerase in hematologic malignancy. Future Oncol 2010; 6:769-89. [PMID: 20465390 DOI: 10.2217/fon.10.42] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Over the past two decades, it has become increasingly apparent that telomerase-mediated telomere maintenance plays a crucial role in hematopoiesis. Supporting evidence is underscored by recent findings of mutations in genes involved in telomerase-mediated telomere maintenance that contribute to the pathogenesis of bone marrow failure syndromes. More recently described telomere-independent functions of telomerase are also likely to contribute to both normal hematopoiesis and hematologic diseases. The high levels of telomerase detected in aggressive leukemias have fueled fervent investigation into diverse approaches to targeting telomerase in hematologic malignancies. Successful preclinical investigations that employed genetic strategies, oligonucleotides, small-molecule inhibitors and immunotherapy have resulted in a rapid translation to clinical trials. Further investigation of telomere-independent functions of telomerase and detailed preclinical studies of telomerase inhibition in both normal and malignant hematopoiesis will be invaluable for refining treatments to effectively and safely exploit telomerase as a therapeutic target in hematologic malignancies.
Collapse
Affiliation(s)
- Michelle F Maritz
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, New South Wales, Australia
| | | | | | | |
Collapse
|
47
|
Berardinelli F, Antoccia A, Cherubini R, De Nadal V, Gerardi S, Cirrone GAP, Tanzarella C, Sgura A. Transient activation of the ALT pathway in human primary fibroblasts exposed to high-LET radiation. Radiat Res 2010; 174:539-49. [PMID: 20726710 DOI: 10.1667/rr2127.1] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
It is well established that high-LET radiations efficiently induce chromosome aberrations. However, data on the effect of protons on telomere maintenance, as involved in genomic stability, are scarce and contradictory. Here we demonstrate that high-LET protons induce telomere lengthening in human primary fibroblasts and that this elongation does not involve the telomerase enzyme, supporting the hypothesis that high-LET radiations are able to activate a telomerase-independent mechanism. In tumor cells that lack telomerase, one or more non-telomerase mechanisms for telomere maintenance are present, which are termed alternative lengthening of telomeres (ALT). Since ALT cells are characterized by recombinational events at telomeres, known as telomeric-sister chromatid exchanges (T-SCE), and colocalization of telomeres and premyelocytic leukemia protein (PML), we analyzed both T-SCE and PML. Our results show that high-LET protons induce a 2.5-fold increase of T-SCE and a colocalization of PML protein and telomeric DNA. Furthermore, our data show that the ALT pathway can be activated in human primary cells after induction of severe DNA damage. Thus, since telomeres are known to be involved in chromosome maintenance, the present work may contribute in the elucidation of the mechanism by which ionizing radiation induces genomic instability.
Collapse
|
48
|
Abstract
Telomerase and the control of telomere length are intimately linked to the process of tumourigenesis in humans. Here I review the evidence that variation at the 5p15.33 locus, which contains theTERTgene (encoding the catalytic subunit of telomerase), might play a role in the determination of cancer risk. Mutations in the coding regions ofTERTcan affect telomerase activity and telomere length, and create severe clinical phenotypes, including bone marrow failure syndromes and a substantive increase in cancer frequency. Variants within theTERTgene have been associated with increased risk of haematological malignancies, including myelodysplastic syndrome and acute myeloid leukaemia as well as chronic lymphocytic leukaemia. Furthermore, there is good evidence from a number of independent genome-wide association studies to implicate variants at the 5p15.33 locus in cancer risk at several different sites: lung cancer, basal cell carcinoma and pancreatic cancer show strong associations, while bladder, prostate and cervical cancer and glioma also show risk alleles in this region. Thus, multiple independent lines of evidence have implicated variation in theTERTgene as a risk factor for cancer. The mechanistic basis of these risk variants is yet to be established; however, the basic biology suggests that telomere length control is a tantalising candidate mechanism underlying cancer risk.
Collapse
|
49
|
O'Connor MS, Carlson ME, Conboy IM. Differentiation rather than aging of muscle stem cells abolishes their telomerase activity. Biotechnol Prog 2009; 25:1130-7. [PMID: 19455648 DOI: 10.1002/btpr.223] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
A general feature of stem cells is the ability to routinely proliferate to build, maintain, and repair organ systems. Accordingly, embryonic and germline, as well as some adult stem cells, produce the telomerase enzyme at various levels of expression. Our results show that, while muscle is a largely postmitotic tissue, the muscle stem cells (satellite cells) that maintain this biological system throughout adult life do indeed display robust telomerase activity. Conversely, primary myoblasts (the immediate progeny of satellite cells) quickly and dramatically downregulate telomerase activity. This work thus suggests that satellite cells, and early transient myoblasts, may be more promising therapeutic candidates for regenerative medicine than traditionally utilized myoblast cultures. Muscle atrophy accompanies human aging, and satellite cells endogenous to aged muscle can be triggered to regenerate old tissue by exogenous molecular cues. Therefore, we also examined whether these aged muscle stem cells would produce tissue that is "young" with respect to telomere maintenance. Interestingly, this work shows that the telomerase activity in muscle stem cells is largely retained into old age wintin inbred "long" telomere mice and in wild-derived short telomere mouse strains, and that age-specific telomere shortening is undetectable in the old differentiated muscle fibers of either strain. Summarily, this work establishes that young and old muscle stem cells, but not necessarily their progeny, myoblasts, are likely to produce tissue with normal telomere maintenance when used in molecular and regenerative medicine approaches for tissue repair.
Collapse
Affiliation(s)
- Matthew S O'Connor
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA
| | | | | |
Collapse
|
50
|
Abstract
Myriad genetic and epigenetic alterations are required to drive normal cells toward malignant transformation. These somatic events commandeer many signaling pathways that cooperate to endow aspiring cancer cells with a full range of biological capabilities needed to grow, disseminate and ultimately kill its host. Cancer genomes are highly rearranged and are characterized by complex translocations and regional copy number alterations that target loci harboring cancer-relevant genes. Efforts to uncover the underlying mechanisms driving genome instability in cancer have revealed a prominent role for telomeres. Telomeres are nucleoprotein structures that protect the ends of eukaryotic chromosomes and are particularly vulnerable due to progressive shortening during each round of DNA replication and, thus, a lifetime of tissue renewal places the organism at risk for increasing chromosomal instability. Indeed, telomere erosion has been documented in aging tissues and hyperproliferative disease states-conditions strongly associated with increased cancer risk. Telomere dysfunction can produce the opposing pathophysiological states of degenerative aging or cancer with the specific outcome dictated by the integrity of DNA damage checkpoint responses. In most advanced cancers, telomerase is reactivated and serves to maintain telomere length and emerging data have also documented the capacity of telomerase to directly regulate cancer-promoting pathways. This review covers the role of telomeres and telomerase in the biology of normal tissue stem/progenitor cells and in the development of cancer.
Collapse
Affiliation(s)
- Steven E Artandi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | |
Collapse
|