151
|
Ait-Aissa K, Heisner JS, Norwood Toro LE, Bruemmer D, Doyon G, Harmann L, Geurts A, Camara AKS, Beyer AM. Telomerase Deficiency Predisposes to Heart Failure and Ischemia-Reperfusion Injury. Front Cardiovasc Med 2019; 6:31. [PMID: 31001540 PMCID: PMC6454001 DOI: 10.3389/fcvm.2019.00031] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 03/07/2019] [Indexed: 12/13/2022] Open
Abstract
Introduction: Elevated levels of mitochondrial reactive oxygen species (ROS) contribute to the development of numerous cardiovascular diseases. TERT, the catalytic subunit of telomerase, has been shown to translocate to mitochondria to suppress ROS while promoting ATP production. Acute overexpression of TERT increases survival and decreases infarct size in a mouse model of myocardial infarct, while decreased telomerase activity predisposes to mitochondrial defects and heart failure. In the present study, we examined the role of TERT on cardiac structure and function under basal conditions and conditions of acute or prolonged stress in a novel rat model of TERT deficiency. Methods: Cardiac structure and function were evaluated via transthoracic echocardiogram. Langendorff preparations were used to test the effects of acute global ischemia reperfusion injury on cardiac function and infarction. Coronary flow and left ventricular pressure were measured during and after ischemia/reperfusion (I/R). Mitochondrial DNA integrity was measured by PCR and mitochondrial respiration was assessed in isolated mitochondria using an Oxygraph. Angiotensin II infusion was used as an established model of systemic stress. Results: No structural changes (echocardiogram) or coronary flow/left ventricle pressure (isolated hearts) were observed in TERT-/- rats at baseline; however, after I/R, coronary flow was significantly reduced in TERT-/- compared to wild type (WT) rats, while diastolic Left Ventricle Pressure was significantly elevated (n = 6 in each group; p < 0.05) in the TERT-/-. Interestingly, infarct size was less in TERT-/- rats compared to WT rats, while mitochondrial respiratory control index decreased and mitochondrial DNA lesions increased in TERT-/- compared to WT. Angiotensin II treatment did not alter cardiac structure or function; however, it augmented the infarct size significantly more in TERT-/- compared to the WT. Conclusion: Absence of TERT activity increases susceptibility to stress like cardiac injury. These results suggest a critical role of telomerase in chronic heart disease.
Collapse
Affiliation(s)
- Karima Ait-Aissa
- Cardiovascular Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - James S. Heisner
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Laura E. Norwood Toro
- Cardiovascular Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Dennis Bruemmer
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Genevieve Doyon
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, United States
| | - Leanne Harmann
- Cardiovascular Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Aron Geurts
- Cardiovascular Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Amadou K. S. Camara
- Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Andreas M. Beyer
- Cardiovascular Center, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
152
|
Demerdash HM, Elyamany AS, Arida E. Impact of direct-acting antivirals on leukocytic DNA telomere length in hepatitis C virus-related hepatic cirrhosis. Eur J Gastroenterol Hepatol 2019; 31:494-498. [PMID: 30444746 DOI: 10.1097/meg.0000000000001306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Direct-acting antiviral (DAAs) represent advancement in the management of hepatitis C virus (HCV)-related hepatic cirrhosis. A high proportion of patients achieve a sustained virologic response; eradication of HCV is coupled with a decreased risk of hepatocellular carcinoma. Recent evidence suggests that shortening of the DNA telomere may be linked to cellular senescence as well as predisposition to malignant transformation. OBJECTIVE This study aimed to assess pretreatment leukocytic DNA telomere length in HCV-related cirrhosis and post viral eradication using DAAs. PATIENTS AND METHODS This study included 24 patients with HCV-related cirrhosis, Child-Pugh A. Whole-blood samples were obtained from patients before treatment and 12 weeks after the end of treatment, as well as from 24 healthy controls. Terminal restriction fragment, corresponding to telomere length, was measured using a nonradioactive Southern blot technique, detected by chemiluminescence. RESULTS DNA telomere length was significantly shorter before treatment compared with 12 weeks after end of treatment in HCV-related cirrhotic patients. Also, it was significantly shorter in patients before treatment compared with healthy individuals. CONCLUSION Telomere elongation in blood leukocytes can be considered a marker of recovery of inflammation after DAAs-induced HCV eradication. Still, the possibility of activation by cancer initiation cannot be excluded.
Collapse
Affiliation(s)
- Hala M Demerdash
- Departments of Clinical Pathology, Alexandria University Hospitals
| | | | - Emad Arida
- Anaesthesia and Intensive Care, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
153
|
Green PD, Sharma NK, Santos JH. Telomerase Impinges on the Cellular Response to Oxidative Stress Through Mitochondrial ROS-Mediated Regulation of Autophagy. Int J Mol Sci 2019; 20:1509. [PMID: 30917518 PMCID: PMC6470917 DOI: 10.3390/ijms20061509] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/15/2019] [Accepted: 03/15/2019] [Indexed: 12/30/2022] Open
Abstract
Telomerase has cellular functions beyond telomere stabilization, including a role in mitochondria. The function of the catalytic component-TERT-in mitochondria is still unknown, but it seems to play a role in the response to oxidative stress. Here, we interrogated the role of the subcellular localization of TERT to the response to hydrogen peroxide (H₂O₂) treatment. Using normal human fibroblasts (NHF) expressing non-tagged wild type (WT) human TERT (hTERT) or nuclear localization and function (nuchTERT), a mutant that we previously described as being competent in telomere elongation, while not being able to localize to mitochondria, we found the differential activation of autophagy as a function of hTERT's subcellular localization. Specifically, we found that only cells expressing the mutant had significant increases in autophagy markers as a response to H₂O₂ challenge. Either the reintroduction of the mitochondrial pool of hTERT or the expression of mitochondrially-targeted catalase in mutant cells blunted the autophagic response under oxidative stress. Interestingly, autophagy activation was also associated with decreased levels of mitochondrial DNA damage. Taken together, these results suggest that the loss of hTERT in mitochondria initiates a signaling cascade that allows for cells to adapt to and cope with the lack of mitochondrial telomerase. Such effects also influence the cellular response to oxidative damage.
Collapse
Affiliation(s)
- Paula D Green
- Department of Pharmacology and Physiology, New Jersey Medical School, Rutgers The State University of New Jersey, 185 South Orange Avenue, Medical Sciences Building, Newark, NJ 07103, USA.
| | - Nilesh K Sharma
- Department of Pharmacology and Physiology, New Jersey Medical School, Rutgers The State University of New Jersey, 185 South Orange Avenue, Medical Sciences Building, Newark, NJ 07103, USA.
- Department of Molecular Biology and Genetics, Dr. D.Y. Patil Vidyapeeth's, Dr. D.Y. Patil Biotechnology and Bioinformatics Institute, Mumbai- Bangalore Highway, Tathawade, Pune 411033, India.
| | - Janine Hertzog Santos
- Department of Pharmacology and Physiology, New Jersey Medical School, Rutgers The State University of New Jersey, 185 South Orange Avenue, Medical Sciences Building, Newark, NJ 07103, USA.
- Current address: National Toxicology Program, National Institute of Environmental Health Sciences (NIEHS), National Institutes of Health, 111 TW Alexander Drive Blg 101, Durham, NC 27709, USA.
| |
Collapse
|
154
|
de Punder K, Heim C, Wadhwa PD, Entringer S. Stress and immunosenescence: The role of telomerase. Psychoneuroendocrinology 2019; 101:87-100. [PMID: 30445409 PMCID: PMC6458519 DOI: 10.1016/j.psyneuen.2018.10.019] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/27/2018] [Accepted: 10/22/2018] [Indexed: 01/04/2023]
Abstract
Chronic stress is associated with the accelerated aging of the immune system and represents a potent risk factor for the development and progression of a wide range of physical and mental disorders. The elucidation of molecular pathways and mechanisms underlying the link between stress and cellular aging is an area of considerable interest and investigation. In this context, telomere biology has emerged as a particularly attractive candidate mechanism. Several studies have linked immune cell telomere length with stress-related conditions and states, and also with several physical and mental disorders. Because the cellular reverse transcriptase enzyme telomerase is the primary regulator of telomere length (by adding telomeric DNA to telomeres and thereby attenuating telomere shortening), the understanding of its regulation and regulatory functions constitutes a prime target for developing strategies to prevent, attenuate or reverse the adverse consequences of immune system aging (immunosenescence). In this review we provide an overview of the mechanistic pathways linking telomerase with stress and cellular aging, with an emphasis on the immune system. We summarize and synthesize the current state of the literature on immune cell telomerase in different stress- and aging-related disease states and provide recommendations for future research directions.
Collapse
Affiliation(s)
- Karin de Punder
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Medical Psychology, Berlin, Germany.
| | - Christine Heim
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Medical Psychology, Berlin, Germany; Department of Biobehavioral Health, College of Health and Human Development, Pennsylvania State University, USA
| | - Pathik D Wadhwa
- Department of Psychiatry & Human Behavior, University of California, Irvine, School of Medicine, Irvine, CA, USA; Department of Obstetrics & Gynecology, University of California, Irvine, School of Medicine, Irvine, CA, USA; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, CA, USA; Department of Epidemiology, University of California, Irvine, School of Medicine, Irvine, CA, USA
| | - Sonja Entringer
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Medical Psychology, Berlin, Germany; Department of Pediatrics, University of California, Irvine, School of Medicine, Irvine, CA, USA; Development, Health and Disease Research Program, University of California, Irvine, School of Medicine, Irvine, CA, USA
| |
Collapse
|
155
|
Exposure to environmental radionuclides associates with tissue-specific impacts on telomerase expression and telomere length. Sci Rep 2019; 9:850. [PMID: 30696885 PMCID: PMC6351625 DOI: 10.1038/s41598-018-37164-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 12/04/2018] [Indexed: 11/08/2022] Open
Abstract
Telomeres, the protective structures at the ends of chromosomes, can be shortened when individuals are exposed to stress. In some species, the enzyme telomerase is expressed in adult somatic tissues, and potentially protects or lengthens telomeres. Telomeres can be damaged by ionizing radiation and oxidative stress, although the effect of chronic exposure to elevated levels of radiation on telomere maintenance is unknown for natural populations. We quantified telomerase expression and telomere length (TL) in different tissues of the bank vole Myodes glareolus, collected from the Chernobyl Exclusion Zone, an environment heterogeneously contaminated with radionuclides, and from uncontaminated control sites elsewhere in Ukraine. Inhabiting the Chernobyl Exclusion Zone was associated with reduced TL in the liver and testis, and upregulation of telomerase in brain and liver. Thus upregulation of telomerase does not appear to associate with longer telomeres but may reflect protective functions other than telomere maintenance or an attempt to maintain shorter telomeres in a stressful environment. Tissue specific differences in the rate of telomere attrition and apparent radiosensitivity weaken the intra-individual correlation in telomere length among tissues in voles exposed to radionuclides. Our data show that ionizing radiation alters telomere homeostasis in wild animal populations in tissue specific ways.
Collapse
|
156
|
Criscuolo F, Smith S, Zahn S, Heidinger BJ, Haussmann MF. Experimental manipulation of telomere length: does it reveal a corner-stone role for telomerase in the natural variability of individual fitness? Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2016.0440. [PMID: 29335364 DOI: 10.1098/rstb.2016.0440] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2017] [Indexed: 12/11/2022] Open
Abstract
Telomeres, the non-coding ends of linear chromosomes, are thought to be an important mechanism of individual variability in performance. Research suggests that longer telomeres are indicative of better health and increased fitness; however, many of these data are correlational and whether these effects are causal are poorly understood. Experimental tests are emerging in medical and laboratory-based studies, but these types of experiments are rare in natural populations, which precludes conclusions at an evolutionary level. At the crossroads between telomere length and fitness is telomerase, an enzyme that can lengthen telomeres. Experimental modulation of telomerase activity is a powerful tool to manipulate telomere length, and to look at the covariation of telomerase, telomeres and individual life-history traits. Here, we review studies that manipulate telomerase activity in laboratory conditions and emphasize the associated physiological and fitness consequences. We then discuss how telomerase's impact on ageing may go beyond telomere maintenance. Based on this overview, we then propose several research avenues for future studies to explore how individual variability in health, reproduction and survival may have coevolved with different patterns of telomerase activity and expression. Such knowledge is of prime importance to fully understand the role that telomere dynamics play in the evolution of animal ageing.This article is part of the theme issue 'Understanding diversity in telomere dynamics'.
Collapse
Affiliation(s)
- F Criscuolo
- Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - S Smith
- Department of Integrative Biology and Evolution, University of Veterinary Medicine, Vienna, Austria
| | - S Zahn
- Université de Strasbourg, CNRS, IPHC UMR 7178, 67000 Strasbourg, France
| | - B J Heidinger
- Biological Sciences Department, North Dakota State University, Stevens Hall, Fargo, ND 58108, USA
| | - M F Haussmann
- Department of Biology, Bucknell University, Lewisburg, PA 17837, USA
| |
Collapse
|
157
|
Barnes RP, Fouquerel E, Opresko PL. The impact of oxidative DNA damage and stress on telomere homeostasis. Mech Ageing Dev 2019; 177:37-45. [PMID: 29604323 PMCID: PMC6162185 DOI: 10.1016/j.mad.2018.03.013] [Citation(s) in RCA: 342] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 12/12/2022]
Abstract
Telomeres are dynamic nucleoprotein-DNA structures that cap and protect linear chromosome ends. Because telomeres shorten progressively with each replication, they impose a functional limit on the number of times a cell can divide. Critically short telomeres trigger cellular senescence in normal cells, or genomic instability in pre-malignant cells, which contribute to numerous degenerative and aging-related diseases including cancer. Therefore, a detailed understanding of the mechanisms of telomere loss and preservation is important for human health. Numerous studies have shown that oxidative stress is associated with accelerated telomere shortening and dysfunction. Oxidative stress caused by inflammation, intrinsic cell factors or environmental exposures, contributes to the pathogenesis of many degenerative diseases and cancer. Here we review the studies demonstrating associations between oxidative stress and accelerated telomere attrition in human tissue, mice and cell culture, and discuss possible mechanisms and cellular pathways that protect telomeres from oxidative damage.
Collapse
|
158
|
Fu W, Chen Z, Bai Y, Wu X, Li G, Chen W, Wang G, Wang S, Li X, He M, Zhang X, Wu T, Guo H. The interaction effects of polycyclic aromatic hydrocarbons exposure and TERT- CLPTM1L variants on longitudinal telomere length shortening: A prospective cohort study. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2018; 242:2100-2110. [PMID: 30097281 DOI: 10.1016/j.envpol.2018.05.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 04/08/2018] [Accepted: 05/08/2018] [Indexed: 06/08/2023]
Abstract
Telomere length (TL) is an index of cellular aging and can predict the incidences of many age-related diseases. Change of TL might be affected by environmental pollution and individual's genetic background. In this cohort study, we aimed to evaluate the associations between polycyclic aromatic hydrocarbons (PAHs) exposure and longitudinal TL shortening, and investigate whether genetic variations in TERT-CLPTM1L can modify these associations. We measured the baseline concentrations of twelve urinary PAH metabolites and genotyped six variants at TERT-CLPTM1L among 1243 coke-oven workers. The relative leukocyte TL was detected in both baseline and follow-up (4 years later) visits. The TL shortening were estimated by TL decline and TL ratio. We found that the urinary level of 1-hydroxypyrene (1-OHP) had significant dose-response relationships with increased TL decline [β(95%CI) = 0.078(0.023, 0.133), P = 0.005] and TL ratio [β(95%CI) = 0.096(0.037, 0.155), P = 0.002]. Besides, urinary 1-hydroxynaphthalene (1-OHNa) was marginally dose-related with elevated TL decline [β(95%CI) = 0.053(-0.001, 0.107), P = 0.055] and TL ratio [β(95%CI) = 0.057(-0.002, 0.116), P = 0.058]. Analyses of TERT-CLPTM1L variants showed that the rs401681 and rs465498 could modify the effect of 1-OHP on increasing TL decline (Pinteraction = 0.012 and 0.035, respectively) and TL ratio (Pinteraction = 0.014 and 0.067, respectively), which were pronounced among rs401681TT and rs465498CC carriers, but not seen among rs401681TC + CC and rs465498CT + TT carriers. In conclusion, elevated exposure to PAHs can accelerate the TL shortening and this effect can be modified by TERT-CLPTM1L variants. These results may add potential evidence for gene-environment interactions on dynamic changes of telomere length. Further studies are warranted to validate these findings and uncover the underlying mechanisms.
Collapse
Affiliation(s)
- Wenshan Fu
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhuowang Chen
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yansen Bai
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiulong Wu
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guyanan Li
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Weilin Chen
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Gege Wang
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Suhan Wang
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaoliang Li
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Meian He
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xiaomin Zhang
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tangchun Wu
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Huan Guo
- Department of Occupational and Environmental Health, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
159
|
Booth SA, Wadley GD, Marques FZ, Wlodek ME, Charchar FJ. Fetal growth restriction shortens cardiac telomere length, but this is attenuated by exercise in early life. Physiol Genomics 2018; 50:956-963. [PMID: 30192712 DOI: 10.1152/physiolgenomics.00042.2018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND AIMS Fetal and postnatal growth restriction cause a predisposition to cardiovascular disease (CVD) in adulthood. Telomeres are repetitive DNA-protein structures that protect chromosome ends, and the loss of these repeats (a reduction in telomere length) is associated with CVD. As exercise preserves telomere length and cardiovascular health, the aim of this study was to determine the effects of growth restriction and exercise training on cardiac telomere length and telomeric genes. METHODS AND RESULTS Pregnant Wistar Kyoto rats underwent bilateral uterine vessel ligation to induce uteroplacental insufficiency and fetal growth restriction ("Restricted"). Sham-operated rats had either intact litters ("Control") or their litters reduced to five pups with slowed postnatal growth ("Reduced"). Control, Restricted, and Reduced male rats were assigned to Sedentary, Early exercise (5-9 wk of age), or Late exercise (20-24 wk of age) groups. Hearts were excised at 24 wk of age for telomere length and gene expression measurements by quantitative PCR. Growth restriction shortened cardiac telomere length ( P < 0.001), but this was rescued by early exercise ( P < 0.001). Early and Late exercise increased cardiac weight index ( P < 0.001), but neither this nor telomere length was associated with expression of the telomeric genes Tert, Terc, Trf2, Pnuts, or Sirt1. DISCUSSION AND CONCLUSIONS Growth restriction shortens cardiac telomere length, reflecting the cardiac pathologies associated with low birth weight. Exercise in early life may offer long-term protective effects on cardiac telomere length, which could help prevent CVD in later life.
Collapse
Affiliation(s)
- S A Booth
- School of Health and Life Sciences, Federation University Australia , Victoria , Australia
| | - G D Wadley
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University , Geelong, Victoria , Australia
| | - F Z Marques
- School of Health and Life Sciences, Federation University Australia , Victoria , Australia.,Heart Failure Research Group, Baker Heart and Diabetes Institute , Melbourne , Australia.,Central Clinical School, Faculty of Medicine Nursing and Health Sciences, Monash University , Melbourne , Australia
| | - M E Wlodek
- Department of Physiology, The University of Melbourne , Parkville, Victoria , Australia
| | - F J Charchar
- School of Health and Life Sciences, Federation University Australia , Victoria , Australia.,Department of Physiology, The University of Melbourne , Parkville, Victoria , Australia.,Department of Cardiovascular Sciences, University of Leicester , Leicester , United Kingdom
| |
Collapse
|
160
|
Ridout KK, Khan M, Ridout SJ. Adverse Childhood Experiences Run Deep: Toxic Early Life Stress, Telomeres, and Mitochondrial DNA Copy Number, the Biological Markers of Cumulative Stress. Bioessays 2018; 40:e1800077. [DOI: 10.1002/bies.201800077] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 06/20/2018] [Indexed: 01/01/2023]
Affiliation(s)
- Kathryn K. Ridout
- Department of Psychiatry; Kaiser Permanente; San Jose CA 95123 USA
- Department of Psychiatry and Human Behavior; Alpert Medical School of Brown University; Providence RI 02906 USA
| | - Mariam Khan
- Oncology Clinical Trials Department; Kaiser Permanente; San Jose CA 95123 USA
| | - Samuel J. Ridout
- Department of Psychiatry; Kaiser Permanente; San Jose CA 95123 USA
| |
Collapse
|
161
|
Jin Y, You L, Kim HJ, Lee HW. Telomerase Reverse Transcriptase Contains a BH3-Like Motif and Interacts with BCL-2 Family Members. Mol Cells 2018; 41:684-694. [PMID: 29937479 PMCID: PMC6078858 DOI: 10.14348/molcells.2018.0206] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2018] [Accepted: 05/21/2018] [Indexed: 12/12/2022] Open
Abstract
Upregulation of human telomerase reverse transcriptase (hTERT) expression is an important factor in the cellular survival and cancer. Although growing evidence suggests that hTERT inhibits cellular apoptosis by telomere-independent functions, the mechanisms involved are not fully understood. Here, we show that hTERT contains a BH3-like motif, a short peptide sequence found in BCL-2 family proteins, and interacts with anti-apoptotic BCL-2 family proteins MCL-1 and BCL-xL, suggesting a functional link between hTERT and the mitochondrial pathway of apoptosis. Additionally, we propose that hTERT can be categorized into the atypical BH3-only proteins that promote cellular survival, possibly due to the non-canonical interaction between hTERT and antiapoptotic proteins. Although the detailed mechanisms underlying the hTERT BH3-like motif functions and interactions between hTERT and BCL-2 family proteins have not been elucidated, this work proposes a possible connection between hTERT and BCL-2 family members and reconsiders the role of the BH3-like motif as an interaction motif.
Collapse
Affiliation(s)
- Young Jin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| | - Long You
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| | - Hye Jeong Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| | - Han-Woong Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722,
Korea
| |
Collapse
|
162
|
Boeck C, Salinas-Manrique J, Calzia E, Radermacher P, von Arnim CAF, Dietrich DE, Kolassa IT, Karabatsiakis A. Targeting the association between telomere length and immuno-cellular bioenergetics in female patients with Major Depressive Disorder. Sci Rep 2018; 8:9419. [PMID: 29925891 PMCID: PMC6010455 DOI: 10.1038/s41598-018-26867-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 05/17/2018] [Indexed: 12/29/2022] Open
Abstract
Major Depressive Disorder (MDD) has been associated with telomere dysfunction and alterations in mitochondrial activity, which seem to be co-regulated in human cells. To investigate this co-regulation in MDD, we assessed telomere length (TL) in peripheral blood mononuclear cells (PBMC) and selected immune cell subsets by quantitative fluorescence in situ hybridization and mitochondrial respiratory activity in PBMC by high-resolution respirometry in a study cohort of 18 MDD patients and 21 non-depressed controls. We provide initial evidence for a differential vulnerability to telomere attrition in selected adaptive immune cell populations. Here we found the highest difference in TL between depressed and control subjects for memory cytotoxic T cells. Depression was associated with reduced mitochondrial activity (mitochondrial bioenergetics), but increased mitochondrial density (mitochondrial biogenesis) in PBMC. Exploratory post-hoc analyses indicated that the changes in TL and immune cell bioenergetics were most pronounced in MDD patients who reported experiences of childhood sexual abuse. Among MDD patients, PBMC TL was as a trend positively associated with mitochondrial density and negatively associated with mitochondrial leak respiration, but not with mitochondrial activity related to biological energy production. These initial findings support the hypothesis of a co-regulation between telomeres and mitochondrial biogenesis but not mitochondrial bioenergetics among MDD patients.
Collapse
Affiliation(s)
- Christina Boeck
- Clinical & Biological Psychology, Institute of Psychology and Education, Ulm University, Ulm, Germany.
| | | | - Enrico Calzia
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany
| | - Peter Radermacher
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Hospital Ulm, Helmholtzstrasse 8/1, 89081, Ulm, Germany
| | | | - Detlef E Dietrich
- Burghof-Klinik, Rinteln, Germany
- Department of Mental Health, Hannover Medical School, Hannover, Germany
| | - Iris-Tatjana Kolassa
- Clinical & Biological Psychology, Institute of Psychology and Education, Ulm University, Ulm, Germany
| | - Alexander Karabatsiakis
- Clinical & Biological Psychology, Institute of Psychology and Education, Ulm University, Ulm, Germany.
| |
Collapse
|
163
|
Zhang H, Hu N. Telomerase reverse transcriptase induced thyroid carcinoma cell proliferation through PTEN/AKT signaling pathway. Mol Med Rep 2018; 18:1345-1352. [PMID: 29901196 PMCID: PMC6072153 DOI: 10.3892/mmr.2018.9119] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 04/20/2018] [Indexed: 02/06/2023] Open
Abstract
Thyroid carcinoma is the most common endocrine malignant tumor in the world, and so, there is a requirement to develop novel molecular targets for thyroid cancer diagnosis and treatment. Telomerase reverse transcriptase (TERT) was revealed to promote cell proliferation in a number of types of cell. To evaluate whether and how TERT functioned on papillary thyroid cancer (PTC) cell proliferation, the present study constructed TERT over‑expression [recombined (r)TERT plasmid group] and interference [small interfering RNA (si)‑TERT group] models by liposome transfection respectively to study the molecular mechanisms. The transfection efficiency was first detected by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blotting to analyze TERT levels compared with the negative control (NC) and control groups. Then MTT and carboxyfluorescein diacetate succinimidyl ester assays were performed to determine living cell proliferation and total cell proliferation respectively. Propidium iodide assay was used to detect alterations in cell cycle progression. RT‑qPCR and western blotting were performed to detect associated factor variation. The results demonstrated that, following the generation of TERT overexpression or silencing PTC cells, the living cells and also total cell proliferation increased significantly in the rTERT group, and decreased significantly in siTERT group, when compared with the NC and control groups. The cell cycle was accelerated in the rTERT group, and blocked in the G1/S transition in the siTERT group. The mRNA and protein levels of P27, P53 and phosphatase and tensin homolog (PTEN) decreased significantly in the rTERP group and increased in the siTERP group, while cyclin dependent kinase 2 and Cyclin D1 increased significantly in the rTERP group and decreased in the siTERP group. The expression of cell division cycle 25A did not alter significantly. The protein levels of β‑catenin and retinoblastoma were also unaltered. Protein kinase B (AKT) was detected once activated by TERT, and there were increased phosphorylated (p)‑AKT protein levels in the rTERT group, and decreased p‑AKT protein levels in the siTERT group. In conclusion, TERT could induce thyroid carcinoma cell proliferation mainly through the PTEN/AKT signaling pathway.
Collapse
Affiliation(s)
- Hao Zhang
- The First Sector of Department of Thyroid Breast Surgery, Northern Branch of Jingmen No. 1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| | - Ning Hu
- The Second Sector of Department of Thyroid Breast Surgery, Southern Branch of Jingmen No. 1 People's Hospital, Jingmen, Hubei 448000, P.R. China
| |
Collapse
|
164
|
Ahmed W, Lingner J. PRDX1 and MTH1 cooperate to prevent ROS-mediated inhibition of telomerase. Genes Dev 2018; 32:658-669. [PMID: 29773556 PMCID: PMC6004070 DOI: 10.1101/gad.313460.118] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/23/2018] [Indexed: 11/29/2022]
Abstract
In this study, Ahmed et al. demonstrate that oxidative damage of telomeres inhibits telomerase activity at chromosome ends in cancer cells. Deletion of two antioxidant enzymes—PRDX1 and MTH1, needed for protecting telomeres from oxidative damage—results in loss of telomeric DNA in an oxygen concentration-dependent manner due to inhibition of telomerase, thus providing new insights into the role of antioxidant systems that are required to protect telomeres from oxidation. Telomerase counteracts telomere shortening and cellular senescence in germ, stem, and cancer cells by adding repetitive DNA sequences to the ends of chromosomes. Telomeres are susceptible to damage by reactive oxygen species (ROS), but the consequences of oxidation of telomeres on telomere length and the mechanisms that protect from ROS-mediated telomere damage are not well understood. In particular, 8-oxoguanine nucleotides at 3′ ends of telomeric substrates inhibit telomerase in vitro, whereas, at internal positions, they suppress G-quadruplex formation and were therefore proposed to promote telomerase activity. Here, we disrupt the peroxiredoxin 1 (PRDX1) and 7,8-dihydro-8-oxoguanine triphosphatase (MTH1) genes in cancer cells and demonstrate that PRDX1 and MTH1 cooperate to prevent accumulation of oxidized guanine in the genome. Concomitant disruption of PRDX1 and MTH1 leads to ROS concentration-dependent continuous shortening of telomeres, which is due to efficient inhibition of telomere extension by telomerase. Our results identify antioxidant systems that are required to protect telomeres from oxidation and are necessary to allow telomere maintenance by telomerase conferring immortality to cancer cells.
Collapse
Affiliation(s)
- Wareed Ahmed
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Joachim Lingner
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
165
|
Richardson GD, Sage A, Bennaceur K, Al Zhrany N, Coelho-Lima J, Dookun E, Draganova L, Saretzki G, Breault DT, Mallat Z, Spyridopoulos I. Telomerase Mediates Lymphocyte Proliferation but Not the Atherosclerosis-Suppressive Potential of Regulatory T-Cells. Arterioscler Thromb Vasc Biol 2018; 38:1283-1296. [PMID: 29599138 PMCID: PMC5965929 DOI: 10.1161/atvbaha.117.309940] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 03/05/2018] [Indexed: 01/05/2023]
Abstract
Supplemental Digital Content is available in the text. Objective— Atherosclerosis is an age-related disease characterized by systemic oxidative stress and low-grade inflammation. The role of telomerase and telomere length in atherogenesis remains contentious. Short telomeres of peripheral leukocytes are predictive for coronary artery disease. Conversely, attenuated telomerase has been demonstrated to be protective for atherosclerosis. Hence, a potential causative role of telomerase in atherogenesis is critically debated. Approach and Results— In this study, we used multiple mouse models to investigate the regulation of telomerase under oxidative stress as well as its impact on atherogenesis in vitro and in vivo. Using primary lymphocytes and myeloid cell cultures, we demonstrate that cultivation under hyperoxic conditions induced oxidative stress resulting in chronic activation of CD4+ cells and significantly reduced CD4+ T-cell proliferation. The latter was telomerase dependent because oxidative stress had no effect on the proliferation of primary lymphocytes isolated from telomerase knockout mice. In contrast, myeloid cell proliferation was unaffected by oxidative stress nor reliant on telomerase. Telomerase reverse transcriptase deficiency had no effect on regulatory T-cell (Treg) numbers in vivo or suppressive function ex vivo. Adoptive transfer of telomerase reverse transcriptase–/– Tregs into Rag2–/– ApoE–/– (recombination activating gene 2/apolipoprotein E) double knockout mice demonstrated that telomerase function was not required for the ability of Tregs to protect against atherosclerosis. However, telomere length was critical for Treg function. Conclusions— Telomerase contributes to lymphocyte proliferation but plays no major role in Treg function, provided that telomere length is not critically short. We suggest that oxidative stress may contribute to atherosclerosis via suppression of telomerase and acceleration of telomere attrition in Tregs.
Collapse
Affiliation(s)
- Gavin David Richardson
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| | - Andrew Sage
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.S., Z.M.)
| | - Karim Bennaceur
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| | - Nayef Al Zhrany
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| | - Jose Coelho-Lima
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| | - Emily Dookun
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| | - Lilia Draganova
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| | - Gabriele Saretzki
- Institute for Cell and Molecular Biosciences, The Ageing Biology Centre, Newcastle University Institute for Ageing, Campus for Ageing and Vitality (G.S.), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, MA (D.T.B.).,Harvard Stem Cell Institute, Cambridge, MA (D.T.B.)
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.S., Z.M.).,INSERM U970, Paris Cardiovascular Research Center, France (Z.M.).,Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Ioakim Spyridopoulos
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| |
Collapse
|
166
|
Chemotherapeutic-Induced Cardiovascular Dysfunction: Physiological Effects, Early Detection-The Role of Telomerase to Counteract Mitochondrial Defects and Oxidative Stress. Int J Mol Sci 2018. [PMID: 29534446 PMCID: PMC5877658 DOI: 10.3390/ijms19030797] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although chemotherapeutics can be highly effective at targeting malignancies, their ability to trigger cardiovascular morbidity is clinically significant. Chemotherapy can adversely affect cardiovascular physiology, resulting in the development of cardiomyopathy, heart failure and microvascular defects. Specifically, anthracyclines are known to cause an excessive buildup of free radical species and mitochondrial DNA damage (mtDNA) that can lead to oxidative stress-induced cardiovascular apoptosis. Therefore, oncologists and cardiologists maintain a network of communication when dealing with patients during treatment in order to treat and prevent chemotherapy-induced cardiovascular damage; however, there is a need to discover more accurate biomarkers and therapeutics to combat and predict the onset of cardiovascular side effects. Telomerase, originally discovered to promote cellular proliferation, has recently emerged as a potential mechanism to counteract mitochondrial defects and restore healthy mitochondrial vascular phenotypes. This review details mechanisms currently used to assess cardiovascular damage, such as C-reactive protein (CRP) and troponin levels, while also unearthing recently researched biomarkers, including circulating mtDNA, telomere length and telomerase activity. Further, we explore a potential role of telomerase in the mitigation of mitochondrial reactive oxygen species and maintenance of mtDNA integrity. Telomerase activity presents a promising indicator for the early detection and treatment of chemotherapy-derived cardiac damage.
Collapse
|
167
|
Entringer S, de Punder K, Buss C, Wadhwa PD. The fetal programming of telomere biology hypothesis: an update. Philos Trans R Soc Lond B Biol Sci 2018; 373:20170151. [PMID: 29335381 PMCID: PMC5784074 DOI: 10.1098/rstb.2017.0151] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2017] [Indexed: 12/17/2022] Open
Abstract
Research on mechanisms underlying fetal programming of health and disease risk has focused primarily on processes that are specific to cell types, organs or phenotypes of interest. However, the observation that developmental conditions concomitantly influence a diverse set of phenotypes, the majority of which are implicated in age-related disorders, raises the possibility that such developmental conditions may additionally exert effects via a common underlying mechanism that involves cellular/molecular ageing-related processes. In this context, we submit that telomere biology represents a process of particular interest in humans because, firstly, this system represents among the most salient antecedent cellular phenotypes for common age-related disorders; secondly, its initial (newborn) setting appears to be particularly important for its long-term effects; and thirdly, its initial setting appears to be plastic and under developmental regulation. We propose that the effects of suboptimal intrauterine conditions on the initial setting of telomere length and telomerase expression/activity capacity may be mediated by the programming actions of stress-related maternal-placental-fetal oxidative, immune, endocrine and metabolic pathways in a manner that may ultimately accelerate cellular dysfunction, ageing and disease susceptibility over the lifespan. This perspectives paper provides an overview of each of the elements underlying this hypothesis, with an emphasis on recent developments, findings and future directions.This article is part of the theme issue 'Understanding diversity in telomere dynamics'.
Collapse
Affiliation(s)
- Sonja Entringer
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Medical Psychology, Berlin, Germany
- Department of Pediatrics, University of California, School of Medicine, Irvine, CA, USA
- Development, Health and Disease Research Program, University of California, School of Medicine, Irvine, CA, USA
| | - Karin de Punder
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Medical Psychology, Berlin, Germany
| | - Claudia Buss
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Institute of Medical Psychology, Berlin, Germany
- Department of Pediatrics, University of California, School of Medicine, Irvine, CA, USA
- Development, Health and Disease Research Program, University of California, School of Medicine, Irvine, CA, USA
| | - Pathik D Wadhwa
- Department of Psychiatry and Human Behavior, University of California, School of Medicine, Irvine, CA, USA
- Department of Obstetrics and Gynecology, University of California, School of Medicine, Irvine, CA, USA
- Department of Pediatrics, University of California, School of Medicine, Irvine, CA, USA
- Department of Epidemiology, University of California, School of Medicine, Irvine, CA, USA
- Development, Health and Disease Research Program, University of California, School of Medicine, Irvine, CA, USA
| |
Collapse
|
168
|
Huang W, Zhou W, Li C, Yang Y, Shang YK, Chen C, Zhang J, Yao R, Wang P, Wen W, Liu HQ, Wang L, Li X, Bian H, Chen ZN. Promoter mutations and cellular distribution of telomerase in non-clear cell and clear cell hepatocellular carcinoma. Oncotarget 2018; 8:26288-26297. [PMID: 28460432 PMCID: PMC5432257 DOI: 10.18632/oncotarget.15458] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 02/07/2017] [Indexed: 12/13/2022] Open
Abstract
Reactivation of telomerase is a critical step in the development of hepatocellular carcinoma (HCC). Here we identified the frequency of mutations in telomerase reverse transcriptase (TERT) promoter was 34% in non-clear cell HCC (NCCHCC, n = 259) and 26.3% in clear cell HCC (CCHCC, n = 57). The mutations were independently associated with poor recurrence-free survival of HCCs. Interestingly immunohistochemical analysis demonstrated a higher positive rate of TERT cytoplasmic localization (95%) than nuclear localization (64%) in HCCs. In NCCHCCs, the mutations correlated with higher TERT nuclear expression and increased telomere-dependent telomerase activity. Higher cytoplasmic expression was found in adjacent tissues compared to tumor tissues, and was associated with tumor well-differentiation and lower level of α-fetoprotein. NCCHCCs with low nuclear as well as high cytoplasmic expression correlated with better prognosis. In CCHCCs, elevated TERT cytoplasmic expression was observed in CCHCCs harboring mutations. Higher TERT cytoplasmic expression was found in tumor tissues compared to adjacent tissues, and was associated with multiple numbers of tumors and poor prognosis of CCHCCs. In conclusion, mutations in TERT promoter disclose the significance of both nuclear and cytoplasmic TERT in HCC. Cytoplasmic TERT should also be considered when determining prognosis and treatment of HCCs.
Collapse
Affiliation(s)
- Wan Huang
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Weiping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Can Li
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Yuan Yang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Yu-Kui Shang
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Changsheng Chen
- Department of Health Statistics, Fourth Military Medical University, Xi'an, China
| | - Jing Zhang
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Rui Yao
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Pei Wang
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Wen Wen
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Han-Qiang Liu
- Department of Nutrition and Food Hygiene, Fourth Military Medical University, Xi'an, China
| | - Ling Wang
- Department of Health Statistics, Fourth Military Medical University, Xi'an, China
| | - Xia Li
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Huijie Bian
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| | - Zhi-Nan Chen
- National Translational Science Center for Molecular Medicine, Department of Cell Biology, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
169
|
Stress, cell senescence and organismal ageing. Mech Ageing Dev 2018; 170:2-9. [DOI: 10.1016/j.mad.2017.07.001] [Citation(s) in RCA: 179] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/17/2017] [Accepted: 07/04/2017] [Indexed: 12/25/2022]
|
170
|
|
171
|
Song YL, Tian KY, Mi WJ, Ding ZJ, Qiu Y, Chen FQ, Zha DJ, Qiu JH. Decreased expression of TERT correlated with postnatal cochlear development and proliferation reduction of cochlear progenitor cells. Mol Med Rep 2018; 17:6077-6083. [PMID: 29436610 DOI: 10.3892/mmr.2018.8565] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 01/17/2018] [Indexed: 11/06/2022] Open
Abstract
Cochlear progenitor cells are considered as one of the best candidates for hair cell regeneration, thus, the regulation of cochlear progenitor cell proliferation has become a focus in this field. Several genes expressed in the inner ear during postnatal development have been demonstrated to be involved in maintaining the proliferative potential of progenitor cells, but the mechanism for regulating the proliferation and differentiation of cochlear progenitor cells remains poorly understood. Telomerase reverse transcriptase (TERT) has rate limiting telomerase activity and the overexpression of TERT has been shown to promote cell proliferation in series of cell lines. The aim of the present study was to evaluate the expression of TERT in the postnatal development of the cochlea and progenitor cells. The results demonstrated that TERT was expressed in the basilar membranes during the first postnatal week. In vitro, TERT expression in progenitor cells reached a maximum at day 4 after culture and decreased as the culture time prolonged or the cell passage number increased. These results led us to hypothesize that TERT may be involved in the development of the cochlea and in maintaining the proliferation ability of progenitor cells.
Collapse
Affiliation(s)
- Yong-Li Song
- Department of Otolaryngology‑Head and Neck Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Ke-Yong Tian
- Department of Otolaryngology‑Head and Neck Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Wen-Juan Mi
- Department of Otolaryngology‑Head and Neck Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Zhong-Jia Ding
- Department of Otolaryngology‑Head and Neck Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yang Qiu
- Department of Otolaryngology‑Head and Neck Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Fu-Quan Chen
- Department of Otolaryngology‑Head and Neck Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Ding-Jun Zha
- Department of Otolaryngology‑Head and Neck Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Jian-Hua Qiu
- Department of Otolaryngology‑Head and Neck Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
172
|
Wei ZZ, Qin QP, Meng T, Deng CX, Liang H, Chen ZF. 5-Bromo-oxoisoaporphine platinum(II) complexes exhibit tumor cell cytotoxcicity via inhibition of telomerase activity and disruption of c-myc G-quadruplex DNA and mitochondrial functions. Eur J Med Chem 2018; 145:360-369. [DOI: 10.1016/j.ejmech.2017.12.092] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 11/20/2017] [Accepted: 12/29/2017] [Indexed: 01/01/2023]
|
173
|
Impact of oxidative stress on telomere biology. Differentiation 2018; 99:21-27. [DOI: 10.1016/j.diff.2017.12.002] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 12/12/2022]
|
174
|
Telomere Biology and Thoracic Aortic Aneurysm. Int J Mol Sci 2017; 19:ijms19010003. [PMID: 29267201 PMCID: PMC5795955 DOI: 10.3390/ijms19010003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 12/13/2017] [Accepted: 12/19/2017] [Indexed: 12/27/2022] Open
Abstract
Ascending aortic aneurysms are mostly asymptomatic and present a great risk of aortic dissection or perforation. Consequently, ascending aortic aneurysms are a source of lethality with increased age. Biological aging results in progressive attrition of telomeres, which are the repetitive DNA sequences at the end of chromosomes. These telomeres play an important role in protection of genomic DNA from end-to-end fusions. Telomere maintenance and telomere attrition-associated senescence of endothelial and smooth muscle cells have been indicated to be part of the pathogenesis of degenerative vascular diseases. This systematic review provides an overview of telomeres, telomere-associated proteins and telomerase to the formation and progression of aneurysms of the thoracic ascending aorta. A better understanding of telomere regulation in the vascular pathology might provide new therapeutic approaches. Measurements of telomere length and telomerase activity could be potential prognostic biomarkers for increased risk of death in elderly patients suffering from an aortic aneurysm.
Collapse
|
175
|
Aeby E, Ahmed W, Redon S, Simanis V, Lingner J. Peroxiredoxin 1 Protects Telomeres from Oxidative Damage and Preserves Telomeric DNA for Extension by Telomerase. Cell Rep 2017; 17:3107-3114. [PMID: 28009281 DOI: 10.1016/j.celrep.2016.11.071] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 11/04/2016] [Accepted: 11/22/2016] [Indexed: 01/19/2023] Open
Abstract
Oxidative damage of telomeres can promote cancer, cardiac failure, and muscular dystrophy. Specific mechanisms protecting telomeres from oxidative damage have not been described. We analyzed telomeric chromatin composition during the cell cycle and show that the antioxidant enzyme peroxiredoxin 1 (PRDX1) is enriched at telomeres during S phase. Deletion of the PRDX1 gene leads to damage of telomeric DNA upon oxidative stress, revealing a protective function of PRDX1 against oxidative damage at telomeres. We also show that the oxidized nucleotide 8-oxo-2'deoxyguanosine-5'-triphosphate (8oxodGTP) causes premature chain termination when incorporated by telomerase and that some DNA substrates terminating in 8oxoG prevent extension by telomerase. Thus, PRDX1 safeguards telomeres from oxygen radicals to counteract telomere damage and preserve telomeric DNA for elongation by telomerase.
Collapse
Affiliation(s)
- Eric Aeby
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Wareed Ahmed
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Sophie Redon
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Viesturs Simanis
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Joachim Lingner
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
176
|
Park HH, Lee KY, Park DW, Choi NY, Lee YJ, Son JW, Kim S, Moon C, Kim HW, Rhyu IJ, Koh SH. Tracking and protection of transplanted stem cells using a ferrocenecarboxylic acid-conjugated peptide that mimics hTERT. Biomaterials 2017; 155:80-91. [PMID: 29169040 DOI: 10.1016/j.biomaterials.2017.11.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/08/2017] [Accepted: 11/12/2017] [Indexed: 02/07/2023]
Abstract
In vivo tracking of transplanted stem cells has been a central aim of stem cell therapy. Although many tracking systems have been introduced, no method has yet been validated for clinical applications. We developed a novel sophisticated peptide (GV1001) that mimics hTERT (human telomerase reverse transcriptase) and analysed its ability to track and protect stem cells after transplantation. Ferrocenecarboxylic acid-conjugated GV1001 (Fe-GV1001) efficiently penetrated stem cells with no adverse effects. Moreover, Fe-GV1001 improved the viability, proliferation, and migration of stem cells under hypoxia. After Fe-GV1001-labelled stem cells were transplanted into the brains of rats after stroke, the labelled cells were easily tracked by MRI. Our findings indicate that Fe-GV1001 can be used for the in vivo tracking of stem cells after transplantation into the brain and can improve the efficacy of stem cell therapy by sustaining and enhancing stem cell characteristics under disease conditions.
Collapse
Affiliation(s)
- Hyun-Hee Park
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Kyu-Yong Lee
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Dong Woo Park
- Department of Radiology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Na-Young Choi
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Young Joo Lee
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea
| | - Jeong-Woo Son
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, 04763, South Korea
| | - Sangjae Kim
- Teloid Inc., 920 Westholme Ave, Los Angeles (City), CA 90024, USA
| | - Chanil Moon
- Department of Neuroscience, GemVax & KAEL Co., Ltd., Seoul, South Korea
| | - Hyun-Wook Kim
- Brain Korea 21 PLUS, KU Medical Science Center for Convergent Translational Research, 73 Inchonro, Seongbuk-gu, Seoul, 136-705, South Korea; Department of Anatomy, College of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul, 136-705, South Korea
| | - Im Joo Rhyu
- Brain Korea 21 PLUS, KU Medical Science Center for Convergent Translational Research, 73 Inchonro, Seongbuk-gu, Seoul, 136-705, South Korea; Department of Anatomy, College of Medicine, Korea University, 73 Inchon-ro, Seongbuk-gu, Seoul, 136-705, South Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University Guri Hospital, 153, Gyeongchun-ro, Guri-si, Gyeonggi-do 11923, South Korea; Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul, 04763, South Korea.
| |
Collapse
|
177
|
Dinami R, Buemi V, Sestito R, Zappone A, Ciani Y, Mano M, Petti E, Sacconi A, Blandino G, Giacca M, Piazza S, Benetti R, Schoeftner S. Epigenetic silencing of miR-296 and miR-512 ensures hTERT dependent apoptosis protection and telomere maintenance in basal-type breast cancer cells. Oncotarget 2017; 8:95674-95691. [PMID: 29221158 PMCID: PMC5707052 DOI: 10.18632/oncotarget.21180] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 08/27/2017] [Indexed: 12/31/2022] Open
Abstract
The catalytic subunit of the telomerase complex, hTERT, ensures unlimited proliferative potential of cancer cells by maintaining telomere function and protecting from apoptosis. Using a miRNA screening approach we identified miR-296-5p and miR-512-5p as miRNAs that target hTERT in breast cancer cells. Ectopic miR-296-5p and miR-512-5p reduce telomerase activity, drive telomere shortening and cause proliferation defects by enhancing senescence and apoptosis in breast cancer cells. In line with the relevance of hTERT expression for human cancer we found that miR-296-5p and miR-512-5p expression is reduced in human breast cancer. Accordingly, high expression of miR-296-5p and miR-512-5p target genes including hTERT is linked with significantly reduced distant metastasis free survival and relapse free survival of basal type breast cancer patients. This suggests relevance of the identified miRNAs in basal type breast cancer. Epigenetic silencing of miR-296 and miR-512 encoding genes is responsible for low levels of miR-296-5p and miR-512-5p expression in basal type breast cancer cells. Disrupting gene silencing results in a dramatic upregulation of miR-296-5p and miR-512-5p levels leading to reduced hTERT expression and increased sensitivity to the induction of apoptosis. Altogether, our data suggest that epigenetic regulatory circuits in basal type breast cancer may contribute to high hTERT levels by silencing miR-296-5p and miR-512-5p expression, thereby contributing to the aggressiveness of basal type breast cancer.
Collapse
Affiliation(s)
- Roberto Dinami
- Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie (LNCIB), Genomic Stability Unit, Trieste 34149, Italy.,Italian National Cancer Institute, Regina Elena, Rome 00144, Italy
| | - Valentina Buemi
- Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie (LNCIB), Genomic Stability Unit, Trieste 34149, Italy.,Department of Life Sciences, Università degli Studi di Trieste, Trieste 34127, Italy
| | - Rosanna Sestito
- Italian National Cancer Institute, Regina Elena, Rome 00144, Italy
| | - Antonina Zappone
- Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie (LNCIB), Genomic Stability Unit, Trieste 34149, Italy.,Department of Life Sciences, Università degli Studi di Trieste, Trieste 34127, Italy
| | - Yari Ciani
- Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie (LNCIB), Bioinformatics and Functional Genomics Unit (BFGU), Trieste 34149, Italy
| | - Miguel Mano
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Molecular Medicine Laboratory, Trieste 34149, Italy
| | - Eleonora Petti
- Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie (LNCIB), Genomic Stability Unit, Trieste 34149, Italy.,Italian National Cancer Institute, Regina Elena, Rome 00144, Italy.,Department of Life Sciences, Università degli Studi di Trieste, Trieste 34127, Italy
| | - Andrea Sacconi
- Italian National Cancer Institute, Regina Elena, Translational Oncogenomics Group, Rome 00144, Italy
| | - Giovanni Blandino
- Italian National Cancer Institute, Regina Elena, Translational Oncogenomics Group, Rome 00144, Italy
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Molecular Medicine Laboratory, Trieste 34149, Italy
| | - Silvano Piazza
- Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie (LNCIB), Bioinformatics and Functional Genomics Unit (BFGU), Trieste 34149, Italy
| | - Roberta Benetti
- Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie (LNCIB), Cancer Epigenetics Unit, Trieste 34149, Italy.,Department of Medical and Biological Sciences, Università degli Studi di Udine, Udine 33100, Italy
| | - Stefan Schoeftner
- Laboratorio Nazionale Consorzio Interuniversitario Biotecnologie (LNCIB), Genomic Stability Unit, Trieste 34149, Italy.,Italian National Cancer Institute, Regina Elena, Rome 00144, Italy.,Department of Life Sciences, Università degli Studi di Trieste, Trieste 34127, Italy
| |
Collapse
|
178
|
Desipramine rescues age-related phenotypes in depression-like rats induced by chronic mild stress. Life Sci 2017; 188:96-100. [PMID: 28842312 DOI: 10.1016/j.lfs.2017.08.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/12/2017] [Accepted: 08/20/2017] [Indexed: 01/21/2023]
Abstract
AIMS Our previous finding demonstrates that major depressive disorder can mediate accelerated aging in rats. Desipramine is a typical tricyclic antidepressant, and can provide neuroprotection and counteract depression-like behaviors. However, whether desipramine can rescue age-related phenotypes in depressed individuals is not understood. In the present study, we investigated the physiological function of desipramine on rescuing the age-related phenotypes in these animals. MAIN METHODS The rats were induced by chronic mild stress paradigm, and the depression-like behaviors of rats were detected by sucrose intake test, open field test (OFT) and forced swimming test (FST). Then the depressed rats were treated by desipramine. KEY FINDINGS Desipramine administration was effective in counteracting depression-like behaviors by increasing the sucrose solution intake, reducing the immobility time in the FST, and increasing total distance travelled and numbers of grid line crossed in the OFT. Moreover, desipramine treatment was able to reduce the oxidative damage to rat liver, and to increase the expression of telomerase reverse transcriptase (TERT), leading to correspondingly restored telomerase activity. SIGNIFICANCE Our findings identify that one function of desipramine may partly be to rescue age-related phenotypes in depressed individuals induced by chronic stress.
Collapse
|
179
|
Hapangama DK, Kamal A, Saretzki G. Implications of telomeres and telomerase in endometrial pathology. Hum Reprod Update 2017; 23:166-187. [PMID: 27979878 PMCID: PMC5850744 DOI: 10.1093/humupd/dmw044] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Accepted: 12/02/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Eukaryotic chromosomal ends are linear and are protected by nucleoprotein complexes known as telomeres. The complex structural anatomy and the diverse functions of telomeres as well as the unique reverse transcriptase enzyme, telomerase that maintains telomeres are under intensive scientific scrutiny. Both are involved in many human diseases including cancer, but also in ageing and chronic disease such as diabetes. Their intricate involvement in many cellular processes and pathways is being dynamically deciphered in many organs including the endometrium. This review summarizes our current knowledge on the topic of telomeres and telomerase and their potential role in providing plausible explanations for endometrial aberrations related to common gynaecological pathologies. OBJECTIVE AND RATIONALE This review outlines the recent major findings in telomere and telomerase functions in the context of endometrial biology. It highlights the contemporary discoveries in hormonal regulation, normal endometrial regeneration, stem cells and common gynaecological diseases such as endometriosis, infertility, recurrent reproductive failure and endometrial cancer (EC). SEARCH METHODS The authors carried out systematic PubMed (Medline) and Ovid searches using the key words: telomerase, telomeres, telomere length, human telomerase reverse transcriptase, telomeric RNA component, with endometrium, hormonal regulation, endometrial stem/progenitor cells, endometrial regeneration, endometriosis, recurrent miscarriage, infertility, endometrial hyperplasia, EC and uterine cancer. Publications used in this review date from 1995 until 31st June 2016. OUTCOMES The human endometrium is a unique somatic organ, which displays dynamic telomerase activity (TA) related to the menstrual cycle. Telomerase is implicated in almost all endometrial pathologies and appears to be crucial to endometrial stem cells. In particular, it is vital for normal endometrial regeneration, providing a distinct route to formulate possible curative, non-hormonal therapies to treat chronic endometrial conditions. Furthermore, our current understanding of telomere maintenance in EC is incomplete. Data derived from other malignancies on the role of telomerase in carcinogenesis cannot be extrapolated to EC because unlike in other cancers, TA is already present in proliferating healthy endometrial cells. WIDER IMPLICATIONS Since telomerase is pivotal to endometrial regeneration, further studies elucidating the role of telomeres, telomerase, their associated proteins and their regulation in normal endometrial regeneration as well as their role in endometrial pathologies are essential. This approach may allow future development of novel treatment strategies that are not only non-hormonal but also potentially curative.
Collapse
Affiliation(s)
- D K Hapangama
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, L8 7SS, UK.,Liverpool Women's Hospital NHS Foundation Trust, Crown Street, Liverpool L8 7SS, UK
| | - A Kamal
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, L8 7SS, UK.,The National Center for Early Detection of Cancer, Oncology Teaching Hospital, Baghdad Medical City, Baghdad, Iraq
| | - G Saretzki
- Institute for Ageing and Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| |
Collapse
|
180
|
Telomerase reverse transcriptase promotes chemoresistance by suppressing cisplatin-dependent apoptosis in osteosarcoma cells. Sci Rep 2017; 7:7070. [PMID: 28765565 PMCID: PMC5539325 DOI: 10.1038/s41598-017-07204-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 06/23/2017] [Indexed: 11/18/2022] Open
Abstract
Cisplatin is one of the most efficacious antimitotic drugs used in the treatment of a range of malignant tumors. However, treatment failures are common due to the development of chemoresistance. In addition to its telomere maintenance function, telomerase plays a pro-survival role, inducing decreased apoptosis and increased resistance against DNA damage. Elucidation of the molecular mechanisms underlying this effect is critical to improve treatment outcomes. Previously, our group showed higher telomerase reverse transcriptase(TERT) expression in cisplatin resistant osteosarcoma cells. In this study, confocal fluorescence microscopy experiments revealed that TERT translocates from the nucleus to mitochondria in cisplatin treated osteosarcoma cells. We observed decreased apoptosis rate and improved mitochondrial function in TERT-overexpressing cells following cisplatin treatment. Based on these results, we further established that TERT inhibits cisplatin-induced apoptosis independently of telomerase reverse transcriptase activity. Moreover, TERT suppressed cisplatin-induced apoptosis and improved mitochondrial function via alleviating intracellular ROS in osteosarcoma cells. Our finding that TERT shuttles from the nucleus to the mitochondrion in response to cisplatin treatment and inhibits cisplatin-induced apoptosis in osteosarcoma cells may be especially important to overcome drug resistance.
Collapse
|
181
|
Pavlov KI, Mukhin VN, Klimenko VM, Anisimov VN. The telomere-telomerase system and mental processes in aging, norm and pathology (Literature review). ADVANCES IN GERONTOLOGY 2017. [DOI: 10.1134/s2079057017020114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
182
|
Booth SA, Charchar FJ. Cardiac telomere length in heart development, function, and disease. Physiol Genomics 2017; 49:368-384. [DOI: 10.1152/physiolgenomics.00024.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Telomeres are repetitive nucleoprotein structures at chromosome ends, and a decrease in the number of these repeats, known as a reduction in telomere length (TL), triggers cellular senescence and apoptosis. Heart disease, the worldwide leading cause of death, often results from the loss of cardiac cells, which could be explained by decreases in TL. Due to the cell-specific regulation of TL, this review focuses on studies that have measured telomeres in heart cells and critically assesses the relationship between cardiac TL and heart function. There are several lines of evidence that have identified rapid changes in cardiac TL during the onset and progression of heart disease as well as at critical stages of development. There are also many factors, such as the loss of telomeric proteins, oxidative stress, and hypoxia, that decrease cardiac TL and heart function. In contrast, antioxidants, calorie restriction, and exercise can prevent both cardiac telomere attrition and the progression of heart disease. TL in the heart is also indicative of proliferative potential and could facilitate the identification of cells suitable for cardiac rejuvenation. Although these findings highlight the involvement of TL in heart function, there are important questions regarding the validity of animal models, as well as several confounding factors, that need to be considered when interpreting results and planning future research. With these in mind, elucidating the telomeric mechanisms involved in heart development and the transition to disease holds promise to prevent cardiac dysfunction and potentiate regeneration after injury.
Collapse
Affiliation(s)
- S. A. Booth
- Faculty of Science and Technology, School of Applied and Biomedical Sciences, Federation University Australia, Balllarat, Australia
| | - F. J. Charchar
- Faculty of Science and Technology, School of Applied and Biomedical Sciences, Federation University Australia, Balllarat, Australia
- Department of Physiology, The University of Melbourne, Melbourne, Australia; and
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
183
|
Tamrin SH, Majedi FS, Tondar M, Sanati-Nezhad A, Hasani-Sadrabadi MM. Electromagnetic Fields and Stem Cell Fate: When Physics Meets Biology. Rev Physiol Biochem Pharmacol 2017; 171:63-97. [PMID: 27515674 DOI: 10.1007/112_2016_4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Controlling stem cell (SC) fate is an extremely important topic in the realm of SC research. A variety of different external cues mainly mechanical, chemical, or electrical stimulations individually or in combination have been incorporated to control SC fate. Here, we will deconstruct the probable relationship between the functioning of electromagnetic (EMF) and SC fate of a variety of different SCs. The electromagnetic (EM) nature of the cells is discussed with the emphasis on the effects of EMF on the determinant factors that directly and/or indirectly influence cell fate. Based on the EM effects on a variety of cellular processes, it is believed that EMFs can be engineered to provide a controlled signal with the highest impact on the SC fate decision. Considering the novelty and broad applications of applying EMFs to change SC fate, it is necessary to shed light on many unclear mechanisms underlying this phenomenon.
Collapse
Affiliation(s)
- Sara Hassanpour Tamrin
- Center of Excellence in Biomaterials, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | | | - Mahdi Tondar
- Department of Biochemistry and Molecular & Cellular Biology, School of Medicine, Georgetown University, Washington, DC, USA
| | - Amir Sanati-Nezhad
- BioMEMS and BioInspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, Center for Bioengineering Research and Education, University of Calgary, Calgary, AB, Canada, T2N1N4.
| | - Mohammad Mahdi Hasani-Sadrabadi
- Department of Chemistry & Biochemistry, and California NanoSystems Institute, University of California at Los Angeles, Los Angeles, CA, 90095, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience and G.W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
184
|
Gastrointestinal cancer cells treatment with bevacizumab activates a VEGF autoregulatory mechanism involving telomerase catalytic subunit hTERT via PI3K-AKT, HIF-1α and VEGF receptors. PLoS One 2017; 12:e0179202. [PMID: 28594907 PMCID: PMC5466359 DOI: 10.1371/journal.pone.0179202] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 05/25/2017] [Indexed: 11/29/2022] Open
Abstract
Background Targeting angiogenesis has been considered a promising treatment of choice for a large number of malignancies, including gastrointestinal cancers. Bevacizumab is an anti-vascular endothelial growth factor (anti-VEGF) being used for this purpose. However, treatment efficacy is largely questioned. Telomerase activity, responsible for cancer cell immortality, is detected in 85–95% of human cancers and is considered a potential regulator of VEGF. The aim of our study was to investigate the interrelationship between VEGF and hTERT in gastrointestinal cancers and to explore cell response to a combined inhibition of telomerase and VEGF. Methods AGS (gastric cancer), Caco-2 (colorectal cancer) and HepG2/C3A (hepatocellular carcinoma), were treated with telomerase inhibitors BIBR-1232 (10μM) and costunolide (10μM), with bevacizumab (Avastin® at 5 ng/ml or 100μg/ml) or with a combination of both types of inhibitors. VEGF and hTERT mRNA levels, and telomerase activity were detected by RT-PCR. VEGF levels were quantified by ELISA. Telomerase was knocked down using hTERT siRNA and hTERT was overexpressed in the telomerase negative cell line, Saos-2 (osteosarcoma), using constructs expressing either wild type hTERT (hTERT-WT) or dominant negative hTERT (hTERT-DN). Tube formation by HUVECs was assessed using ECMatrix™ (EMD Millipore). Results Our results showed that telomerase regulates VEGF expression and secretion through its catalytic subunit hTERT in AGS, Caco2, and HepG2/C3A, independent of its catalytic activity. Interestingly, VEGF inhibition with bevacizumab (100μg/ml) increased hTERT expression 42.3% in AGS, 94.1% in Caco2, and 52.5% in HepG2/C3A, and increased telomerase activity 30-fold in AGS, 10.3-fold in Caco2 and 8-fold in HepG2/C3A. A further investigation showed that VEGF upregulates hTERT expression in a mechanism that implicates the PI3K/AKT/mTOR pathway and HIF-1α. Moreover, bevacizumab treatment increased VEGFR1 and VEGFR2 expression in cancer cells and human umbilical vein endothelial cells (HUVECs) through hTERT. Thus, the combination of bevacizumab with telomerase inhibitors decreased VEGF expression and secretion by cancer cells, inhibited VEGFR1 and VEGFR2 upregulation, and reduced tube formation by HUVECs. Conclusions Taken together, our results suggest that bevacizumab treatment activates a VEGF autoregulatory mechanism involving hTERT and VEGF receptors and that an inhibition of this pathway could improve tumor cell response to anti-VEGF treatment.
Collapse
|
185
|
Moskalev AA, Proshkina EN, Belyi AA, Solovyev IA. Genetics of aging and longevity. RUSSIAN JOURNAL OF GENETICS: APPLIED RESEARCH 2017; 7:369-384. [DOI: 10.1134/s2079059717040074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
186
|
Trachana V, Petrakis S, Fotiadis Z, Siska EK, Balis V, Gonos ES, Kaloyianni M, Koliakos G. Human mesenchymal stem cells with enhanced telomerase activity acquire resistance against oxidative stress-induced genomic damage. Cytotherapy 2017; 19:808-820. [PMID: 28454681 DOI: 10.1016/j.jcyt.2017.03.078] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 03/25/2017] [Accepted: 03/25/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Human mesenchymal stem cells (MSC) are important tools for several cell-based therapies. However, their use in such therapies requires in vitro expansion during which MSCs quickly reach replicative senescence. Replicative senescence has been linked to macromolecular damage, and especially oxidative stress-induced DNA damage. Recent studies on the other hand, have implicated telomerase in the cellular response to oxidative damage, suggesting that telomerase has a telomere-length independent function that promotes survival. METHODS Here, we studied the DNA damage accumulation and repair during in vitro expansion as well as after acute external oxidative exposure of control MSCs and MSCs that overexpress the catalytic subunit of telomerase (hTERT MSCs). RESULTS We showed that hTERT MSCs at high passages have a significant lower percentage of DNA lesions as compared to control cells of the same passages. Additionally, less damage was accumulated due to external oxidative insult in the nuclei of hTERT overexpressing cells as compared to the control cells. Moreover, we demonstrated that oxidative stress leads to diverse nucleus malformations, such as multillobular nuclei or donut-shaped nuclei, in the control cells whereas hTERT MSCs showed significant resistance to the formation of such defects. Finally, hTERT MSCs were found to possess higher activities of the basic antioxidant enzymes, superoxide dismutase and catalase, than control MSCs. DISCUSSION On the basis of these results, we propose that hTERT enhancement confers resistance to genomic damage due to the amelioration of the cell's basic antioxidant machinery.
Collapse
Affiliation(s)
- Varvara Trachana
- Laboratory of Biology, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41500 Larisa, Greece.
| | - Spyros Petrakis
- Biohellenika Biotechnology Company, 57001 Thessaloniki, Greece
| | - Zisis Fotiadis
- Department of Zoology, School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Evangelia K Siska
- Biohellenika Biotechnology Company, 57001 Thessaloniki, Greece; Department of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Vasileios Balis
- Department of Zoology, School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Efstathios S Gonos
- National Hellenic Research Foundation, 48 Vas. Konstantinou Str, 11635 Athens, Greece
| | - Martha Kaloyianni
- Department of Zoology, School of Biology, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - George Koliakos
- Biohellenika Biotechnology Company, 57001 Thessaloniki, Greece; Department of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
187
|
Ozturk MB, Li Y, Tergaonkar V. Current Insights to Regulation and Role of Telomerase in Human Diseases. Antioxidants (Basel) 2017; 6:antiox6010017. [PMID: 28264499 PMCID: PMC5384180 DOI: 10.3390/antiox6010017] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 02/16/2017] [Accepted: 02/21/2017] [Indexed: 12/31/2022] Open
Abstract
The telomerase ribonucleoprotein complex has a pivotal role in regulating the proliferation and senescence of normal somatic cells as well as cancer cells. This complex is comprised mainly of telomerase reverse transcriptase (TERT), telomerase RNA component (TERC) and other associated proteins that function to elongate telomeres localized at the end of the chromosomes. While reactivation of telomerase is a major hallmark of most cancers, together with the synergistic activation of other oncogenic signals, deficiency in telomerase and telomeric proteins might lead to aging and senescence-associated disorders. Therefore, it is critically important to understand the canonical as well as non-canonical functions of telomerase through TERT to develop a therapeutic strategy against telomerase-related diseases. In this review, we shed light on the regulation and function of telomerase, and current therapeutic strategies against telomerase in cancer and age-related diseases.
Collapse
Affiliation(s)
- Mert Burak Ozturk
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore.
| | - Yinghui Li
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore.
| | - Vinay Tergaonkar
- Division of Cancer Genetics and Therapeutics, Laboratory of NFκB Signaling, Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore 117597, Singapore.
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide SA 5000, Australia.
| |
Collapse
|
188
|
Pestana A, Vinagre J, Sobrinho-Simões M, Soares P. TERT biology and function in cancer: beyond immortalisation. J Mol Endocrinol 2017; 58:R129-R146. [PMID: 28057768 DOI: 10.1530/jme-16-0195] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 01/05/2017] [Indexed: 12/23/2022]
Abstract
Evasion of replicative senescence and proliferation without restriction, sometimes designated as immortalisation, is one of the hallmarks of cancer that may be attained through reactivation of telomerase in somatic cells. In contrast to most normal cells in which there is lack of telomerase activity, upregulation of TERT transcription/activity is detected in 80-90% of malignant tumours. In several types of cancer, there is a relationship between the presence of TERT promoter mutations, TERT mRNA expression and clinicopathological features, but the biological bridge between the occurrence of TERT promoter mutations and the aggressive/invasive features displayed by the tumours remains unidentified. We and others have associated the presence of TERT promoter mutations with metastisation/survival in several types of cancer. In follicular cell-derived thyroid cancer, such mutations are associated with worse prognostic features (age of patients, tumour size and tumour stage) as well as with distant metastases, worse response to treatment and poorer survival. In this review, we analyse the data reported in several studies that imply TERT transcription reactivation/activity with cell proliferation, tumour invasion and metastisation. A particular attention is given to the putative connections between TERT transcriptional reactivation and signalling pathways frequently altered in cancer, such as c-MYC, NF-κB and B-Catenin.
Collapse
Affiliation(s)
- Ana Pestana
- Institute of Molecular Pathology and ImmunologyUniversity of Porto (IPATIMUP), Porto, Portugal
- Institute for Research and Innovation in Health (I3S)University of Porto, Porto, Portugal
| | - João Vinagre
- Institute of Molecular Pathology and ImmunologyUniversity of Porto (IPATIMUP), Porto, Portugal
- Institute for Research and Innovation in Health (I3S)University of Porto, Porto, Portugal
| | - Manuel Sobrinho-Simões
- Institute of Molecular Pathology and ImmunologyUniversity of Porto (IPATIMUP), Porto, Portugal
- Institute for Research and Innovation in Health (I3S)University of Porto, Porto, Portugal
- Medical FacultyUniversity of Porto, Porto, Portugal
- Department of PathologyCentro Hospitalar S. João, Porto, Portugal
- Department of PathologyMedical Faculty, University of Porto, Porto, Portugal
| | - Paula Soares
- Institute of Molecular Pathology and ImmunologyUniversity of Porto (IPATIMUP), Porto, Portugal
- Institute for Research and Innovation in Health (I3S)University of Porto, Porto, Portugal
- Medical FacultyUniversity of Porto, Porto, Portugal
- Department of PathologyMedical Faculty, University of Porto, Porto, Portugal
| |
Collapse
|
189
|
Miao GY, Zhou X, Zhang X, Xie Y, Sun C, Liu Y, Gan L, Zhang H. Telomere-Mitochondrion Links Contribute to Induction of Senescence in MCF-7 Cells after Carbon-Ion Irradiation. Asian Pac J Cancer Prev 2017; 17:1993-8. [PMID: 27221886 DOI: 10.7314/apjcp.2016.17.4.1993] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The effects of carbon-ion irradiation on cancer cell telomere function have not been comprehensively studied. In our previous report cancer cells with telomere dysfunction were more sensitive to carbon-ion irradiation, but the underlying mechanisms remained unclear. Here we found that telomerase activity was suppressed by carbon-ion irradiation via hTERT down-regulation. Inhibition of telomere activity by MST-312 further increased cancer cell radiosensitivity to carbon-ion radiation. hTERT suppression caused by either carbon-ion irradiation or MST-312 impaired mitochondrial function, as indicated by decreased membrane potential, mtDNA copy number, mitochondrial mass, total ATP levels and elevated reactive oxygen species (ROS). PGC-1α expression was repressed after carbion-ion irradiation, and hTERT inhibition by MST-312 could further exacerbate this effect. Lowering the mitochondrial ROS level by MitoTEMPO could partially counteract the induction of cellular senescence induced by carbon-ion radiation and MST-312 incubation. Taken together, the current data suggest that telomere-mitochondrion links play a role in the induction of senescence in MCF-7 cells after carbon-ion irradiation.
Collapse
Affiliation(s)
- Guo-Ying Miao
- Department of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, Gansu Province, China E-mail :
| | | | | | | | | | | | | | | |
Collapse
|
190
|
Bashash D, Zareii M, Safaroghli-Azar A, Omrani MD, Ghaffari SH. Inhibition of telomerase using BIBR1532 enhances doxorubicin-induced apoptosis in pre-B acute lymphoblastic leukemia cells. ACTA ACUST UNITED AC 2017; 22:330-340. [PMID: 28054503 DOI: 10.1080/10245332.2016.1275426] [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] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Interest into targeting telomerase in cancer has increased by the recent disclosure that elevated telomerase activity is associated with disease recurrence and poor outcome in cancers. In addition, cellular acquisition of unlimited replicative potential, which is closely related to the maintenance of telomeres mostly via the reactivation of telomerase, has been shown to confer loss of sensitivity to a wide range of anti-neoplastic agents. METHODS To evaluate whether telomerase inhibition using non-nucleosidic inhibitor of telomerase BIBR1532 could enhance cytotoxic effect of doxorubicin in acute lymphoblastic leukemia, Nalm-6 pre-B ALL cells were subjected to combination treatment and subsequent cell viability, growth kinetics, caspase-3 activity, and transcriptional alteration of p73, p21, FOXO3a, c-Myc, hTERT, and other apoptosis-related target genes were investigated. RESULTS Combination of BIBR1532 with doxorubicin produced a synergistic anticancer effect probably through induction of p73. Transcription factor p73 not only suppressed the proliferative capacity of the cells through induction of p21-mediated G1 arrest, but also down-regulated the mRNA level of hTERT and c-Myc. Our results also report that BIBR1532 induced a caspase-dependent apoptosis, at least partially, through heightened ROS levels, and noteworthy enhanced the pro-oxidant property of doxorubicin. In harmony, transcriptional repression of survivin could be a probable underlying mechanism for the induction of apoptosis through shifting the ratio of death promoters to death repressors via alteration of Bax and Bcl2 expression. CONCLUSIONS Overall, it seems that combination of BIBR1532 and doxorubicin could be a novel therapeutic strategy for acute lymphoblastic leukemia that may be clinically accessible in the near future.
Collapse
Affiliation(s)
- Davood Bashash
- a Department of Hematology and Blood Banking, School of Allied Medical Sciences , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Mohadeseh Zareii
- a Department of Hematology and Blood Banking, School of Allied Medical Sciences , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Ava Safaroghli-Azar
- a Department of Hematology and Blood Banking, School of Allied Medical Sciences , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Mir Davood Omrani
- b Department of Medical Genetics, Faculty of Medicine , Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Seyed H Ghaffari
- c Hematology, Oncology and Stem Cell Transplantation Research Center , Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
191
|
Zhu Z, Tran H, Mathahs MM, Moninger TO, Schmidt WN. HCV Induces Telomerase Reverse Transcriptase, Increases Its Catalytic Activity, and Promotes Caspase Degradation in Infected Human Hepatocytes. PLoS One 2017; 12:e0166853. [PMID: 28056029 PMCID: PMC5215869 DOI: 10.1371/journal.pone.0166853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 10/17/2016] [Indexed: 01/09/2023] Open
Abstract
Introduction Telomerase repairs the telomeric ends of chromosomes and is active in nearly all malignant cells. Hepatitis C virus (HCV) is known to be oncogenic and potential interactions with the telomerase system require further study. We determined the effects of HCV infection on human telomerase reverse transcriptase (TERT) expression and enzyme activity in primary human hepatocytes and continuous cell lines. Results Primary human hepatocytes and Huh-7.5 hepatoma cells showed early de novo TERT protein expression 2–4 days after infection and these events coincided with increased TERT promoter activation, TERT mRNA, and telomerase activity. Immunoprecipitation studies demonstrated that NS3-4A protease-helicase, in contrast to core or NS5A, specifically bound to the C-terminal region of TERT through interactions between helicase domain 2 and protease sequences. Increased telomerase activity was noted when NS3-4A was transfected into cells, when added to reconstituted mixtures of TERT and telomerase RNA, and when incubated with high molecular weight telomerase ‘holoenzyme’ complexes. The NS3-4A catalytic effect on telomerase was inhibited with primuline or danoprevir, agents that are known to inhibit NS3 helicase and protease activities respectively. In HCV infected cells, NS3-4A could be specifically recovered with telomerase holoenzyme complexes in contrast to NS5A or core protein. HCV infection also activated the effector caspase 7 which is known to target TERT. Activation coincided with the appearance of lower molecular weight carboxy-terminal fragment(s) of TERT, chiefly sized at 45 kD, which could be inhibited with pancaspase or caspase 7 inhibitors. Conclusions HCV infection induces TERT expression and stimulates telomerase activity in addition to triggering Caspase activity that leads to increased TERT degradation. These activities suggest multiple points whereby the virus can influence neoplasia. The NS3-4A protease-helicase can directly bind to TERT, increase telomerase activity, and thus potentially influence telomere repair and host cell neoplastic behavior.
Collapse
Affiliation(s)
- Zhaowen Zhu
- Department of Internal Medicine and Research Service, Veterans Affairs Medical Center, Iowa City, IA, United States of America
- Department of Internal Medicine Roy G. and Lucille A. Carver College of Medicine, University of Iowa Iowa City, IA, United States of America
| | - Huy Tran
- Department of Internal Medicine Roy G. and Lucille A. Carver College of Medicine, University of Iowa Iowa City, IA, United States of America
| | - M. Meleah Mathahs
- Department of Internal Medicine and Research Service, Veterans Affairs Medical Center, Iowa City, IA, United States of America
| | - Thomas O. Moninger
- Central Microscopy Research Facility Roy G. and Lucille A. Carver College of Medicine, University of Iowa Iowa City, IA, United States of America
| | - Warren N. Schmidt
- Department of Internal Medicine and Research Service, Veterans Affairs Medical Center, Iowa City, IA, United States of America
- Department of Internal Medicine Roy G. and Lucille A. Carver College of Medicine, University of Iowa Iowa City, IA, United States of America
- * E-mail:
| |
Collapse
|
192
|
Abstract
Telomerase is an enzyme that maintains telomeres in dividing cells using a template on its inherent RNA component. Additionally, the protein part TERT (Telomerase Reverse Transcriptase) has various non-canonical functions. For example, it can localize to mitochondria under increased stress and protect cells in vitro from oxidative stress, DNA damage and apoptosis. Recently it has been demonstrated that TERT protein persists in adult neurons in the brain and data emerge suggesting that it might have a protective function in these post-mitotic cells as well. We have recently published that TERT protein accumulated in mitochondria from brain tissue of mice that have undergone short-term dietary restriction (DR) and rapamycin treatment. This localization correlated to lower levels of oxidative stress in these brain mitochondria. Since rapamycin treatment decreases mTOR signaling which is also thought to play an important role for the beneficial effects of DR, we conclude that the mTOR pathway might be involved in the TERT localization and its effects in brain mitochondria in vivo. These data are in line with previous findings from our group about increased mitochondrial localization of TERT in Alzheimer's disease (AD) brains and a protective function of TERT protein in neurons in vitro against pathological tau.
Collapse
Affiliation(s)
- Satomi Miwa
- Ageing Biology Centre and Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Gabriele Saretzki
- Ageing Biology Centre and Institute for Cell and Molecular Biosciences, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
193
|
Robinson NJ, Schiemann WP. Means to the ends: The role of telomeres and telomere processing machinery in metastasis. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1866:320-329. [PMID: 27768860 PMCID: PMC5138103 DOI: 10.1016/j.bbcan.2016.10.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 10/12/2016] [Accepted: 10/15/2016] [Indexed: 12/29/2022]
Abstract
Despite significant clinical advancements, cancer remains a leading cause of mortality throughout the world due largely to the process of metastasis and the dissemination of cancer cells from their primary tumor of origin to distant secondary sites. The clinical burden imposed by metastasis is further compounded by a paucity of information regarding the factors that mediate metastatic progression. Linear chromosomes are capped by structures known as telomeres, which dictate cellular lifespan in humans by shortening progressively during successive cell divisions. Although telomere shortening occurs in nearly all somatic cells, telomeres may be elongated via two seemingly disjoint pathways: (i) telomerase-mediated extension, and (ii) homologous recombination-based alternative lengthening of telomeres (ALT). Both telomerase and ALT are activated in various human cancers, with more recent evidence implicating both pathways as potential mediators of metastasis. Here we review the known roles of telomere homeostasis in metastasis and posit a mechanism whereby metastatic activity is determined by a dynamic fluctuation between ALT and telomerase, as opposed to the mere activation of a generic telomere elongation program. Additionally, the pleiotropic nature of the telomere processing machinery makes it an attractive therapeutic target for metastasis, and as such, we also explore the therapeutic implications of our proposed mechanism.
Collapse
Affiliation(s)
- Nathaniel J Robinson
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - William P Schiemann
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
194
|
Miwa S, Czapiewski R, Wan T, Bell A, Hill KN, von Zglinicki T, Saretzki G. Decreased mTOR signalling reduces mitochondrial ROS in brain via accumulation of the telomerase protein TERT within mitochondria. Aging (Albany NY) 2016; 8:2551-2567. [PMID: 27777385 PMCID: PMC5115906 DOI: 10.18632/aging.101089] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/02/2016] [Indexed: 01/11/2023]
Abstract
Telomerase in its canonical function maintains telomeres in dividing cells. In addition, the telomerase protein TERT has non-telomeric functions such as shuttling to mitochondria resulting in a decreased oxidative stress, DNA damage and apoptosis. TERT protein persists in adult neurons and can co-localise to mitochondria under various stress conditions. We show here that TERT expression decreased in mouse brain during aging while release of reactive oxygen species (ROS) from the mitochondrial electron transport chain increased. Dietary restriction (DR) caused accumulation of TERT protein in mouse brain mitochondria correlating to decreased ROS release and improved learning and spatial short-term memory. Decreased mTOR signalling is a mediator of DR. Accordingly, feeding mice with rapamycin increased brain mitochondrial TERT and reduced ROS release. Importantly, the beneficial effects of rapamycin on mitochondrial function were absent in brains and fibroblasts from first generation TERT -/- mice, and when TERT shuttling was inhibited by the Src kinase inhibitor bosutinib. Taken together, our data suggests that the mTOR signalling pathway impinges on the mitochondrial localisation of TERT protein, which might in turn contribute to the protection of the brain by DR or rapamycin against age-associated mitochondrial ROS increase and cognitive decline.
Collapse
Affiliation(s)
- Satomi Miwa
- Institute for Cell and Molecular Biosciences, Newcastle Institute for Ageing, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Rafal Czapiewski
- Institute for Cell and Molecular Biosciences, Newcastle Institute for Ageing, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Tengfei Wan
- Institute for Cell and Molecular Biosciences, Newcastle Institute for Ageing, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Amy Bell
- Institute for Cell and Molecular Biosciences, Newcastle Institute for Ageing, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Kirsten N. Hill
- Institute for Cell and Molecular Biosciences, Newcastle Institute for Ageing, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Thomas von Zglinicki
- Institute for Cell and Molecular Biosciences, Newcastle Institute for Ageing, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Gabriele Saretzki
- Institute for Cell and Molecular Biosciences, Newcastle Institute for Ageing, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| |
Collapse
|
195
|
Lionaki E, Gkikas I, Tavernarakis N. Differential Protein Distribution between the Nucleus and Mitochondria: Implications in Aging. Front Genet 2016; 7:162. [PMID: 27695477 PMCID: PMC5025450 DOI: 10.3389/fgene.2016.00162] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/01/2016] [Indexed: 01/05/2023] Open
Abstract
The coordination of nuclear and mitochondrial genomes plays a pivotal role in maintenance of mitochondrial biogenesis and functionality during stress and aging. Environmental and cellular inputs signal to nucleus and/or mitochondria to trigger interorganellar compensatory responses. Loss of this tightly orchestrated coordination results in loss of cellular homeostasis and underlies various pathologies and age-related diseases. Several signaling cascades that govern interorganellar communication have been revealed up to now, and have been classified as part of the anterograde (nucleus to mitochondria) or retrograde (mitochondrial to nucleus) response. Many of these molecular pathways rely on the dual distribution of nuclear or mitochondrial components under basal or stress conditions. These dually localized components usually engage in specific tasks in their primary organelle of function, whilst upon cellular stimuli, they appear in the other organelle where they engage in the same or a different task, triggering a compensatory stress response. In this review, we focus on protein factors distributed between the nucleus and mitochondria and activated to exert their functions upon basal or stress conditions. We further discuss implications of bi-organellar targeting in the context of aging.
Collapse
Affiliation(s)
- Eirini Lionaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas Heraklion, Greece
| | - Ilias Gkikas
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas Heraklion, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-HellasHeraklion, Greece; Department of Basic Sciences, Faculty of Medicine, University of CreteHeraklion, Greece
| |
Collapse
|
196
|
Ait-Aissa K, Ebben JD, Kadlec AO, Beyer AM. Friend or foe? Telomerase as a pharmacological target in cancer and cardiovascular disease. Pharmacol Res 2016; 111:422-433. [PMID: 27394166 PMCID: PMC5026584 DOI: 10.1016/j.phrs.2016.07.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/01/2016] [Accepted: 07/02/2016] [Indexed: 12/20/2022]
Abstract
Aging, cancer, and chronic disease have remained at the forefront of basic biological research for decades. Within this context, significant attention has been paid to the role of telomerase, the enzyme responsible for lengthening telomeres, the nucleotide sequences located at the end of chromosomes found in the nucleus. Alterations in telomere length and telomerase activity are a common denominator to the underlying pathology of these diseases. While nuclear-specific, telomere-lengthening effects of telomerase impact cellular/organismal aging and cancer development, non-canonical, extra-nuclear, and non-telomere-lengthening contributions of telomerase have only recently been described and their exact physiological implications are ill defined. Although the mechanism remains unclear, recent reports reveal that the catalytic subunit of telomerase, telomerase reverse transcriptase (TERT), regulates levels of mitochondrial-derived reactive oxygen species (mtROS), independent of its established role in the nucleus. Telomerase inhibition has been the target of chemotherapy (directed or indirectly) for over a decade now, yet no telomerase inhibitor is FDA approved and few are currently in late-stage clinical trials, possibly due to underappreciation of the distinct extra-nuclear functions of telomerase. Moreover, evaluation of telomerase-specific therapies is largely limited to the context of chemotherapy, despite reports of the beneficial effects of telomerase activation in the cardiovascular system in relation to such processes as endothelial dysfunction and myocardial infarction. Thus, there is a need for better understanding of telomerase-focused cell and organism physiology, as well as development of telomerase-specific therapies in relation to cancer and extension of these therapies to cardiovascular pathologies. This review will detail findings related to telomerase and evaluate its potential to serve as a therapeutic target.
Collapse
Affiliation(s)
- Karima Ait-Aissa
- Department of Medicine
- Department of Physiology, Cardiovascular Center
| | - Johnathan D. Ebben
- Department of Pharmacology & Toxicology
- Cancer Center, Medical College of Wisconsin
| | - Andrew O. Kadlec
- Department of Medicine
- Department of Physiology, Cardiovascular Center
| | - Andreas M. Beyer
- Department of Medicine
- Department of Physiology, Cardiovascular Center
| |
Collapse
|
197
|
Nephrotoxicity in rabbits after long-term nandrolone decanoate administration. Toxicol Lett 2016; 259:21-27. [DOI: 10.1016/j.toxlet.2016.06.1122] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 06/21/2016] [Accepted: 06/23/2016] [Indexed: 12/23/2022]
|
198
|
Ameer SS, Xu Y, Engström K, Li H, Tallving P, Nermell B, Boemo A, Parada LA, Peñaloza LG, Concha G, Harari F, Vahter M, Broberg K. Exposure to Inorganic Arsenic Is Associated with Increased Mitochondrial DNA Copy Number and Longer Telomere Length in Peripheral Blood. Front Cell Dev Biol 2016; 4:87. [PMID: 27597942 PMCID: PMC4992680 DOI: 10.3389/fcell.2016.00087] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 08/08/2016] [Indexed: 01/03/2023] Open
Abstract
Background: Exposure to inorganic arsenic (iAs) through drinking water causes cancer. Alterations in mitochondrial DNA copy number (mtDNAcn) and telomere length in blood have been associated with cancer risk. We elucidated if arsenic exposure alters mtDNAcn and telomere length in individuals with different arsenic metabolizing capacity. Methods: We studied two groups in the Salta province, Argentina, one in the Puna area of the Andes (N = 264, 89% females) and one in Chaco (N = 169, 75% females). We assessed arsenic exposure as the sum of arsenic metabolites [iAs, methylarsonic acid (MMA), dimethylarsinic acid (DMA)] in urine (U-As) using high-performance liquid chromatography coupled with hydride generation and inductively coupled plasma mass spectrometry. Efficiency of arsenic metabolism was expressed as percentage of urinary metabolites. MtDNAcn and telomere length were determined in blood by real-time PCR. Results: Median U-As was 196 (5–95 percentile: 21–537) μg/L in Andes and 80 (5–95 percentile: 15–1637) μg/L in Chaco. The latter study group had less-efficient metabolism, with higher %iAs and %MMA in urine compared with the Andean group. U-As was significantly associated with increased mtDNAcn (log2 transformed to improve linearity) in Chaco (β = 0.027 per 100 μg/L, p = 0.0085; adjusted for age and sex), but not in Andes (β = 0.025, p = 0.24). U-As was also associated with longer telomere length in Chaco (β = 0.016, p = 0.0066) and Andes (β = 0.0075, p = 0.029). In both populations, individuals with above median %iAs showed significantly higher mtDNAcn and telomere length compared with individuals with below median %iAs. Conclusions: Arsenic was associated with increased mtDNAcn and telomere length, particularly in individuals with less-efficient arsenic metabolism, a group who may have increased risk for arsenic-related cancer.
Collapse
Affiliation(s)
- Syeda S Ameer
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University Lund, Sweden
| | - YiYi Xu
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University Lund, Sweden
| | - Karin Engström
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund UniversityLund, Sweden; Unit of Metals and Health, Institute of Environmental Medicine, Karolinska InstitutetStockholm, Sweden
| | - Huiqi Li
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University Lund, Sweden
| | - Pia Tallving
- Department of Laboratory Medicine, Division of Occupational and Environmental Medicine, Lund University Lund, Sweden
| | - Barbro Nermell
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet Stockholm, Sweden
| | - Analia Boemo
- Facultad de Ciencias Exactas and Consejo de Investigación, Universidad Nacional de Salta Salta, Argentina
| | - Luis A Parada
- Institute of Experimental Pathology - UNSa - CONICET Salta, Argentina
| | - Lidia G Peñaloza
- Facultad de Ciencias Exactas and Consejo de Investigación, Universidad Nacional de Salta Salta, Argentina
| | - Gabriela Concha
- Risk Benefit Assessment Unit, Science Department, National Food Agency Uppsala, Sweden
| | - Florencia Harari
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet Stockholm, Sweden
| | - Marie Vahter
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet Stockholm, Sweden
| | - Karin Broberg
- Unit of Metals and Health, Institute of Environmental Medicine, Karolinska Institutet Stockholm, Sweden
| |
Collapse
|
199
|
Radan L, Hughes CS, Teichroeb JH, Postovit LM, Betts DH. Delivering Antisense Morpholino Oligonucleotides to Target Telomerase Splice Variants in Human Embryonic Stem Cells. Methods Mol Biol 2016; 1341:133-42. [PMID: 25822725 DOI: 10.1007/7651_2015_226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Morpholino oligonucleotides (MO) are an innovative tool that provides a means for examining and modifying gene expression outcomes by antisense interaction with targeted RNA transcripts. The site-specific nature of their binding facilitates focused modulation to alter splice variant expression patterns. Here we describe the steric-blocking of human telomerase reverse transcriptase (hTERT) Δα and Δβ splice variants using MO to examine cellular outcomes related to pluripotency and differentiation in human embryonic stem cells.
Collapse
Affiliation(s)
- Lida Radan
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | - Chris S Hughes
- British Columbia Cancer Research Center, Vancouver, BC, Canada
| | - Jonathan H Teichroeb
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| | | | - Dean H Betts
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
200
|
Muzza M, Colombo C, Cirello V, Perrino M, Vicentini L, Fugazzola L. Oxidative stress and the subcellular localization of the telomerase reverse transcriptase (TERT) in papillary thyroid cancer. Mol Cell Endocrinol 2016; 431:54-61. [PMID: 27164443 DOI: 10.1016/j.mce.2016.05.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/04/2016] [Accepted: 05/04/2016] [Indexed: 01/23/2023]
Abstract
During hormonogenesis, thyrocytes are physiologically exposed to high levels of oxidative stress (OS) which could either be involved in the pathogenesis of thyroid cancer or exert a cytotoxic effect. We analyzed the oxidative status of papillary thyroid cancer (PTC) both directly, by measuring H2O2 generation by NADPH oxidases (NOXs), and indirectly, by evaluating the antioxidant activity of glutathione peroxidase (GPX), which neutralizes H2O2 excess, and the lipid peroxidation (LP). Moreover, we investigated the subcellular localization of telomerase reverse transcriptase (TERT), and the H2O2 levels in the mitochondria of tumor and normal tissues. The calcium-dependent and independent H2O2 generation activity was significantly higher in tumors than in normal tissues. The GPX activity was higher in PTCs than in normal tissues, and, consistently, no differences were found in LP levels. Moreover, while TERT nuclear expression was similar in tumor and normal tissues, the mitochondrial localization was significantly higher in tumors. At the mitochondrial level, no differences were found in H2O2 generation between tumor and normal tissues. In conclusion, present data demonstrate that the intracellular H2O2 generation by NOXs is significantly higher in PTCs than in normal thyroid tissues. The increased GPX activity found in tumors counteracts the potential cytotoxic effects of high OS exposure. The significantly higher mitochondrial localization of TERT in tumors is consistent with its shuttling from the nucleus upon exposure to high OS. Finally, mitochondrial OS was not significantly different in tumors and normal tissues, supporting the postulated role of mitochondrial TERT in the control of local H2O2 production.
Collapse
Affiliation(s)
- Marina Muzza
- Endocrine Unit, Fondazione IRCCS Ca' Granda, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Carla Colombo
- Endocrine Unit, Fondazione IRCCS Ca' Granda, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Valentina Cirello
- Endocrine Unit, Fondazione IRCCS Ca' Granda, Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Michela Perrino
- Endocrine Unit, Fondazione IRCCS Ca' Granda, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | | | - Laura Fugazzola
- Endocrine Unit, Fondazione IRCCS Ca' Granda, Milan, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy.
| |
Collapse
|