101
|
Akincilar SC, Chan CHT, Ng QF, Fidan K, Tergaonkar V. Non-canonical roles of canonical telomere binding proteins in cancers. Cell Mol Life Sci 2021; 78:4235-4257. [PMID: 33599797 PMCID: PMC8164586 DOI: 10.1007/s00018-021-03783-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 12/28/2020] [Accepted: 01/29/2021] [Indexed: 02/06/2023]
Abstract
Reactivation of telomerase is a major hallmark observed in 90% of all cancers. Yet paradoxically, enhanced telomerase activity does not correlate with telomere length and cancers often possess short telomeres; suggestive of supplementary non-canonical roles that telomerase might play in the development of cancer. Moreover, studies have shown that aberrant expression of shelterin proteins coupled with their release from shortening telomeres can further promote cancer by mechanisms independent of their telomeric role. While targeting telomerase activity appears to be an attractive therapeutic option, this approach has failed in clinical trials due to undesirable cytotoxic effects on stem cells. To circumvent this concern, an alternative strategy could be to target the molecules involved in the non-canonical functions of telomeric proteins. In this review, we will focus on emerging evidence that has demonstrated the non-canonical roles of telomeric proteins and their impact on tumorigenesis. Furthermore, we aim to address current knowledge gaps in telomeric protein functions and propose future research approaches that can be undertaken to achieve this.
Collapse
Affiliation(s)
- Semih Can Akincilar
- 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), Proteos, 61, Biopolis Drive, Singapore, 138673, Singapore
| | - Claire Hian Tzer Chan
- 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), Proteos, 61, Biopolis Drive, Singapore, 138673, Singapore
| | - Qin Feng Ng
- 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), Proteos, 61, Biopolis Drive, Singapore, 138673, Singapore
| | - Kerem Fidan
- 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), Proteos, 61, Biopolis Drive, 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), Proteos, 61, Biopolis Drive, Singapore, 138673, Singapore.
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117593, Singapore.
| |
Collapse
|
102
|
Zia A, Farkhondeh T, Pourbagher-Shahri AM, Samarghandian S. The Roles of mitochondrial dysfunction and Reactive Oxygen Species in Aging and Senescence. Curr Mol Med 2021; 22:37-49. [PMID: 33602082 DOI: 10.2174/1566524021666210218112616] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/16/2020] [Accepted: 12/23/2020] [Indexed: 11/22/2022]
Abstract
The aging process deteriorates organs' function at different levels, causing its progressive decline to resist stress, damage, and disease. In addition to alterations in metabolic control and gene expression, the rate of aging has been connected with the generation of high amounts of Reactive Oxygen Species (ROS). The essential perspective in free radical biology is that reactive oxygen species (ROS) and free radicals are toxic, mostly cause direct biological damage to targets, and are thus a major cause of oxidative stress. Different enzymatic and non-enzymatic compounds in the cells have roles in neutralizing this toxicity. Oxidative damage in aging is mostly high in particular molecular targets, such as mitochondrial DNA and aconitase, and oxidative stress in mitochondria can cause tissue aging across intrinsic apoptosis. Mitochondria's function and morphology are impaired through aging, following a decrease in the membrane potential by an increase in peroxide generation and size of the organelles. Telomeres may be the significant trigger of replicative senescence. Oxidative stress accelerates telomere loss, whereas antioxidants slow it down. Oxidative stress is a crucial modulator of telomere shortening, and that telomere-driven replicative senescence is mainly a stress response. The age-linked mitochondrial DNA mutation and protein dysfunction aggregate in some organs like the brain and skeletal muscle, thus contributing considerably to these post-mitotic tissues' aging. The aging process is mostly due to accumulated damage done by harmful species in some macromolecules such proteins, DNA, and lipids. The degradation of non-functional, oxidized proteins is a crucial part of the antioxidant defenses of cells, in which the clearance of these proteins occurs through autophagy in the cells, which is known as mitophagy for mitochondria.
Collapse
Affiliation(s)
- Aliabbas Zia
- Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran. Iran
| | - Tahereh Farkhondeh
- Medical Toxicology and Drug Abuse Research Center (MTDRC), Birjand University of Medical Sciences, Birjand. Iran
| | | | - Saeed Samarghandian
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur. Iran
| |
Collapse
|
103
|
Song H, Chen X, Jiao Q, Qiu Z, Shen C, Zhang G, Sun Z, Zhang H, Luo QY. HIF-1α-Mediated Telomerase Reverse Transcriptase Activation Inducing Autophagy Through Mammalian Target of Rapamycin Promotes Papillary Thyroid Carcinoma Progression During Hypoxia Stress. Thyroid 2021; 31:233-246. [PMID: 32772829 DOI: 10.1089/thy.2020.0023] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Background: It is important to properly understand the molecular mechanisms of aggressive tumors among papillary thyroid carcinomas (PTCs) that are often the most indolent. Hypoxia inducible factor-1α (HIF-1α), induced by hypoxia, plays pivotal roles in the development and metastasis of the many tumors, including PTCs. Upregulation of telomerase reverse transcriptase (TERT) activity is found in highly invasive PTCs. Further, previous studies have reported that autophagy serves as a protective mechanism to facilitate PTC cell survival. We, therefore, hypothesized that there was a link between HIF-1α, TERT, and autophagy in promoting PTC progression. Methods: Immunohistochemistry staining was conducted to evaluate the expressions of HIF-1α, TERT, and autophagy marker, LC3-II, in matched PTC tumors and corresponding nontumor tissues. Two PTC cell lines (TPC-1 and BCPAP) were used in subsequent cytological function studies. Cell viability, proliferation, apoptosis, migration, and invasion were assessed during hypoxia, genetic enhancement and inhibition of TERT, and chemical and genetic inhibition of autophagy. The protein expression levels of the corresponding biomarkers were determined by Western blotting, and autophagy flow was detected. We characterized the molecular mechanism of PTC cell progression. Results: The protein expression levels of HIF-1α, TERT, and LC3-II were upregulated in PTCs and were significantly correlated with high tumor-node-metastasis stage. Further, an in vitro study indicated that HIF-1α induced by hypoxia functioned as a transcriptional activator by binding with sequences potentially located in the TERT promoter and was positively correlated with the malignant behavior of PTC cell lines. Overexpression of TERT inhibited the kinase activity of mammalian target of rapamycin (mTOR), resulting in the activation of autophagy. Functionally, TERT-induced autophagy provided a survival advantage to PTC cells during hypoxia stress. Conclusions: We identified a novel molecular mechanism involving the HIF-1α/TERT axis, which promoted PTC progression by inducing autophagy through mTOR during hypoxia stress. These findings may provide a basis for the new treatment of aggressive PTCs.
Collapse
Affiliation(s)
- Hongjun Song
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Xiaoyue Chen
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Qiong Jiao
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhongling Qiu
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Chentian Shen
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Guoqiang Zhang
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhenkui Sun
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Huizhen Zhang
- Department of Pathology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| |
Collapse
|
104
|
Hoffmann J, Richardson G, Haendeler J, Altschmied J, Andrés V, Spyridopoulos I. Telomerase as a Therapeutic Target in Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2021; 41:1047-1061. [PMID: 33504179 DOI: 10.1161/atvbaha.120.315695] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Shortened telomeres have been linked to numerous chronic diseases, most importantly coronary artery disease, but the underlying mechanisms remain ill defined. Loss-of-function mutations and deletions in telomerase both accelerate telomere shortening but do not necessarily lead to a clinical phenotype associated with atherosclerosis, questioning the causal role of telomere length in cardiac pathology. The differential extranuclear functions of the 2 main components of telomerase, telomerase reverse transcriptase and telomerase RNA component, offer important clues about the complex relationship between telomere length and cardiovascular pathology. In this review, we critically discuss relevant preclinical models, genetic disorders, and clinical studies to elucidate the impact of telomerase in cardiovascular disease and its potential role as a therapeutic target. We suggest that the antioxidative function of mitochondrial telomerase reverse transcriptase might be atheroprotective, making it a potential target for clinical trials. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Jedrzej Hoffmann
- Department of Medicine, Cardiology, Goethe University Hospital, Frankfurt, Germany (J.H.)
| | - Gavin Richardson
- Institute of Biosciences, Newcastle University, United Kingdom (G.R.)
| | - Judith Haendeler
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty and University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Germany (J.H., J.A.)
| | - Joachim Altschmied
- Institute of Clinical Chemistry and Laboratory Diagnostic, Medical Faculty and University Hospital Duesseldorf and Heinrich-Heine-University Duesseldorf, Germany (J.H., J.A.).,IUF - Leibniz Research Institute for Environmental Medicine (J.A.)
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (V.A.).,Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Spain (V.A.)
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Newcastle University, United Kingdom (I.S.).,Freeman Hospital, Cardiothoracic Centre, Newcastle Upon Tyne Hospital Trust, United Kingdom (I.S.)
| |
Collapse
|
105
|
Abstract
PURPOSE One of the most important serious malignancies is gastric cancer (GC) with a high mortality globally. In this way, beside the environmental factors, genetic parameter has a remarkable effective fluctuation in GC. Correspondingly, telomeres are nucleoprotein structures measuring the length of telomeres and they have special potential in diagnosis of various types of cancers. Defect protection of the telomeric length initiates the instability of the genome during cancer, including gastric cancer. The most common way of maintaining telomere length is the function of the telomerase enzyme that replicates the TTAGGG to the end of the 3' chromosome. METHODS In this review, we want to discuss the alterations of hTERT repression on the modification of TERRA gene expression in conjunction with the importance of telomere and telomerase in GC. RESULTS The telomerase enzyme contains two essential components called telomerase reverse transcriptase (hTERT) and RNA telomerase (hTR, hTERC). Deregulation of hTERT plays a key role in the multistage process of tumorigenicity and anticancer drug resistance. The direct relationship between telomerase activity and hTERT has led to hTERT to be considered a key target for cancer treatment. Recent results show that telomeres are transcribed into telomeric repeat-containing RNA (TERRA) in mammalian cells and are long noncoding RNAs (lncRNAs) identified in different tissues. In addition, most chemotherapy methods have a lot of side effects on normal cells. CONCLUSION Telomere and telomerase are useful therapeutic goal. According to the main roles of hTERT in tumorigenesis, growth, migration, and cancer invasion, hTERT and regulatory mechanisms that control the expression of hTERT are attractive therapeutic targets for cancer treatment.
Collapse
|
106
|
Telomerase therapy attenuates cardiotoxic effects of doxorubicin. Mol Ther 2021; 29:1395-1410. [PMID: 33388418 PMCID: PMC8058493 DOI: 10.1016/j.ymthe.2020.12.035] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 11/17/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022] Open
Abstract
Doxorubicin is one of the most potent chemotherapeutic agents. However, its clinical use is restricted due to the severe risk of cardiotoxicity, partially attributed to elevated production of reactive oxygen species (ROS). Telomerase canonically maintains telomeres during cell division but is silenced in adult hearts. In non-dividing cells such as cardiomyocytes, telomerase confers pro-survival traits, likely owing to the detoxification of ROS. Therefore, we hypothesized that pharmacological overexpression of telomerase may be used as a therapeutic strategy for the prevention of doxorubicin-induced cardiotoxicity. We used adeno-associated virus (AAV)-mediated gene therapy for long-term expression of telomerase in in vitro and in vivo models of doxorubicin-induced cardiotoxicity. Overexpression of telomerase protected the heart from doxorubicin-mediated apoptosis and rescued cardiac function, which was accompanied by preserved cardiomyocyte size. At the mechanistic level, we observed altered mitochondrial morphology and dynamics in response to telomerase expression. Complementary in vitro experiments confirmed the anti-apoptotic effects of telomerase overexpression in human induced pluripotent stem cell-derived cardiomyocytes after doxorubicin treatment. Strikingly, elevated levels of telomerase translocated to the mitochondria upon doxorubicin treatment, which helped to maintain mitochondrial function. Thus, telomerase gene therapy could be a novel preventive strategy for cardiotoxicity by chemotherapy agents such as the anthracyclines.
Collapse
|
107
|
Um S, Ha J, Choi SJ, Oh W, Jin HJ. Prospects for the therapeutic development of umbilical cord blood-derived mesenchymal stem cells. World J Stem Cells 2020; 12:1511-1528. [PMID: 33505598 PMCID: PMC7789129 DOI: 10.4252/wjsc.v12.i12.1511] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/23/2020] [Accepted: 11/12/2020] [Indexed: 02/06/2023] Open
Abstract
Umbilical cord blood (UCB) is a primitive and abundant source of mesenchymal stem cells (MSCs). UCB-derived MSCs have a broad and efficient therapeutic capacity to treat various diseases and disorders. Despite the high latent self-renewal and differentiation capacity of these cells, the safety, efficacy, and yield of MSCs expanded for ex vivo clinical applications remains a concern. However, immunomodulatory effects have emerged in various disease models, exhibiting specific mechanisms of action, such as cell migration and homing, angiogenesis, anti-apoptosis, proliferation, anti-cancer, anti-fibrosis, anti-inflammation and tissue regeneration. Herein, we review the current literature pertaining to the UCB-derived MSC application as potential treatment strategies, and discuss the concerns regarding the safety and mass production issues in future applications.
Collapse
Affiliation(s)
- Soyoun Um
- Research Team for Immune Cell Therapy, Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Jueun Ha
- Research Team for Osteoarthritis, Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Soo Jin Choi
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Wonil Oh
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| | - Hye Jin Jin
- Biomedical Research Institute, MEDIPOST Co., Ltd., Seongnam 13494, South Korea
| |
Collapse
|
108
|
Vedelek B, Maddali AK, Davenova N, Vedelek V, Boros IM. TERT promoter alterations could provide a solution for Peto's paradox in rodents. Sci Rep 2020; 10:20815. [PMID: 33257697 PMCID: PMC7704627 DOI: 10.1038/s41598-020-77648-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 11/13/2020] [Indexed: 12/19/2022] Open
Abstract
Cancer is a genetic disease caused by changes in gene expression resulting from somatic mutations and epigenetic changes. Although the probability of mutations is proportional with cell number and replication cycles, large bodied species do not develop cancer more frequently than smaller ones. This notion is known as Peto's paradox, and assumes stronger tumor suppression in larger animals. One of the possible tumor suppressor mechanisms involved could be replicative senescence caused by telomere shortening in the absence of telomerase activity. We analysed telomerase promoter activity and transcription factor binding in mammals to identify the key element of telomerase gene inactivation. We found that the GABPA transcription factor plays a key role in TERT regulation in somatic cells of small rodents, but its binding site is absent in larger beavers. Protein binding and reporter gene assays verify different use of this site in different species. The presence or absence of the GABPA TF site in TERT promoters of rodents correlates with TERT promoter activity; thus it could determine whether replicative senescence plays a tumor suppressor role in these species, which could be in direct relation with body mass. The GABPA TF binding sites that contribute to TERT activity in somatic cells of rodents are analogous to those mutated in human tumors, which activate telomerase by a non-ALT mechanism.
Collapse
Affiliation(s)
- Balázs Vedelek
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre, Szeged, Hungary
| | - Asha Kiran Maddali
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
- Institute of Genetics, Biological Research Centre, Szeged, Hungary
| | - Nurgul Davenova
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - Viktor Vedelek
- Department of Genetics, University of Szeged, Szeged, Hungary
| | - Imre M Boros
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary.
- Institute of Biochemistry, Biological Research Centre, Szeged, Hungary.
| |
Collapse
|
109
|
Increased telomerase improves motor function and alpha-synuclein pathology in a transgenic mouse model of Parkinson's disease associated with enhanced autophagy. Prog Neurobiol 2020; 199:101953. [PMID: 33188884 PMCID: PMC7938226 DOI: 10.1016/j.pneurobio.2020.101953] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 10/21/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023]
Abstract
Telomerase activators (TA) increase Tert expression in brains of a PD mouse model. Activator treatment improves PD motor symptoms: gait and balance. Activators reduce different forms of alpha-synuclein in brains of transgenic mice. Decreased autophagy markers LC3 and p62 suggest a better protein degradation. Our preclinical data suggest a use of TA to ameliorate PD-like symptoms.
Protective effects of the telomerase protein TERT have been shown in neurons and brain. We previously demonstrated that TERT protein can accumulate in mitochondria of Alzheimer’s disease (AD) brains and protect from pathological tau in primary mouse neurons. This prompted us to employ telomerase activators in order to boost telomerase expression in a mouse model of Parkinson’s disease (PD) overexpressing human wild type α-synuclein. Our aim was to test whether increased Tert expression levels were able to ameliorate PD symptoms and to activate protein degradation. We found increased Tert expression in brain for both activators which correlated with a substantial improvement of motor functions such as gait and motor coordination while telomere length in the analysed region was not changed. Interestingly, only one activator (TA-65) resulted in a decrease of reactive oxygen species from brain mitochondria. Importantly, we demonstrate that total, phosphorylated and aggregated α-synuclein were significantly decreased in the hippocampus and neocortex of activator-treated mice corresponding to enhanced markers of autophagy suggesting an improved degradation of toxic alpha-synuclein. We conclude that increased Tert expression caused by telomerase activators is associated with decreased α-synuclein protein levels either by activating autophagy or by preventing or delaying impairment of degradation mechanisms during disease progression. This encouraging preclinical data could be translated into novel therapeutic options for neurodegenerative disorders such as PD.
Collapse
|
110
|
Telomeres and telomerase in risk assessment of cardiovascular diseases. Exp Cell Res 2020; 397:112361. [PMID: 33171154 DOI: 10.1016/j.yexcr.2020.112361] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/02/2020] [Indexed: 01/14/2023]
Abstract
Telomeres are repetitive nucleoprotein structures located at the ends of chromosomes. Reduction in the number of repetitions causes cell senescence. Cells with high proliferative potential age with each replication cycle. Post-mitotic cells (e.g. cardiovascular cells) have a different aging mechanism. During the aging of cardiovascular system cells, permanent DNA damage occurs in the telomeric regions caused by mitochondrial dysfunction, which is a phenomenon independent of cell proliferation and telomere length. Mitochondrial dysfunction is accompanied by increased production of reactive oxygen species and development of inflammation. This phenomenon in the cells of blood vessels can lead to atherosclerosis development. Telomere damage in cardiomyocytes leads to the activation of the DNA damage response system, histone H2A.X phosphorylation, p53 activation and p21 and p16 protein synthesis, resulting in the SASP phenotype (senescence-associated secretory phenotype), increased inflammation and cardiac dysfunction. Cardiovascular cells show the activity of the TERT subunit of telomerase, an enzyme that prevents telomere shortening. It turns out that disrupting the activity of this enzyme can also contribute to the formation of cardiovascular diseases. Measurements of telomere length according to the "blood-muscle" model may help in the future to assess the risk of cardiovascular complications in people undergoing cardiological procedures, as well as to assess the effectiveness of some drugs.
Collapse
|
111
|
Affiliation(s)
- Yongkang Zou
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Yu-sheng Cong
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, Zhejiang 311121, China
| | - Junzhi Zhou
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
112
|
Maier R, Bawamia B, Bennaceur K, Dunn S, Marsay L, Amoah R, Kasim A, Filby A, Austin D, Hancock H, Spyridopoulos I. Telomerase Activation to Reverse Immunosenescence in Elderly Patients With Acute Coronary Syndrome: Protocol for a Randomized Pilot Trial. JMIR Res Protoc 2020; 9:e19456. [PMID: 32965237 PMCID: PMC7542409 DOI: 10.2196/19456] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/30/2020] [Accepted: 07/07/2020] [Indexed: 12/23/2022] Open
Abstract
Background Inflammation plays a key role in the pathophysiology of coronary heart disease (CHD) and its acute manifestation, acute coronary syndrome (ACS). Aging is associated with a decline of the immune system, a process known as immunosenescence. This is characterized by an increase in highly proinflammatory T cells that are involved in CHD progression, plaque destabilization, and myocardial ischemia–reperfusion injury. Telomere dysfunction has been implicated in immunosenescence of T lymphocytes. Telomerase is the enzyme responsible for maintaining telomeres during cell divisions. It has a protective effect on cells under oxidative stress and helps regulate flow-mediated dilation in microvasculature. Objective The TACTIC (Telomerase ACTivator to reverse Immunosenescence in Acute Coronary Syndrome) trial will investigate whether a telomerase activator, TA-65MD, can reduce the proportion of senescent T cells in patients with ACS with confirmed CHD. It will also assess the effect of TA-65MD on decreasing telomere shortening, reducing oxidative stress, and improving endothelial function. Methods The study was designed as a single-center, randomized, double-blind, parallel-group, placebo-controlled phase II trial. Recruitment started in January 2019. A total of 90 patients, aged 65 years or older, with treated ACS who have had CHD confirmed by angiography will be enrolled. They will be randomized to one of two groups: TA-65MD oral therapy (8 mg twice daily) or placebo taken for 12 months. The primary outcome is the effect on immunosenescence determined by a decrease in the proportion of CD8+ TEMRA (T effector memory cells re-expressing CD45RA [CD45 expressing exon A]) cells at 12 months. Secondary outcomes include leukocyte telomere length, endothelial function, cardiac function as measured by echocardiography and NT-proBNP (N-terminal fragment of the prohormone brain-type natriuretic peptide), systemic inflammation, oxidative stress, and telomerase activity. Results The study received National Health Service (NHS) ethics approval on August 9, 2018; Medicines and Healthcare products Regulatory Agency approval on October 19, 2018; and NHS Health Research Authority approval on October 22, 2018. The trial began recruiting participants in January 2019 and completed recruitment in March 2020; the trial is due to report results in 2021. Conclusions This pilot trial in older patients with CHD will explore outcomes not previously investigated outside in vitro or preclinical models. The robust design ensures that bias has been minimized. Should the results indicate reduced frequency of immunosenescent CD8+ T cells as well as improvements in telomere length and endothelial function, we will plan a larger, multicenter trial in patients to determine if TA-65MD is beneficial in the treatment of CHD in elderly patients. Trial Registration ISRCTN Registry ISRCTN16613292; http://www.isrctn.com/ISRCTN16613292 and European Union Drug Regulating Authorities Clinical Trials Database (EudraCT), European Union Clinical Trials Register 2017-002876-26; https://tinyurl.com/y4m2so8g International Registered Report Identifier (IRRID) DERR1-10.2196/19456
Collapse
Affiliation(s)
- Rebecca Maier
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Bilal Bawamia
- James Cook University Hospital, Middlesbrough, United Kingdom
| | - Karim Bennaceur
- Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Sarah Dunn
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Leanne Marsay
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Roland Amoah
- Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Adetayo Kasim
- Wolfson Research Institute for Health and Wellbeing, Durham University, Durham, United Kingdom
| | - Andrew Filby
- Flow Cytometry Core Facility, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - David Austin
- James Cook University Hospital, Middlesbrough, United Kingdom
| | - Helen Hancock
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Ioakim Spyridopoulos
- Institute of Genetic Medicine, Newcastle University, Newcastle Upon Tyne, United Kingdom
| |
Collapse
|
113
|
Liao Y, Gao Y, Chang A, Li Z, Wang H, Cao J, Gu W, Tang R. Melatonin synergizes BRAF-targeting agent dabrafenib for the treatment of anaplastic thyroid cancer by inhibiting AKT/hTERT signalling. J Cell Mol Med 2020; 24:12119-12130. [PMID: 32935463 PMCID: PMC7579709 DOI: 10.1111/jcmm.15854] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 08/07/2020] [Accepted: 08/17/2020] [Indexed: 12/30/2022] Open
Abstract
As a selective inhibitor of BRAF kinase, dabrafenib has shown potent anti‐tumour activities in patients with BRAFV600E mutant anaplastic thyroid cancer. However, the resistance of thyroid cancer cells to dabrafenib limited its therapeutic effect. The effects of melatonin and dabrafenib as monotherapy or in combination on the proliferation, cell cycle arrest, apoptosis, migration and invasion of anaplastic thyroid cancer cells were examined. The molecular mechanism involved in drug combinations was also revealed. Melatonin enhanced dabrafenib‐mediated inhibition of cell proliferation, migration and invasion, and promoted dabrafenib‐induced apoptosis and cell cycle arrest in anaplastic thyroid cancer cells. Molecular mechanistic studies further uncovered that melatonin synergized with dabrafenib to inhibit AKT and EMT signalling pathways. Furthermore, melatonin and dabrafenib synergistically inhibited the expression of hTERT, and the inhibition of cell viability and the induction of cell cycle arrest mediated by the combination of these two drugs were reversed by hTERT overexpression. Taken together, our results demonstrated that melatonin synergized the anti‐tumour effect of dabrafenib in human anaplastic thyroid cancer cells by inhibiting multiple signalling pathways, and provided new insights in exploring the potential therapeutic targets for the treatment of anaplastic thyroid cancer.
Collapse
Affiliation(s)
- Yina Liao
- Shanghai Center for Thyroid Disease, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yao Gao
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - An Chang
- Department of Drug Administration, First affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Zongjuan Li
- The Second Affiliated Hospital and Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Huayu Wang
- Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Cao
- Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Gu
- Department of Endocrinology, Children's Hospital of Nanjing Medical University, Nanjing, China
| | - Ranran Tang
- Nanjing Maternity and Child Health Care Hospital, Women's Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
114
|
Zou Y, Cong YS, Zhou J. Implications of telomerase reverse transcriptase in tumor metastasis. BMB Rep 2020; 53:458-465. [PMID: 32731912 PMCID: PMC7526981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/08/2020] [Accepted: 07/14/2020] [Indexed: 08/03/2024] Open
Abstract
Metastasis is the main culprit of the great majority of cancerrelated deaths. However, the complicated process of the invasion-metastasis cascade remains the least understood aspect of cancer biology. Telomerase plays a pivotal role in bypassing cellular senescence and sustaining the cancer progression by maintaining telomere homeostasis and genomic integrity. Telomerase reverse transcriptase (TERT) exerts a series of fundamental functions that are independent of its enzymatic cellular activity, including proliferation, inflammation, epithelia-mesenchymal transition (EMT), angiogenesis, DNA repair, and gene expression. Accumulating evidence indicates that TERT may facilitate most steps of the invasion-metastasis cascade. In this review, we summarize important advances that have revealed some of the mechanisms by which TERT facilitates tumor metastasis, providing an update on the non-canonical functions of telomerase beyond telomere maintaining. [BMB Reports 2020; 53(9): 458-465].
Collapse
Affiliation(s)
- Yongkang Zou
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Yu-sheng Cong
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, Zhejiang 311121, China
| | - Junzhi Zhou
- Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- Institute of Aging Research, Hangzhou Normal University School of Medicine, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
115
|
Pańczyszyn A, Boniewska-Bernacka E, Goc A. The role of telomeres and telomerase in the senescence of postmitotic cells. DNA Repair (Amst) 2020; 95:102956. [PMID: 32937289 DOI: 10.1016/j.dnarep.2020.102956] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
Abstract
Senescence is a process related to the stopping of divisions and changes leading the cell to the SASP phenotype. Permanent senescence of many SASP cells contributes to faster aging of the body and development of age-related diseases due to the release of pro-inflammatory factors. Both mitotically active and non-dividing cells can undergo senescence as a result of activation of different molecular pathways. Telomeres, referred to as the molecular clock, direct the dividing cell into the aging pathway when reaching a critical length. In turn, the senescence of postmitotic cells depends not on the length of telomeres, but their functionality. Dysfunctional telomeres are responsible for triggering the signaling of DNA damage response (DDR). Telomerase subunits in post-mitotic cells translocate between the nucleus, cytoplasm and mitochondria, participating in the regulation of their activity. Among other things, they contribute to the reduction of reactive oxygen species generation, which leads to telomere dysfunction and, consequently, senescence. Some proteins of the shelterin complex also play a protective role by inhibiting senescence-initiating kinases and limiting ROS production by mitochondria.
Collapse
Affiliation(s)
- Anna Pańczyszyn
- University of Opole, Institute of Medical Sciences, Department of Biology and Genetics, Opole 45-040, Pl.Kopernika 11a, Poland.
| | - Ewa Boniewska-Bernacka
- University of Opole, Institute of Medical Sciences, Department of Biology and Genetics, Opole 45-040, Pl.Kopernika 11a, Poland.
| | - Anna Goc
- University of Opole, Institute of Medical Sciences, Department of Biology and Genetics, Opole 45-040, Pl.Kopernika 11a, Poland.
| |
Collapse
|
116
|
Hsieh AY, Kimmel E, Pick N, Sauvé L, Brophy J, Kakkar F, Bitnun A, Murray MC, Côté HC. Inverse relationship between leukocyte telomere length attrition and blood mitochondrial DNA content loss over time. Aging (Albany NY) 2020; 12:15196-15221. [PMID: 32703912 PMCID: PMC7467389 DOI: 10.18632/aging.103703] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 07/06/2020] [Indexed: 05/16/2023]
Abstract
Leukocyte telomere length (LTL) and whole blood mitochondrial DNA (WB mtDNA) content are aging markers impacted by chronic diseases such as human immunodeficiency virus (HIV) infection. We characterized the relationship between these two markers in 312 women ≥12 years of age living with HIV and 300 HIV-negative controls. We found no relationship between the two markers cross-sectionally. In multivariable models, age, ethnicity, HIV, and tobacco smoking were independently associated with shorter LTL, and the former three with lower WB mtDNA. Longitudinally, among a subgroup of 228 HIV participants and 68 HIV-negative controls with ≥2 biospecimens ≥1 year apart, an inverted pattern was observed between the rates of change in LTL and WB mtDNA content per year, whereby faster decline of one was associated with the preservation of the other. Furthermore, if HIV viral control was not maintained between visits, increased rates of both LTL attrition and WB mtDNA loss were observed. We describe a novel relationship between two established aging markers, whereby rates of change in LTL and WB mtDNA are inversely related. Our findings highlight the importance of maintaining HIV viral control, the complementary longitudinal relationship between the two markers, and the need to consider both in aging studies.
Collapse
Affiliation(s)
- Anthony Y.Y. Hsieh
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver V6T 2B5, British Columbia, Canada
- Centre for Blood Research, University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| | - Elana Kimmel
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver V6T 2B5, British Columbia, Canada
- Centre for Blood Research, University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
| | - Neora Pick
- Oak Tree Clinic, BC Women's Hospital, Vancouver V6H 3N1, British Columbia, Canada
- Women's Health Research Institute, Vancouver V6H 2N9, British Columbia, Canada
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver V5Z 1M9, British Columbia, Canada
| | - Laura Sauvé
- Oak Tree Clinic, BC Women's Hospital, Vancouver V6H 3N1, British Columbia, Canada
- Women's Health Research Institute, Vancouver V6H 2N9, British Columbia, Canada
- Department of Pediatrics, University of British Columbia, Vancouver V6H 0B3, British Columbia, Canada
| | - Jason Brophy
- Department of Pediatrics, Children's Hospital of Eastern Ontario, University of Ottawa, Ottawa K1H 8L1, Ontario, Canada
| | - Fatima Kakkar
- Department of Pediatrics, Centre Hospitalier Universtaire Sainte-Justine, Université de Montréal, Montréal H3T 1C5, Quebec, Canada
| | - Ari Bitnun
- Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto M5G 1X8, Ontario, Canada
| | - Melanie C.M. Murray
- Oak Tree Clinic, BC Women's Hospital, Vancouver V6H 3N1, British Columbia, Canada
- Women's Health Research Institute, Vancouver V6H 2N9, British Columbia, Canada
- Department of Medicine, Division of Infectious Diseases, University of British Columbia, Vancouver V5Z 1M9, British Columbia, Canada
| | - Hélène C.F. Côté
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver V6T 2B5, British Columbia, Canada
- Centre for Blood Research, University of British Columbia, Vancouver V6T 1Z3, British Columbia, Canada
- Women's Health Research Institute, Vancouver V6H 2N9, British Columbia, Canada
| |
Collapse
|
117
|
Rosen J, Jakobs P, Ale-Agha N, Altschmied J, Haendeler J. Non-canonical functions of Telomerase Reverse Transcriptase - Impact on redox homeostasis. Redox Biol 2020; 34:101543. [PMID: 32502898 PMCID: PMC7267725 DOI: 10.1016/j.redox.2020.101543] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/11/2020] [Accepted: 04/13/2020] [Indexed: 02/08/2023] Open
Abstract
Telomerase consists of the catalytic subunit Telomerase Reverse Transcriptase (TERT) and the Telomerase RNA Component. Its canonical function is the prevention of telomere erosion. Over the last years it became evident that TERT is also present in tissues with low replicative potential. Important non-canonical functions of TERT are protection against apoptosis and maintenance of the cellular redox homeostasis in cancer as well as in somatic tissues. Intriguingly, TERT and reactive oxygen species (ROS) are interdependent on each other, with TERT being regulated by changes in the redox balance and itself controlling ROS levels in the cytosol and in the mitochondria. The latter is achieved because TERT is present in the mitochondria, where it protects mitochondrial DNA and maintains levels of anti-oxidative enzymes. Since numerous diseases are associated with oxidative stress, increasing the mitochondrial TERT level could be of therapeutic value.
Collapse
Affiliation(s)
- Julia Rosen
- Environmentally-induced Cardiovascular Degeneration, Institute of Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Philipp Jakobs
- Environmentally-induced Cardiovascular Degeneration, Institute of Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Niloofar Ale-Agha
- Environmentally-induced Cardiovascular Degeneration, Institute of Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Joachim Altschmied
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| | - Judith Haendeler
- Environmentally-induced Cardiovascular Degeneration, Institute of Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| |
Collapse
|
118
|
Human telomerase reverse transcriptase positively regulates mitophagy by inhibiting the processing and cytoplasmic release of mitochondrial PINK1. Cell Death Dis 2020; 11:425. [PMID: 32513926 PMCID: PMC7280311 DOI: 10.1038/s41419-020-2641-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 12/18/2022]
Abstract
Mutations in the phosphatase and tensin homologue-induced putative kinase 1 (PINK1) gene have been linked to an early-onset autosomal recessive form of familial Parkinson′s disease (PD). PINK1, a mitochondrial serine/threonine-protein kinase, plays an important role in clearing defective mitochondria by mitophagy – the selective removal of mitochondria through autophagy. Evidence suggests that alteration of the PINK1 pathway contributes to the pathogenesis of PD, but the mechanisms by which the PINK1 pathway regulates mitochondrial quality control through mitophagy remain unclear. Human telomerase reverse transcriptase (hTERT) is a catalytic subunit of telomerase that functions in telomere maintenance as well as several non-telomeric activities. For example, hTERT has been associated with cellular immortalization, cell growth control, and mitochondrial regulation. We determined that hTERT negatively regulates the cleavage and cytosolic processing of PINK1 and enhances its mitochondrial localization by inhibiting mitochondrial processing peptidase β (MPPβ). Consequently, hTERT promotes mitophagy following carbonyl cyanide m-chlorophenylhydrazone (CCCP)-induced mitochondrial dysfunction and improves the function of damaged mitochondria by modulating PINK1. These findings suggest that hTERT positively regulates PINK1 function, leading to increased mitophagy following mitochondrial damage.
Collapse
|
119
|
Hughes WE, Beyer AM, Gutterman DD. Vascular autophagy in health and disease. Basic Res Cardiol 2020; 115:41. [PMID: 32506214 DOI: 10.1007/s00395-020-0802-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 05/26/2020] [Indexed: 12/14/2022]
Abstract
Homeostasis is maintained within organisms through the physiological recycling process of autophagy, a catabolic process that is intricately involved in the mobilization of nutrients during starvation, recycling of cellular cargo, as well as initiation of cellular death pathways. Specific to the cardiovascular system, autophagy responds to both chemical (e.g. free radicals) and mechanical stressors (e.g. shear stress). It is imperative to note that autophagy is not a static process, and measurement of autophagic flux provides a more comprehensive investigation into the role of autophagy. The overarching themes emerging from decades of autophagy research are that basal levels of autophagic flux are critical, physiological stressors may increase or decrease autophagic flux, and more importantly, aberrant deviations from basal autophagy may elicit detrimental effects. Autophagy has predominantly been examined within cardiac or vascular smooth muscle tissue within the context of disease development and progression. Autophagic flux within the endothelium holds an important role in maintaining vascular function, demonstrated by the necessary role for intact autophagic flux for shear-induced release of nitric oxide however the underlying mechanisms have yet to be elucidated. Within this review, we theorize that autophagy itself does not solely control vascular homeostasis, rather, it works in concert with mitochondria, telomerase, and lipids to maintain physiological function. The primary emphasis of this review is on the role of autophagy within the human vasculature, and the integrative effects with physiological processes and diseases as they relate to the vascular structure and function.
Collapse
Affiliation(s)
- William E Hughes
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI, 53213, USA.
| | - Andreas M Beyer
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI, 53213, USA
| | - David D Gutterman
- Department of Medicine, Cardiovascular Center, Medical College of Wisconsin, 8701 West Watertown Plank Road, Milwaukee, WI, 53213, USA
| |
Collapse
|
120
|
Krupinska K, Blanco NE, Oetke S, Zottini M. Genome communication in plants mediated by organelle-n-ucleus-located proteins. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190397. [PMID: 32362260 PMCID: PMC7209962 DOI: 10.1098/rstb.2019.0397] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
An increasing number of eukaryotic proteins have been shown to have a dual localization in the DNA-containing organelles, mitochondria and plastids, and/or the nucleus. Regulation of dual targeting and relocation of proteins from organelles to the nucleus offer the most direct means for communication between organelles as well as organelles and nucleus. Most of the mitochondrial proteins of animals have functions in DNA repair and gene expression by modelling of nucleoid architecture and/or chromatin. In plants, such proteins can affect replication and early development. Most plastid proteins with a confirmed or predicted second location in the nucleus are associated with the prokaryotic core RNA polymerase and are required for chloroplast development and light responses. Few plastid–nucleus-located proteins are involved in pathogen defence and cell cycle control. For three proteins, it has been clearly shown that they are first targeted to the organelle and then relocated to the nucleus, i.e. the nucleoid-associated proteins HEMERA and Whirly1 and the stroma-located defence protein NRIP1. Relocation to the nucleus can be experimentally demonstrated by plastid transformation leading to the synthesis of proteins with a tag that enables their detection in the nucleus or by fusions with fluoroproteins in different experimental set-ups. This article is part of the theme issue ‘Retrograde signalling from endosymbiotic organelles’.
Collapse
Affiliation(s)
- Karin Krupinska
- Institute of Botany, Christian-Albrechts-University of Kiel, Olshausenstraße 40, 24098 Kiel, Germany
| | - Nicolás E Blanco
- Centre of Photosynthetic and Biochemical Studies, Faculty of Biochemical Science and Pharmacy, National University of Rosario (CEFOBI/UNR-CONICET), Rosario, Argentina
| | - Svenja Oetke
- Institute of Botany, Christian-Albrechts-University of Kiel, Olshausenstraße 40, 24098 Kiel, Germany
| | - Michela Zottini
- Department of Biology, University of Padova, Via U. Bassi 58B, 35131 Padova, Italy
| |
Collapse
|
121
|
Vecoli C, Borghini A, Andreassi MG. The molecular biomarkers of vascular aging and atherosclerosis: telomere length and mitochondrial DNA 4977 common deletion. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 784:108309. [PMID: 32430098 DOI: 10.1016/j.mrrev.2020.108309] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022]
Abstract
Age is the dominant risk factor for the most prevalent atherosclerotic vascular diseases, including coronary artery disease, myocardial infarction, cerebrovascular disease and stroke. In human, telomere erosion and mitochondrial DNA (mtDNA) damage play a central role in the mechanisms leading to cellular aging decline. This review summarizes the most relevant findings on the role of telomere shortening and the common mtDNA4977 deletion in the progression and evolution of atherosclerosis by combining insight from experimental models and human clinical studies. The current evidence shows a link between telomere erosion and compromised mitochondrial function and provides a new perspective regarding their potential role as clinical biomarkers and therapeutic targets.
Collapse
|
122
|
Stellos K, Spyridopoulos I. Exercise, telomerase activity, and cardiovascular disease prevention. Eur Heart J 2020; 40:47-49. [PMID: 30496530 DOI: 10.1093/eurheartj/ehy707] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Konstantinos Stellos
- Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.,Freeman Hospital, Newcastle Upon Tyne Hospital Trust, Newcastle Upon Tyne, UK
| | - Ioakim Spyridopoulos
- Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK.,Freeman Hospital, Newcastle Upon Tyne Hospital Trust, Newcastle Upon Tyne, UK
| |
Collapse
|
123
|
Ding X, Liu X, Wang F, Wang F, Geng X. Role of Senescence and Neuroprotective Effects of Telomerase in Neurodegenerative Diseases. Rejuvenation Res 2020; 23:150-158. [DOI: 10.1089/rej.2018.2115] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Xuelu Ding
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin, China
| | - Xuewen Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin, China
| | - Feng Wang
- Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Fei Wang
- Department of Neurology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xin Geng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, Tianjin, China
| |
Collapse
|
124
|
Robin JD, Jacome Burbano M, Peng H, Croce O, Thomas JL, Laberthonniere C, Renault V, Lototska L, Pousse M, Tessier F, Bauwens S, Leong W, Sacconi S, Schaeffer L, Magdinier F, Ye J, Gilson E. Mitochondrial function in skeletal myofibers is controlled by a TRF2-SIRT3 axis over lifetime. Aging Cell 2020; 19:e13097. [PMID: 31991048 PMCID: PMC7059141 DOI: 10.1111/acel.13097] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 11/23/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022] Open
Abstract
Telomere shortening follows a developmentally regulated process that leads to replicative senescence of dividing cells. However, whether telomere changes are involved in postmitotic cell function and aging remains elusive. In this study, we discovered that the level of the TRF2 protein, a key telomere-capping protein, declines in human skeletal muscle over lifetime. In cultured human myotubes, TRF2 downregulation did not trigger telomere dysfunction, but suppressed expression of the mitochondrial Sirtuin 3 gene (SIRT3) leading to mitochondrial respiration dysfunction and increased levels of reactive oxygen species. Importantly, restoring the Sirt3 level in TRF2-compromised myotubes fully rescued mitochondrial functions. Finally, targeted ablation of the Terf2 gene in mouse skeletal muscle leads to mitochondrial dysfunction and sirt3 downregulation similarly to those of TRF2-compromised human myotubes. Altogether, these results reveal a TRF2-SIRT3 axis controlling muscle mitochondrial function. We propose that this axis connects developmentally regulated telomere changes to muscle redox metabolism.
Collapse
Affiliation(s)
- Jérôme D. Robin
- Université Côte d'Azur CNRS Inserm Institut for Research on Cancer and Aging, Nice (IRCAN) Medical School of Nice Nice France
- Marseille Medical Genetics (MMG) U1251 Aix Marseille University Marseille France
| | - Maria‐Sol Jacome Burbano
- Université Côte d'Azur CNRS Inserm Institut for Research on Cancer and Aging, Nice (IRCAN) Medical School of Nice Nice France
| | - Han Peng
- International Research Laboratory in “Hematology, Cancer and Aging” Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Nice University Pôle Sino‐Français de Recherche en Sciences du Vivant et Génomique Shanghai Ruijin Hospital Shanghai China
| | - Olivier Croce
- Université Côte d'Azur CNRS Inserm Institut for Research on Cancer and Aging, Nice (IRCAN) Medical School of Nice Nice France
| | - Jean Luc Thomas
- Neuromuscular Differentiation Group Institut NeuroMyoGene (INMG) UMR5310 Inserm U1217 Ecole Normale Supérieure de Lyon Lyon France
| | | | - Valerie Renault
- Université Côte d'Azur CNRS Inserm Institut for Research on Cancer and Aging, Nice (IRCAN) Medical School of Nice Nice France
| | - Liudmyla Lototska
- Université Côte d'Azur CNRS Inserm Institut for Research on Cancer and Aging, Nice (IRCAN) Medical School of Nice Nice France
| | - Mélanie Pousse
- Université Côte d'Azur CNRS Inserm Institut for Research on Cancer and Aging, Nice (IRCAN) Medical School of Nice Nice France
| | - Florent Tessier
- Université Côte d'Azur CNRS Inserm Institut for Research on Cancer and Aging, Nice (IRCAN) Medical School of Nice Nice France
| | - Serge Bauwens
- Université Côte d'Azur CNRS Inserm Institut for Research on Cancer and Aging, Nice (IRCAN) Medical School of Nice Nice France
| | - Waiian Leong
- International Research Laboratory in “Hematology, Cancer and Aging” Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Nice University Pôle Sino‐Français de Recherche en Sciences du Vivant et Génomique Shanghai Ruijin Hospital Shanghai China
| | - Sabrina Sacconi
- Université Côte d'Azur CNRS Inserm Institut for Research on Cancer and Aging, Nice (IRCAN) Medical School of Nice Nice France
- Peripheral Nervous System, Muscle and ALS Neuromuscular & ALS Center of Reference FHU Oncoage Pasteur 2 Nice University Hospital Nice France
| | - Laurent Schaeffer
- Neuromuscular Differentiation Group Institut NeuroMyoGene (INMG) UMR5310 Inserm U1217 Ecole Normale Supérieure de Lyon Lyon France
| | - Frédérique Magdinier
- Marseille Medical Genetics (MMG) U1251 Aix Marseille University Marseille France
| | - Jing Ye
- International Research Laboratory in “Hematology, Cancer and Aging” Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Nice University Pôle Sino‐Français de Recherche en Sciences du Vivant et Génomique Shanghai Ruijin Hospital Shanghai China
| | - Eric Gilson
- Université Côte d'Azur CNRS Inserm Institut for Research on Cancer and Aging, Nice (IRCAN) Medical School of Nice Nice France
- International Research Laboratory in “Hematology, Cancer and Aging” Shanghai Jiao Tong University School of Medicine/Ruijin Hospital/CNRS/Inserm/Nice University Pôle Sino‐Français de Recherche en Sciences du Vivant et Génomique Shanghai Ruijin Hospital Shanghai China
- Department of Medical Genetics Archet 2 Hospital FHU Oncoage CHU of Nice Nice France
| |
Collapse
|
125
|
Wu L, Fidan K, Um JY, Ahn KS. Telomerase: Key regulator of inflammation and cancer. Pharmacol Res 2020; 155:104726. [PMID: 32109579 DOI: 10.1016/j.phrs.2020.104726] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 12/11/2022]
Abstract
The telomerase holoenzyme, which has a highly conserved role in maintaining telomere length, has long been regarded as a high-profile target in cancer therapy due to the high dependency of the majority of cancer cells on constitutive and elevated telomerase activity for sustained proliferation and immortality. In this review, we present the salient findings in the telomerase field with special focus on the association of telomerase with inflammation and cancer. The elucidation of extra-telomeric roles of telomerase in inflammation, reactive oxygen species (ROS) generation, and cancer development further complicated the design of anti-telomerase therapy. Of note, the discovery of the unique mechanism that underlies reactivation of the dormant telomerase reverse transcriptase TERT promoter in somatic cells not only enhanced our understanding of the critical role of TERT in carcinogenesis but also opens up new intervention ideas that enable the differential targeting of cancer cells only. Despite significant effort invested in developing telomerase-targeted therapeutics, devising efficacious cancer-specific telomerase/TERT inhibitors remains an uphill task. The latest discoveries of the telomere-independent functionalities of telomerase in inflammation and cancer can help illuminate the path of developing specific anti-telomerase/TERT therapeutics against cancer cells.
Collapse
Affiliation(s)
- Lele Wu
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore 138673, Singapore
| | - Kerem Fidan
- 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
| | - Jae-Young Um
- College of Korean Medicine, Kyung Hee University, #47, Kyungheedae-gil, Dongdaemoon-gu, Seoul 130-701, Republic of Korea
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, #47, Kyungheedae-gil, Dongdaemoon-gu, Seoul 130-701, Republic of Korea.
| |
Collapse
|
126
|
Qin H, Yan Z, Du Z, Ren J, Qu X. Developing Enzyme‐Responsive Nanomedicine for Inhibition of hTERT Mitochondrial Translocation. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.201900203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Hongshuang Qin
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource UtilizationChangchun Institute of Applied ChemistryChinese Academy of Sciences Changchun Jilin 130022 China
| | - Zhengqing Yan
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource UtilizationChangchun Institute of Applied ChemistryChinese Academy of Sciences Changchun Jilin 130022 China
- University of Chinese Academy of Sciences Beijing 100039 China
| | - Zhi Du
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource UtilizationChangchun Institute of Applied ChemistryChinese Academy of Sciences Changchun Jilin 130022 China
- University of Chinese Academy of Sciences Beijing 100039 China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource UtilizationChangchun Institute of Applied ChemistryChinese Academy of Sciences Changchun Jilin 130022 China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource UtilizationChangchun Institute of Applied ChemistryChinese Academy of Sciences Changchun Jilin 130022 China
| |
Collapse
|
127
|
Victorelli S, Passos JF. Telomeres: beacons of autocrine and paracrine DNA damage during skin aging. Cell Cycle 2020; 19:532-540. [PMID: 32065062 DOI: 10.1080/15384101.2020.1728016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Cellular senescence is an irreversible cell cycle arrest, which can be triggered by a number of stressors, including telomere damage. Among many other phenotypic changes, senescence is accompanied by increased secretion of pro-inflammatory molecules, also known as the senescence-associated secretory phenotype (SASP). It is thought that accumulation of senescent cells contributes to age-associated tissue dysfunction partly by inducing senescence in neighboring cells through mechanisms involving SASP factors. Here, we will review evidence suggesting that telomeres can become dysfunctional irrespectively of shortening, and that this may be a mechanism-driving senescence in post-mitotic or slow dividing cells. Furthermore, we review recent evidence that supports that senescent melanocytes induce paracrine telomere damage during skin aging, which may be the mechanism responsible for propagation of senescent cells. We propose that telomeres are sensors of imbalances in the cellular milieu and act as beacons of stress, contributing to autocrine and paracrine senescence.
Collapse
Affiliation(s)
- Stella Victorelli
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, USA
| | - João F Passos
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, USA.,Institute for Cell and Molecular Biosciences, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
128
|
Smith-Sonneborn J. Telomerase Biology Associations Offer Keys to Cancer and Aging Therapeutics. Curr Aging Sci 2020; 13:11-21. [PMID: 31544708 PMCID: PMC7403649 DOI: 10.2174/1874609812666190620124324] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/07/2019] [Accepted: 05/24/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Although telomerase has potential for age-related disease intervention, the overexpression of telomerase in about 90% of cancers, and in HIV virus reservoirs, cautions against se in anti-aging telomerase therapeutics. While multiple reviews document the canonical function of telomerase for maintenance of telomeres, as well as an increasing numbers of reviews that reveal new non-canonical functions of telomerase, there was no systematic review that focuses on the array of associates of the subunit of Telomerase Reverse transcriptase protein (TERT) as pieces of the puzzle to assemble a picture of the how specific TERT complexes uniquely impact aging and age-related diseases and more can be expected. METHODS A structured search of bibliographic data on TERT complexes was undertaken using databases from the National Center for Biotechnology Information Pubmed with extensive access to biomedical and genomic information in order to obtain a unique documented and cited overview of TERT complexes that may uniquely impact aging and age-related diseases. RESULTS The TERT associations include proper folding, intracellular TERT transport, metabolism, mitochondrial ROS (Reactive Oxygen Species) regulation, inflammation, cell division, cell death, and gene expression, in addition to the well-known telomere maintenance. While increase of cell cycle inhibitors promote aging, in cancer, the cell cycle check-point regulators are ambushed in favor of cell proliferation, while cytoplasmic TERT protects a cell cycle inhibitor in oxidative stress. The oncogene cMyc regulates gene expression for overexpression of TERT, and reduction of cell cycle inhibitors-the perfect storm for cancer promotion. TERT binds with the oncogene RMRP RNA, and TERT-RMRP function can regulate levels of that oncogene RNA, and TERT in a TBN complex can regulate heterochromatin. Telomerase benefit and novel function in neurology and cardiology studies open new anti- aging hope. GV1001, a 16 amino acid peptide of TERT that associates with Heat Shock Proteins (HSP's), bypasses the cell membrane with remarkable anti disease potential. CONCLUSIONS TERT "associates" are anti-cancer targets for downregulation, but upregulation in antiaging therapy. The overview revealed that unique TERT associations that impact all seven pillars of aging identified by the Trans-NIH Geroscience Initiative that influence aging and urge research for appropriate targeted telomerase supplements/ stimulation, and inclusion in National Institute on Aging Intervention Testing Program. The preference for use of available "smart drugs", targeted to only cancer, not off-target anti- aging telomerase is implied by the multiplicity of TERT associates functions.
Collapse
Affiliation(s)
- Joan Smith-Sonneborn
- Department Zoology and Physiology, University of Wyoming, Laramie, Wyoming, WY, USA
| |
Collapse
|
129
|
Hematopoietic cellular aging is not accelerated during the first 2 years of life in children born preterm. Pediatr Res 2020; 88:903-909. [PMID: 32170191 PMCID: PMC7086539 DOI: 10.1038/s41390-020-0833-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/10/2020] [Accepted: 02/19/2020] [Indexed: 11/08/2022]
Abstract
BACKGROUND Prematurity in itself and exposure to neonatal intensive care triggers inflammatory processes and oxidative stress, leading to risk for disease later in life. The effects on cellular aging processes are incompletely understood. METHODS Relative telomere length (RTL) was measured by qPCR in this longitudinal cohort study with blood samples taken at birth and at 2 years of age from 60 children (16 preterm and 44 term). Viral respiratory infections the first year were evaluated. Epigenetic biological DNA methylation (DNAm) age was predicted based on methylation array data in 23 children (11 preterm and 12 term). RTL change/year and DNAm age change/year was compared in preterm and term during the 2 first years of life. RESULTS Preterm infants had longer telomeres than term born at birth and at 2 years of age, but no difference in telomere attrition rate could be detected. Predicted epigenetic DNAm age was younger in preterm infants, but rate of DNAm aging was similar in both groups. CONCLUSIONS Despite early exposure to risk factors for accelerated cellular aging, children born preterm exhibited preserved telomeres. Stress during the neonatal intensive care period did not reflect accelerated epigenetic DNAm aging. Early-life aging was not explained by preterm birth. IMPACT Preterm birth is associated with elevated disease risk later in life. Preterm children often suffer from inflammation early in life. Stress-related telomere erosion during neonatal intensive care has been proposed. Inflammation-accelerated biological aging in preterm is unknown. We find no accelerated aging due to prematurity or infections during the first 2 years of life.
Collapse
|
130
|
Lundberg M, Millischer V, Backlund L, Martinsson L, Stenvinkel P, Sellgren CM, Lavebratt C, Schalling M. Lithium and the Interplay Between Telomeres and Mitochondria in Bipolar Disorder. Front Psychiatry 2020; 11:586083. [PMID: 33132941 PMCID: PMC7553080 DOI: 10.3389/fpsyt.2020.586083] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/31/2020] [Indexed: 01/06/2023] Open
Abstract
Bipolar disorder is a severe psychiatric disorder which affects more than 1% of the world's population and is a leading cause of disability among young people. For the past 50 years, lithium has been the drug of choice for maintenance treatment of bipolar disorder due to its potent ability to prevent both manic and depressive episodes as well as suicide. However, though lithium has been associated with a multitude of effects within different cellular pathways and biological systems, its specific mechanism of action in stabilizing mood remains largely elusive. Mitochondrial dysfunction and telomere shortening have been implicated in both the pathophysiology of bipolar disorder and as targets of lithium treatment. Interestingly, it has in recent years become clear that these phenomena are intimately linked, partly through reactive oxygen species signaling and the subcellular translocation and non-canonical actions of telomerase reverse transcriptase. In this review, we integrate the current understanding of mitochondrial dysfunction, oxidative stress and telomere shortening in bipolar disorder with documented effects of lithium. Moreover, we propose that lithium's mechanism of action is intimately connected with the interdependent regulation of mitochondrial bioenergetics and telomere maintenance.
Collapse
Affiliation(s)
- Martin Lundberg
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Vincent Millischer
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Lena Backlund
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Lina Martinsson
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Healthcare Services, Region Stockholm, Stockholm, Sweden
| | - Peter Stenvinkel
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Carl M Sellgren
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Healthcare Services, Region Stockholm, Stockholm, Sweden.,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Schalling
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
131
|
Cell cycle-dependent and -independent telomere shortening accompanies murine brain aging. Aging (Albany NY) 2019; 10:3397-3420. [PMID: 30472697 PMCID: PMC6286833 DOI: 10.18632/aging.101655] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 11/15/2018] [Indexed: 12/16/2022]
Abstract
Replication-based telomere shortening during lifetime is species- and tissue-specific, however, its impact on healthy aging is unclear. In particular, the contribution of telomere truncation to the aging process of the CNS, where replicative senescence alone fails to explain organ aging due to low to absent mitotic activity of intrinsic populations, is undefined. Here, we assessed changes in relative telomere length in non-replicative and replicative neural brain populations and telomerase activity as a function of aging in C57BL/6 mice. Telomeres in neural cells and sub-selected neurons shortened with aging in a cell cycle-dependent and -independent manner, with preponderance in replicative moieties, implying that proliferation accelerates, but is not prerequisite for telomere shortening. Consistent with this telomere erosion, telomerase activity and nuclear TERT protein were not induced with aging. Knockdown of the Rela subunit of NF-κB, which controls both telomerase enzyme and subcellular TERT protein allocation, did also not influence telomerase activity or telomere length, in spite of its naive up-regulation selectively under aging conditions. We conclude that telomere instability is intrinsic to physiological brain aging beyond cell replication, and appears to occur independently of a functional interplay with NF-κB, but rather as a failure to induce or relocate telomerase.
Collapse
|
132
|
Månsson KNT, Lindqvist D, Yang LL, Svanborg C, Isung J, Nilsonne G, Bergman-Nordgren L, El Alaoui S, Hedman-Lagerlöf E, Kraepelien M, Högström J, Andersson G, Boraxbekk CJ, Fischer H, Lavebratt C, Wolkowitz OM, Furmark T. Improvement in indices of cellular protection after psychological treatment for social anxiety disorder. Transl Psychiatry 2019; 9:340. [PMID: 31852887 PMCID: PMC6920472 DOI: 10.1038/s41398-019-0668-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 11/11/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022] Open
Abstract
Telomere attrition is a hallmark of cellular aging and shorter telomeres have been reported in mood and anxiety disorders. Telomere shortening is counteracted by the enzyme telomerase and cellular protection is also provided by the antioxidant enzyme glutathione peroxidase (GPx). Here, telomerase, GPx, and telomeres were investigated in 46 social anxiety disorder (SAD) patients in a within-subject design with repeated measures before and after cognitive behavioral therapy. Treatment outcome was assessed by the Liebowitz Social Anxiety Scale (self-report), administered three times before treatment to control for time and regression artifacts, and posttreatment. Venipunctures were performed twice before treatment, separated by 9 weeks, and once posttreatment. Telomerase activity and telomere length were measured in peripheral blood mononuclear cells and GPx activity in plasma. All patients contributed with complete data. Results showed that social anxiety symptom severity was significantly reduced from pretreatment to posttreatment (Cohen's d = 1.46). There were no significant alterations in telomeres or cellular protection markers before treatment onset. Telomere length and telomerase activity did not change significantly after treatment, but an increase in telomerase over treatment was associated with reduced social anxiety. Also, lower pretreatment telomerase activity predicted subsequent symptom improvement. GPx activity increased significantly during treatment, and increases were significantly associated with symptom improvement. The relationships between symptom improvement and putative protective enzymes remained significant also after controlling for body mass index, sex, duration of SAD, smoking, concurrent psychotropic medication, and the proportion of lymphocytes to monocytes. Thus, indices of cellular protection may be involved in the therapeutic mechanisms of psychological treatment for anxiety.
Collapse
Affiliation(s)
- Kristoffer N. T. Månsson
- 0000 0004 1937 0626grid.4714.6Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden ,0000 0004 1936 9377grid.10548.38Department of Psychology, Stockholm University, Stockholm, Sweden ,0000 0004 1936 9457grid.8993.bDepartment of Psychology, Uppsala University, Uppsala, Sweden
| | - Daniel Lindqvist
- 0000 0001 0930 2361grid.4514.4Department of Clinical Sciences Lund, Psychiatry, , Lund University, Lund, Sweden
| | - Liu L. Yang
- 0000 0004 1937 0626grid.4714.6Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden ,0000 0000 9241 5705grid.24381.3cCenter for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Cecilia Svanborg
- 0000 0004 1937 0626grid.4714.6Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Josef Isung
- 0000 0004 1937 0626grid.4714.6Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Gustav Nilsonne
- 0000 0004 1937 0626grid.4714.6Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden ,0000 0004 1936 9377grid.10548.38Stress Research Institute, Stockholm University, Stockholm, Sweden
| | - Lise Bergman-Nordgren
- 0000 0004 1937 0626grid.4714.6Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Samir El Alaoui
- 0000 0004 1937 0626grid.4714.6Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Erik Hedman-Lagerlöf
- 0000 0004 1937 0626grid.4714.6Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Martin Kraepelien
- 0000 0004 1937 0626grid.4714.6Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Jens Högström
- 0000 0004 1937 0626grid.4714.6Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden
| | - Gerhard Andersson
- 0000 0004 1937 0626grid.4714.6Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet, & Stockholm Health Care Services, Region Stockholm, Stockholm, Sweden ,0000 0001 2162 9922grid.5640.7Department of Behavioural Sciences and Learning, Linköping University, Linköping, Sweden
| | - Carl-Johan Boraxbekk
- 0000 0001 1034 3451grid.12650.30Centre for Demographic and Ageing Research, Umeå University, Umeå, Sweden ,0000 0004 0646 7373grid.4973.9Center for Magnetic Resonance (DRCMR), Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital, Hvidovre, Denmark
| | - Håkan Fischer
- 0000 0004 1936 9377grid.10548.38Department of Psychology, Stockholm University, Stockholm, Sweden
| | - Catharina Lavebratt
- 0000 0004 1937 0626grid.4714.6Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden ,0000 0000 9241 5705grid.24381.3cCenter for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Owen M. Wolkowitz
- 0000 0001 2297 6811grid.266102.1Department of Psychiatry, University of California, San Francisco, CA USA
| | - Tomas Furmark
- 0000 0004 1936 9457grid.8993.bDepartment of Psychology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
133
|
Martens A, Schmid B, Akintola O, Saretzki G. Telomerase Does Not Improve DNA Repair in Mitochondria upon Stress but Increases MnSOD Protein under Serum-Free Conditions. Int J Mol Sci 2019; 21:ijms21010027. [PMID: 31861522 PMCID: PMC6981674 DOI: 10.3390/ijms21010027] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/12/2019] [Accepted: 12/17/2019] [Indexed: 02/08/2023] Open
Abstract
Telomerase is best known for its function in maintaining telomeres but has also multiple additional, non-canonical functions. One of these functions is the decrease of oxidative stress and DNA damage due to localisation of the telomerase protein TERT into mitochondria under oxidative stress. However, the exact molecular mechanisms behind these protective effects are still not well understood. We had shown previously that overexpression of human telomerase reverse transcriptase (hTERT) in human fibroblasts results in a decrease of mitochondrial DNA (mtDNA) damage after oxidative stress. MtDNA damage caused by oxidative stress is removed via the base excision repair (BER) pathway. Therefore we aimed to analyse whether telomerase is able to improve this pathway. We applied different types of DNA damaging agents such as irradiation, arsenite treatment (NaAsO2) and treatment with hydrogen peroxide (H2O2). Using a PCR-based assay to evaluate mtDNA damage, we demonstrate that overexpression of hTERT in MRC-5 fibroblasts protects mtDNA from H2O2 and NaAsO2 induced damage, compared with their isogenic telomerase-negative counterparts. However, overexpression of hTERT did not seem to increase repair of mtDNA after oxidative stress, but promoted increased levels of manganese superoxide dismutase (MnSOD) and forkhead-box-protein O3 (FoxO3a) proteins during incubation in serum free medium as well as under oxidative stress, while no differences were found in protein levels of catalase. Together, our results suggest that rather than interfering with mitochondrial DNA repair mechanisms, such as BER, telomerase seems to increase antioxidant defence mechanisms to prevent mtDNA damage and to increase cellular resistance to oxidative stress. However, the result has to be reproduced in additional cellular systems in order to generalise our findings.
Collapse
Affiliation(s)
- Alexander Martens
- The Ageing Biology Centre, Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Bianca Schmid
- The Ageing Biology Centre, Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Olasubomi Akintola
- The Ageing Biology Centre, Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Gabriele Saretzki
- The Ageing Biology Centre, Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| |
Collapse
|
134
|
Ségal-Bendirdjian E, Geli V. Non-canonical Roles of Telomerase: Unraveling the Imbroglio. Front Cell Dev Biol 2019; 7:332. [PMID: 31911897 PMCID: PMC6914764 DOI: 10.3389/fcell.2019.00332] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022] Open
Abstract
Telomerase plays a critical role in stem cell function and tissue regeneration that depends on its ability to elongate telomeres. For nearly two decades, it turned out that TERT regulates a broad spectrum of functions including signal transduction, gene expression regulation, and protection against oxidative damage that are independent of its telomere elongation activity. These conclusions that were mainly obtained in cell lines overexpressing telomerase were further strengthened by in vivo models of ectopic expression of telomerase or models of G1 TERT knockout mice without detectable telomere dysfunction. However, the later models were questioned due to the presence of aberrantly shortened telomere in the germline of the parents TERT+/- that were used to create the G1 TERT -/- mice. The physiological relevance of the functions associated with overexpressed telomerase raised also some concerns due to artifactual situations and localizations and complications to quantify the level of TERT. Another concern with non-canonical functions of TERT was the difficulty to separate a direct TERT-related function from secondary effects. Despite these concerns, more and more evidence accumulates for non-canonical roles of telomerase that are non-obligatory extra-telomeric. Here, we review these non-canonical roles of the TERT subunit of telomerase. Also, we emphasize recent results that link TERT to mitochondria and protection to reactive oxygen species suggesting a protective role of TERT in neurons. Throughout this review, we dissect some controversies regarding the non-canonical functions of telomerase and provide some insights to explain these discrepancies. Finally, we discuss the importance of understanding these alternative functions of telomerase for the development of anticancer strategies.
Collapse
Affiliation(s)
- Evelyne Ségal-Bendirdjian
- INSERM UMR-S 1124, Team: Cellular Homeostasis, Cancer and Therapies, INSERM US36, CNRS UMS 2009, BioMedTech Facilities, Université de Paris, Paris, France
| | - Vincent Geli
- Marseille Cancer Research Center, U1068 INSERM, UMR 7258 CNRS, Aix Marseille University, Institut Paoli-Calmettes, Equipe labellisée Ligue, Marseille, France
| |
Collapse
|
135
|
Abstract
Stress exposure can leave long-term footprints within the organism, like in telomeres (TLs), protective chromosome caps that shorten during cell replication and following exposure to stressors. Short TLs are considered to indicate lower fitness prospects, but why TLs shorten under stressful conditions is not understood. Glucocorticoid hormones (GCs) increase upon stress exposure and are thought to promote TL shortening by increasing oxidative damage. However, evidence that GCs are pro-oxidants and oxidative stress is causally linked to TL attrition is mixed . Based on new biochemical findings, we propose the metabolic telomere attrition hypothesis: during times of substantially increased energy demands, TLs are shortened as part of the transition into an organismal 'emergency state', which prioritizes immediate survival functions over processes with longer-term benefits. TL attrition during energy shortages could serve multiple roles including amplified signalling of cellular energy debt to re-direct critical resources to immediately important processes. This new view of TL shortening as a strategy to resolve major energetic trade-offs can improve our understanding of TL dynamics. We suggest that TLs are master regulators of cell homeostasis and propose future research avenues to understand the interactions between energy homeostasis, metabolic regulators and TL.
Collapse
Affiliation(s)
- Stefania Casagrande
- 1 Research Group Evolutionary Physiology, Max Planck Institute for Ornithology , 82319 Seewiesen , Germany
| | - Michaela Hau
- 1 Research Group Evolutionary Physiology, Max Planck Institute for Ornithology , 82319 Seewiesen , Germany.,2 Department of Biology, University of Konstanz , D-78457 Konstanz , Germany
| |
Collapse
|
136
|
Mitochondrial Dysfunction in Aging and Cancer. J Clin Med 2019; 8:jcm8111983. [PMID: 31731601 PMCID: PMC6912717 DOI: 10.3390/jcm8111983] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/01/2019] [Accepted: 11/13/2019] [Indexed: 12/14/2022] Open
Abstract
Aging is a major risk factor for developing cancer, suggesting that these two events may represent two sides of the same coin. It is becoming clear that some mechanisms involved in the aging process are shared with tumorigenesis, through convergent or divergent pathways. Increasing evidence supports a role for mitochondrial dysfunction in promoting aging and in supporting tumorigenesis and cancer progression to a metastatic phenotype. Here, a summary of the current knowledge of three aspects of mitochondrial biology that link mitochondria to aging and cancer is presented. In particular, the focus is on mutations and changes in content of the mitochondrial genome, activation of mitochondria-to-nucleus signaling and the newly discovered mitochondria-telomere communication.
Collapse
|
137
|
Hou J, Yin W, Li P, Hu C, Zhang Y, Wang X, Wang G, Gao E, Zhang J, Wang L, Li T, Wang L, Yu Z, Yuan J. Seasonal modification of the associations of exposure to polycyclic aromatic hydrocarbons or phthalates of cellular aging. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 182:109384. [PMID: 31272023 DOI: 10.1016/j.ecoenv.2019.109384] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 06/04/2019] [Accepted: 06/24/2019] [Indexed: 05/06/2023]
Abstract
Exposure to polycyclic aromatic hydrocarbons (PAHs) and phthalates link to oxidative stress and inflammatory response, which exert cellular aging. However, modification effect of seasonal factor on the association of PAHs or phthalates exposure with relative telomere length (RTL) or mitochondrial DNA copy number (mtDNA-CN) has remained unclear. In this pilot study, 106 subjects were from an urban population (n = 1240) who lived in the two districts in Wuhan city, China. Participants completed physical examinations and provided 191 blood samples for RTL and mtDNA-CN analysis and 627 urine samples for monohydroxylated-PAHs (OH-PAHs) and phthalate metabolites measurements in the winter and summer seasons. We assessed the associations of urinary OH-PAHs or phthalates metabolites with RTL or mtDNA-CN by linear regression analysis and linear mixed-effect models. We found that urinary OH-PAHs were positively associated with mtDNA-CN at lag 2 day and 3-day moving average, but negatively related to RTL at lag 0, lag 1 and lag 2 day and 3-day moving average (p < 0.05). Urinary phthalate metabolites were negatively associated with mtDNA lag 0, lag 1 and lag 2 day and 3-day moving average, but positively related to RTL at lag 0 day (p < 0.05). Seasonal factor modified the association of urinary OH-PAHs with mtDNA-CN as well as urinary phthalate metabolites with RTL. In vitro experiment showed that under certain conditions, benzo[a]pyrene increased mtDNA-CN at 48 h and di (2-ethylhexyl) phthalate did RTL at 24 h in HepG2 cells. Seasonal variations in the metabolisms of PAHs or phthalates in human body may affect the relation of PAHs or phthalates exposure with cellular aging.
Collapse
Affiliation(s)
- Jian Hou
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China
| | - Wenjun Yin
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China
| | - Pei Li
- State Key Laboratory of Organic Geochemistry, Guangdong Key Laboratory of Environment and Resources, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou, 510640, China
| | - Chen Hu
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China
| | - Youjian Zhang
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China
| | - Xian Wang
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China
| | - Guiyang Wang
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China
| | - Erwei Gao
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China
| | - Jiafei Zhang
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China
| | - Lu Wang
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China
| | - Tian Li
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China
| | - Lin Wang
- Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China
| | - Zhiqiang Yu
- State Key Laboratory of Organic Geochemistry, Guangdong Key Laboratory of Environment and Resources, Guangzhou Institute of Geochemistry, Chinese Academy of Sciences, Guangzhou, 510640, China
| | - Jing Yuan
- Department of Occupational and Environmental Health, PR China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental Health (Incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13, Wuhan, 430030, Hubei, PR China.
| |
Collapse
|
138
|
Increased Gene Expression of RUNX2 and SOX9 in Mesenchymal Circulating Progenitors Is Associated with Autophagy during Physical Activity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8426259. [PMID: 31737174 PMCID: PMC6815530 DOI: 10.1155/2019/8426259] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/22/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022]
Abstract
Lack of physical exercise is considered an important risk factor for chronic diseases. On the contrary, physical exercise reduces the morbidity rates of obesity, diabetes, bone disease, and hypertension. In order to gain novel molecular and cellular clues, we analyzed the effects of physical exercise on differentiation of mesenchymal circulating progenitor cells (M-CPCs) obtained from runners. We also investigated autophagy and telomerase-related gene expression to evaluate the involvement of specific cellular functions in the differentiation process. We performed cellular and molecular analyses in M-CPCs, obtained by a depletion method, of 22 subjects before (PRE RUN) and after (POST RUN) a half marathon performance. In order to prove our findings, we performed also in vitro analyses by testing the effects of runners' sera on a human bone marrow-derived mesenchymal stem (hBM-MSC) cell line. PCR array analyses of PRE RUN versus POST RUN M-CPC total RNAs put in evidence several genes which appeared to be modulated by physical activity. Our results showed that physical exercise promotes differentiation. Osteogenesis-related genes as RUNX2, MSX1, and SPP1 appeared to be upregulated after the run; data showed also increased levels of BMP2 and BMP6 expressions. SOX9, COL2A1, and COMP gene enhanced expression suggested the induction of chondrocytic differentiation as well. The expression of telomerase-associated genes and of two autophagy-related genes, ATG3 and ULK1, was also affected and correlated positively with MSC differentiation. These data highlight an attractive cellular scenario, outlining the role of autophagic response to physical exercise and suggesting new insights into the benefits of physical exercise in counteracting chronic degenerative conditions.
Collapse
|
139
|
Billard P, Poncet DA. Replication Stress at Telomeric and Mitochondrial DNA: Common Origins and Consequences on Ageing. Int J Mol Sci 2019; 20:ijms20194959. [PMID: 31597307 PMCID: PMC6801922 DOI: 10.3390/ijms20194959] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/04/2019] [Accepted: 10/05/2019] [Indexed: 12/12/2022] Open
Abstract
Senescence is defined as a stress-induced durable cell cycle arrest. We herein revisit the origin of two of these stresses, namely mitochondrial metabolic compromise, associated with reactive oxygen species (ROS) production, and replicative senescence, activated by extreme telomere shortening. We discuss how replication stress-induced DNA damage of telomeric DNA (telDNA) and mitochondrial DNA (mtDNA) can be considered a common origin of senescence in vitro, with consequences on ageing in vivo. Unexpectedly, mtDNA and telDNA share common features indicative of a high degree of replicative stress, such as G-quadruplexes, D-loops, RNA:DNA heteroduplexes, epigenetic marks, or supercoiling. To avoid these stresses, both compartments use similar enzymatic strategies involving, for instance, endonucleases, topoisomerases, helicases, or primases. Surprisingly, many of these replication helpers are active at both telDNA and mtDNA (e.g., RNAse H1, FEN1, DNA2, RecQ helicases, Top2α, Top2β, TOP3A, DNMT1/3a/3b, SIRT1). In addition, specialized telomeric proteins, such as TERT (telomerase reverse transcriptase) and TERC (telomerase RNA component), or TIN2 (shelterin complex), shuttle from telomeres to mitochondria, and, by doing so, modulate mitochondrial metabolism and the production of ROS, in a feedback manner. Hence, mitochondria and telomeres use common weapons and cooperate to resist/prevent replication stresses, otherwise producing common consequences, namely senescence and ageing.
Collapse
Affiliation(s)
- Pauline Billard
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, 69008 Lyon, France.
- Institut de Biopathologie moléculaire, Centre de Bio-Pathologie Est, Groupement hospitalier Est, Hospices Civils de Lyon, 69500 Bron, France.
| | - Delphine A Poncet
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, 69008 Lyon, France.
- Institut de Biopathologie moléculaire, Centre de Bio-Pathologie Est, Groupement hospitalier Est, Hospices Civils de Lyon, 69500 Bron, France.
| |
Collapse
|
140
|
Mustofa MS, Suyatna FD, Sadikin M, Pujianto DA, Royhan A, Suwardji K. Soybean extract increases telomerase reverse transcriptase protein expression in pancreatic β-cells of diabetes mellitus-induced rats. MEDICAL JOURNAL OF INDONESIA 2019. [DOI: 10.13181/mji.v28i3.1732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
BACKGROUND A number of studies on the benefits of soybean (Glycine max (L.) Merr) in the treatment of diabetes mellitus (DM) have already been conducted; however, the effects of soybean extracts on telomerase reverse transcriptase (TERT) expression in improving telomerase activity in pancreatic cells is currently unknown. This study was aimed to evaluate the effects of soybean extracts on TERT protein expression in the pancreatic β-cells of rats with DM.
METHODS Sixty male Sprague-Dawley rats were randomly divided into six groups: (1) negative control (N); (2) DM rats induced by alloxan (DMA); (3) DM rats with glibenclamide (DMG; positive control); (4) DM rats with 1 mg/kgBW/day soybean extract (DM1E); (5) DM rats with 5 mg/kgBW/day soybean extract (DM5E); (6) DM rats with 25 mg/kgBW/day soybean extract (DM25E). The treatments were carried out over 28 days. The measured variables included fasting blood glucose (FBG) level, TERT protein expression, and the number of pancreatic β-cells.
RESULTS All parameters were measured against the diabetes control group. The FBG levels in rats DM1E, DM5E, and DM25E were significantly reduced on the 28th day (p < 0.05). TERT protein expression and the number of pancreatic β-cells (DM25E) also showed significant improvements compared to DM rats (p < 0.05).
CONCLUSIONS Soybean extracts can increase TERT protein expression in pancreatic β-cells in diabetes-induced rats.
Collapse
|
141
|
Mitochondria in the signaling pathways that control longevity and health span. Ageing Res Rev 2019; 54:100940. [PMID: 31415807 PMCID: PMC7479635 DOI: 10.1016/j.arr.2019.100940] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/09/2019] [Accepted: 08/06/2019] [Indexed: 12/26/2022]
Abstract
Genetic and pharmacological intervention studies have identified evolutionarily conserved and functionally interconnected networks of cellular energy homeostasis, nutrient-sensing, and genome damage response signaling pathways, as prominent regulators of longevity and health span in various species. Mitochondria are the primary sites of ATP production and are key players in several other important cellular processes. Mitochondrial dysfunction diminishes tissue and organ functional performance and is a commonly considered feature of the aging process. Here we review the evidence that through reciprocal and multilevel functional interactions, mitochondria are implicated in the lifespan modulation function of these pathways, which altogether constitute a highly dynamic and complex system that controls the aging process. An important characteristic of these pathways is their extensive crosstalk and apparent malleability to modification by non-invasive pharmacological, dietary, and lifestyle interventions, with promising effects on lifespan and health span in animal models and potentially also in humans.
Collapse
|
142
|
Wood EM, Young AJ. Telomere attrition predicts reduced survival in a wild social bird, but short telomeres do not. Mol Ecol 2019; 28:3669-3680. [PMID: 31332860 PMCID: PMC6772082 DOI: 10.1111/mec.15181] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 06/04/2019] [Accepted: 06/26/2019] [Indexed: 01/02/2023]
Abstract
Attempts to understand the causes of variation in senescence trajectories would benefit greatly from biomarkers that reflect the progressive declines in somatic integrity (SI) that lead to senescence. While telomere length has attracted considerable interest in this regard, sources of variation in telomere length potentially unrelated to declines in SI could, in some contexts, leave telomere attrition rates a more effective biomarker than telomere length alone. Here, we investigate whether telomere length and telomere attrition rates predict the survival of wild white‐browed sparrow‐weaver nestlings (Plocepasser mahali). Our analyses of telomere length reveal counterintuitive patterns: telomere length soon after hatching negatively predicted nestling survival to fledging, a pattern that appears to be driven by differentially high in‐nest predation of broods with longer telomeres. Telomere length did not predict survival outside this period: neither hatchling telomere length nor telomere length in the mid‐nestling period predicted survival from fledging to adulthood. Our analyses using within‐individual telomere attrition rates, by contrast, revealed the expected relationships: nestlings that experienced a higher rate of telomere attrition were less likely to survive to adulthood, regardless of their initial telomere length and independent of effects of body mass. Our findings support the growing use of telomeric traits as biomarkers of SI, but lend strength to the view that longitudinal assessments of within‐individual telomere attrition since early life may be a more effective biomarker in some contexts than telomere length alone.
Collapse
Affiliation(s)
- Emma M Wood
- Centre for Ecology & Conservation, University of Exeter, Penryn, UK
| | - Andrew J Young
- Centre for Ecology & Conservation, University of Exeter, Penryn, UK
| |
Collapse
|
143
|
González-Giraldo Y, Garzón-Benitez AV, Forero DA, Barreto GE. TERT inhibition leads to reduction of IL-6 expression induced by palmitic acid and interferes with the protective effects of tibolone in an astrocytic cell model. J Neuroendocrinol 2019; 31:e12768. [PMID: 31278797 DOI: 10.1111/jne.12768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 07/02/2019] [Accepted: 07/03/2019] [Indexed: 01/04/2023]
Abstract
Although it has been shown that telomerase has neuroprotective effects, mainly as a result of its non-canonical functions in neuronal cells, its role with respect to glial cells remains unknown. There is growing evidence indicating that telomerase plays an important role with respect to inflammation, especially in the regulation of pro-inflammatory cytokine gene expression. The present study aimed to evaluate the role of telomerase in an astrocyte cell model treated with palmitic acid (PA) and tibolone. Cell death, reactive oxygen species production and interleukin-6 expression were evaluated under telomerase inhibition with the BIBR1532 compound in T98G cells treated with tibolone and PA, using fluorometry, flow cytometry, enzyme-linked immunosorbent assays and the quantitative polymerase chain reaction. The results obtained showed that telomerase protein was increased by PA after 36 hours, alone or in combination with tibolone, and that its activity was affected by PA. Telomerase inhibition reduced interleukin-6 expression and it interfered with the protective effects of tibolone on cell death. Moreover, tibolone increased Tyr707 phosphorylation in PA-treated cells. In the present study, we provide novel findings about the regulation of telomerase by PA and tibolone. Telomerase was involved in inflammation by PA and in protective effects of tibolone. Therefore, we conclude that telomerase could play a dual role in these cells.
Collapse
Affiliation(s)
- Yeimy González-Giraldo
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá DC, Colombia
| | - Angie V Garzón-Benitez
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá DC, Colombia
| | - Diego A Forero
- Laboratory of Neuropsychiatric Genetics, Biomedical Sciences Research Group, School of Medicine, Universidad Antonio Nariño, Bogotá, Colombia
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá DC, Colombia
| |
Collapse
|
144
|
Mitochondrial Homeostasis and Cellular Senescence. Cells 2019; 8:cells8070686. [PMID: 31284597 PMCID: PMC6678662 DOI: 10.3390/cells8070686] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 07/02/2019] [Accepted: 07/05/2019] [Indexed: 01/07/2023] Open
Abstract
Cellular senescence refers to a stress response aiming to preserve cellular and, therefore, organismal homeostasis. Importantly, deregulation of mitochondrial homeostatic mechanisms, manifested as impaired mitochondrial biogenesis, metabolism and dynamics, has emerged as a hallmark of cellular senescence. On the other hand, impaired mitostasis has been suggested to induce cellular senescence. This review aims to provide an overview of homeostatic mechanisms operating within mitochondria and a comprehensive insight into the interplay between cellular senescence and mitochondrial dysfunction.
Collapse
|
145
|
Identifying Pig Mitochondrial TSS: Structure and Functional Features. Mitochondrion 2019; 49:19-24. [PMID: 31279875 DOI: 10.1016/j.mito.2019.07.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 04/12/2019] [Accepted: 07/03/2019] [Indexed: 11/23/2022]
Abstract
The transcription start sites (TSSs) of porcine mitochondrial genome were firstly identified in this study, including heavy-strand promoter 1 and 2 (HSP1 and HSP2) harbored at nt 903 and nt 1369 in H strand, respectively, and light-strand promoter (LSP) located at nt 166 in L strand. HSP1 structure and expression features were investigated by analyzing mtDNA copy number, expression of 11 nucleoplasmic genes, mtDNA methylation levels, and gene expression levels of methyl-modifying enzymes, DNMT1 and TETs. The mtDNA copy number presented large differences among 15 organs/tissues, and the largest disparity, nearly 17 times, was found between pancreas (~1890 relative copy numbers) and spleen (~110 relative copy numbers, P < .01). The expression levels of HSP1 strand in these organs/tissues presented similar trends with mtDNA copy number (P < .05), and all of 11 nucleoplasmic genes (POLG, POLRMT, TERT, TFAM, TFB1M, TFB2M, NRF-1, PPARα, ESRRA, SP1 and TUFM) detected in this study displayed significantly higher expression values in pancreas than those in spleen (P < .05). Besides, bisulfite sequencing showed that all cytosine residues in the detected region (D-loop) existed methylation with different levels, and the methylation level in spleen was significantly higher than that in pancreas (P < .05). Unlike nuclear DNA, the tested region contained four types of methylation mode (CA, CC, CT, and CG). In addition, the expression of TET1 in pancreas was significantly higher than that in spleen (P < .05). Collectively, our findings indicated that mtDNA TSSs had correlation to mtDNA copy number, expression of nucleoplasmic gene, and mtDNA methylation level.
Collapse
|
146
|
Nie J, Li J, Cheng L, Deng Y, Li Y, Yan Z, Duan L, Niu Q, Tang D. Prenatal polycyclic aromatic hydrocarbons metabolites, cord blood telomere length, and neonatal neurobehavioral development. ENVIRONMENTAL RESEARCH 2019; 174:105-113. [PMID: 31055168 DOI: 10.1016/j.envres.2019.04.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 02/27/2019] [Accepted: 04/21/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Prenatal exposure to polycyclic aromatic hydrocarbon (PAH) is a potential risk factor for child neurobehavioral development. Telomere length (TL) has important implications for health over the life course. OBJECTIVE In this study, we aimed to investigate whether prenatal urinary PAH metabolites were associated with adverse neonatal neurobehavioral development and altered cord blood TL and to explore whether the change of TL was a predictor of neonatal neurobehavioral development. METHOD We enrolled 283 nonsmoking pregnant women in Taiyuan city. Eleven PAH metabolites were measured in maternal urine samples. TL in cord blood was measured by real time quantitative polymerase chain reaction. Neonatal behavioral neurological assessment (NBNA) tests were conducted when the infants were three days old. Multiple linear regression models were used to analyze the associations of maternal urinary PAH metabolites with NBNA scores and cord blood TL, and restricted cubic spline models were further used to examine the shapes of dose-response relationships. A mediation analysis was also conducted. RESULT We observed dose-response associations of maternal urinary 2-hydroxyfluorene (2-OHFlu) and 2-hydroxyphenanthrene (2-OH Phe) with decreased active tone scores, sum of NBNA scores, and cord blood TL (P for trend<0.05). Each 1 unit increase in urinary levels of Ln (2-OH Flu) or Ln (2-OH Phe) was associated with a 0.092 or 0.135 decrease in the active tone scores and a 0.174 or 0.199 decrease in the sum of NBNA scores. Mediation analysis showed TL could explained 21.74% of the effect of sum of NBNA scores change related to prenatal exposure to 2-OH Phe (P for mediator = 0.047). CONCLUSION Our data indicates maternal urinary specific PAH metabolites are inversely associated with neonatal neurobehavioral development and cord blood TL. TL mediates the associations of 2-OH Phe with neonatal neurobehavioral development and partly explains the effect of 2-OH Phe on neonatal neurobehavioral development.
Collapse
Affiliation(s)
- Jisheng Nie
- Department of Occupational and Environmental Health, School of Public Health, Shanxi Medical University, Xinjiannan Road 56, Taiyuan, 030001, China; Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722W. 168th Street, New York, NY, 10032, USA.
| | - Jinyu Li
- Department of Occupational and Environmental Health, School of Public Health, Shanxi Medical University, Xinjiannan Road 56, Taiyuan, 030001, China.
| | - Lin Cheng
- Department of Occupational and Environmental Health, School of Public Health, Shanxi Medical University, Xinjiannan Road 56, Taiyuan, 030001, China.
| | - Yunjun Deng
- Department of Occupational and Environmental Health, School of Public Health, Shanxi Medical University, Xinjiannan Road 56, Taiyuan, 030001, China.
| | - Yanning Li
- Department of Occupational and Environmental Health, School of Public Health, Shanxi Medical University, Xinjiannan Road 56, Taiyuan, 030001, China.
| | - Zhiwei Yan
- Department of Occupational and Environmental Health, School of Public Health, Shanxi Medical University, Xinjiannan Road 56, Taiyuan, 030001, China.
| | - Lei Duan
- Department of Occupational and Environmental Health, School of Public Health, Shanxi Medical University, Xinjiannan Road 56, Taiyuan, 030001, China.
| | - Qiao Niu
- Department of Occupational and Environmental Health, School of Public Health, Shanxi Medical University, Xinjiannan Road 56, Taiyuan, 030001, China.
| | - Deliang Tang
- Department of Occupational and Environmental Health, School of Public Health, Shanxi Medical University, Xinjiannan Road 56, Taiyuan, 030001, China; Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, 722W. 168th Street, New York, NY, 10032, USA.
| |
Collapse
|
147
|
A telomerase with novel non-canonical roles: TERT controls cellular aggregation and tissue size in Dictyostelium. PLoS Genet 2019; 15:e1008188. [PMID: 31237867 PMCID: PMC6592521 DOI: 10.1371/journal.pgen.1008188] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/10/2019] [Indexed: 11/19/2022] Open
Abstract
Telomerase, particularly its main subunit, the reverse transcriptase, TERT, prevents DNA erosion during eukaryotic chromosomal replication, but also has poorly understood non-canonical functions. Here, in the model social amoeba Dictyostelium discoideum, we show that the protein encoded by tert has telomerase-like motifs, and regulates, non-canonically, important developmental processes. Expression levels of wild-type (WT) tert were biphasic, peaking at 8 and 12 h post-starvation, aligning with developmental events, such as the initiation of streaming (~7 h) and mound formation (~10 h). In tert KO mutants, however, aggregation was delayed until 16 h. Large, irregular streams formed, then broke up, forming small mounds. The mound-size defect was not induced when a KO mutant of countin (a master size-regulating gene) was treated with TERT inhibitors, but anti-countin antibodies did rescue size in the tert KO. Although, conditioned medium (CM) from countin mutants failed to rescue size in the tert KO, tert KO CM rescued the countin KO phenotype. These and additional observations indicate that TERT acts upstream of smlA/countin: (i) the observed expression levels of smlA and countin, being respectively lower and higher (than WT) in the tert KO; (ii) the levels of known size-regulation intermediates, glucose (low) and adenosine (high), in the tert mutant, and the size defect's rescue by supplemented glucose or the adenosine-antagonist, caffeine; (iii) the induction of the size defect in the WT by tert KO CM and TERT inhibitors. The tert KO's other defects (delayed aggregation, irregular streaming) were associated with changes to cAMP-regulated processes (e.g. chemotaxis, cAMP pulsing) and their regulatory factors (e.g. cAMP; acaA, carA expression). Overexpression of WT tert in the tert KO rescued these defects (and size), and restored a single cAMP signaling centre. Our results indicate that TERT acts in novel, non-canonical and upstream ways, regulating key developmental events in Dictyostelium.
Collapse
|
148
|
Bignoux MJ, Cuttler K, Otgaar TC, Ferreira E, Letsolo BT, Weiss SF. LRP::FLAG Rescues Cells from Amyloid-β-Mediated Cytotoxicity Through Increased TERT Levels and Telomerase Activity. J Alzheimers Dis 2019; 69:729-741. [DOI: 10.3233/jad-190075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Monique J. Bignoux
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Wits 2050, Johannesburg, South Africa
| | - Katelyn Cuttler
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Wits 2050, Johannesburg, South Africa
| | - Tyrone C. Otgaar
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Wits 2050, Johannesburg, South Africa
| | - Eloise Ferreira
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Wits 2050, Johannesburg, South Africa
| | - Boitelo T. Letsolo
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Wits 2050, Johannesburg, South Africa
| | - Stefan F.T. Weiss
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, Wits 2050, Johannesburg, South Africa
| |
Collapse
|
149
|
Abstract
Telomeres are specialised structures at the end of linear chromosomes. They consist of tandem repeats of the hexanucleotide sequence TTAGGG, as well as a protein complex called shelterin. Together, they form a protective loop structure against chromosome fusion and degradation. Shortening or damage to telomeres and opening of the loop induce an uncapped state that triggers a DNA damage response resulting in senescence or apoptosis.Average telomere length, usually measured in human blood lymphocytes, was thought to be a biomarker for ageing, survival and mortality. However, it becomes obvious that regulation of telomere length is very complex and involves multiple processes. For example, the "end replication problem" during DNA replication as well as oxidative stress are responsible for the shortening of telomeres. In contrast, telomerase activity can potentially counteract telomere shortening when it is able to access and interact with telomeres. However, while highly active during development and in cancer cells, the enzyme is down-regulated in most human somatic cells with a few exceptions such as human lymphocytes. In addition, telomeres can be transcribed, and the transcription products called TERRA are involved in telomere length regulation.Thus, telomere length and their integrity are regulated at many different levels, and we only start to understand this process under conditions of increased oxidative stress, inflammation and during diseases as well as the ageing process.This chapter aims to describe our current state of knowledge on telomeres and telomerase and their regulation in order to better understand their role for the ageing process.
Collapse
|
150
|
Gasinska A, Jaszczynski J, Rychlik U, Łuczynska E, Pogodzinski M, Palaczynski M. Prognostic Significance of Serum PSA Level and Telomerase, VEGF and GLUT-1 Protein Expression for the Biochemical Recurrence in Prostate Cancer Patients after Radical Prostatectomy. Pathol Oncol Res 2019; 26:1049-1056. [PMID: 30989489 PMCID: PMC7242245 DOI: 10.1007/s12253-019-00659-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/01/2019] [Indexed: 02/07/2023]
Abstract
The aim of the study was to evaluate prognosis for biochemical recurrence (BR) by analysing the pathological and biological characteristics of prostate cancer (PCa) after radical prostatectomy (RP). There were 130 men with clinically localized PCa in whom pretreatment serum PSA level and Ki-67, prostate specific membrane antigen (PSMA), glucose transporter-1 (GLUT-1), vascular endothelial growth factor (VEGF), microvessel density (MVD) and human telomerase reverse transcriptase (hTERT) proteins expression, based on number of immunohistochemically positive cells (labelling index), were retrospectively studied. In order to assess the prognostic significance of analysed variables in univariate and multivariate Cox analysis, patients were dichotomized based on cut-off points chosen by receiver operating characteristic (ROC) curves. There were 83 males (63.8%) at pT stage 1–2 and 47 (36.1%) at pT stage 3–4, respectively, with median (range) age of 62.8 years (49–77), and median follow-up of 78.5 months (12–148). In 42 (32.3%) men BR was found. In univariate analysis, tumour biological features: PSA ≤ 8 ng/mL (p = 0.006), Ki-67LI ≤ 12.7% (p = 0.015), VEGFLI>11.0% (p = 0.030), and hTERTLI>6.7% (p = 0.016), but not clinicopathological parameters, appeared to be positive prognosticators for BRFS. In the Cox analysis, Ki-67 lost its significance, and clinicopathological parameters appeared to be nonsignificant. The independent negative prognostic factors for BRFS were: PSA > 8.0 ng/mL, (Hazard ratio = 2.75, p = 0.003), GLUT-1 > 19.1% (HR = 2.1, p = 0.032), VEGF≤11.0% (HR = 1, p = 0.024) and hTERT≤6.7% (HR = 1, p = 0.017). High PSA level, and GLUT-1 expression and lower VEGF and nuclear hTERT expression may indicate the great role of hypoxia in BR induction in PCa.
Collapse
Affiliation(s)
- Anna Gasinska
- Department of Tumour Pathology, Oncology Center, Maria Sklodowska - Curie Institute, Cracow Branch, Garncarska 11, 31-115, Cracow, Poland.
| | - Janusz Jaszczynski
- Department of Surgery, Oncology Center, Maria Sklodowska - Curie Institute, Cracow Branch, Cracow, Poland
| | - Urszula Rychlik
- Department of Clinical Biochemistry, Oncology Center, Maria Sklodowska-Curie Institute, Cracow Branch, Cracow, Poland
| | - Elżbieta Łuczynska
- Department of Radiology, Oncology Center, Maria Sklodowska-Curie Institute, Cracow Branch, Cracow, Poland
| | - Marek Pogodzinski
- Department of Surgery, Oncology Center, Maria Sklodowska - Curie Institute, Cracow Branch, Cracow, Poland
| | - Mikolaj Palaczynski
- Department of Surgery, Oncology Center, Maria Sklodowska - Curie Institute, Cracow Branch, Cracow, Poland
| |
Collapse
|