51
|
Tsoukalas D, Fragkiadaki P, Docea AO, Alegakis AK, Sarandi E, Thanasoula M, Spandidos DA, Tsatsakis A, Razgonova MP, Calina D. Discovery of potent telomerase activators: Unfolding new therapeutic and anti-aging perspectives. Mol Med Rep 2019; 20:3701-3708. [PMID: 31485647 PMCID: PMC6755196 DOI: 10.3892/mmr.2019.10614] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022] Open
Abstract
Telomere length, a marker of cellular aging, decreases with age and it has been associated with aging‑related diseases. Environmental factors, including diet and lifestyle factors, affect the rate of telomere shortening which can be reversed by telomerase. Telomerase activation by natural molecules has been suggested to be an anti‑aging modulator that can play a role in the treatment of aging‑related diseases. This study aimed to investigate the effect of natural compounds on telomerase activity in human peripheral blood mononuclear cells (PBMCs). The tested compounds included Centella asiatica extract formulation (08AGTLF), Astragalus extract formulation (Nutrient 4), TA‑65 (containing Astragalus membranaceus extract), oleanolic acid (OA), maslinic acid (MA), and 3 multi‑nutrient formulas (Nutrients 1, 2 and 3) at various concentrations. The mean absorbance values of telomerase activity measured following treatment with some of the above‑mentioned formulations were statistically significantly higher compared to those of the untreated cells. In particular, in order of importance with respect to telomerase activation from highest to lowest, 08AGTLF, OA, Nutrient 4, TA‑65, MA, Nutrient 3 and Nutrient 2, triggered statistically significant increase in telomerase activity compared to the untreated cells. 08AGTLF reached the highest levels of telomerase activity reported to date, at least to our knowledge, increasing telomerase activity by 8.8 folds compared to untreated cells, while Nutrient 4 and OA were also potent activators (4.3‑fold and 5.9‑fold increase, respectively). On the whole, this study indicates that the synergistic effect of nutrients and natural compounds can activate telomerase and produce more potent formulations. Human clinical studies using these formulations are required to evaluate their mode of action. This would reveal the health benefits of telomerase activation through natural molecules and would shed new light onto the treatment of aging‑related diseases.
Collapse
Affiliation(s)
- Dimitris Tsoukalas
- Metabolomic Μedicine, Health Clinics for Autoimmune and Chronic Diseases, 10674 Athens, Greece
| | - Persefoni Fragkiadaki
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Anca Oana Docea
- Department of Toxicology, University of Medicine and Pharmacy, Faculty of Pharmacy, 200349 Craiova, Romania
| | - Athanasios K Alegakis
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | - Evangelia Sarandi
- Metabolomic Μedicine, Health Clinics for Autoimmune and Chronic Diseases, 10674 Athens, Greece
| | - Maria Thanasoula
- Metabolomic Μedicine, Health Clinics for Autoimmune and Chronic Diseases, 10674 Athens, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, 71003 Heraklion, Greece
| | | | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy, Faculty of Pharmacy, 200349 Craiova, Romania
| |
Collapse
|
52
|
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
|
53
|
Kokubun T, Saitoh SI, Miura S, Ishida T, Takeishi Y. Telomerase Plays a Pivotal Role in Collateral Growth Under Ischemia by Suppressing Age-Induced Oxidative Stress, Expression of p53, and Pro-Apoptotic Proteins. Int Heart J 2019; 60:736-745. [PMID: 31105157 DOI: 10.1536/ihj.18-564] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Aging is not only a major risk factor for impaired collateral growth under ischemia but also shortens the telomere length, which is regulated by telomerase. We examined the role of telomerase activity during impaired collateral growth during aging in ischemic skeletal muscle. Unilateral hind limb ischemia was generated in old, young, and old mice chronically administered a telomerase activator. In old mice, blood flow recovery and capillary density development in ischemic hind limbs were reduced compared to those in young mice, and these changes were restored to equal levels by administration of TA-65, a telomerase activator. During the early phase of ischemic muscle changes in old mice, telomerase reverse transcriptase expression and telomerase activity were both low compared to those in young mice and old mice treated with TA-65. Levels of reactive oxygen species (ROS), DNA double-strand breaks, and expression of p53, p16, and Bax/Bcl-2 were all elevated in ischemic muscles of old mice compared to those in the muscles of young mice and old mice treated with TA-65 treatment; these factors were maintained at low levels equivalent to those seen in young mice during the experiment. Expression of HIF1α/vascular endothelial growth factor (VEGF) and PGC1α were decreased in old mice compared to those in young mice and old mice treated with TA-65. Collateral growth under ischemic conditions is impaired in aged animals due to low telomerase activity, increased ROS, resultant DNA damage, and expression of tumor suppressor and pro-apoptotic proteins. These data suggest that telomerase activation enhances collateral growth and rescues ischemic tissue in old individuals.
Collapse
Affiliation(s)
- Tomoki Kokubun
- Department of Cardiovascular Medicine, Fukushima Medical University
| | - Shu-Ichi Saitoh
- Department of Cardiovascular Medicine, Ohara General Hospital
| | - Shunsuke Miura
- Department of Cardiovascular Medicine, Fukushima Medical University
| | - Takafumi Ishida
- Department of Cardiovascular Medicine, Fukushima Medical University
| | | |
Collapse
|
54
|
Libertini G, Ferrara N, Rengo G, Corbi G. Elimination of Senescent Cells: Prospects According to the Subtelomere-Telomere Theory. BIOCHEMISTRY (MOSCOW) 2019; 83:1477-1488. [PMID: 30878023 DOI: 10.1134/s0006297918120064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell senescence is an artificially reversible condition activated by various factors and characterized by replicative senescence and typical general alteration of cell functions, including extra-cellular secretion. The number of senescent cells increases with age and contributes strongly to the manifestations of aging. For these reasons, research is under way to obtain "senolytic" compounds, defined as drugs that eliminate senescent cells and therefore reduce aging-associated decay, as already shown in some experiments on animal models. This objective is analyzed in the context of the programmed aging paradigm, as described by the mechanisms of the subtelomere-telomere theory. In this regard, positive effects of the elimination of senescent cells and limits of this method are discussed. For comparison, positive effects and limits of telomerase activation are also analyzed, as well of the combined action of the two methods and the possible association of opportune gene modifications. Ethical issues associated with the use of these methods are outlined.
Collapse
Affiliation(s)
- G Libertini
- Federico II University, Department of Translational Medical Sciences, Naples, 80138, Italy.
| | - N Ferrara
- Federico II University, Department of Translational Medical Sciences, Naples, 80138, Italy
| | - G Rengo
- Federico II University, Department of Translational Medical Sciences, Naples, 80138, Italy
| | - G Corbi
- Federico II University, Department of Translational Medical Sciences, Naples, 80138, Italy
| |
Collapse
|
55
|
Hillson O, Gonzalez S, Rallis C. Prospects of Pharmacological Interventions to Organismal Aging. Biomol Concepts 2018; 9:200-215. [DOI: 10.1515/bmc-2018-0018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/04/2018] [Indexed: 12/25/2022] Open
Abstract
AbstractIntense research in the areas of cellular and organismal aging using diverse laboratory model systems has enriched our knowledge in the processes and the signalling pathways involved in normal and pathological conditions. The field finds itself in a position to take decisive steps towards clinical applications and interventions not only for targeted age-related diseases such as cardiovascular conditions and neurodegeneration but also for the modulation of health span and lifespan of a whole organism. Beyond nutritional interventions such as dietary restriction without malnutrition and various regimes of intermittent fasting, accumulating evidence provides promise for pharmacological interventions. The latter, mimic caloric or dietary restriction, tune cellular and organismal stress responses, affect the metabolism of microbiome with subsequent effects on the host or modulate repair pathways, among others. In this mini review, we summarise some of the evidence on drugs that can alter organismal lifespan and the prospects they might offer for promoting healthspan and delaying age-related diseases.
Collapse
Affiliation(s)
- Olivia Hillson
- School of Health, Sport and Bioscience, University of East London, Water Lane, E15 4LZ, London, United Kingdom
| | - Suam Gonzalez
- School of Health, Sport and Bioscience, University of East London, Water Lane, E15 4LZ, London, United Kingdom
| | - Charalampos Rallis
- School of Health, Sport and Bioscience, University of East London, Water Lane, E15 4LZ, London, United Kingdom
| |
Collapse
|
56
|
Lin H, Jiang B, Chen C, Song Y, Yang M, Huang H, Chen G. Microbial transformation of the anti-aging agent cycloastragenol by Mucor racemosus. Nat Prod Res 2018; 33:3103-3108. [DOI: 10.1080/14786419.2018.1519822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Haijun Lin
- School of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Baocheng Jiang
- School of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Chen Chen
- School of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Yan Song
- School of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Min Yang
- School of Pharmacy, Nantong University, Nantong 226001, PR China
| | - Huilian Huang
- Key Laboratory of Modern Preparation of TCM, Jiangxi University of Traditional Chinese Medicine, ministry of education, Nanchang 330004, PR China
| | - Guangtong Chen
- School of Pharmacy, Nantong University, Nantong 226001, PR China
| |
Collapse
|
57
|
Lopes AC, Oliveira PF, Sousa M. Shedding light into the relevance of telomeres in human reproduction and male factor infertility†. Biol Reprod 2018; 100:318-330. [DOI: 10.1093/biolre/ioy215] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 09/05/2018] [Accepted: 10/01/2018] [Indexed: 12/20/2022] Open
Affiliation(s)
- Ana Catarina Lopes
- Laboratory of Cell Biology, Department of Microscopy, and Multidisciplinary Unit for Biomedical Research (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- Department of Life Sciences, Faculty of Sciences and Technology, New University of Lisbon (FCT-UNL), Campus Caparica, Caparica, Portugal
| | - Pedro F Oliveira
- Laboratory of Cell Biology, Department of Microscopy, and Multidisciplinary Unit for Biomedical Research (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- Department of Genetics, Faculty of Medicine, University of Porto, Porto, Portugal
- i3S- Institute of Research and Innovation in Health, University of Porto, Porto, Portugal
| | - Mário Sousa
- Laboratory of Cell Biology, Department of Microscopy, and Multidisciplinary Unit for Biomedical Research (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Porto, Portugal
- Centre for Reproductive Genetics Professor Alberto Barros, Porto, Portugal
| |
Collapse
|
58
|
Yu Y, Zhou L, Yang Y, Liu Y. Cycloastragenol: An exciting novel candidate for age-associated diseases. Exp Ther Med 2018; 16:2175-2182. [PMID: 30186456 PMCID: PMC6122403 DOI: 10.3892/etm.2018.6501] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 06/25/2018] [Indexed: 12/26/2022] Open
Abstract
Cycloastragenol (CAG) is a triterpenoid saponin compound and a hydrolysis product of the main active ingredient in Astragalus membranaceus (Fisch.) Bunge. An increasing body of evidence has indicated that CAG has a wide spectrum of pharmacological functions, which are attracting attention in the research community. The aim of the present review paper was to review and elucidate the advanced study of CAG. The focus was on advanced studies of CAG in English and Chinese databases; the literature was collected and reviewed to summarize the latest efficacy, pharmacokinetics and adverse reactions of CAG. Extensive pharmacological effects have been attributed to CAG, including telomerase activation, telomere elongation, anti-inflammatory and anti-oxidative properties; CAG has also been reported to improve lipid metabolism. Clinical research has demonstrated that CAG activates telomerase in humans and ameliorates various biomarkers. CAG is absorbed through the intestinal epithelium via passive diffusion and undergoes first-pass hepatic metabolism. Within a certain dose range, oral CAG is relatively safe; however, underlying mechanisms associated with CAG are not clear, and thus, we should be aware of potential adverse reactions associated with CAG. According to existing studies and clinical trials, CAG is safe and has broad application prospects. However, further studies are required to fully understand its efficacy and potential adverse reactions, and to ensure the proper use of CAG is applied to treat diseases clinically.
Collapse
Affiliation(s)
- Yongjie Yu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Limin Zhou
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Yajun Yang
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| | - Yuyu Liu
- Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China.,Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, Guangdong 524023, P.R. China
| |
Collapse
|
59
|
Stress, cell senescence and organismal ageing. Mech Ageing Dev 2018; 170:2-9. [DOI: 10.1016/j.mad.2017.07.001] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/17/2017] [Accepted: 07/04/2017] [Indexed: 12/25/2022]
|
60
|
Vidacek NŠ, Nanic L, Ravlic S, Sopta M, Geric M, Gajski G, Garaj-Vrhovac V, Rubelj I. Telomeres, Nutrition, and Longevity: Can We Really Navigate Our Aging? J Gerontol A Biol Sci Med Sci 2017; 73:39-47. [PMID: 28510637 DOI: 10.1093/gerona/glx082] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 04/25/2017] [Indexed: 01/08/2023] Open
Abstract
Telomeres are dynamic chromosome-end structures that serve as guardians of genome stability. They are known to be one of the major determinants of aging and longevity in higher mammals. Studies have demonstrated a direct correlation between telomere length and life expectancy, stress, DNA damage, and onset of aging-related diseases. This review discusses the most important factors that influence our telomeres. Various genetic and environmental factors such as diet, physical activity, obesity, and stress are known to influence health and longevity as well as telomere dynamics. Individuals currently have the opportunity to modulate the dynamics of their aging and health span, monitor these processes, and even make future projections by following their telomere dynamics. As telomeres react to positive as well as negative health factors, we should be able to directly influence our telomere metabolism, slow their deterioration, and diminish our aging and perhaps extend our life and health span.
Collapse
Affiliation(s)
| | - Lucia Nanic
- Department of Molecular Biology, Ruder Boškovic Institute, Zagreb, Croatia
| | - Sanda Ravlic
- Department of Molecular Biology, Ruder Boškovic Institute, Zagreb, Croatia
| | - Mary Sopta
- Department of Molecular Biology, Ruder Boškovic Institute, Zagreb, Croatia
| | - Marko Geric
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Goran Gajski
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Vera Garaj-Vrhovac
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Zagreb, Croatia
| | - Ivica Rubelj
- Department of Molecular Biology, Ruder Boškovic Institute, Zagreb, Croatia
| |
Collapse
|
61
|
Abstract
AbstractRecently, short telomeres have become a widely accepted cellular hallmark of aging. Telomere lengths in a single cell are heterogeneous and it is believed that the shortest telomere in a cell drives the induction of senescence. Hence, measuring the shortest telomere lengths (not just average) can provide more information about aging, cancer progression and telomere related diseases.Chronic exposure to DNA damaging agents, oxidative stress, inflammation, smoking, alcohol, exposure to acute and chronic stress promote telomere shortening and earlier onset of cell aging. Healthy life style including Mediterranean diet, moderate exercise, managing stress (breathing, meditation, yoga), spending time with loved ones and lots of laughter will help us to keep our telomeres long and safe.
Collapse
|
62
|
Derevyanko A, Whittemore K, Schneider RP, Jiménez V, Bosch F, Blasco MA. Gene therapy with the TRF1 telomere gene rescues decreased TRF1 levels with aging and prolongs mouse health span. Aging Cell 2017; 16:1353-1368. [PMID: 28944611 PMCID: PMC5676056 DOI: 10.1111/acel.12677] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2017] [Indexed: 12/18/2022] Open
Abstract
The shelterin complex protects telomeres by preventing them from being degraded and recognized as double‐strand DNA breaks. TRF1 is an essential component of shelterin, with important roles in telomere protection and telomere replication. We previously showed that TRF1 deficiency in the context of different mouse tissues leads to loss of tissue homeostasis owing to impaired stem cell function. Here, we show that TRF1 levels decrease during organismal aging both in mice and in humans. We further show that increasing TRF1 expression in both adult (1‐year‐old) and old (2‐year‐old) mice using gene therapy can delay age‐associated pathologies. To this end, we used the nonintegrative adeno‐associated serotype 9 vector (AAV9), which transduces the majority of mouse tissues allowing for moderate and transient TRF1 overexpression. AAV9‐TRF1 gene therapy significantly prevented age‐related decline in neuromuscular function, glucose tolerance, cognitive function, maintenance of subcutaneous fat, and chronic anemia. Interestingly, although AAV9‐TRF1 treatment did not significantly affect median telomere length, we found a lower abundance of short telomeres and of telomere‐associated DNA damage in some tissues. Together, these findings suggest that rescuing naturally decreased TRF1 levels during mouse aging using AAV9‐TRF1 gene therapy results in an improved mouse health span.
Collapse
Affiliation(s)
- Aksinya Derevyanko
- Telomeres and Telomerase Group Molecular Oncology Program Spanish National Cancer Centre (CNIO) Melchor Fernández Almagro 3 Madrid E‐28029 Spain
| | - Kurt Whittemore
- Telomeres and Telomerase Group Molecular Oncology Program Spanish National Cancer Centre (CNIO) Melchor Fernández Almagro 3 Madrid E‐28029 Spain
| | - Ralph P. Schneider
- Telomeres and Telomerase Group Molecular Oncology Program Spanish National Cancer Centre (CNIO) Melchor Fernández Almagro 3 Madrid E‐28029 Spain
| | - Verónica Jiménez
- Center of Animal Biotechnology and Gene Therapy Department of Biochemistry and Molecular Biology School of Veterinary Medicine Universitat Autònoma de Barcelona Bellaterra 08193 Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) Madrid Spain
| | - Fàtima Bosch
- Center of Animal Biotechnology and Gene Therapy Department of Biochemistry and Molecular Biology School of Veterinary Medicine Universitat Autònoma de Barcelona Bellaterra 08193 Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM) Madrid Spain
| | - Maria A. Blasco
- Telomeres and Telomerase Group Molecular Oncology Program Spanish National Cancer Centre (CNIO) Melchor Fernández Almagro 3 Madrid E‐28029 Spain
| |
Collapse
|
63
|
Liu P, Zhao H, Luo Y. Anti-Aging Implications of Astragalus Membranaceus (Huangqi): A Well-Known Chinese Tonic. Aging Dis 2017; 8:868-886. [PMID: 29344421 PMCID: PMC5758356 DOI: 10.14336/ad.2017.0816] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/16/2017] [Indexed: 12/20/2022] Open
Abstract
Owing to a dramatic increase in average life expectancy and the Family Planning program of the 1970s - 1990s, China is rapidly becoming an aging society. Therefore, the investigation of healthspan-extending drugs becomes more urgent. Astragalus membranaceus (Huangqi) is a major medicinal herb that has been commonly used in many herbal formulations in the practice of traditional Chinese medicine (TCM) to treat a wide variety of diseases and body disorders, or marketed as life-prolonging extracts for human use in China, for more than 2000 years. The major components of Astragalus membranaceus are polysaccharides, flavonoids, and saponins. Pharmacological research indicates that the extract component of Astragalus membranaceus can increase telomerase activity, and has antioxidant, anti-inflammatory, immunoregulatory, anticancer, hypolipidemic, antihyperglycemic, hepatoprotective, expectorant, and diuretic effects. A proprietary extract of the dried root of Astragalus membranaceus, called TA-65, was associated with a significant age-reversal effect in the immune system. Our review focuses on the function and the underlying mechanisms of Astragalus membranaceus in lifespan extension, anti-vascular aging, anti-brain aging, and anti-cancer effects, based on experimental and clinical studies.
Collapse
Affiliation(s)
- Ping Liu
- 1Cerebrovascular Diseases Research Institute, and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Haiping Zhao
- 1Cerebrovascular Diseases Research Institute, and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Yumin Luo
- 1Cerebrovascular Diseases Research Institute, and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China.,2Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,3Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| |
Collapse
|
64
|
Hastings WJ, Shalev I, Belsky DW. Translating Measures of Biological Aging to Test Effectiveness of Geroprotective Interventions: What Can We Learn from Research on Telomeres? Front Genet 2017; 8:164. [PMID: 29213278 PMCID: PMC5702647 DOI: 10.3389/fgene.2017.00164] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 10/16/2017] [Indexed: 11/13/2022] Open
Abstract
Intervention studies in animals suggest molecular changes underlying age-related disease and disability can be slowed or reversed. To speed translation of these so-called "geroprotective" therapies to prevent age-related disease and disability in humans, biomarkers are needed that can track changes in the rate of human aging over the course of intervention trials. Algorithm methods that measure biological processes of aging from combinations of DNA methylation marks or clinical biomarkers show promise. To identify next steps for establishing utility of these algorithm-based measures of biological aging for geroprotector trials, we considered the history a candidate biomarker of aging that has received substantial research attention, telomere length. Although telomere length possesses compelling biology to recommend it as a biomarker of aging, mixed research findings have impeded clinical and epidemiologic translation. Strengths of telomeres that should be established for algorithm biomarkers of aging are correlation with chronological age across the lifespan, prediction of disease, disability, and early death, and responsiveness to risk and protective exposures. Key challenges in telomere research that algorithm biomarkers of aging must address are measurement precision and reliability, establishing links between longitudinal rates of change across repeated measurements and aging outcomes, and clarity over whether the biomarker is a causal mechanism of aging. These strengths and challenges suggest a research agenda to advance translation of algorithm-based aging biomarkers: establish validity in young-adult and midlife individuals; test responsiveness to exposures that shorten or extend healthy lifespan; and conduct repeated-measures longitudinal studies to test differential rates of change.
Collapse
Affiliation(s)
- Waylon J Hastings
- Department of Biobehavioral Health, Pennsylvania State University, State College, PA, United States
| | - Idan Shalev
- Department of Biobehavioral Health, Pennsylvania State University, State College, PA, United States
| | - Daniel W Belsky
- Department of Population Health Sciences, Duke University School of Medicine, Durham, NC, United States.,Center for the Study of Aging and Human Development, Duke University, Durham, NC, United States
| |
Collapse
|
65
|
Affiliation(s)
- Komal Saraswat
- Department of Biochemistry, University of Allahabad, Allahabad, India
| | | |
Collapse
|
66
|
Heiss C, Spyridopoulos I, Haendeler J. Interventions to slow cardiovascular aging: Dietary restriction, drugs and novel molecules. Exp Gerontol 2017; 109:108-118. [PMID: 28658611 DOI: 10.1016/j.exger.2017.06.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 06/16/2017] [Accepted: 06/21/2017] [Indexed: 02/05/2023]
Abstract
Cardiovascular aging is a highly dynamic process. Despite the fact that cardiovascular function and structure change with age, they can still be modulated even in aged humans. The most prominent approaches to improve age-dependent vascular changes include dietary restriction and pharmacologic agents interacting with signaling pathways implicated in this context. These include inhibition of TOR, glycolysis, and GH/IGF-1, activation of sirtuins, and AMPK, as well as modulators of inflammation, epigenetic pathways, and telomeres. Promising nutritional approaches include Mediterranean diet and novel dietary bioactives including flavanols, anthocyanins, and lignins. Many plant bioactives improve cardiovascular parameters implied in vascular healthy aging including endothelial function, arterial stiffness, blood pressure, cholesterol, and glycemic control. However, the mechanism of action of most bioactives is not established and it remains to be elucidated whether they act as dietary restriction mimetics or via other modes of action. Even more importantly, whether these interventions can slow or even reverses components of cardiovascular aging itself and can increase healthspan or longevity in humans needs to be determined.
Collapse
Affiliation(s)
- Christian Heiss
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, University Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany.
| | - Ioakim Spyridopoulos
- Institute of Genetic Medicine, Medical Faculty, Newcastle University, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK.
| | - Judith Haendeler
- Central Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, University of Duesseldorf, Moorenstr. 5, 40225 Duesseldorf, Germany; IUF-Leibniz Research Institute for Environmental Medicine, Auf'm Hennekamp 50, 40225 Duesseldorf, Germany.
| |
Collapse
|
67
|
Sun C, Jiang M, Zhang L, Yang J, Zhang G, Du B, Ren Y, Li X, Yao J. Cycloastragenol mediates activation and proliferation suppression in concanavalin A-induced mouse lymphocyte pan-activation model. Immunopharmacol Immunotoxicol 2017; 39:131-139. [DOI: 10.1080/08923973.2017.1300170] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Chenghong Sun
- Linyi Key Laboratory for Immunopharmacology and Immunotoxicology of Natural Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- State Key Laboratory of Generic Pharmaceutical Technology for Chinese Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- Center for New Drug Pharmacology, Lunan Pharmaceutical Group Corporation, Linyi, PR China
| | - Mingmin Jiang
- Linyi Key Laboratory for Immunopharmacology and Immunotoxicology of Natural Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- State Key Laboratory of Generic Pharmaceutical Technology for Chinese Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- Center for New Drug Pharmacology, Lunan Pharmaceutical Group Corporation, Linyi, PR China
| | - Li Zhang
- State Key Laboratory of Generic Pharmaceutical Technology for Chinese Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
| | - Jian Yang
- Linyi Key Laboratory for Immunopharmacology and Immunotoxicology of Natural Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- State Key Laboratory of Generic Pharmaceutical Technology for Chinese Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- Center for New Drug Pharmacology, Lunan Pharmaceutical Group Corporation, Linyi, PR China
| | - Guimin Zhang
- State Key Laboratory of Generic Pharmaceutical Technology for Chinese Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
| | - Bingyuan Du
- Linyi Key Laboratory for Immunopharmacology and Immunotoxicology of Natural Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- State Key Laboratory of Generic Pharmaceutical Technology for Chinese Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- Center for New Drug Pharmacology, Lunan Pharmaceutical Group Corporation, Linyi, PR China
| | - Yushan Ren
- Linyi Key Laboratory for Immunopharmacology and Immunotoxicology of Natural Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- State Key Laboratory of Generic Pharmaceutical Technology for Chinese Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- Center for New Drug Pharmacology, Lunan Pharmaceutical Group Corporation, Linyi, PR China
| | - Xin Li
- Linyi Key Laboratory for Immunopharmacology and Immunotoxicology of Natural Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- State Key Laboratory of Generic Pharmaceutical Technology for Chinese Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- Center for New Drug Pharmacology, Lunan Pharmaceutical Group Corporation, Linyi, PR China
| | - Jingchun Yao
- Linyi Key Laboratory for Immunopharmacology and Immunotoxicology of Natural Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- State Key Laboratory of Generic Pharmaceutical Technology for Chinese Medicine, Lunan Pharmaceutical Group Corporation, Linyi, China
- Center for New Drug Pharmacology, Lunan Pharmaceutical Group Corporation, Linyi, PR China
| |
Collapse
|
68
|
Libertini G, Ferrara N. Possible interventions to modify aging. BIOCHEMISTRY (MOSCOW) 2016; 81:1413-1428. [DOI: 10.1134/s0006297916120038] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
69
|
Therapeutic Manipulation of Ageing: Repurposing Old Dogs and Discovering New Tricks. EBioMedicine 2016; 14:24-31. [PMID: 27889480 PMCID: PMC5161440 DOI: 10.1016/j.ebiom.2016.11.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/14/2016] [Accepted: 11/14/2016] [Indexed: 01/04/2023] Open
Abstract
Ageing is a leading risk factor for many debilitating diseases. While age-related diseases have been the subject of over a century of intense investigation, until recently, physiological ageing was considered unavoidable. Pharmacological and genetic studies have since shown that ageing is a malleable process and that its abrogation can prevent its associated diseases. This review summarises a sample of the most promising efforts to deliver the products of ageing research to the clinic. Current efforts include the use of clinically approved drugs that have since been repurposed, as well as the development of novel therapeutics, to target ageing. Furthermore, ongoing research has sought reliable biomarkers of ageing that will accelerate the development of such therapeutics. Development of these technologies will improve quality of late-life and help relieve the enormous stress placed on state healthcare systems by a rapidly ageing global population. Thus, for both medical and socioeconomic reasons, it is imperative that ageing is made to yield to intervention.
Collapse
|
70
|
Vaiserman AM, Lushchak OV, Koliada AK. Anti-aging pharmacology: Promises and pitfalls. Ageing Res Rev 2016; 31:9-35. [PMID: 27524412 DOI: 10.1016/j.arr.2016.08.004] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 08/06/2016] [Accepted: 08/09/2016] [Indexed: 12/12/2022]
Abstract
Life expectancy has grown dramatically in modern times. This increase, however, is not accompanied by the same increase in healthspan. Efforts to extend healthspan through pharmacological agents targeting aging-related pathological changes are now in the spotlight of geroscience, the main idea of which is that delaying of aging is far more effective than preventing the particular chronic disorders. Currently, anti-aging pharmacology is a rapidly developing discipline. It is a preventive field of health care, as opposed to conventional medicine which focuses on treating symptoms rather than root causes of illness. A number of pharmacological agents targeting basic aging pathways (i.e., calorie restriction mimetics, autophagy inducers, senolytics etc.) are now under investigation. This review summarizes the literature related to advances, perspectives and challenges in the field of anti-aging pharmacology.
Collapse
Affiliation(s)
| | - Oleh V Lushchak
- Vasyl Stefanyk Precarpathian National University, Ivano-Frankivsk, Ukraine
| | | |
Collapse
|
71
|
da Costa JP, Vitorino R, Silva GM, Vogel C, Duarte AC, Rocha-Santos T. A synopsis on aging-Theories, mechanisms and future prospects. Ageing Res Rev 2016; 29:90-112. [PMID: 27353257 PMCID: PMC5991498 DOI: 10.1016/j.arr.2016.06.005] [Citation(s) in RCA: 216] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 06/23/2016] [Accepted: 06/23/2016] [Indexed: 12/31/2022]
Abstract
Answering the question as to why we age is tantamount to answering the question of what is life itself. There are countless theories as to why and how we age, but, until recently, the very definition of aging - senescence - was still uncertain. Here, we summarize the main views of the different models of senescence, with a special emphasis on the biochemical processes that accompany aging. Though inherently complex, aging is characterized by numerous changes that take place at different levels of the biological hierarchy. We therefore explore some of the most relevant changes that take place during aging and, finally, we overview the current status of emergent aging therapies and what the future holds for this field of research. From this multi-dimensional approach, it becomes clear that an integrative approach that couples aging research with systems biology, capable of providing novel insights into how and why we age, is necessary.
Collapse
Affiliation(s)
- João Pinto da Costa
- CESAM and Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal.
| | - Rui Vitorino
- Department of Medical Sciences, Institute for Biomedicine-iBiMED, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal; Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| | - Gustavo M Silva
- Department of Biology, Center for Genomics and Systems Biology, NY, NY 10003, USA
| | - Christine Vogel
- Department of Biology, Center for Genomics and Systems Biology, NY, NY 10003, USA
| | - Armando C Duarte
- CESAM and Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| | - Teresa Rocha-Santos
- CESAM and Department of Chemistry, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
72
|
Jäger K, Walter M. Therapeutic Targeting of Telomerase. Genes (Basel) 2016; 7:genes7070039. [PMID: 27455328 PMCID: PMC4962009 DOI: 10.3390/genes7070039] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 06/16/2016] [Accepted: 06/24/2016] [Indexed: 12/20/2022] Open
Abstract
Telomere length and cell function can be preserved by the human reverse transcriptase telomerase (hTERT), which synthesizes the new telomeric DNA from a RNA template, but is normally restricted to cells needing a high proliferative capacity, such as stem cells. Consequently, telomerase-based therapies to elongate short telomeres are developed, some of which have successfully reached the stage I in clinical trials. Telomerase is also permissive for tumorigenesis and 90% of all malignant tumors use telomerase to obtain immortality. Thus, reversal of telomerase upregulation in tumor cells is a potential strategy to treat cancer. Natural and small-molecule telomerase inhibitors, immunotherapeutic approaches, oligonucleotide inhibitors, and telomerase-directed gene therapy are useful treatment strategies. Telomerase is more widely expressed than any other tumor marker. The low expression in normal tissues, together with the longer telomeres in normal stem cells versus cancer cells, provides some degree of specificity with low risk of toxicity. However, long term telomerase inhibition may elicit negative effects in highly-proliferative cells which need telomerase for survival, and it may interfere with telomere-independent physiological functions. Moreover, only a few hTERT molecules are required to overcome senescence in cancer cells, and telomerase inhibition requires proliferating cells over a sufficient number of population doublings to induce tumor suppressive senescence. These limitations may explain the moderate success rates in many clinical studies. Despite extensive studies, only one vaccine and one telomerase antagonist are routinely used in clinical work. For complete eradication of all subpopulations of cancer cells a simultaneous targeting of several mechanisms will likely be needed. Possible technical improvements have been proposed including the development of more specific inhibitors, methods to increase the efficacy of vaccination methods, and personalized approaches. Telomerase activation and cell rejuvenation is successfully used in regenerative medicine for tissue engineering and reconstructive surgery. However, there are also a number of pitfalls in the treatment with telomerase activating procedures for the whole organism and for longer periods of time. Extended cell lifespan may accumulate rare genetic and epigenetic aberrations that can contribute to malignant transformation. Therefore, novel vector systems have been developed for a 'mild' integration of telomerase into the host genome and loss of the vector in rapidly-proliferating cells. It is currently unclear if this technique can also be used in human beings to treat chronic diseases, such as atherosclerosis.
Collapse
Affiliation(s)
- Kathrin Jäger
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin 13353, Germany.
| | - Michael Walter
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin 13353, Germany.
- Labor Berlin-Charité Vivantes Services GmbH, Sylter Str. 2, Berlin 13353, Germany.
| |
Collapse
|
73
|
Salvador L, Singaravelu G, Harley CB, Flom P, Suram A, Raffaele JM. A Natural Product Telomerase Activator Lengthens Telomeres in Humans: A Randomized, Double Blind, and Placebo Controlled Study. Rejuvenation Res 2016; 19:478-484. [PMID: 26950204 PMCID: PMC5178008 DOI: 10.1089/rej.2015.1793] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
TA-65 is a dietary supplement based on an improved formulation of a small molecule telomerase activator that was discovered in a systematic screening of natural product extracts from traditional Chinese medicines. This study summarizes the findings on telomere length (TL) changes from a randomized, double blind, placebo controlled study of TA-65 over a 1 year period. The study was conducted on 117 relatively healthy cytomegalovirus-positive subjects aged 53–87 years old. Subjects taking the low dose of TA-65 (250 U) significantly increased TL over the 12 months period (530 ± 180 bp; p = 0.005), whereas subjects in the placebo group significantly lost TL (290 ± 100 bp; p = 0.01). The high dose of TA-65 (1000 U) showed a trend of improvements in TL compared with that of the placebo group; however, the improvements did not reach statistical significance. TL changes in the low-dose group were similar for both median and 20th percentile TLs. The findings suggest that TA-65 can lengthen telomeres in a statistically and possibly clinically significant manner.
Collapse
Affiliation(s)
| | | | | | - Peter Flom
- 4 Peter Flom Consulting , New York, New York
| | | | | |
Collapse
|
74
|
Abstract
Telomeres, the protective ends of linear chromosomes, shorten throughout an individual's lifetime. Telomere shortening is a hallmark of molecular aging and is associated with premature appearance of diseases associated with aging. Here, we discuss the role of telomere shortening as a direct cause for aging and age-related diseases. In particular, we draw attention to the fact that telomere length influences longevity. Furthermore, we discuss intrinsic and environmental factors that can impact on human telomere erosion. Finally, we highlight recent advances in telomerase-based therapeutic strategies for the treatment of diseases associated with extremely short telomeres owing to mutations in telomerase, as well as age-related diseases, and ultimately aging itself.
Collapse
Affiliation(s)
- Christian Bär
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| | - Maria A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre (CNIO), Madrid, Spain
| |
Collapse
|
75
|
Zhang J, Rane G, Dai X, Shanmugam MK, Arfuso F, Samy RP, Lai MKP, Kappei D, Kumar AP, Sethi G. Ageing and the telomere connection: An intimate relationship with inflammation. Ageing Res Rev 2016; 25:55-69. [PMID: 26616852 DOI: 10.1016/j.arr.2015.11.006] [Citation(s) in RCA: 251] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 11/16/2015] [Accepted: 11/19/2015] [Indexed: 12/19/2022]
Abstract
Telomeres are the heterochromatic repeat regions at the ends of eukaryotic chromosomes, whose length is considered to be a determinant of biological ageing. Normal ageing itself is associated with telomere shortening. Here, critically short telomeres trigger senescence and eventually cell death. This shortening rate may be further increased by inflammation and oxidative stress and thus affect the ageing process. Apart from shortened or dysfunctional telomeres, cells undergoing senescence are also associated with hyperactivity of the transcription factor NF-κB and overexpression of inflammatory cytokines such as TNF-α, IL-6, and IFN-γ in circulating macrophages. Interestingly, telomerase, a reverse transcriptase that elongates telomeres, is involved in modulating NF-κB activity. Furthermore, inflammation and oxidative stress are implicated as pre-disease mechanisms for chronic diseases of ageing such as neurodegenerative diseases, cardiovascular disease, and cancer. To date, inflammation and telomere shortening have mostly been studied individually in terms of ageing and the associated disease phenotype. However, the interdependent nature of the two demands a more synergistic approach in understanding the ageing process itself and for developing new therapeutic approaches. In this review, we aim to summarize the intricate association between the various inflammatory molecules and telomeres that together contribute to the ageing process and related diseases.
Collapse
|
76
|
Are Anxiety Disorders Associated with Accelerated Aging? A Focus on Neuroprogression. Neural Plast 2015; 2016:8457612. [PMID: 26881136 PMCID: PMC4736204 DOI: 10.1155/2016/8457612] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/05/2015] [Accepted: 10/08/2015] [Indexed: 12/16/2022] Open
Abstract
Anxiety disorders (AnxDs) are highly prevalent throughout the lifespan, with detrimental effects on daily-life functioning, somatic health, and quality of life. An emerging perspective suggested that AnxDs may be associated with accelerated aging. In this paper, we explored the association between AnxDs and hallmarks of accelerated aging, with a specific focus on neuroprogression. We reviewed animal and human findings that suggest an overlap between processes of impaired neurogenesis, neurodegeneration, structural, functional, molecular, and cellular modifications in AnxDs, and aging. Although this research is at an early stage, our review suggests a link between anxiety and accelerated aging across multiple processes involved in neuroprogression. Brain structural and functional changes that accompany normal aging were more pronounced in subjects with AnxDs than in coevals without AnxDs, including reduced grey matter density, white matter alterations, impaired functional connectivity of large-scale brain networks, and poorer cognitive performance. Similarly, molecular correlates of brain aging, including telomere shortening, Aβ accumulation, and immune-inflammatory and oxidative/nitrosative stress, were overrepresented in anxious subjects. No conclusions about causality or directionality between anxiety and accelerated aging can be drawn. Potential mechanisms of this association, limitations of the current research, and implications for treatments and future studies are discussed.
Collapse
|
77
|
Carnero A, Blanco-Aparicio C, Kondoh H, Lleonart ME, Martinez-Leal JF, Mondello C, Ivana Scovassi A, Bisson WH, Amedei A, Roy R, Woodrick J, Colacci A, Vaccari M, Raju J, Al-Mulla F, Al-Temaimi R, Salem HK, Memeo L, Forte S, Singh N, Hamid RA, Ryan EP, Brown DG, Wise JP, Wise SS, Yasaei H. Disruptive chemicals, senescence and immortality. Carcinogenesis 2015; 36 Suppl 1:S19-37. [PMID: 26106138 PMCID: PMC4565607 DOI: 10.1093/carcin/bgv029] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 08/04/2014] [Accepted: 08/05/2014] [Indexed: 12/16/2022] Open
Abstract
Carcinogenesis is thought to be a multistep process, with clonal evolution playing a central role in the process. Clonal evolution involves the repeated 'selection and succession' of rare variant cells that acquire a growth advantage over the remaining cell population through the acquisition of 'driver mutations' enabling a selective advantage in a particular micro-environment. Clonal selection is the driving force behind tumorigenesis and possesses three basic requirements: (i) effective competitive proliferation of the variant clone when compared with its neighboring cells, (ii) acquisition of an indefinite capacity for self-renewal, and (iii) establishment of sufficiently high levels of genetic and epigenetic variability to permit the emergence of rare variants. However, several questions regarding the process of clonal evolution remain. Which cellular processes initiate carcinogenesis in the first place? To what extent are environmental carcinogens responsible for the initiation of clonal evolution? What are the roles of genotoxic and non-genotoxic carcinogens in carcinogenesis? What are the underlying mechanisms responsible for chemical carcinogen-induced cellular immortality? Here, we explore the possible mechanisms of cellular immortalization, the contribution of immortalization to tumorigenesis and the mechanisms by which chemical carcinogens may contribute to these processes.
Collapse
Affiliation(s)
- Amancio Carnero
- *To whom correspondence should be addressed. Tel: +34955923111; Fax: +34955923101;
| | - Carmen Blanco-Aparicio
- Spanish National Cancer Research Center, Experimental Therapuetics Department, Melchor Fernandez Almagro, 3, 28029 Madrid, Spain
| | - Hiroshi Kondoh
- Department of Geriatric Medicine, Kyoto University Hospital, 54 Kawaharacho, Shogoin, Sakyo-ku Kyoto 606-8507, Japan
| | - Matilde E. Lleonart
- Institut De Recerca Hospital Vall D’Hebron, Passeig Vall d’Hebron, 119–129, 08035 Barcelona, Spain
| | | | - Chiara Mondello
- Istituto di Genetica Molecolare, CNR, Via Abbiategrasso 207, 27100 Pavia, Italy
| | - A. Ivana Scovassi
- Istituto di Genetica Molecolare, CNR, Via Abbiategrasso 207, 27100 Pavia, Italy
| | - William H. Bisson
- Environmental and Molecular Toxicology, Environmental Health Science Center, Oregon State University, Corvallis, OR 97331, USA
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Firenze, Italy, Florence 50134, Italy
| | - Rabindra Roy
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Jordan Woodrick
- Molecular Oncology Program, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Annamaria Colacci
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Monica Vaccari
- Center for Environmental Carcinogenesis and Risk Assessment, Environmental Protection and Health Prevention Agency, Bologna 40126, Italy
| | - Jayadev Raju
- Toxicology Research Division, Bureau of Chemical Safety Food Directorate, Health Products and Food Branch Health Canada, Ottawa, Ontario K1A0K9, Canada
| | - Fahd Al-Mulla
- Department of Pathology, Kuwait University, Safat 13110, Kuwait
| | | | - Hosni K. Salem
- Urology Department, kasr Al-Ainy School of Medicine, Cairo University, El Manial, Cairo 12515, Egypt
| | - Lorenzo Memeo
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Stefano Forte
- Mediterranean Institute of Oncology, Viagrande 95029, Italy
| | - Neetu Singh
- Centre for Advanced Research, King George’s Medical University, Chowk, Lucknow, Uttar Pradesh 226003, India
| | - Roslida A. Hamid
- Department of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor 43400, Malaysia
| | - Elizabeth P. Ryan
- Department of Environmental and Radiological Health Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - Dustin G. Brown
- Department of Environmental and Radiological Health Sciences, Colorado State University/Colorado School of Public Health, Fort Collins, CO 80523-1680, USA
| | - John Pierce Wise
- The Wise Laboratory of Environmental and Genetic Toxicology, Maine Center for Toxicology and Environmental Health, Department of Applied Medical Sciences, University of Southern Maine, 96 Falmouth Street, Portland, ME 04104, USA and
| | - Sandra S. Wise
- The Wise Laboratory of Environmental and Genetic Toxicology, Maine Center for Toxicology and Environmental Health, Department of Applied Medical Sciences, University of Southern Maine, 96 Falmouth Street, Portland, ME 04104, USA and
| | - Hemad Yasaei
- Brunel Institute of Cancer Genetics and Pharmacogenomics, Health and Environment Theme, Institute of Environment, Health and Societies, Brunel University London, Kingston Lane, Uxbridge, UB8 3PH, UK
| |
Collapse
|
78
|
Feng LM, Ji S, Qiao X, Li ZW, Lin XH, Ye M. Biocatalysis of Cycloastragenol by Syncephalastrum racemosum
and Alternaria alternata
to Discover Anti-Aging Derivatives. Adv Synth Catal 2015. [DOI: 10.1002/adsc.201401158] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
79
|
Bedir E, Kula C, Öner Ö, Altaş M, Tağ Ö, Öngen G. Microbial transformation of Astragalus sapogenins using Cunninghamella blakesleeana NRRL 1369 and Glomerella fusarioides ATCC 9552. ACTA ACUST UNITED AC 2015. [DOI: 10.1016/j.molcatb.2015.01.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
80
|
Ramunas J, Yakubov E, Brady JJ, Corbel SY, Holbrook C, Brandt M, Stein J, Santiago JG, Cooke JP, Blau HM. Transient delivery of modified mRNA encoding TERT rapidly extends telomeres in human cells. FASEB J 2015; 29:1930-9. [PMID: 25614443 DOI: 10.1096/fj.14-259531] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 12/31/2014] [Indexed: 12/13/2022]
Abstract
Telomere extension has been proposed as a means to improve cell culture and tissue engineering and to treat disease. However, telomere extension by nonviral, nonintegrating methods remains inefficient. Here we report that delivery of modified mRNA encoding TERT to human fibroblasts and myoblasts increases telomerase activity transiently (24-48 h) and rapidly extends telomeres, after which telomeres resume shortening. Three successive transfections over a 4 d period extended telomeres up to 0.9 kb in a cell type-specific manner in fibroblasts and myoblasts and conferred an additional 28 ± 1.5 and 3.4 ± 0.4 population doublings (PDs), respectively. Proliferative capacity increased in a dose-dependent manner. The second and third transfections had less effect on proliferative capacity than the first, revealing a refractory period. However, the refractory period was transient as a later fourth transfection increased fibroblast proliferative capacity by an additional 15.2 ± 1.1 PDs, similar to the first transfection. Overall, these treatments led to an increase in absolute cell number of more than 10(12)-fold. Notably, unlike immortalized cells, all treated cell populations eventually stopped increasing in number and expressed senescence markers to the same extent as untreated cells. This rapid method of extending telomeres and increasing cell proliferative capacity without risk of insertional mutagenesis should have broad utility in disease modeling, drug screening, and regenerative medicine.
Collapse
Affiliation(s)
- John Ramunas
- *Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Clinical Sciences Research Center, Stanford University School of Medicine, Stanford, California, USA; Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, California, USA; SpectraCell Laboratories, Inc., Houston, Texas, USA; and Department of Mechanical Engineering, Stanford University, Stanford, California, USA
| | - Eduard Yakubov
- *Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Clinical Sciences Research Center, Stanford University School of Medicine, Stanford, California, USA; Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, California, USA; SpectraCell Laboratories, Inc., Houston, Texas, USA; and Department of Mechanical Engineering, Stanford University, Stanford, California, USA
| | - Jennifer J Brady
- *Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Clinical Sciences Research Center, Stanford University School of Medicine, Stanford, California, USA; Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, California, USA; SpectraCell Laboratories, Inc., Houston, Texas, USA; and Department of Mechanical Engineering, Stanford University, Stanford, California, USA
| | - Stéphane Y Corbel
- *Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Clinical Sciences Research Center, Stanford University School of Medicine, Stanford, California, USA; Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, California, USA; SpectraCell Laboratories, Inc., Houston, Texas, USA; and Department of Mechanical Engineering, Stanford University, Stanford, California, USA
| | - Colin Holbrook
- *Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Clinical Sciences Research Center, Stanford University School of Medicine, Stanford, California, USA; Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, California, USA; SpectraCell Laboratories, Inc., Houston, Texas, USA; and Department of Mechanical Engineering, Stanford University, Stanford, California, USA
| | - Moritz Brandt
- *Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Clinical Sciences Research Center, Stanford University School of Medicine, Stanford, California, USA; Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, California, USA; SpectraCell Laboratories, Inc., Houston, Texas, USA; and Department of Mechanical Engineering, Stanford University, Stanford, California, USA
| | - Jonathan Stein
- *Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Clinical Sciences Research Center, Stanford University School of Medicine, Stanford, California, USA; Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, California, USA; SpectraCell Laboratories, Inc., Houston, Texas, USA; and Department of Mechanical Engineering, Stanford University, Stanford, California, USA
| | - Juan G Santiago
- *Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Clinical Sciences Research Center, Stanford University School of Medicine, Stanford, California, USA; Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, California, USA; SpectraCell Laboratories, Inc., Houston, Texas, USA; and Department of Mechanical Engineering, Stanford University, Stanford, California, USA
| | - John P Cooke
- *Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Clinical Sciences Research Center, Stanford University School of Medicine, Stanford, California, USA; Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, California, USA; SpectraCell Laboratories, Inc., Houston, Texas, USA; and Department of Mechanical Engineering, Stanford University, Stanford, California, USA
| | - Helen M Blau
- *Baxter Laboratory for Stem Cell Biology, Department of Microbiology and Immunology, Institute for Stem Cell Biology and Regenerative Medicine, Clinical Sciences Research Center, Stanford University School of Medicine, Stanford, California, USA; Falk Cardiovascular Research Center, Stanford University School of Medicine, Stanford, California, USA; SpectraCell Laboratories, Inc., Houston, Texas, USA; and Department of Mechanical Engineering, Stanford University, Stanford, California, USA
| |
Collapse
|
81
|
Modulators of cellular senescence: mechanisms, promises, and challenges from in vitro studies with dietary bioactive compounds. Nutr Res 2014; 34:1017-35. [DOI: 10.1016/j.nutres.2014.02.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Revised: 02/24/2014] [Accepted: 02/26/2014] [Indexed: 12/11/2022]
|
82
|
Bennaceur K, Atwill M, Al Zhrany N, Hoffmann J, Keavney B, Breault D, Richardson G, von Zglinicki T, Saretzki G, Spyridopoulos I. Atorvastatin induces T cell proliferation by a telomerase reverse transcriptase (TERT) mediated mechanism. Atherosclerosis 2014; 236:312-20. [PMID: 25127175 DOI: 10.1016/j.atherosclerosis.2014.07.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 07/18/2014] [Accepted: 07/21/2014] [Indexed: 12/12/2022]
Abstract
UNLABELLED Statins are one of the most potent drugs in delaying age-related inflammatory changes in the arterial vessel wall, slowing down the progression of atherosclerosis. Statins have also been shown to abrogate telomere-attributed cardiovascular risk. The goal of our study was to explore a potential effect of atorvastatin on telomerase activity in peripheral blood mononuclear cells (PBMCs) and T-lymphocytes (T cells). METHODS AND RESULTS Treatment with pharmacologically relevant concentrations (0.1-0.3 μM) of atorvastatin resulted in a 6-fold increase of telomerase activity (TA) (p < 0.0001) in human and mouse PBMCs and CD4 T cells, translating into moderate proliferation of T lymphocytes. In contrast, high doses of atorvastatin (2-5 μM) or the addition of LDL cholesterol completely inhibited proliferation, thereby abrogating telomerase activity. The proliferative effect of atorvastatin was ablated by the absense of the catalytic subunit of telomerase, telomerase reverse transcriptase (TERT). Using transgenic GFP-mTert reporter mice, we observed a decrease in telomerase-positive lymphocytes from 30% to 15% during the first 5 months of age (p < 0.01). This suggests that the decrease in immune cell turnover during normal development and maturation is mirrored by a reduction in telomerase activity in lymphocytes in-vivo. CONCLUSION Atorvastatin and cholesterol have opposing effects on telomerase in mononuclear cells and T-lymphocytes. Our study suggests a link between cholesterol metabolism and telomere-related cardiovascular risk.
Collapse
Affiliation(s)
| | - Mark Atwill
- Institute of Genetic Medicine at Newcastle University, UK
| | | | | | | | - David Breault
- Department of Pediatrics, Childrens Hospital and Harvard Stem Cell Institute, Cambridge, Boston, MA, USA
| | | | | | | | | |
Collapse
|
83
|
Taka T, Changtam C, Thaichana P, Kaewtunjai N, Suksamrarn A, Lee TR, Tuntiwechapikul W. Curcuminoid derivatives enhance telomerase activity in an in vitro TRAP assay. Bioorg Med Chem Lett 2014; 24:5242-6. [PMID: 25305686 DOI: 10.1016/j.bmcl.2014.09.059] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 09/01/2014] [Accepted: 09/20/2014] [Indexed: 10/24/2022]
Abstract
The length of telomeres controls the life span of eukaryotic cells. Telomerase maintains the length of telomeres in certain eukaryotic cells, such as germline cells and stem cells, and allows these cells to evade replicative senescence. Here, we report for the first time a number of curcuminoid derivatives that enhance telomerase activity in an in vitro TRAP assay. A preliminary analysis of structure-activity relationships found that the minimal requirement for this enhanced telomerase activity is a curcuminoid core with at least one n-pentylpyridine side chain, while curcuminoids with two such side chains exhibit even greater activity. The finding here might lead to a new class of telomerase activators that act directly or indirectly on telomerase, rather than through the reactivation of the telomerase reverse transcriptase (TERT) gene associated with other telomerase activators found in the literature.
Collapse
Affiliation(s)
- Thanachai Taka
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chatchawan Changtam
- Division of Physical Science, Faculty of Science and Technology, Huachiew Chalermprakiet University, Samutprakarn 10540, Thailand
| | - Pak Thaichana
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Navakoon Kaewtunjai
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Apichart Suksamrarn
- Department of Chemistry, Faculty of Science, Ramkhamhaeng University, Bangkok 10240, Thailand
| | - T Randall Lee
- Department of Chemistry and the Texas Center for Superconductivity, University of Houston, Houston, TX 77204-5003, USA
| | - Wirote Tuntiwechapikul
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
84
|
Harley CB, Liu W, Flom PL, Raffaele JM. A natural product telomerase activator as part of a health maintenance program: metabolic and cardiovascular response. Rejuvenation Res 2014; 16:386-95. [PMID: 23808324 DOI: 10.1089/rej.2013.1430] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
A short average telomere length is associated with low telomerase activity and certain degenerative diseases. Studies in animals and with human cells confirm a causal mechanism for cell or tissue dysfunction triggered by critically short telomeres, suggesting that telomerase activation may be an approach to health maintenance. Previously, we reported on positive immune remodeling in humans taking a commercial health maintenance program, PattonProtocol-1, composed of TA-65® (a natural product-derived telomerase activator) and other dietary supplements. In over a 5-year period and an estimated 7000 person-years of use, no adverse events or effects have been attributed to TA-65 by physicians licensed to sell the product. Here we report on changes in metabolic markers measured at baseline (n=97-107 subjects) and every 3-6 months (n=27-59 subjects) during the first 12 months of study. Rates of change per year from baseline determined by a multi-level model were -3.72 mg/dL for fasting glucose (p=0.02), -1.32 mIU/mL for insulin (p=0.01), -13.2 and -11.8 mg/dL for total cholesterol and low-density lipoprotein cholesterol (LDL-C) (p=0.002, p=0.002, respectively), -17.3 and -4.2 mmHg for systolic and diastolic blood pressure (p=0.007 and 0.001, respectively), and -3.6 μmole/L homocysteine (p=0.001). In a subset of individuals with bone mineral density (BMD) measured at baseline and 12 months, density increased 2.0% in the spine (p=0.003). We conclude that in addition to apparent positive immune remodeling, PattonProtocol-1 may improve markers of metabolic, bone, and cardiovascular health.
Collapse
|
85
|
Boccardi V, Paolisso G. Telomerase activation: a potential key modulator for human healthspan and longevity. Ageing Res Rev 2014; 15:1-5. [PMID: 24561251 DOI: 10.1016/j.arr.2013.12.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 12/18/2013] [Accepted: 12/23/2013] [Indexed: 12/12/2022]
Abstract
The elderly population is increasing progressively. Along with this increase the number of age related diseases, such as cardiovascular, neurodegenerative diseases, metabolic impairment and cancer, is also on the rise thereby negatively impacting the burden on health care systems. Telomere shortening and dysfunction results in cellular senescence, an irreversible proliferative arrest that has been suggested to promote organismal aging and disabling age-related diseases. Given that telomerase, the enzyme responsible for maintaining telomere lengths, is not expressed at levels sufficient to prevent telomere shortening in most of our cells, telomeres progressively erode with advancing age. Telomerase activation, therefore, might serve as a viable therapeutic strategy to delay the onset of cellular senescence, tissue dysfunction and organismal decline. Here we analyze the more recent findings in telomerase activation as a potential key modulator for human healthspan and longevity.
Collapse
|
86
|
Reichert S, Bize P, Arrivé M, Zahn S, Massemin S, Criscuolo F. Experimental increase in telomere length leads to faster feather regeneration. Exp Gerontol 2014; 52:36-8. [DOI: 10.1016/j.exger.2014.01.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 01/16/2014] [Indexed: 10/25/2022]
|
87
|
Cukusic Kalajzic A, Skrobot Vidacek N, Huzak M, Ivankovic M, Rubelj I. Telomere Q-PNA-FISH--reliable results from stochastic signals. PLoS One 2014; 9:e92559. [PMID: 24643066 PMCID: PMC3958560 DOI: 10.1371/journal.pone.0092559] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 02/24/2014] [Indexed: 11/18/2022] Open
Abstract
Structural and functional analysis of telomeres is very important for understanding basic biological functions such as genome stability, cell growth control, senescence and aging. Recently, serious concerns have been raised regarding the reliability of current telomere measurement methods such as Southern blot and quantitative polymerase chain reaction. Since telomere length is associated with age related pathologies, including cardiovascular disease and cancer, both at the individual and population level, accurate interpretation of measured results is a necessity. The telomere Q-PNA-FISH technique has been widely used in these studies as well as in commercial analysis for the general population. A hallmark of telomere Q-PNA-FISH is the wide variation among telomere signals which has a major impact on obtained results. In the present study we introduce a specific mathematical and statistical analysis of sister telomere signals during cell culture senescence which enabled us to identify high regularity in their variations. This phenomenon explains the reproducibility of results observed in numerous telomere studies when the Q-PNA-FISH technique is used. In addition, we discuss the molecular mechanisms which probably underlie the observed telomere behavior.
Collapse
Affiliation(s)
| | | | - Miljenko Huzak
- Department of Mathematics, University of Zagreb, Zagreb, Croatia
| | - Milena Ivankovic
- Department of Molecular Biology, Ruder Boskovic Institute, Zagreb, Croatia
| | - Ivica Rubelj
- Department of Molecular Biology, Ruder Boskovic Institute, Zagreb, Croatia
- * E-mail:
| |
Collapse
|
88
|
Dietary safety of cycloastragenol from Astragalus spp.: Subchronic toxicity and genotoxicity studies. Food Chem Toxicol 2014; 64:322-34. [DOI: 10.1016/j.fct.2013.11.041] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 11/13/2013] [Accepted: 11/27/2013] [Indexed: 12/18/2022]
|
89
|
Hartwig F, Bertoldi D, Larangeira M, Wagner MS. Up-regulating telomerase and tumor suppressors: focusing on anti-aging interventions at the population level. Aging Dis 2014; 5:17-26. [PMID: 24490113 DOI: 10.14336/ad.2014.050017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 10/06/2013] [Accepted: 10/07/2013] [Indexed: 12/15/2022] Open
Abstract
Most human populations are undergoing a demographic transition regarding their age structure. This transition is reflected in chronic non-communicable diseases featuring among the main contributors to burden of disease. Considering that the aging process is a major risk factor for such conditions, understanding the mechanisms underlying aging and age-related diseases is critical to develop strategies to impact human health at population and/or individual-levels. Two different aspects of aging process (namely, telomere shortening and DNA damage accumulation) were shown to interact in positively impacting mice median survival. However, strategies aimed at translating such knowledge into actual human health benefits have not yet been discussed. In this manuscript, we present potential exposures that are suited for population-level interventions, and contextualize the roles of population (based on behavioral exposures) and individual-level (based on small-molecule administration) anti-aging interventions in different levels of disease prevention. We suggest that exposures such as moderate wine consumption, reducing calorie intake and active lifestyle are potentially useful for primordial and primary prevention, while small-molecules that activate telomerase and/or tumor suppression responses are more suited for secondary and tertiary prevention (although important for primary prevention in specific population subgroups). We also indicate the need of studying the impacts, on aging and age-related diseases, of different combinations of these exposures in well-conducted randomized controlled trials, and propose Mendelian randomization as a valuable alternative to gather information in human populations regarding the effects of potential anti-aging interventions.
Collapse
Affiliation(s)
| | - Daniel Bertoldi
- Biotechnology Baccalaureate Course, Federal University of Pelotas, Brazil
| | - Martin Larangeira
- Biotechnology Baccalaureate Course, Federal University of Pelotas, Brazil
| | | |
Collapse
|
90
|
Gopalakrishnan S, Cheung NK, Yip BW, Au DW. Medaka fish exhibits longevity gender gap, a natural drop in estrogen and telomere shortening during aging: a unique model for studying sex-dependent longevity. Front Zool 2013; 10:78. [PMID: 24364913 PMCID: PMC3878272 DOI: 10.1186/1742-9994-10-78] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 12/17/2013] [Indexed: 01/07/2023] Open
Abstract
Introduction Females having a longer telomere and lifespan than males have been documented in many animals. Such linkage however has never been reported in fish. Progressive shortening of telomere length is an important aging mechanism. Mounting in vitro evidence has shown that telomere shortening beyond a critical length triggered replicative senescence or cell death. Estrogen has been postulated as a key factor contributing to maintenance of telomere and sex-dependent longevity in animals. This postulation remains unproven due to the lack of a suitable animal system for testing. Here, we introduce a teleost model, the Japanese medaka Oryzias latipes, which shows promise for research into the molecular mechanism(s) controlling sex difference in aging. Results Using the medaka, we demonstrate for the first time in teleost that (i) sex differences (female > male) in telomere length and longevity also exist in fish, and (ii) a natural, ‘menopause’-like decline of plasma estrogen was evident in females during aging. Estrogen levels significantly correlated with telomerase activity as well as telomere length in female organs (not in males), suggesting estrogen could modulate telomere length via telomerase activation in a sex -specific manner. A hypothetical in vivo ‘critical’ terminal restriction fragment (TRF, representing telomere) length of approximately 4 kb was deduced in medaka liver for prediction of organismal mortality, which is highly comparable with that for human cells. An age conversion model was also established to enable age translation between medaka (in months) and human (in years). These novel tools are useful for future research on comparative biology of aging using medaka. Conclusion The striking similarity in estrogen profile between aging female O. latipes and women enables studying the influence of “postmenopausal” decline of estrogen on telomere and longevity without the need of invasive ovariectomy. Medaka fish is advantageous for studying the direct effect of increased estrogen on telomere length and longevity without the breast cancer complications reported in rodents. The findings strongly support the notion that O. latipes is a unique non-mammalian model for validation of estrogenic influence on telomere and longevity in vertebrates. This laboratory model fish is of potential significance for deciphering the ostensibly conserved mechanism(s) of sex-associated longevity in vertebrates.
Collapse
Affiliation(s)
| | | | | | - Doris Wt Au
- State Key Laboratory in Marine Pollution, Department of Biology and Chemistry, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR.
| |
Collapse
|
91
|
|
92
|
Bernardes de Jesus B, Blasco MA. Telomerase at the intersection of cancer and aging. Trends Genet 2013; 29:513-20. [PMID: 23876621 DOI: 10.1016/j.tig.2013.06.007] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2013] [Revised: 06/07/2013] [Accepted: 06/18/2013] [Indexed: 12/16/2022]
Abstract
Although cancer and aging have been studied as independent diseases, mounting evidence suggests that cancer is an aging-associated disease and that cancer and aging share many molecular pathways. In particular, recent studies validated telomerase activation as a potential therapeutic target for age-related diseases; in addition, abnormal telomerase expression and telomerase mutations have been associated with many different types of human tumor. Here, we revisit the connection between telomerase and cancer and aging in light of recent findings supporting a role for telomerase not only in telomere elongation, but also in metabolic fitness and Wnt activation. Understanding the physiological impact of telomerase regulation is fundamental given the therapeutic strategies that are being developed that involve telomerase modulation.
Collapse
Affiliation(s)
- Bruno Bernardes de Jesus
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, Madrid, E-28029, Spain
| | | |
Collapse
|
93
|
Mendelsohn AR, Larrick JW. Ectopic expression of telomerase safely increases health span and life span. Rejuvenation Res 2013; 15:435-8. [PMID: 22877566 DOI: 10.1089/rej.2012.1359] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The absence of telomerase from somatic cells of mammals has significant consequences for aging. First, it limits the number of potential cell divisions and in so doing sets limits on both life span and cancer cell proliferation. Second, shortened telomeres are known to result in physiological dysfunction, including playing a role in human diseases such as Werner syndrome and ataxia telangiectasia. Ectopic expression of the catalytic subunit of telomerase, telomerase reverse transcriptase (TERT), has been reported to extend life span by as much as 40% in cancer-resistant mice. On the other hand, ectopic expression of TERT promotes cancer in normal mice. However, transient induction of TERT by an astragalus-derived compound increases health span without an apparent increase in cancer incidence. Ectopic expression of TERT using adeno-associated virus serotype 9 (AAV9)-based gene therapy in adult mice increases both health span and life span without increasing cancer incidence. Available evidence suggests that increases in life span may require both elongated telomeres and the continuous presence of telomerase to stimulate the WNT/β-catenin signaling pathway. The recent observation that WNT/β-catenin signaling can stimulate TERT expression raises the possibility of a positive feedback loop between TERT and WNT/β-catenin. Such a positive feedback loop implies that safety must be carefully considered in the development of drugs that stimulate telomerase activity.
Collapse
|
94
|
Kuban M, Öngen G, Khan IA, Bedir E. Microbial transformation of cycloastragenol. PHYTOCHEMISTRY 2013; 88:99-104. [PMID: 23357596 DOI: 10.1016/j.phytochem.2012.12.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 12/11/2012] [Accepted: 12/17/2012] [Indexed: 06/01/2023]
Abstract
The microbial transformation of cycloastragenol by the fungi Cunninghamella blakesleeana NRRL 1369 and Glomerella fusarioides ATCC 9552, and the bacterium Mycobacterium sp. NRRL 3805 were investigated. Both fungi mainly provided hydroxylated metabolites together with products formed by cyclization, dehydrogenation and Baeyer-Villiger oxidation resulting in a ring cleavage. The bacteria yielded only a single oxidation product, namely, 3-oxo-cycloastragenol. Structures of the metabolites were elucidated by 1-D ((1)H,(13)C), 2-D NMR (COSY, HMBC, HMQC) and HRMS analyses.
Collapse
Affiliation(s)
- Melis Kuban
- Ege University, Faculty of Engineering, Department of Bioengineering, 35100 Bornova, İzmir, Turkey
| | | | | | | |
Collapse
|
95
|
Molgora B, Bateman R, Sweeney G, Finger D, Dimler T, Effros RB, Valenzuela HF. Functional assessment of pharmacological telomerase activators in human T cells. Cells 2013; 2:57-66. [PMID: 24709644 PMCID: PMC3972662 DOI: 10.3390/cells2010057] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 01/07/2013] [Accepted: 01/08/2013] [Indexed: 12/13/2022] Open
Abstract
Telomeres are structures at the ends of chromosomes that shorten during cell division and eventually signal an irreversible state of growth arrest known as cellular senescence. To delay this cellular aging, human T cells, which are critical in the immune control over infections and cancer, activate the enzyme telomerase, which binds and extends the telomeres. Several different extracts from the Astragalus membranaceus root have been documented to activate telomerase activity in human T cells. The objective of this research was to compare two extracts from Astragalus membranaceus, TA-65 and HTA, for their effects on both telomerase and proliferative activity of human CD4 and CD8 T cells. Our results demonstrate that, TA-65 increased telomerase activity significantly (1.3 to 3.3-fold relative to controls) in T cell cultures from six donors tested, whereas HTA only increased telomerase levels in two out of six donors. We also demonstrate that TA-65 activates telomerase by a MAPK- specific pathway. Finally, we determine that during a three-day culture period, only the T cells treated with the TA-65 extract showed a statistically significant increase in proliferative activity. Our results underscore the importance of comparing multiple telomerase activators within the same experiment, and of including functional assays in addition to measuring telomerase activity.
Collapse
Affiliation(s)
- Brenda Molgora
- Department of Biology, Whittier College,13406 Philadelphia Street, P.O. Box 634, Whittier 90608, CA, USA.
| | - Riley Bateman
- Department of Biology, Whittier College,13406 Philadelphia Street, P.O. Box 634, Whittier 90608, CA, USA.
| | - Greg Sweeney
- Department of Biology, Whittier College,13406 Philadelphia Street, P.O. Box 634, Whittier 90608, CA, USA.
| | - Danielle Finger
- Department of Biology, Whittier College,13406 Philadelphia Street, P.O. Box 634, Whittier 90608, CA, USA.
| | - Taylor Dimler
- Department of Biology, Whittier College,13406 Philadelphia Street, P.O. Box 634, Whittier 90608, CA, USA.
| | - Rita B Effros
- Department of Pathology and Lab Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Avenue, Los Angeles 90095-1732, CA, USA.
| | - Hector F Valenzuela
- Department of Biology, Whittier College,13406 Philadelphia Street, P.O. Box 634, Whittier 90608, CA, USA.
| |
Collapse
|
96
|
Cai Z, Yan LJ, Ratka A. Telomere Shortening and Alzheimer’s Disease. Neuromolecular Med 2012; 15:25-48. [DOI: 10.1007/s12017-012-8207-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 11/02/2012] [Indexed: 10/27/2022]
|
97
|
Bernardes de Jesus B, Blasco MA. Potential of telomerase activation in extending health span and longevity. Curr Opin Cell Biol 2012; 24:739-43. [PMID: 23085234 DOI: 10.1016/j.ceb.2012.09.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 09/10/2012] [Accepted: 09/18/2012] [Indexed: 11/16/2022]
Abstract
The progressive increase in the elderly population worldwide has resulted in higher numbers of individuals affected by age-associated diseases, such as neurodegenerative and heart diseases, metabolic impairment, or cancer, with the subsequent burden for national health systems. Therapeutic interventions aimed to increase the quality of life at advanced age are visualized as important demands for the future, both at the level of individuals and society. Novel advances in telomerase function from several independent laboratories have resulted in potential new therapeutic strategies which appear as promising new venues to prevent cellular and tissue dysfunction and organismal decline, thereby increasing the so-called "health span". Here, we analyze these recent advances.
Collapse
Affiliation(s)
- Bruno Bernardes de Jesus
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, Madrid, E-28029, Spain
| | | |
Collapse
|
98
|
Hartwig FP, Nedel F, Collares TV, Tarquinio SBC, Nör JE, Demarco FF. Telomeres and Tissue Engineering: The Potential Roles of TERT in VEGF-mediated Angiogenesis. Stem Cell Rev Rep 2012; 8:1275-81. [DOI: 10.1007/s12015-012-9414-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
99
|
|
100
|
Abstract
A major goal in cancer and aging research is to discriminate the biochemical modifications that happen locally that could account for the healthiness or malignancy of tissues. Senescence is one general antiproliferative cellular process that acts as a strong barrier for cancer progression, playing a crucial role in aging. Here, we focus on the current methods to assess cellular senescence, discriminating the advantages and disadvantages of several senescence biomarkers.
Collapse
Affiliation(s)
- Bruno Bernardes de Jesus
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Centre, Madrid, Spain
| | | |
Collapse
|