1
|
Lin W, Huang D, Li M, Ren Y, Zheng X, Wu B, Miao Y. WHIRLY proteins, multi-layer regulators linking the nucleus and organelles in developmental and stress-induced senescence of plants. ANNALS OF BOTANY 2024; 134:521-536. [PMID: 38845347 PMCID: PMC11523626 DOI: 10.1093/aob/mcae092] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 06/03/2024] [Indexed: 11/01/2024]
Abstract
Plant senescence is an integrated programme of plant development that aims to remobilize nutrients and energy from senescing tissues to developing organs under developmental and stress-induced conditions. Upstream in the regulatory network, a small family of single-stranded DNA/RNA-binding proteins known as WHIRLYs occupy a central node, acting at multiple regulatory levels and via trans-localization between the nucleus and organelles. In this review, we summarize the current progress on the role of WHIRLY members in plant development and stress-induced senescence. WHIRLY proteins can be traced back in evolution to green algae. WHIRLY proteins trade off the balance of plant developmental senescence and stress-induced senescence through maintaining organelle genome stability via R-loop homeostasis, repressing the transcription at a configuration condition, and recruiting RNA to impact organelle RNA editing and splicing, as evidenced in several species. WHIRLY proteins also act as retrograde signal transducers between organelles and the nucleus through protein modification and stromule or vesicle trafficking. In addition, WHIRLY proteins interact with hormones, reactive oxygen species and environmental signals to orchestrate cell fate in an age-dependent manner. Finally, prospects for further research and promotion to improve crop production under environmental constraints are highlighted.
Collapse
Affiliation(s)
- Wenfang Lin
- Fujian Provincial Key Laboratory of Plant Functional Biology, Fujian Agriculture and Forestry University, 350002 Fuzhou, China
| | - Dongmei Huang
- Department of Biochemistry and Molecular Biology, Xiamen Medical College, Xiamen 361023, China
| | - Mengsi Li
- Fujian Provincial Key Laboratory of Plant Functional Biology, Fujian Agriculture and Forestry University, 350002 Fuzhou, China
| | - Yujun Ren
- Fujian Provincial Key Laboratory of Plant Functional Biology, Fujian Agriculture and Forestry University, 350002 Fuzhou, China
| | - Xiangzi Zheng
- Fujian Provincial Key Laboratory of Plant Functional Biology, Fujian Agriculture and Forestry University, 350002 Fuzhou, China
| | - Binghua Wu
- Fujian Provincial Key Laboratory of Plant Functional Biology, Fujian Agriculture and Forestry University, 350002 Fuzhou, China
| | - Ying Miao
- Fujian Provincial Key Laboratory of Plant Functional Biology, Fujian Agriculture and Forestry University, 350002 Fuzhou, China
| |
Collapse
|
2
|
Zheng YL, Wu X, Williams M, Verhulst S, Lin J, Takahashi Y, Ma JX, Wang Y. High-throughput single telomere analysis using DNA microarray and fluorescent in situ hybridization. Nucleic Acids Res 2024; 52:e96. [PMID: 39291738 PMCID: PMC11514468 DOI: 10.1093/nar/gkae812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 08/30/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024] Open
Abstract
The human telomere system is highly dynamic. Both short and long leucocyte average telomere lengths (aTL) are associated with an increased risk of cancer and early death, illustrating the complex relationship between TL and human health and the importance of assessing TL distributions with single TL analysis. A DNA microarray and telomere fluorescent in situ hybridization (DNA-array-FISH) approach was developed to measure the base-pair (bp) lengths of single telomeres. On average 32000 telomeres were measured per DNA sample with one microarray chip assaying 96 test DNA samples. Various telomere parameters, i.e. aTL and the frequency of short/long telomeres, were computed to delineate TL distribution. The intra-assay and inter-assay coefficient of variations of aTL ranged from 1.37% to 3.98%. The correlation coefficient (r) of aTL in repeated measurements ranged from 0.91 to 1.00, demonstrating high measurement precision. aTLs measured by DNA-array-FISH predicted aTLs measured by terminal restriction fragment (TRF) analysis with r ranging 0.87-0.99. A new accurate and high-throughput method has been developed to measure the bp lengths of single telomeres. The large number of single TL data provides an opportunity for an in-depth analysis of telomere dynamics and the complex relationship between telomere and age-related diseases.
Collapse
Affiliation(s)
- Yun-Ling Zheng
- Cancer Prevention and Control Program, Department of Oncology, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, USA
| | - Xingjia Wu
- Cancer Prevention and Control Program, Department of Oncology, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, USA
| | - Madeline Williams
- Cancer Prevention and Control Program, Department of Oncology, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, USA
| | - Simon Verhulst
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, The Netherlands
| | - Jue Lin
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA 94143, USA
| | - Yusuke Takahashi
- Department of Biochemistry, Wake Forest School of Medicine, NC 27157, USA
| | - Jian-Xing Ma
- Department of Biochemistry, Wake Forest School of Medicine, NC 27157, USA
| | - Ying Wang
- TelohealthDx, LLC, Clarksburg, MD 20871, USA
| |
Collapse
|
3
|
Lasho T, Patnaik MM. Adaptive and Maladaptive Clonal Hematopoiesis in Telomere Biology Disorders. Curr Hematol Malig Rep 2024; 19:35-44. [PMID: 38095828 DOI: 10.1007/s11899-023-00719-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/24/2023] [Indexed: 01/30/2024]
Abstract
PURPOSE OF REVIEW Telomere biology disorders (TBDs) are germline-inherited conditions characterized by reduction in telomerase function, accelerated shortening of telomeres, predisposition to organ-failure syndromes, and increased risk of neoplasms, especially myeloid malignancies. In normal cells, critically short telomeres trigger apoptosis and/or cellular senescence. However, the evolutionary mechanism by which TBD-related telomerase-deficient cells can overcome this fitness constraint remains elusive. RECENT FINDINGS Preliminary data suggests the existence of adaptive somatic mosaic states characterized by variants in TBD-related genes and maladaptive somatic mosaic states that attempt to overcome hematopoietic fitness constraints by alternative methods leading to clonal hematopoiesis. TBDs are both rare and highly heterogeneous in presentation, and the association of TBD with malignant transformation is unclear. Understanding the clonal complexity and mechanisms behind TBD-associated molecular signatures that lead to somatic adaptation in the setting of defective hematopoiesis will help inform therapy and treatment for this set of diseases.
Collapse
Affiliation(s)
- Terra Lasho
- Division of Hematology, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN, 55905, USA
| | - Mrinal M Patnaik
- Division of Hematology, Mayo Clinic Rochester, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
4
|
Ghanim GE, Sekne Z, Balch S, van Roon AMM, Nguyen THD. 2.7 Å cryo-EM structure of human telomerase H/ACA ribonucleoprotein. Nat Commun 2024; 15:746. [PMID: 38272871 PMCID: PMC10811338 DOI: 10.1038/s41467-024-45002-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 01/03/2024] [Indexed: 01/27/2024] Open
Abstract
Telomerase is a ribonucleoprotein (RNP) enzyme that extends telomeric repeats at eukaryotic chromosome ends to counterbalance telomere loss caused by incomplete genome replication. Human telomerase is comprised of two distinct functional lobes tethered by telomerase RNA (hTR): a catalytic core, responsible for DNA extension; and a Hinge and ACA (H/ACA) box RNP, responsible for telomerase biogenesis. H/ACA RNPs also have a general role in pseudouridylation of spliceosomal and ribosomal RNAs, which is critical for the biogenesis of the spliceosome and ribosome. Much of our structural understanding of eukaryotic H/ACA RNPs comes from structures of the human telomerase H/ACA RNP. Here we report a 2.7 Å cryo-electron microscopy structure of the telomerase H/ACA RNP. The significant improvement in resolution over previous 3.3 Å to 8.2 Å structures allows us to uncover new molecular interactions within the H/ACA RNP. Many disease mutations are mapped to these interaction sites. The structure also reveals unprecedented insights into a region critical for pseudouridylation in canonical H/ACA RNPs. Together, our work advances understanding of telomerase-related disease mutations and the mechanism of pseudouridylation by eukaryotic H/ACA RNPs.
Collapse
Affiliation(s)
| | - Zala Sekne
- MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | | | | | | |
Collapse
|
5
|
Lim CJ. Telomere C-Strand Fill-In Machinery: New Insights into the Human CST-DNA Polymerase Alpha-Primase Structures and Functions. Subcell Biochem 2024; 104:73-100. [PMID: 38963484 DOI: 10.1007/978-3-031-58843-3_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Telomeres at the end of eukaryotic chromosomes are extended by a specialized set of enzymes and telomere-associated proteins, collectively termed here the telomere "replisome." The telomere replisome acts on a unique replicon at each chromosomal end of the telomeres, the 3' DNA overhang. This telomere replication process is distinct from the replisome mechanism deployed to duplicate the human genome. The G-rich overhang is first extended before the complementary C-strand is filled in. This overhang is extended by telomerase, a specialized ribonucleoprotein and reverse transcriptase. The overhang extension process is terminated when telomerase is displaced by CTC1-STN1-TEN1 (CST), a single-stranded DNA-binding protein complex. CST then recruits DNA polymerase α-primase to complete the telomere replication process by filling in the complementary C-strand. In this chapter, the recent structure-function insights into the human telomere C-strand fill-in machinery (DNA polymerase α-primase and CST) will be discussed.
Collapse
Affiliation(s)
- Ci Ji Lim
- Department of Biochemistry, College of Agricultural and Life Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
6
|
Aitova A, Berezhnoy A, Tsvelaya V, Gusev O, Lyundup A, Efimov AE, Agapov I, Agladze K. Biomimetic Cardiac Tissue Models for In Vitro Arrhythmia Studies. Biomimetics (Basel) 2023; 8:487. [PMID: 37887618 PMCID: PMC10604593 DOI: 10.3390/biomimetics8060487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/26/2023] [Accepted: 10/03/2023] [Indexed: 10/28/2023] Open
Abstract
Cardiac arrhythmias are a major cause of cardiovascular mortality worldwide. Many arrhythmias are caused by reentry, a phenomenon where excitation waves circulate in the heart. Optical mapping techniques have revealed the role of reentry in arrhythmia initiation and fibrillation transition, but the underlying biophysical mechanisms are still difficult to investigate in intact hearts. Tissue engineering models of cardiac tissue can mimic the structure and function of native cardiac tissue and enable interactive observation of reentry formation and wave propagation. This review will present various approaches to constructing cardiac tissue models for reentry studies, using the authors' work as examples. The review will highlight the evolution of tissue engineering designs based on different substrates, cell types, and structural parameters. A new approach using polymer materials and cellular reprogramming to create biomimetic cardiac tissues will be introduced. The review will also show how computational modeling of cardiac tissue can complement experimental data and how such models can be applied in the biomimetics of cardiac tissue.
Collapse
Affiliation(s)
- Aleria Aitova
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, 129110 Moscow, Russia
- Almetyevsk State Oil Institute, 423450 Almetyevsk, Russia
| | - Andrey Berezhnoy
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, 129110 Moscow, Russia
- Almetyevsk State Oil Institute, 423450 Almetyevsk, Russia
| | - Valeriya Tsvelaya
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, 129110 Moscow, Russia
- Almetyevsk State Oil Institute, 423450 Almetyevsk, Russia
| | - Oleg Gusev
- Regulatory Genomics Research Center, Institute of Fundamental Medicine and Biology, Kazan Federal University, 420018 Kazan, Russia
- Life Improvement by Future Technologies (LIFT) Center, 143025 Moscow, Russia
- Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo 113-8421, Japan
| | | | - Anton E. Efimov
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| | - Igor Agapov
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 123182 Moscow, Russia
| | - Konstantin Agladze
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, 129110 Moscow, Russia
| |
Collapse
|
7
|
Garger D, Meinel M, Dietl T, Hillig C, Garzorz‐Stark N, Eyerich K, de Angelis MH, Eyerich S, Menden MP. The impact of the cardiovascular component and somatic mutations on ageing. Aging Cell 2023; 22:e13957. [PMID: 37608601 PMCID: PMC10577550 DOI: 10.1111/acel.13957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/27/2023] [Accepted: 07/20/2023] [Indexed: 08/24/2023] Open
Abstract
Mechanistic insight into ageing may empower prolonging the lifespan of humans; however, a complete understanding of this process is still lacking despite a plethora of ageing theories. In order to address this, we investigated the association of lifespan with eight phenotypic traits, that is, litter size, body mass, female and male sexual maturity, somatic mutation, heart, respiratory, and metabolic rate. In support of the somatic mutation theory, we analysed 15 mammalian species and their whole-genome sequencing deriving somatic mutation rate, which displayed the strongest negative correlation with lifespan. All remaining phenotypic traits showed almost equivalent strong associations across this mammalian cohort, however, resting heart rate explained additional variance in lifespan. Integrating somatic mutation and resting heart rate boosted the prediction of lifespan, thus highlighting that resting heart rate may either directly influence lifespan, or represents an epiphenomenon for additional lower-level mechanisms, for example, metabolic rate, that are associated with lifespan.
Collapse
Affiliation(s)
- Daniel Garger
- Computational Health Center, Helmholtz MunichNeuherbergGermany
- Faculty of BiologyLudwig Maximilian UniversityMartinsriedGermany
| | - Martin Meinel
- Computational Health Center, Helmholtz MunichNeuherbergGermany
- Faculty of BiologyLudwig Maximilian UniversityMartinsriedGermany
- Department of Dermatology and AllergyTechnical University of MunichMunichGermany
| | - Tamina Dietl
- Computational Health Center, Helmholtz MunichNeuherbergGermany
- Faculty of BiologyLudwig Maximilian UniversityMartinsriedGermany
| | - Christina Hillig
- Computational Health Center, Helmholtz MunichNeuherbergGermany
- Department of MathematicsTechnical University of MunichMunichGermany
| | - Natalie Garzorz‐Stark
- Department of Dermatology and AllergyTechnical University of MunichMunichGermany
- Division of Dermatology and Venereology, Department of Medicine Solna, and Center for molecular medicineKarolinska InstitutetStockholmSweden
| | - Kilian Eyerich
- Division of Dermatology and Venereology, Department of Medicine Solna, and Center for molecular medicineKarolinska InstitutetStockholmSweden
- Department of Dermatology and Venerology, Medical SchoolUniversity of FreiburgFreiburgGermany
| | - Martin Hrabě de Angelis
- Institute of Experimental GeneticsHelmholtz MunichNeuherbergGermany
- Chair of Experimental Genetics, TUM School of Life SciencesTechnical University MunichFreisingGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
| | - Stefanie Eyerich
- Center for Allergy and Environment (ZAUM)Technical University MunichMunichGermany
- Institute for Allergy ResearchHelmholtz Munich, NeuherbergNeuherbergGermany
| | - Michael P. Menden
- Computational Health Center, Helmholtz MunichNeuherbergGermany
- Faculty of BiologyLudwig Maximilian UniversityMartinsriedGermany
- German Center for Diabetes Research (DZD)NeuherbergGermany
- Department of Biochemistry and PharmacologyUniversity of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
8
|
Romero-Haro AA, Figuerola J, Alonso-Alvarez C. Low Antioxidant Glutathione Levels Lead to Longer Telomeres: A Sex-Specific Link to Longevity? Integr Org Biol 2023; 5:obad034. [PMID: 37753451 PMCID: PMC10519275 DOI: 10.1093/iob/obad034] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/22/2023] [Accepted: 08/29/2023] [Indexed: 09/28/2023] Open
Abstract
Telomeres are repetitive DNA sequences at the end of chromosomes that protect them from degradation. They have been the focus of intense research because short telomeres would predict accelerated ageing and reduced longevity in vertebrates. Oxidative stress is considered a physiological driver of the telomere shortening and, consequently, short lifespan. Among molecules fighting against oxidative stress, glutathione is involved in many antioxidant pathways. Literature supports that oxidative stress may trigger a compensatory "hormetic" response increasing glutathione levels and telomere length. Here, we tested the link between total glutathione concentration and telomere length in captive birds (zebra finches; Taeniopygia guttata). Total glutathione levels were experimentally decreased during birds' growth using a specific inhibitor of glutathione synthesis (buthionine sulfoximine; BSO). We monitored the birds' reproductive performance in an outdoor aviary during the first month of life, and their longevity for almost 9 years. Among control individuals, erythrocyte glutathione levels during development positively predicted erythrocyte telomere length in adulthood. However, BSO-treated females, but not males, showed longer telomeres than control females in adulthood. This counterintuitive finding suggests that females mounted a compensatory response. Such compensation agrees with precedent findings in the same population where the BSO treatment increased growth and adult body mass in females but not males. BSO did not influence longevity or reproductive output in any sex. However, early glutathione levels and adult telomere length interactively predicted longevity only among control females. Those females with "naturally" low (non-manipulated) glutathione levels at the nestling age but capable of producing longer telomeres in adulthood seem to live longer. The results suggest that the capability to mount a hormetic response triggered by low early glutathione levels can improve fitness via telomere length. Overall, the results may indicate a sex-specific link between glutathione and telomere values. Telomerase activity and sexual steroids (estrogens) are good candidates to explain the sex-biased mechanism underlying the early-life impact of oxidative stress on adult telomere length.
Collapse
Affiliation(s)
- A A Romero-Haro
- Instituto de Investigación en Recursos Cinegéticos (IREC), CSIC-UCLM-JCCM, Ronda de Toledo 12, 13071Ciudad Real, 41092 Sevilla, Spain
| | - J Figuerola
- Estación Biológica de Doñana—CSIC, Sevilla, 28029 Madrid, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - C Alonso-Alvarez
- Evolutionary Ecology Department, National Museum of Natural Sciences (MNCN-CSIC), C/José Gutiérrez Abascal 2, 28006 Madrid, Spain
- Instituto Pirenaico de Ecología (IPE-CSIC) Avda. Nuestra Señora de la Victoria, 16. 22700 Jaca, Huesca, Spain
| |
Collapse
|
9
|
Chaney C, Wiley KS. The variable associations between PFASs and biological aging by sex and reproductive stage in NHANES 1999-2018. ENVIRONMENTAL RESEARCH 2023; 227:115714. [PMID: 36965790 DOI: 10.1016/j.envres.2023.115714] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/31/2023] [Accepted: 03/16/2023] [Indexed: 05/08/2023]
Abstract
BACKGROUND Per- and polyfluoroalkyl substances (PFASs) are endocrine disrupting chemicals that have myriad effects on human physiology. Estrogenic PFASs may influence biological aging by mimicking the activity of endogenous estrogens, which can decrease inflammation and oxidative stress and enhance telomerase activity. We hypothesized that PFAS exposure would be differentially associated with measures of biological aging based on biological sex and reproductive stage. METHODS We analyzed associations between serum PFAS levels and measures of biological aging for pre- and postmenopausal women and men (n = 3193) using data from the 2003 to 2018 waves of the National Health and Nutrition Examination Survey. Examining PFASs both individually and in mixture models, we investigated four measures of clinical aging (Homeostatic Dysregulation, the Klemera-Doubal Method, Phenotypic Age Acceleration, and Allostatic Load), oxidative stress, and telomere length. RESULTS PFOA and PFOS were negatively associated with Phenotypic Age Acceleration (e.g. decelerated aging) for men B = -0.22, 95% CI: -0.32, -0.12; B = -0.04, 95% CI: -0.06, -0.03) , premenopausal women (B = -0.58, 95% CI: -0.83, -0.32; B = -0.15, 95% CI: -0.20, -0.09), and postmenopausal women (B= -0.22, 95% CI: -0.43, -0.01; B = -0.05, 95% CI: -0.08, -0.02). In mixture models, we found net negative effects for Phenotypic Age Acceleration and Allostatic Load for men, premenopausal women, and postmenopausal women. We also found significant mixture effects for the antioxidants bilirubin and albumin among the three sample groups. We found no evidence to support effects on telomere length. DISCUSSION Our findings suggest that PFAS exposure may be inversely associated with some measures of biological aging at the relatively low levels of exposure in this sample, regardless of reproductive stage and sex, which does not support our hypothesis. This research provides insights into how PFAS exposure may variably influence aging measures depending on the physiological process investigated.
Collapse
Affiliation(s)
- C Chaney
- Department of Anthropology, Yale University, New Haven, CT, USA.
| | - K S Wiley
- Department of Anthropology, University of California, Los Angeles, CA, USA; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| |
Collapse
|
10
|
Telomere Length Changes in Cancer: Insights on Carcinogenesis and Potential for Non-Invasive Diagnostic Strategies. Genes (Basel) 2023; 14:genes14030715. [PMID: 36980987 PMCID: PMC10047978 DOI: 10.3390/genes14030715] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/08/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Telomere dynamics play a crucial role in the maintenance of chromosome integrity; changes in telomere length may thus contribute to the development of various diseases including cancer. Understanding the role of telomeric DNA in carcinogenesis and detecting the presence of cell-free telomeric DNA (cf-telDNA) in body fluids offer a potential biomarker for novel cancer screening and diagnostic strategies. Liquid biopsy is becoming increasingly popular due to its undeniable benefits over conventional invasive methods. However, the organization and function of cf-telDNA in the extracellular milieu are understudied. This paper provides a review based on 3,398,017 cancer patients, patients with other conditions, and control individuals with the aim to shed more light on the inconsistent nature of telomere lengthening/shortening in oncological contexts. To gain a better understanding of biological factors (e.g., telomerase activation, alternative lengthening of telomeres) affecting telomere homeostasis across different types of cancer, we summarize mechanisms responsible for telomere length maintenance. In conclusion, we compare tissue- and liquid biopsy-based approaches in cancer assessment and provide a brief outlook on the methodology used for telomere length evaluation, highlighting the advances of state-of-the-art approaches in the field.
Collapse
|
11
|
Al-Karmalawy AA, Nafie MS, Shaldam MA, Elmaaty AA, Antar SA, El-Hamaky AA, Saleh MA, Elkamhawy A, Tawfik HO. Ligand-Based Design on the Dog-Bone-Shaped BIBR1532 Pharmacophoric Features and Synthesis of Novel Analogues as Promising Telomerase Inhibitors with In Vitro and In Vivo Evaluations. J Med Chem 2023; 66:777-792. [PMID: 36525642 DOI: 10.1021/acs.jmedchem.2c01668] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Telomerase is an outstanding biological target for cancer treatment. BIBR1532 is a non-nucleoside selective telomerase inhibitor; however, it experiences ineligible pharmacokinetics. Herein, we aimed to design new BIBR1532-based analogues as promising telomerase inhibitors. Therefore, two novel series of pyridazine-linked to cyclopenta[b]thiophene (8a-f) and tetrahydro-1-benzothiophene (9a-f) were synthesized. A quantitative real-time polymerase chain reaction was utilized to investigate the telomerase inhibitory activity of candidates. Notably, 8e and 9e exhibited the best inhibition profiles. Moreover, 8e showed strong antitumor effects against both MCF-7 and A549 cancer cell lines. The effects of 8e on the cell cycle and apoptosis were measured. Besides, 8e was evaluated for its in vivo antitumor activity using solid Ehrlich carcinoma. The reduction in both the tumor weight and volume was greater than doxorubicin. Also, molecular docking and ADME studies were performed. Finally, a SAR study was conducted to gain further insights into the different telomerase inhibition potentials upon variable structural modifications.
Collapse
Affiliation(s)
- Ahmed A Al-Karmalawy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ahram Canadian University, 6th of October City, Giza 12566, Egypt
| | - Mohamed S Nafie
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Ayman Abo Elmaaty
- Department of Medicinal Chemistry, Faculty of Pharmacy, Port Said University, Port Said 42526, Egypt
| | - Samar A Antar
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, Horus University, New Damietta 34518, Egypt.,Center for Vascular and Heart Research, Fralin Biomedical Research Institute, Virginia Tech, Roanoke, Virginia 24016, United States
| | - Anwar A El-Hamaky
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Mohamed A Saleh
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, The United Arab Emirates.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed Elkamhawy
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea.,Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
12
|
Targeting the "hallmarks of aging" to slow aging and treat age-related disease: fact or fiction? Mol Psychiatry 2023; 28:242-255. [PMID: 35840801 PMCID: PMC9812785 DOI: 10.1038/s41380-022-01680-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 06/20/2022] [Accepted: 06/27/2022] [Indexed: 01/09/2023]
Abstract
Aging is a major risk factor for a number of chronic diseases, including neurodegenerative and cerebrovascular disorders. Aging processes have therefore been discussed as potential targets for the development of novel and broadly effective preventatives or therapeutics for age-related diseases, including those affecting the brain. Mechanisms thought to contribute to aging have been summarized under the term the "hallmarks of aging" and include a loss of proteostasis, mitochondrial dysfunction, altered nutrient sensing, telomere attrition, genomic instability, cellular senescence, stem cell exhaustion, epigenetic alterations and altered intercellular communication. We here examine key claims about the "hallmarks of aging". Our analysis reveals important weaknesses that preclude strong and definitive conclusions concerning a possible role of these processes in shaping organismal aging rate. Significant ambiguity arises from the overreliance on lifespan as a proxy marker for aging, the use of models with unclear relevance for organismal aging, and the use of study designs that do not allow to properly estimate intervention effects on aging rate. We also discuss future research directions that should be taken to clarify if and to what extent putative aging regulators do in fact interact with aging. These include multidimensional analytical frameworks as well as designs that facilitate the proper assessment of intervention effects on aging rate.
Collapse
|
13
|
Vedder O, Moiron M, Bichet C, Bauch C, Verhulst S, Becker PH, Bouwhuis S. Telomere length is heritable and genetically correlated with lifespan in a wild bird. Mol Ecol 2022; 31:6297-6307. [PMID: 33460462 DOI: 10.1111/mec.15807] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/18/2020] [Accepted: 01/08/2021] [Indexed: 01/31/2023]
Abstract
Telomeres are protective caps at the end of eukaryotic chromosomes that shorten with age and in response to stressful or resource-demanding conditions. Their length predicts individual health and lifespan across a wide range of animals, but whether the observed positive association between telomere length and lifespan is environmentally induced, or set at conception due to a shared genetic basis, has not been tested in wild animals. We applied quantitative genetic "animal models" to longitudinal telomere measurements collected over a 10-year period from individuals of a wild seabird (common tern; Sterna hirundo) with known pedigree. We found no variation in telomere shortening with age among individuals at the phenotypic and genetic level, and only a small permanent environmental effect on adult telomere length. Instead, we found telomere length to be highly heritable and strongly positively genetically correlated with lifespan. Such heritable differences between individuals that are set at conception may present a hitherto underappreciated component of variation in somatic state.
Collapse
Affiliation(s)
- Oscar Vedder
- Institute of Avian Research, Wilhelmshaven, Germany.,Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Maria Moiron
- Institute of Avian Research, Wilhelmshaven, Germany.,CEFE, Univ Montpellier, CNRS, EPHE, IRD, Univ Paul Valéry Montpellier 3, Montpellier, France
| | | | - Christina Bauch
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | - Simon Verhulst
- Groningen Institute for Evolutionary Life Sciences, University of Groningen, Groningen, the Netherlands
| | | | | |
Collapse
|
14
|
Tudorancea IM, Ciorpac M, Stanciu GD, Caratașu C, Săcărescu A, Ignat B, Burlui A, Rezuș E, Creangă I, Alexa-Stratulat T, Tudorancea I, Tamba BI. The Therapeutic Potential of the Endocannabinoid System in Age-Related Diseases. Biomedicines 2022; 10:2492. [PMID: 36289755 PMCID: PMC9599275 DOI: 10.3390/biomedicines10102492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/24/2022] [Accepted: 10/03/2022] [Indexed: 11/25/2022] Open
Abstract
The endocannabinoid system (ECS) dynamically regulates many aspects of mammalian physiology. ECS has gained substantial interest since growing evidence suggests that it also plays a major role in several pathophysiological conditions due to its ability to modulate various underlying mechanisms. Furthermore, cannabinoids, as components of the cannabinoid system (CS), have proven beneficial effects such as anti-inflammatory, immunomodulatory, neuromodulatory, antioxidative, and cardioprotective effects. In this comprehensive review, we aimed to describe the complex interaction between CS and most common age-related diseases such as neuro-degenerative, oncological, skeletal, and cardiovascular disorders, together with the potential of various cannabinoids to ameliorate the progression of these disorders. Since chronic inflammation is postulated as the pillar of all the above-mentioned medical conditions, we also discuss in this paper the potential of CS to ameliorate aging-associated immune system dysregulation.
Collapse
Affiliation(s)
- Ivona Maria Tudorancea
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
| | - Mitică Ciorpac
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
| | - Gabriela Dumitrița Stanciu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
| | - Cătălin Caratașu
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
| | - Alina Săcărescu
- Department of Medical Specialties II, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității, 700115 Iași, Romania
- Department of Neurology, Clinical Rehabilitation Hospital, 14 Pantelimon Halipa, 700661 Iași, Romania
| | - Bogdan Ignat
- Department of Neurology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Alexandra Burlui
- Department of Rheumatology and Rehabilitation, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Rehabilitation Hospital, 700661 Iași, Romania
| | - Elena Rezuș
- Department of Rheumatology and Rehabilitation, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Clinical Rehabilitation Hospital, 700661 Iași, Romania
| | - Ioana Creangă
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
- Oncology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
| | - Teodora Alexa-Stratulat
- Oncology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Oncology Department, Regional Institute of Oncology, 700483 Iași, Romania
| | - Ionuț Tudorancea
- Department of Morpho-Functional Sciences II, Discipline of Physiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iași, Romania
- Cardiology Clinic “St. Spiridon” County Clinical Emergency Hospital, 700111 Iași, Romania
| | - Bogdan Ionel Tamba
- Advanced Research and Development Center for Experimental Medicine (CEMEX), “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
- Department of Pharmacology, Clinical Pharmacology and Algesiology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 Universității Street, 700115 Iași, Romania
| |
Collapse
|
15
|
Ogłuszka M, Lipiński P, Starzyński RR. Effect of Omega-3 Fatty Acids on Telomeres-Are They the Elixir of Youth? Nutrients 2022; 14:nu14183723. [PMID: 36145097 PMCID: PMC9504755 DOI: 10.3390/nu14183723] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/17/2022] Open
Abstract
Telomeres are complexes consisting of tandem repeat DNA combined with associated proteins that play a key role in protecting the ends of chromosomes and maintaining genome stability. They are considered a biological clock, as they shorten in parallel with aging. Furthermore, short telomeres are associated with several age-related diseases. However, the variability in telomere shortening independent of chronological age suggests that it is a modifiable factor. In fact, it is regulated inter alia by genetic damage, cell division, aging, oxidative stress, and inflammation. A key question remains: how can we prevent accelerated telomere attrition and subsequent premature replicative senescence? A number of studies have explored the possible impact of omega-3 fatty acids on telomere shortening. This review summarizes published cross-sectional studies, randomized controlled trials, and rodent studies investigating the role of omega-3 fatty acids in telomere biology. It also covers a broad overview of the mechanism, currently favored in the field, that explains the impact of omega-3 fatty acids on telomeres—the food compound’s ability to modulate oxidative stress and inflammation. Although the results of the studies performed to date are not consistent, the vast majority indicate a beneficial effect of omega-3 fatty acids on telomere length.
Collapse
Affiliation(s)
- Magdalena Ogłuszka
- Department of Genomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, 05-552 Jastrzębiec, Poland
| | - Paweł Lipiński
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, 05-552 Jastrzębiec, Poland
| | - Rafał R. Starzyński
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, 05-552 Jastrzębiec, Poland
- Correspondence:
| |
Collapse
|
16
|
Tawfik HO, El-Hamaky AA, El-Bastawissy EA, Shcherbakov KA, Veselovsky AV, Gladilina YA, Zhdanov DD, El-Hamamsy MH. New Genetic Bomb Trigger: Design, Synthesis, Molecular Dynamics Simulation, and Biological Evaluation of Novel BIBR1532-Related Analogs Targeting Telomerase against Non-Small Cell Lung Cancer. Pharmaceuticals (Basel) 2022; 15:ph15040481. [PMID: 35455478 PMCID: PMC9025901 DOI: 10.3390/ph15040481] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 02/06/2023] Open
Abstract
Telomeres serve a critical function in cell replication and proliferation at every stage of the cell cycle. Telomerase is a ribonucleoprotein, responsible for maintaining the telomere length and chromosomal integrity of frequently dividing cells. Although it is silenced in most human somatic cells, telomere restoration occurs in cancer cells because of telomerase activation or alternative telomere lengthening. The telomerase enzyme is a universal anticancer target that is expressed in 85–95% of cancers. BIBR1532 is a selective non-nucleoside potent telomerase inhibitor that acts by direct noncompetitive inhibition. Relying on its structural features, three different series were designed, and 30 novel compounds were synthesized and biologically evaluated as telomerase inhibitors using a telomeric repeat amplification protocol (TRAP) assay. Target compounds 29a, 36b, and 39b reported the greatest inhibitory effect on telomerase enzyme with IC50 values of 1.7, 0.3, and 2.0 μM, respectively, while BIBR1532 displayed IC50 = 0.2 μM. Compounds 29a, 36b, and 39b were subsequently tested using a living-cell TRAP assay and were able to penetrate the cell membrane and inhibit telomerase inside living cancer cells. Compound 36b was tested for cytotoxicity against 60 cancer cell lines using the NCI (USA) procedure, and the % growth was minimally impacted, indicating telomerase enzyme selectivity. To investigate the interaction of compound 36b with the telomerase allosteric binding site, molecular docking and molecular dynamics simulations were used.
Collapse
Affiliation(s)
- Haytham O. Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (A.A.E.-H.); (E.A.E.-B.); (M.H.E.-H.)
- Correspondence: (H.O.T.); (D.D.Z.)
| | - Anwar A. El-Hamaky
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (A.A.E.-H.); (E.A.E.-B.); (M.H.E.-H.)
| | - Eman A. El-Bastawissy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (A.A.E.-H.); (E.A.E.-B.); (M.H.E.-H.)
| | - Kirill A. Shcherbakov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (K.A.S.); (A.V.V.); (Y.A.G.)
| | - Alexander V. Veselovsky
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (K.A.S.); (A.V.V.); (Y.A.G.)
| | - Yulia A. Gladilina
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (K.A.S.); (A.V.V.); (Y.A.G.)
| | - Dmitry D. Zhdanov
- Laboratory of Medical Biotechnology, Institute of Biomedical Chemistry, Pogodinskaya St. 10/8, 119121 Moscow, Russia; (K.A.S.); (A.V.V.); (Y.A.G.)
- Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklaya St. 6, 117198 Moscow, Russia
- Correspondence: (H.O.T.); (D.D.Z.)
| | - Mervat H. El-Hamamsy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt; (A.A.E.-H.); (E.A.E.-B.); (M.H.E.-H.)
| |
Collapse
|
17
|
Vegetables and Their Bioactive Compounds as Anti-Aging Drugs. Molecules 2022; 27:molecules27072316. [PMID: 35408714 PMCID: PMC9000296 DOI: 10.3390/molecules27072316] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 12/27/2022] Open
Abstract
Aging is a continuous process over time that is mainly related to natural alterations in mechanical–biological processes. This phenomenon is due to several factors, including the time and energy of biological processes. Aging can be attributed to biological factors such as oxidative stress, cell longevity, and stem cell senescence. Currently, aging is associated with several diseases, such as neurodegenerative diseases, cancer, and other diseases related to oxidative stress. In addition, certain natural molecules, including those derived from vegetables, have shown the ability to delay the aging process. Their effects are linked to different mechanisms of action, such as tissue regeneration and the activation of longevity and anti-senescence genes. The present work discusses the impact of vegetables, and bioactive compounds isolated from vegetables, against the physiological and pathological aging process and accompanying human diseases.
Collapse
|
18
|
Hamdan Y, Mazini L, Malka G. Exosomes and Micro-RNAs in Aging Process. Biomedicines 2021; 9:968. [PMID: 34440172 PMCID: PMC8393989 DOI: 10.3390/biomedicines9080968] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/06/2021] [Accepted: 07/15/2021] [Indexed: 12/14/2022] Open
Abstract
Exosomes are the main actors of intercellular communications and have gained great interest in the new cell-free regenerative medicine. These nanoparticles are secreted by almost all cell types and contain lipids, cytokines, growth factors, messenger RNA, and different non-coding RNA, especially micro-RNAs (mi-RNAs). Exosomes' cargo is released in the neighboring microenvironment but is also expected to act on distant tissues or organs. Different biological processes such as cell development, growth and repair, senescence, migration, immunomodulation, and aging, among others, are mediated by exosomes and principally exosome-derived mi-RNAs. Moreover, their therapeutic potential has been proved and reinforced by their use as biomarkers for disease diagnostics and progression. Evidence has increasingly shown that exosome-derived mi-RNAs are key regulators of age-related diseases, and their involvement in longevity is becoming a promising issue. For instance, mi-RNAs such as mi-RNA-21, mi-RNA-29, and mi-RNA-34 modulate tissue functionality and regeneration by targeting different tissues and involving different pathways but might also interfere with long life expectancy. Human mi-RNAs profiling is effectively related to the biological fluids that are reported differently between young and old individuals. However, their underlying mechanisms modulating cell senescence and aging are still not fully understood, and little was reported on the involvement of mi-RNAs in cell or tissue longevity. In this review, we summarize exosome biogenesis and mi-RNA synthesis and loading mechanism into exosomes' cargo. Additionally, we highlight the molecular mechanisms of exosomes and exosome-derived mi-RNA regulation in the different aging processes.
Collapse
Affiliation(s)
| | - Loubna Mazini
- Institute of Biological Sciences, Université Mohammed VI Polytechnique, Lot 660 Hay Moulay Rachid, Ben Guerir 3150, Morocco; (Y.H.); (G.M.)
| | | |
Collapse
|
19
|
Wang C, Songyang Z, Huang Y. TRIM28 inhibits alternative lengthening of telomere phenotypes by protecting SETDB1 from degradation. Cell Biosci 2021; 11:149. [PMID: 34330324 PMCID: PMC8325274 DOI: 10.1186/s13578-021-00660-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 07/15/2021] [Indexed: 01/04/2023] Open
Abstract
Background About 10–15% of tumor cells extend telomeres through the alternative lengthening of telomeres (ALT) mechanism, which is a recombination-dependent replication pathway. It is generally believed that ALT cells are related to the chromatin modification of telomeres. However, the mechanism of ALT needs to be further explored. Results Here we found that TRIM28/KAP1 is preferentially located on the telomeres of ALT cells and interacts with telomeric shelterin/telosome complex. Knocking down TRIM28 in ALT cells delayed cell growth, decreased the level of C-circle which is one kind of extrachromosomal circular telomeric DNA, increased the frequency of ALT-associated promyelocytic leukemia bodies (APBs), led to telomere prolongation and increased the telomere sister chromatid exchange in ALT cells. Mechanistically, TRIM28 protects telomere histone methyltransferase SETDB1 from degradation, thus maintaining the H3K9me3 heterochromatin state of telomere DNA. Conclusions Our work provides a model that TRIM28 inhibits alternative lengthening of telomere phenotypes by protecting SETDB1 from degradation. In general, our results reveal the mechanism of telomere heterochromatin maintenance and its effect on ALT, and TRIM28 may serve as a target for the treatment of ALT tumor cells. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00660-y.
Collapse
Affiliation(s)
- Chuanle Wang
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research and SYSU-BCM Joint Research Center, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China
| | - Zhou Songyang
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research and SYSU-BCM Joint Research Center, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China.,Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.,Verna and Marrs Mclean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.,Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Yan Huang
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research and SYSU-BCM Joint Research Center, School of Life Sciences, Sun Yat-Sen University, Guangzhou, 510275, China.
| |
Collapse
|
20
|
Zheng X, Wezel F, Azoitei A, Meessen S, Wang W, Najjar G, Wang X, Kraus JM, Kestler HA, John A, Zengerling F, Bolenz C, Günes C. Shorter Leukocyte Telomere Length Is Associated with Worse Survival of Patients with Bladder Cancer and Renal Cell Carcinoma. Cancers (Basel) 2021; 13:3774. [PMID: 34359672 PMCID: PMC8345040 DOI: 10.3390/cancers13153774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/19/2021] [Accepted: 07/22/2021] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Telomeres are protein-DNA complexes at the tips of linear chromosomes. They protect the DNA from end-to-end fusion and exonucleolytic degradation. Shortening of telomeric DNA during aging can generate dysfunctional telomeres, promoting tumorigenesis. More recent data indicate that both short and long telomeres of peripheral blood leukocyte (PBL) cells can serve as prognostic biomarkers for cancer risk and may be associated with survival of patients with solid cancers. Telomere length in PBL cells could also be a potential prognostic biomarker for survival in bladder cancer (BC) or renal cell carcinoma (RCC). METHODS The relative telomere length (RTL) of PBL cells was assessed in patients with BC (n = 144) and RCC (n = 144) by using qPCR. A control population of patients without malignant disease (NC, n = 73) was included for comparison. The correlation and association of RTL with histopathological parameters and overall survival (OS) were evaluated. RESULTS Patients with BC and RCC had significantly shorter telomeres compared to patients without malignant disease. Within the cancer cohorts, multivariate analysis revealed that short RTL is an independent predictor of worse survival in BC (p = 0.039) and RCC (p = 0.041). CONCLUSION Patients with BC and RCC had significantly shorter telomeres compared to the normal population. Shorter RTL in BC and RCC was an independent predictor of reduced survival.
Collapse
Affiliation(s)
- Xi Zheng
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Felix Wezel
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Anca Azoitei
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Sabine Meessen
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Wenya Wang
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Gregoire Najjar
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Xue Wang
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Johann M. Kraus
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (J.M.K.); (H.A.K.)
| | - Hans A. Kestler
- Institute of Medical Systems Biology, Ulm University, 89081 Ulm, Germany; (J.M.K.); (H.A.K.)
| | - Axel John
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Friedemann Zengerling
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Christian Bolenz
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| | - Cagatay Günes
- Department of Urology, Ulm University Hospital, 89081 Ulm, Germany; (X.Z.); (F.W.); (A.A.); (S.M.); (W.W.); (G.N.); (X.W.); (A.J.); (F.Z.); (C.B.)
| |
Collapse
|
21
|
Kemp BR, Ferraro KF. Are Biological Consequences of Childhood Exposures Detectable in Telomere Length Decades Later? J Gerontol A Biol Sci Med Sci 2021; 76:7-14. [PMID: 31956916 DOI: 10.1093/gerona/glaa019] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Indexed: 12/13/2022] Open
Abstract
Negative early-life exposures have been linked to a host of poor adult health outcomes, but are such early exposures associated with cellular senescence decades later? This study uses data from the Health and Retirement Study to examine the association between six childhood exposure domains (eg, socioeconomic disadvantage, risky parental behavior) and a biomarker of aging, telomere length, among 4,935 respondents. Telomere length is obtained from DNA of cells found in saliva and is measured as the telomere repeat copy number to single gene copy number ratio (T/S). Men who as children were exposed to risky parental behaviors or who reported risky adolescent behaviors have shorter telomeres (b = -0.031, p = .052; b = -0.041, p = .045, respectively); however, these relationships are attenuated after adjusting for adult risks and resources. Among women, parental substance abuse is associated with shorter telomeres even after adjusting for adult risks and resources (b = -0.041, p = .005). In addition, men and women whose mother lived at least until the age of 85 have longer telomeres than those without a long-lived mother (b = 0.021, p = .045; b = 0.032, p = .005, respectively). Taken together, the ways in which early-life exposures are associated with adult telomeres vary for men and women.
Collapse
Affiliation(s)
| | - Kenneth F Ferraro
- Center on Aging and the Life Course, Purdue University, West Lafayette, Indiana.,Department of Sociology, Purdue University, West Lafayette, Indiana
| |
Collapse
|
22
|
Fernandes SG, Dsouza R, Khattar E. External environmental agents influence telomere length and telomerase activity by modulating internal cellular processes: Implications in human aging. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 85:103633. [PMID: 33711516 DOI: 10.1016/j.etap.2021.103633] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/30/2021] [Accepted: 03/05/2021] [Indexed: 06/12/2023]
Abstract
External environment affects cellular physiological processes and impact the stability of our genome. The most important structural components of our linear chromosomes which endure the impact by these agents, are the chromosomal ends called telomeres. Telomeres preserve the integrity of our genome by preventing end to end fusions and telomeric loss through by inhibiting DNA damage response (DDR) activation. This is accomplished by the presence of a six membered shelterin complex at telomeres. Further, telomeres cannot be replicated by normal DNA polymerase and require a special enzyme called telomerase which is expressed only in stem cells, few immune cells and germ cells. Telomeres are rich in guanine content and thus become extremely prone to damage arising due to physiological processes like oxidative stress and inflammation. External environmental factors which includes various physical, biological and chemical agents also affect telomere homeostasis by increasing oxidative stress and inflammation. In the present review, we highlight the effect of these external factors on telomerase activity and telomere length. We also discuss how the external agents affect the physiological processes, thus modulating telomere stability. Further, we describe its implication in the development of aging and its related pathologies.
Collapse
Affiliation(s)
- Stina George Fernandes
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India
| | - Rebecca Dsouza
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India
| | - Ekta Khattar
- Sunandan Divatia School of Science, SVKM's NMIMS (Deemed to be University), Vile Parle West, Mumbai, 400056, India.
| |
Collapse
|
23
|
Stone RC, Aviv A, Paus R. Telomere Dynamics and Telomerase in the Biology of Hair Follicles and their Stem Cells as a Model for Aging Research. J Invest Dermatol 2021; 141:1031-1040. [PMID: 33509633 DOI: 10.1016/j.jid.2020.12.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023]
Abstract
In this review, we propose that telomere length dynamics play an important but underinvestigated role in the biology of the hair follicle (HF), a prototypic, cyclically remodeled miniorgan that shows an intriguing aging pattern in humans. Whereas the HF pigmentary unit ages quickly, its epithelial stem cell (ESC) component and regenerative capacity are surprisingly aging resistant. Telomerase-deficient mice with short telomeres display an aging phenotype of hair graying and hair loss that is attributed to impaired HF ESC mobilization. Yet, it remains unclear whether the function of telomerase and telomeres in murine HF biology translate to the human system. Therefore, we propose new directions for future telomere research of the human HF. Such research may guide the development of novel treatments for selected disorders of human hair growth or pigmentation (e.g., chemotherapy-induced alopecia, telogen effluvium, androgenetic alopecia, cicatricial alopecia, graying). It might also increase the understanding of the global role of telomeres in aging-related human disease.
Collapse
Affiliation(s)
- Rivka C Stone
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA.
| | - Abraham Aviv
- The Center of Human Development and Aging, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Ralf Paus
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA; Centre for Dermatology Research, University of Manchester, Manchester, United Kingdom; Monasterium Laboratory, Münster, Germany
| |
Collapse
|
24
|
Uyar B, Palmer D, Kowald A, Murua Escobar H, Barrantes I, Möller S, Akalin A, Fuellen G. Single-cell analyses of aging, inflammation and senescence. Ageing Res Rev 2020; 64:101156. [PMID: 32949770 PMCID: PMC7493798 DOI: 10.1016/j.arr.2020.101156] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 02/09/2023]
Abstract
Single-cell gene expression (transcriptomics) data are becoming robust and abundant, and are increasingly used to track organisms along their life-course. This allows investigation into how aging affects cellular transcriptomes, and how changes in transcriptomes may underlie aging, including chronic inflammation (inflammaging), immunosenescence and cellular senescence. We compiled and tabulated aging-related single-cell datasets published to date, collected and discussed relevant findings, and inspected some of these datasets ourselves. We specifically note insights that cannot (or not easily) be based on bulk data. For example, in some datasets, the fraction of cells expressing p16 (CDKN2A), one of the most prominent markers of cellular senescence, was reported to increase, in addition to its upregulated mean expression over all cells. Moreover, we found evidence for inflammatory processes in most datasets, some of these driven by specific cells of the immune system. Further, single-cell data are specifically useful to investigate whether transcriptional heterogeneity (also called noise or variability) increases with age, and many (but not all) studies in our review report an increase in such heterogeneity. Finally, we demonstrate some stability of marker gene expression patterns across closely similar studies and suggest that single-cell experiments may hold the key to provide detailed insights whenever interventions (countering aging, inflammation, senescence, disease, etc.) are affecting cells depending on cell type.
Collapse
Affiliation(s)
- Bora Uyar
- Bioinformatics and Omics Data Science Platform, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Daniel Palmer
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Axel Kowald
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Hugo Murua Escobar
- Rostock University Medical Center, Department of Hematology, Oncology and Palliative Medicine, Department of Medicine III, Rostock, Germany
| | - Israel Barrantes
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Steffen Möller
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany
| | - Altuna Akalin
- Bioinformatics and Omics Data Science Platform, Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Georg Fuellen
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock, Germany.
| |
Collapse
|
25
|
Molecular Mechanisms to Target Cellular Senescence in Hepatocellular Carcinoma. Cells 2020; 9:cells9122540. [PMID: 33255630 PMCID: PMC7761055 DOI: 10.3390/cells9122540] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) has emerged as a major cause of cancer-related death and is the most common type of liver cancer. Due to the current paucity of drugs for HCC therapy there is a pressing need to develop new therapeutic concepts. In recent years, the role of Serum Response Factor (SRF) and its coactivators, Myocardin-Related Transcription Factors A and B (MRTF-A and -B), in HCC formation and progression has received considerable attention. Targeting MRTFs results in HCC growth arrest provoked by oncogene-induced senescence. The induction of senescence acts as a tumor-suppressive mechanism and therefore gains consideration for pharmacological interventions in cancer therapy. In this article, we describe the key features and the functional role of senescence in light of the development of novel drug targets for HCC therapy with a focus on MRTFs.
Collapse
|
26
|
Bhattacharya M, Bhaumik P, Ghosh P, Majumder P, Kumar Dey S. Telomere Length Inheritance and Shortening in Trisomy 21. Fetal Pediatr Pathol 2020; 39:390-400. [PMID: 31514556 DOI: 10.1080/15513815.2019.1661049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Objective: Trisomy 21 is a genetic disorder that shows premature aging symptoms. As an aging marker, telomere length is therefore of importance in trisomy families. Methods: We included 63 maternally originated trisomy 21 and 77 control families with infants in the first data set; 48 trisomy 21 and 60 control children in the second set; and 14 paternally originated trisomy 21 families in the third data set. We used Southern blot to measure the telomere length. Results: (1) Offsprings' telomere length increased with parents' age (p < .0001). (2) Trisomy 21 infants had longer telomere than the controls (p < .0001). (3) Post-birth, the telomere attrition rate was higher in cases than in controls (58 bps/year vs. 38 bps/year). Conclusion: (1) Our data supports the older parents-longer gamete telomere hypothesis. (2) Trisomy 21 patients are born with longer telomeres, (3) with advancing trisomy 21 age, the telomere shortens more quickly than euploids.
Collapse
Affiliation(s)
- Mandar Bhattacharya
- Human Genetics, Maulana Abul Kalam Azad University of Technology, Kolkata, India
| | - Pranami Bhaumik
- Maulana Abul Kalam Azad University of Technology, Kolkata, India
| | - Priyanka Ghosh
- Maulana Abul Kalam Azad University of Technology, Kolkata, India
| | - Poulami Majumder
- Maulana Abul Kalam Azad University of Technology, Kolkata, India
| | | |
Collapse
|
27
|
Karakaya E, Akdur A, Ayvazoğlu Soy E, Moray G, Haberal M. Success Rate of Grafts With Multiple Renal Vessels in 3136 Kidney Transplants. EXP CLIN TRANSPLANT 2020; 19:14-19. [PMID: 32967599 DOI: 10.6002/ect.2020.0339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
OBJECTIVES Multiple renal vessels are often detected in living and deceased organ donors. In the past, transplant with multiple renal vessel grafts has been a contraindication because of high vascular and urological complication rates. However, improvements in vascular reconstruction and anastomosis techniques have allowed graft function to be maintained for many years. Here, we retrospectively evaluated transplant of multiple renal vessel grafts and graft survival and postoperative vascular and urological complications. MATERIALS AND METHODS From November 1975 to July 2020, there were 3136 renal transplants (716 deceased donors, 2420 living donors) performed in our center. There were 2167 living donors and 643 deceased donors with single renal vessel grafts and 253 living donors and 73 deceased donors with multiple renal vessel grafts. For anastomoses, external iliac, internal iliac, common iliac, and inferior epigastric arteries and external iliac veins were used. Cold ischemia time, anastomosis time, postoperative vascular and urological complications, acute tubular necrosis, creatinine clearance, serum creatinine levels, graft rejection episodes, and graft and patient survival rates were evaluated. RESULTS With regard to creatinine clearance, cold ischemia and anastomosis time, acute tubular necrosis, rejection episodes, and 1-, 2-, and 5-year posttransplant serum creatinine levels, there were no significant differences between the groups. Graft survival rates in the single renal vessel group were 92.9% at 1 year posttransplant and 78.3% at 5 years posttransplant; rates in the multiple renal vessel group were 93.1% at 1 year and 79.7% at 5 years. The corresponding patient survival rates were 95.5% (1 year) and 92.9% (5 years) for the single renal vessel group and 96.9% (1 year) and 87.2% (5 years) for the multiple renal vessel group. CONCLUSIONS Improved anastomosis and recon struction techniques have allowed the safe transplant of multiple renal vessel grafts that may remain functional for many years.
Collapse
Affiliation(s)
- Emre Karakaya
- From the Department of General Surgery, Baskent University, Ankara, Turkey
| | | | | | | | | |
Collapse
|
28
|
Aberrant Telomere Length in Circulating Cell-Free DNA as Possible Blood Biomarker with High Diagnostic Performance in Endometrial Cancer. Pathol Oncol Res 2020; 26:2281-2289. [PMID: 32462419 DOI: 10.1007/s12253-020-00819-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 05/12/2020] [Indexed: 02/07/2023]
Abstract
To investigate the diagnostic performance of relative telomere length (RTL) in cell-free DNA (cfDNA) for endometrioid endometrial cancer (EC). We measured RTL in cfDNA of 40 EC patients (65 ± 12 years) and 31 healthy controls (HC) (63 ± 13 years), excluding in both groups other oncologic and severe non-oncologic diseases to limit confounders. Circulating cfDNA was extracted from serum using the QIAamp DNA Blood Mini kit (Qiagen, Hilden, Germany). After the quantitative real-time polymerase chain reaction, telomere repeat copy number to single-gene copy number ratio was calculated. RTL in cfDNA was found to be significantly lower in EC patients than in HC (p < 0.0001). The diagnostic performance of cfDNA RTL was estimated with receiver operating characteristics (ROC) curve analysis, which showed a diagnostic accuracy for EC of 0.87 (95% CI: 0.79-0.95, p < 0.0001). The cutoff cfDNA RTL value of 2.505 (T/S copy ratio) reported a sensitivity of 80.0% (95% CI: 64.35-90.95) and a specificity of 80.65% (95% CI: 62.53-92.55). Significant differences of RTL among EC stages or grades (p = 0.85 and p = 0.89, respectively) were not observed. Our results suggest that cfDNA RTL analysis may be a diagnostic tool for EC detection since the early stage, whilst its diagnostic performance seems unsatisfactory for cancer progression, staging, and grading. However, further studies are needed to confirm these preliminary findings. In particular, future investigations should focus on high-risk patients (such as those with atypical endometrial hyperplasia) that may benefit from this tool, because TL shortening is not specific for EC and is influenced by other oncologic and non-oncologic diseases.
Collapse
|
29
|
Baginski M, Serbakowska K. In silico design of telomerase inhibitors. Drug Discov Today 2020; 25:1213-1222. [PMID: 32387261 DOI: 10.1016/j.drudis.2020.04.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/23/2020] [Accepted: 04/29/2020] [Indexed: 12/31/2022]
Abstract
Telomerase is a reverse transcriptase enzyme involved in DNA synthesis at the end of linear chromosomes. Unlike in most other cells, telomerase is reactivated most cancerous cells and, therefore, has become a promising new anticancer target. Despite extensive research, direct telomerase inhibitors have yet not been introduced to the clinics because of the complexity of this enzyme. Structures of this protein from simple organisms and human homology models are currently available and have been used in structure-based drug design efforts to find potential inhibitors. Different is silico strategies have been applied and different chemical groups have been explored. Here, we provide an overview of recent discoveries.
Collapse
Affiliation(s)
- Maciej Baginski
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland.
| | - Katarzyna Serbakowska
- Department of Pharmaceutical Technology and Biochemistry, Faculty of Chemistry, Gdansk University of Technology, 80-233 Gdansk, Poland
| |
Collapse
|
30
|
Nalobin D, Alipkina S, Gaidamaka A, Glukhov A, Khuchua Z. Telomeres and Telomerase in Heart Ontogenesis, Aging and Regeneration. Cells 2020; 9:cells9020503. [PMID: 32098394 PMCID: PMC7072777 DOI: 10.3390/cells9020503] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 12/14/2022] Open
Abstract
The main purpose of the review article is to assess the contributions of telomere length and telomerase activity to the cardiac function at different stages of development and clarify their role in cardiac disorders. It has been shown that the telomerase complex and telomeres are of great importance in many periods of ontogenesis due to the regulation of the proliferative capacity of heart cells. The review article also discusses the problems of heart regeneration and the identification of possible causes of dysfunction of telomeres and telomerase.
Collapse
Affiliation(s)
- Denis Nalobin
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russian
- Correspondence: ; Tel.: +7-916-939-0990
| | - Svetlana Alipkina
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russian
| | - Anna Gaidamaka
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russian
| | - Alexander Glukhov
- Faculty of Biology, Lomonosov Moscow State University, 119991 Moscow, Russian
- Department of Biochemistry, Sechenov First Moscow State Medical University, 119991 Moscow, Russian
| | - Zaza Khuchua
- Department of Biochemistry, Sechenov First Moscow State Medical University, 119991 Moscow, Russian
- Institute of Chemical Biology Ilia State University, 0162 Tbilisi, Georgia
- Division of Molecular and Cardiovascular Biology, Cincinnati Children’s Medical Center, Cincinnati, OH 45229, USA
| |
Collapse
|
31
|
Tateishi-Karimata H, Banerjee D, Ohyama T, Matsumoto S, Miyoshi D, Nakano SI, Sugimoto N. Hydroxyl groups in cosolutes regulate the G-quadruplex topology of telomeric DNA. Biochem Biophys Res Commun 2020; 525:S0006-291X(20)30313-2. [PMID: 32081425 DOI: 10.1016/j.bbrc.2020.02.045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 02/08/2020] [Indexed: 01/12/2023]
Abstract
Telomeric G-quadruplex topology has the ability to regulate telomerase activity, which counteracts the shortening of telomere with successive cell divisions, thereby causing genomic longevity. However, the detailed mechanism of G-quadruplexes topologies formed by telomeric sequences requires further investigation. In this study, we quantitatively investigated the effect of cosolutes, particularly the varying number of hydroxyl groups, on the structural transition between hybrid type and parallel G-quadruplexes formed by telomeric DNA sequences. Cosolutes with one or no hydroxyl groups in the vicinal position more efficiently induced the transition to parallel G-quadruplex from hybrid G-quadruplex than those with more hydroxyl groups. We also examined the effect of cosolute structures on the hydration of G-quadruplex formation; the results indicated that cosolutes with fewer hydroxyl groups lead to the release of greater amount of water during G-quadruplex formation. Molecular dynamics results showed that the parallel G-quadruplex was more dehydrated than the hybrid type G-quadruplex. Generally, a dehydrated structure is favored under crowding condition. Thus, depending on the surrounding cosolutes, the G-quadruplex topology can be controlled by the G-quadruplex hydration state.
Collapse
Affiliation(s)
| | - Dipanwita Banerjee
- Frontier Institute for Biomolecular Engineering Research (FIBER), Konan University, Japan
| | - Tatsuya Ohyama
- Frontier Institute for Biomolecular Engineering Research (FIBER), Konan University, Japan
| | - Saki Matsumoto
- Frontier Institute for Biomolecular Engineering Research (FIBER), Konan University, Japan
| | - Daisuke Miyoshi
- Graduate School of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Japan
| | - Shu-Ich Nakano
- Graduate School of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Japan
| | - Naoki Sugimoto
- Frontier Institute for Biomolecular Engineering Research (FIBER), Konan University, Japan; Graduate School of Frontiers of Innovative Research in Science and Technology (FIRST), Konan University, Japan.
| |
Collapse
|
32
|
Martínez-Cué C, Rueda N. Cellular Senescence in Neurodegenerative Diseases. Front Cell Neurosci 2020; 14:16. [PMID: 32116562 PMCID: PMC7026683 DOI: 10.3389/fncel.2020.00016] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 01/21/2020] [Indexed: 01/10/2023] Open
Abstract
Cellular senescence is a homeostatic biological process characterized by a permanent state of cell cycle arrest that can contribute to the decline of the regenerative potential and function of tissues. The increased presence of senescent cells in different neurodegenerative diseases suggests the contribution of senescence in the pathophysiology of these disorders. Although several factors can induce senescence, DNA damage, oxidative stress, neuroinflammation, and altered proteostasis have been shown to play a role in its onset. Oxidative stress contributes to accelerated aging and cognitive dysfunction stages affecting neurogenesis, neuronal differentiation, connectivity, and survival. During later life stages, it is implicated in the progression of cognitive decline, synapse loss, and neuronal degeneration. Also, neuroinflammation exacerbates oxidative stress, synaptic dysfunction, and neuronal death through the harmful effects of pro-inflammatory cytokines on cell proliferation and maturation. Both oxidative stress and neuroinflammation can induce DNA damage and alterations in DNA repair that, in turn, can exacerbate them. Another important feature associated with senescence is altered proteostasis. Because of the disruption in the function and balance of the proteome, senescence can modify the proper synthesis, folding, quality control, and degradation rate of proteins producing, in some diseases, misfolded proteins or aggregation of abnormal proteins. There is an extensive body of literature that associates cellular senescence with several neurodegenerative disorders including Alzheimer’s disease (AD), Down syndrome (DS), and Parkinson’s disease (PD). This review summarizes the evidence of the shared neuropathological events in these neurodegenerative diseases and the implication of cellular senescence in their onset or aggravation. Understanding the role that cellular senescence plays in them could help to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Carmen Martínez-Cué
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| | - Noemí Rueda
- Department of Physiology and Pharmacology, Faculty of Medicine, University of Cantabria, Santander, Spain
| |
Collapse
|
33
|
McCartney DL, Zhang F, Hillary RF, Zhang Q, Stevenson AJ, Walker RM, Bermingham ML, Boutin T, Morris SW, Campbell A, Murray AD, Whalley HC, Porteous DJ, Hayward C, Evans KL, Chandra T, Deary IJ, McIntosh AM, Yang J, Visscher PM, McRae AF, Marioni RE. An epigenome-wide association study of sex-specific chronological ageing. Genome Med 2019; 12:1. [PMID: 31892350 PMCID: PMC6938636 DOI: 10.1186/s13073-019-0693-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 11/15/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Advanced age is associated with cognitive and physical decline and is a major risk factor for a multitude of disorders. There is also a gap in life expectancy between males and females. DNA methylation differences have been shown to be associated with both age and sex. Here, we investigate age-by-sex differences in blood-based DNA methylation in an unrelated cohort of 2586 individuals between the ages of 18 and 87 years, with replication in a further 4450 individuals between the ages of 18 and 93 years. METHODS Linear regression models were applied, with stringent genome-wide significance thresholds (p < 3.6 × 10-8) used in both the discovery and replication data. A second, highly conservative mixed linear model method that better controls the false-positive rate was also applied, using the same genome-wide significance thresholds. RESULTS Using the linear regression method, 52 autosomal and 597 X-linked CpG sites, mapping to 251 unique genes, replicated with concordant effect size directions in the age-by-sex interaction analysis. The site with the greatest difference mapped to GAGE10, an X-linked gene. Here, DNA methylation levels remained stable across the male adult age range (DNA methylation by age r = 0.02) but decreased across female adult age range (DNA methylation by age r = - 0.61). One site (cg23722529) with a significant age-by-sex interaction also had a quantitative trait locus (rs17321482) that is a genome-wide significant variant for prostate cancer. The mixed linear model method identified 11 CpG sites associated with the age-by-sex interaction. CONCLUSION The majority of differences in age-associated DNA methylation trajectories between sexes are present on the X chromosome. Several of these differences occur within genes that have been implicated in sexually dimorphic traits.
Collapse
Affiliation(s)
- Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Futao Zhang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Robert F Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Qian Zhang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Anna J Stevenson
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Rosie M Walker
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK
| | - Mairead L Bermingham
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Thibaud Boutin
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Stewart W Morris
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Archie Campbell
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Alison D Murray
- Aberdeen Biomedical Imaging Centre, University of Aberdeen, Aberdeen, Scotland, UK
| | - Heather C Whalley
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, Scotland, UK
| | - David J Porteous
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Kathryn L Evans
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK
| | - Tamir Chandra
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
| | - Ian J Deary
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK
- Department of Psychology, University of Edinburgh, Edinburgh, Scotland, UK
| | - Andrew M McIntosh
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK
- Division of Psychiatry, Royal Edinburgh Hospital, University of Edinburgh, Edinburgh, Scotland, UK
| | - Jian Yang
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- Institute for Advanced Research, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Peter M Visscher
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Allan F McRae
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, Scotland, UK.
- Centre for Cognitive Ageing and Cognitive Epidemiology, University of Edinburgh, Edinburgh, Scotland, UK.
| |
Collapse
|
34
|
Abstract
Decades of research into stress responses have highlighted large variation among individuals, populations, and species, and the sources of this variation have been a center of research across disciplines. The most common measure of the vertebrate stress response is glucocorticoids. However, the predictive power of glucocorticoid responses to fitness is surprisingly low. This is partly because the hormone levels rapidly change in response to stressor exposure and elevated levels at one time point can indicate either that glucocorticoids are helping the organism cope with the stressor or that dysregulation of hormone release is harming the organism. Meaning, the fitness consequences of the stressor depends on how efficient the stress responses are at negating the harmful impacts of stressors to cells and tissues. To encompass the idea of the efficiency of stress responses and to integrate cellular and organismal stress responses, a new theoretical model called the Damage-Fitness Model was developed. The model focuses on the downstream effects of stress responses and predicts that the accumulation of damage in cells and tissues (e.g., persistent damage to proteins, lipids, and DNA) negatively impacts fitness components. In this mini-review, we examine evidence supporting the Damage-Fitness Model and explore new directions forward.
Collapse
Affiliation(s)
- Haruka Wada
- Department of Biological Sciences, Auburn University, Auburn, AL 36849, USA
| | - Britt Heidinger
- Department of Biological Sciences, North Dakota State University, Fargo, ND 58108, USA
| |
Collapse
|
35
|
Crassini K, Stevenson WS, Mulligan SP, Best OG. Molecular pathogenesis of chronic lymphocytic leukaemia. Br J Haematol 2019; 186:668-684. [DOI: 10.1111/bjh.16102] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Kyle Crassini
- Northern Blood Research Centre Kolling Institute of Medical Research SydneyNSWAustralia
| | - William S. Stevenson
- Northern Blood Research Centre Kolling Institute of Medical Research SydneyNSWAustralia
| | - Stephen P. Mulligan
- Northern Blood Research Centre Kolling Institute of Medical Research SydneyNSWAustralia
- School of Life and Environmental Science University of Sydney Sydney NSW Australia
| | - O. Giles Best
- Northern Blood Research Centre Kolling Institute of Medical Research SydneyNSWAustralia
- School of Life and Environmental Science University of Sydney Sydney NSW Australia
| |
Collapse
|
36
|
Abstract
Many recent advances have emerged in the telomere and telomerase fields. This Timeline article highlights the key advances that have expanded our views on the mechanistic underpinnings of telomeres and telomerase and their roles in ageing and disease. Three decades ago, the classic view was that telomeres protected the natural ends of linear chromosomes and that telomerase was a specific telomere-terminal transferase necessary for the replication of chromosome ends in single-celled organisms. While this concept is still correct, many diverse fields associated with telomeres and telomerase have substantially matured. These areas include the discovery of most of the key molecular components of telomerase, implications for limits to cellular replication, identification and characterization of human genetic disorders that result in premature telomere shortening, the concept that inhibiting telomerase might be a successful therapeutic strategy and roles for telomeres in regulating gene expression. We discuss progress in these areas and conclude with challenges and unanswered questions in the field.
Collapse
Affiliation(s)
- Jerry W Shay
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Woodring E Wright
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
37
|
Benetos A, Aviv A. Ancestry, Telomere Length, and Atherosclerosis Risk. ACTA ACUST UNITED AC 2019; 10:CIRCGENETICS.117.001718. [PMID: 28615296 DOI: 10.1161/circgenetics.117.001718] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Athanase Benetos
- From the Département de Médecine Gériatrique, CHRU de Nancy, The Institut national de la santé et de la recherche médicale, Université de Lorraine, France (A.B.); and Center of Human Development and Aging, New Jersey Medical School, Rutgers University, Newark (A.A.).
| | - Abraham Aviv
- From the Département de Médecine Gériatrique, CHRU de Nancy, The Institut national de la santé et de la recherche médicale, Université de Lorraine, France (A.B.); and Center of Human Development and Aging, New Jersey Medical School, Rutgers University, Newark (A.A.)
| |
Collapse
|
38
|
Ramos-Ibeas P, Pericuesta E, Peral-Sanchez I, Heras S, Laguna-Barraza R, Pérez-Cerezales S, Gutiérrez-Adán A. Longitudinal analysis of somatic and germ-cell telomere dynamics in outbred mice. Mol Reprod Dev 2019; 86:1033-1043. [PMID: 31209959 DOI: 10.1002/mrd.23218] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 05/15/2019] [Accepted: 05/16/2019] [Indexed: 11/10/2022]
Abstract
Although telomere length (TL) shortens with age in most tissues, an age-related increase in length has been described in sperm through a mechanism that is not yet fully understood. Changes in TL with age in the same individual have not been explored. This longitudinal study examines TL dynamics in somatic tissue and gametes during an entire lifespan in an outbred mouse population (from 8 to up to 114 weeks of age). Our findings indicate a reduced life expectancy in males compared to females (84.75 ± 9.23 vs. 113.16 ± 0.20 weeks) and significant variability in TL dynamics between individuals. While with aging, a clear reduction in TL was produced in somatic cells and oocytes, telomeres in sperm cells significantly lengthened. Finally, we found evidence indicating that telomere elongation in sperm during aging may be dependent on different mechanisms, such as the survival of spermatogonia with longer telomeres and the alternative lengthening of telomeres mechanism in meiotic and postmeiotic spermatogenic cells.
Collapse
Affiliation(s)
| | - Eva Pericuesta
- Departamento de Reproducción Animal, INIA, Madrid, Spain
| | | | - Sonia Heras
- Departamento de Reproducción Animal, INIA, Madrid, Spain
| | | | | | | |
Collapse
|
39
|
Geng L, Liu Z, Zhang W, Li W, Wu Z, Wang W, Ren R, Su Y, Wang P, Sun L, Ju Z, Chan P, Song M, Qu J, Liu GH. Chemical screen identifies a geroprotective role of quercetin in premature aging. Protein Cell 2019; 10:417-435. [PMID: 30069858 PMCID: PMC6538594 DOI: 10.1007/s13238-018-0567-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 06/25/2018] [Indexed: 12/18/2022] Open
Abstract
Aging increases the risk of various diseases. The main goal of aging research is to find therapies that attenuate aging and alleviate aging-related diseases. In this study, we screened a natural product library for geroprotective compounds using Werner syndrome (WS) human mesenchymal stem cells (hMSCs), a premature aging model that we recently established. Ten candidate compounds were identified and quercetin was investigated in detail due to its leading effects. Mechanistic studies revealed that quercetin alleviated senescence via the enhancement of cell proliferation and restoration of heterochromatin architecture in WS hMSCs. RNA-sequencing analysis revealed the transcriptional commonalities and differences in the geroprotective effects by quercetin and Vitamin C. Besides WS hMSCs, quercetin also attenuated cellular senescence in Hutchinson-Gilford progeria syndrome (HGPS) and physiological-aging hMSCs. Taken together, our study identifies quercetin as a geroprotective agent against accelerated and natural aging in hMSCs, providing a potential therapeutic intervention for treating age-associated disorders.
Collapse
Affiliation(s)
- Lingling Geng
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zunpeng Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weiqi Zhang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Wei Li
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zeming Wu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Wei Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ruotong Ren
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yao Su
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Peichang Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Liang Sun
- The MOH Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Beijing, 100730, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, 510632, China
| | - Piu Chan
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Guang-Hui Liu
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
40
|
Cui H, Liang W, Wang D, Guo K, Zhang Y. Establishment and Characterization of an Immortalized Porcine Oral Mucosal Epithelial Cell Line as a Cytopathogenic Model for Porcine Circovirus 2 Infection. Front Cell Infect Microbiol 2019; 9:171. [PMID: 31165052 PMCID: PMC6536654 DOI: 10.3389/fcimb.2019.00171] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/06/2019] [Indexed: 12/22/2022] Open
Abstract
Porcine circovirus 2 (PCV2) is a major etiological agent for porcine circovirus-associated diseases and causes enormous economic losses in domestic and overseas swine production. However, there are currently no suitable cell models to study the cytopathic effects (CPE) of PCV2 in vitro, which severely restricts the study of PCV2 pathogenesis. In the present study, we established an immortalized porcine oral mucosal epithelial cell line (hTERT-POMEC) by introducing the hTERT gene into primary porcine oral mucosal epithelial cells (POMECs) derived from a neonatal, unsuckled piglet. The hTERT-POMEC cells have a homogeneous cobblestone-like morphology and retain the basic physiological properties of primary POMECs. No chromosome abnormality and tumorigenicity transformation was observed in immortalized hTERT-POMECs. Viral infection assays demonstrated that PCV2 propagated and caused CPE in hTERT-POMECs. We conclude that the immortalized cell line hTERT-POMEC is a crucial tool for further research into the pathogenesis of PCV2.
Collapse
Affiliation(s)
- Hongjie Cui
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Wulong Liang
- School of Life Science, Shanxi University, Taiyuan, China
| | - Dahui Wang
- School of Agriculture and Forestry Engineering, Tongren University, Tongren, China
| | - Kangkang Guo
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Yanming Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
41
|
Samavat H, Xun X, Jin A, Wang R, Koh WP, Yuan JM. Association between prediagnostic leukocyte telomere length and breast cancer risk: the Singapore Chinese Health Study. Breast Cancer Res 2019; 21:50. [PMID: 30995937 PMCID: PMC6471852 DOI: 10.1186/s13058-019-1133-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 03/27/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Telomeres and telomerase play key roles in the chromosomal maintenance and stability. Recent epidemiological studies have shown that longer telomeres are associated with increased risk of several cancer types. However, epidemiological data for telomere length and risk of breast cancer are sparse. METHODS We prospectively studied the association between telomere length and risk of breast cancer in 14,305 middle-aged or older Chinese women of the Singapore Chinese Health Study including 442 incident breast cancer cases after 12.3 years of follow-up. Relative telomere length in peripheral blood leukocytes was quantified using a validated monochrome multiple quantitative polymerase chain reaction method. The Cox proportional hazard regression method was used to estimate hazard ratios (HRs) and the corresponding 95% confidence intervals (CIs) for breast cancer associated with longer telomeres after adjustment for potential confounders. RESULTS Longer telomeres were significantly associated with higher risk of breast cancer in a dose-dependent manner (Ptrend = 0.006); the highest quartile of telomere length was associated with a statistically significant 47% higher risk of breast cancer compared with the lowest quartile of telomere length after the adjustment for age and other known risk factors for breast cancer (HRQ4 vs Q1 = 1.47, 95% CI = 1.11, 1.94). CONCLUSIONS The findings of the present study support the hypothesis that longer telomeres may be a risk factor for breast cancer. Telomere length in peripheral blood leukocytes may be developed as a biomarker for breast cancer risk prediction.
Collapse
Affiliation(s)
- Hamed Samavat
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Cancer Pavilion, Suite 4C, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA. .,Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Xiaoshuang Xun
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Aizhen Jin
- Heath Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
| | - Renwei Wang
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Woon-Puay Koh
- Heath Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Jian-Min Yuan
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, UPMC Cancer Pavilion, Suite 4C, 5150 Centre Avenue, Pittsburgh, PA, 15232, USA.,Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
42
|
Ventura A, Pellegrini C, Cardelli L, Rocco T, Ciciarelli V, Peris K, Fargnoli MC. Telomeres and Telomerase in Cutaneous Squamous Cell Carcinoma. Int J Mol Sci 2019; 20:ijms20061333. [PMID: 30884806 PMCID: PMC6470499 DOI: 10.3390/ijms20061333] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 03/08/2019] [Accepted: 03/11/2019] [Indexed: 11/16/2022] Open
Abstract
The role of telomere biology and telomerase activation in skin cancers has been investigated in melanoma and basal cell carcinoma but limited evidence is available for cutaneous squamous cell carcinoma (cSCC). We will review the current knowledge on the role of telomere and telomerase pathway in cSCC pathogenesis. At the somatic level, both long and short telomere lengths have been described in cSCC. This telomere dichotomy is probably related to two different mechanisms of tumour initiation which determines two tumour subtypes. Telomere shortening is observed during the invasive progression from in situ forms of cSCC, such as Bowen's disease (BD) and actinic keratosis (AK), to invasive cSCC. At the germline level, controversial results have been reported on the association between constitutive telomere length and risk of cSCC. Approximately 75⁻85% of cSCC tumours are characterized by a high level of telomerase activity. Telomerase activation has been also reported in AKs and BD and in sun-damaged skin, thus supporting the hypothesis that UV modulates telomerase activity in the skin. Activating TERT promoter mutations have been identified in 32⁻70% of cSCCs, with the majority showing the UV-signature. No significant correlation was observed between TERT promoter mutations and cSCC clinico-pathological features. However, TERT promoter mutations have been recently suggested to be independent predictors of an adverse outcome. The attention on telomere biology and telomerase activity in cSCC is increasing for the potential implications in the development of effective tools for prognostic assessment and of therapeutic strategies in patients with cutaneous cSCC.
Collapse
Affiliation(s)
- Alessandra Ventura
- Department of Dermatology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, 67100 L'Aquila, Italy.
| | - Cristina Pellegrini
- Department of Dermatology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, 67100 L'Aquila, Italy.
| | - Ludovica Cardelli
- Department of Dermatology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, 67100 L'Aquila, Italy.
| | - Tea Rocco
- Department of Dermatology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, 67100 L'Aquila, Italy.
| | - Valeria Ciciarelli
- Department of Dermatology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, 67100 L'Aquila, Italy.
| | - Ketty Peris
- Institute of Dermatology, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Università Cattolica del Sacro Cuore, 00186 Rome, Italy.
| | - Maria Concetta Fargnoli
- Department of Dermatology, Department of Applied Clinical Sciences and Biotechnologies, University of L'Aquila, 67100 L'Aquila, Italy.
| |
Collapse
|
43
|
Nassrally MS, Lau A, Wise K, John N, Kotecha S, Lee KL, Brooks RF. Cell cycle arrest in replicative senescence is not an immediate consequence of telomere dysfunction. Mech Ageing Dev 2019; 179:11-22. [PMID: 30710559 DOI: 10.1016/j.mad.2019.01.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 12/19/2018] [Accepted: 01/28/2019] [Indexed: 11/15/2022]
Abstract
In replicative senescence, cells with critically-short telomeres activate a DNA-damage response leading to cell-cycle arrest, while those without telomere dysfunction would be expected to cycle normally. However, population growth declines more gradually than such a simple binary switch between cycling and non-cycling states would predict. We show here that late-passage cultures of human fibroblasts are not a simple mixture of cycling and non-cycling cells. Rather, although some cells had short cycle times comparable to those of younger cells, others continued to divide but with greatly extended cycle times, indicating a more-gradual approach to permanent arrest. Remarkably, in late passage cells, the majority showed prominent DNA-damage foci positive for 53BP1, yet many continued to divide. Evidently, the DNA-damage-response elicited by critically-short telomeres is not initially strong enough for complete cell-cycle arrest. A similar continuation of the cell cycle in the face of an active DNA-damage response was also seen in cells treated with a low dose of doxorubicin sufficient to produce multiple 53BP1 foci in all nuclei. Cell cycle checkpoint engagement in response to DNA damage is thus weaker than generally supposed, explaining why an accumulation of dysfunctional telomeres is needed before marked cell cycle elongation or permanent arrest is achieved.
Collapse
Affiliation(s)
- M Shamim Nassrally
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK
| | - Ashley Lau
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK
| | - Katherine Wise
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK
| | - Noah John
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK
| | - Sanjeev Kotecha
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK
| | - Kar Lai Lee
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK
| | - Robert F Brooks
- King's College London, Faculty of Life Sciences & Medicine, Department of Anatomy, Guy's Campus, LONDON SE1 1UL, UK; St George's, University of London, Molecular and Clinical Sciences Research Institute, Mailpoint J2A, Cranmer Terrace, London, SW17 0RE, UK.
| |
Collapse
|
44
|
Aviv A, Shay JW. Reflections on telomere dynamics and ageing-related diseases in humans. Philos Trans R Soc Lond B Biol Sci 2019; 373:rstb.2016.0436. [PMID: 29335375 PMCID: PMC5784057 DOI: 10.1098/rstb.2016.0436] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2017] [Indexed: 12/24/2022] Open
Abstract
Epidemiological studies have principally relied on measurements of telomere length (TL) in leucocytes, which reflects TL in other somatic cells. Leucocyte TL (LTL) displays vast variation across individuals—a phenomenon already observed in newborns. It is highly heritable, longer in females than males and in individuals of African ancestry than European ancestry. LTL is also longer in offspring conceived by older men. The traditional view regards LTL as a passive biomarker of human ageing. However, new evidence suggests that a dynamic interplay between selective evolutionary forces and TL might result in trade-offs for specific health outcomes. From a biological perspective, an active role of TL in ageing-related human diseases could occur because short telomeres increase the risk of a category of diseases related to restricted cell proliferation and tissue degeneration, including cardiovascular disease, whereas long telomeres increase the risk of another category of diseases related to increased proliferative growth, including major cancers. To understand the role of telomere biology in ageing-related diseases, it is essential to expand telomere research to newborns and children and seek further insight into the underlying causes of the variation in TL due to ancestry and geographical location. This article is part of the theme issue ‘Understanding diversity in telomere dynamics’.
Collapse
Affiliation(s)
- Abraham Aviv
- The Center of Human Development and Aging, Rutgers, The State University of New Jersey, New Jersey Medical School, Newark, NJ 07103, USA
| | - Jerry W Shay
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA.,Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| |
Collapse
|
45
|
Jiang CM, Liu X, Li CX, Qian HC, Chen D, Lai CQ, Shen LR. Anti-senescence effect and molecular mechanism of the major royal jelly proteins on human embryonic lung fibroblast (HFL-I) cell line. J Zhejiang Univ Sci B 2018; 19:960-972. [PMID: 30507079 PMCID: PMC6305251 DOI: 10.1631/jzus.b1800257] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 09/09/2018] [Accepted: 09/09/2018] [Indexed: 12/15/2022]
Abstract
Royal jelly (RJ) from honeybee has been widely used as a health promotion supplement. The major royal jelly proteins (MRJPs) have been identified as the functional component of RJ. However, the question of whether MRJPs have anti-senescence activity for human cells remains. Human embryonic lung fibroblast (HFL-I) cells were cultured in media containing no MRJPs (A), MRJPs at 0.1 mg/ml (B), 0.2 mg/ml (C), or 0.3 mg/ml (D), or bovine serum albumin (BSA) at 0.2 mg/ml (E). The mean population doubling levels of cells in media B, C, D, and E were increased by 12.4%, 31.2%, 24.0%, and 10.4%, respectively, compared with that in medium A. The cells in medium C also exhibited the highest relative proliferation activity, the lowest senescence, and the longest telomeres. Moreover, MRJPs up-regulated the expression of superoxide dismutase-1 (SOD1) and down-regulated the expression of mammalian target of rapamycin (MTOR), catenin beta like-1 (CTNNB1), and tumor protein p53 (TP53). Raman spectra analysis showed that there were two unique bands related to DNA synthesis materials, amide carbonyl group vibrations and aromatic hydrogens. These results suggest that MRJPs possess anti-senescence activity for the HFL-I cell line, and provide new knowledge illustrating the molecular mechanism of MRJPs as anti-senescence factors.
Collapse
Affiliation(s)
- Chen-min Jiang
- College of Biosystems Engineering and Food Science, Zhejiang University / Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs / Zhejiang Key Laboratory for Agri-Food Processing, Hangzhou 310058, China
| | - Xin Liu
- College of Biosystems Engineering and Food Science, Zhejiang University / Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs / Zhejiang Key Laboratory for Agri-Food Processing, Hangzhou 310058, China
| | - Chun-xue Li
- College of Biosystems Engineering and Food Science, Zhejiang University / Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs / Zhejiang Key Laboratory for Agri-Food Processing, Hangzhou 310058, China
| | - Hao-cheng Qian
- College of Biosystems Engineering and Food Science, Zhejiang University / Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs / Zhejiang Key Laboratory for Agri-Food Processing, Hangzhou 310058, China
| | - Di Chen
- College of Biosystems Engineering and Food Science, Zhejiang University / Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs / Zhejiang Key Laboratory for Agri-Food Processing, Hangzhou 310058, China
| | - Chao-qiang Lai
- USDA ARS Nutritional Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts 02111, the United States
| | - Li-rong Shen
- College of Biosystems Engineering and Food Science, Zhejiang University / Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs / Zhejiang Key Laboratory for Agri-Food Processing, Hangzhou 310058, China
| |
Collapse
|
46
|
Association of Telomere Length With Chromosomal Damage Among Chinese Workers Exposed to Vinyl Chloride Monomer. J Occup Environ Med 2018; 59:e252-e256. [PMID: 29215482 DOI: 10.1097/jom.0000000000001177] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE To explore the relationship between relative telomere length (RTL) and chromosomal damage represented by micronucleus (MN) frequencies among vinyl chloride monomer (VCM) -exposed workers. METHODS A group of 126 VCM-exposed workers, 60 internal controls, and 25 external controls were examined for RTL by Quantitative polymerase chain reaction and MN frequencies by cytokinesis-block micronucleus test. Cumulative exposure dose was used to estimate the exposure of VCM-exposed workers. RESULTS The RTL were significantly shorter in exposed workers and internal controls than in external controls. The exposed workers had significantly increased MN frequencies than both control groups. Additionally, MN frequencies were negatively associated with RTL in VCM-exposed group. CONCLUSIONS VCM exposure may alter telomere length, which could be a potential biomarker of susceptibility to chromosomal damage.
Collapse
|
47
|
Liu MY, Nemes A, Zhou QG. The Emerging Roles for Telomerase in the Central Nervous System. Front Mol Neurosci 2018; 11:160. [PMID: 29867352 PMCID: PMC5964194 DOI: 10.3389/fnmol.2018.00160] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 04/26/2018] [Indexed: 12/11/2022] Open
Abstract
Telomerase, a specialized ribonucleoprotein enzyme complex, maintains telomere length at the 3′ end of chromosomes, and functions importantly in stem cells, cancer and aging. Telomerase exists in neural stem cells (NSCs) and neural progenitor cells (NPCs), at a high level in the developing and adult brains of humans and rodents. Increasing studies have demonstrated that telomerase in NSCs/NPCs plays important roles in cell proliferation, neuronal differentiation, neuronal survival and neuritogenesis. In addition, recent works have shown that telomerase reverse transcriptase (TERT) can protect newborn neurons from apoptosis and excitotoxicity. However, to date, the link between telomerase and diseases in the central nervous system (CNS) is not well reviewed. Here, we analyze the evidence and summarize the important roles of telomerase in the CNS. Understanding the roles of telomerase in the nervous system is not only important to gain further insight into the process of the neural cell life cycle but would also provide novel therapeutic applications in CNS diseases such as neurodegenerative condition, mood disorders, aging and other ailments.
Collapse
Affiliation(s)
- Meng-Ying Liu
- Department of Clinical Pharmacology, Pharmacy College, Nanjing Medical University, Nanjing, China.,The Affiliated Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital, Nanjing, China
| | - Ashley Nemes
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Qi-Gang Zhou
- Department of Clinical Pharmacology, Pharmacy College, Nanjing Medical University, Nanjing, China.,Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
48
|
Leão R, Apolónio JD, Lee D, Figueiredo A, Tabori U, Castelo-Branco P. Mechanisms of human telomerase reverse transcriptase (hTERT) regulation: clinical impacts in cancer. J Biomed Sci 2018. [PMID: 29526163 PMCID: PMC5846307 DOI: 10.1186/s12929-018-0422-8] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background Limitless self-renewal is one of the hallmarks of cancer and is attained by telomere maintenance, essentially through telomerase (hTERT) activation. Transcriptional regulation of hTERT is believed to play a major role in telomerase activation in human cancers. Main body The dominant interest in telomerase results from its role in cancer. The role of telomeres and telomere maintenance mechanisms is well established as a major driving force in generating chromosomal and genomic instability. Cancer cells have acquired the ability to overcome their fate of senescence via telomere length maintenance mechanisms, mainly by telomerase activation. hTERT expression is up-regulated in tumors via multiple genetic and epigenetic mechanisms including hTERT amplifications, hTERT structural variants, hTERT promoter mutations and epigenetic modifications through hTERT promoter methylation. Genetic (hTERT promoter mutations) and epigenetic (hTERT promoter methylation and miRNAs) events were shown to have clinical implications in cancers that depend on hTERT activation. Knowing that telomeres are crucial for cellular self-renewal, the mechanisms responsible for telomere maintenance have a crucial role in cancer diseases and might be important oncological biomarkers. Thus, rather than quantifying TERT expression and its correlation with telomerase activation, the discovery and the assessment of the mechanisms responsible for TERT upregulation offers important information that may be used for diagnosis, prognosis, and treatment monitoring in oncology. Furthermore, a better understanding of these mechanisms may promote their translation into effective targeted cancer therapies. Conclusion Herein, we reviewed the underlying mechanisms of hTERT regulation, their role in oncogenesis, and the potential clinical applications in telomerase-dependent cancers.
Collapse
Affiliation(s)
- Ricardo Leão
- Division of Urology, Department of Surgery Princess Margaret Cancer Centre, University Health Network, 610 University Ave 3-130, Toronto, ON, M5G 2M9, Canada. .,Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON, M5G 1X8, Canada. .,Faculty of Medicine, University of Coimbra, R. Larga, 3004-504, Coimbra, Coimbra, Portugal. .,Department of Urology, Coimbra University Hospital, Coimbra, Portugal.
| | - Joana Dias Apolónio
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Edifício 2 - Ala Norte, 8005-139, Faro, Portugal.,Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal.,Algarve Biomedical Center, Campus Gambelas, Faro, Portugal
| | - Donghyun Lee
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON, M5G 1X8, Canada
| | - Arnaldo Figueiredo
- Faculty of Medicine, University of Coimbra, R. Larga, 3004-504, Coimbra, Coimbra, Portugal.,Department of Urology, Coimbra University Hospital, Coimbra, Portugal
| | - Uri Tabori
- Arthur and Sonia Labatt Brain Tumor Research Center, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, ON, M5G 1X8, Canada.,Division of Haematology/Oncology, The Hospital for Sick Children, 555 University Avenue, Toronto, M5G 1X8ON, Canada
| | - Pedro Castelo-Branco
- Regenerative Medicine Program, Department of Biomedical Sciences and Medicine, University of Algarve, Edifício 2 - Ala Norte, 8005-139, Faro, Portugal.,Centre for Biomedical Research (CBMR), University of Algarve, Faro, Portugal.,Algarve Biomedical Center, Campus Gambelas, Faro, Portugal
| |
Collapse
|
49
|
Lu AT, Xue L, Salfati EL, Chen BH, Ferrucci L, Levy D, Joehanes R, Murabito JM, Kiel DP, Tsai PC, Yet I, Bell JT, Mangino M, Tanaka T, McRae AF, Marioni RE, Visscher PM, Wray NR, Deary IJ, Levine ME, Quach A, Assimes T, Tsao PS, Absher D, Stewart JD, Li Y, Reiner AP, Hou L, Baccarelli AA, Whitsel EA, Aviv A, Cardona A, Day FR, Wareham NJ, Perry JRB, Ong KK, Raj K, Lunetta KL, Horvath S. GWAS of epigenetic aging rates in blood reveals a critical role for TERT. Nat Commun 2018; 9:387. [PMID: 29374233 PMCID: PMC5786029 DOI: 10.1038/s41467-017-02697-5] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/19/2017] [Indexed: 02/02/2023] Open
Abstract
DNA methylation age is an accurate biomarker of chronological age and predicts lifespan, but its underlying molecular mechanisms are unknown. In this genome-wide association study of 9907 individuals, we find gene variants mapping to five loci associated with intrinsic epigenetic age acceleration (IEAA) and gene variants in three loci associated with extrinsic epigenetic age acceleration (EEAA). Mendelian randomization analysis suggests causal influences of menarche and menopause on IEAA and lipoproteins on IEAA and EEAA. Variants associated with longer leukocyte telomere length (LTL) in the telomerase reverse transcriptase gene (TERT) paradoxically confer higher IEAA (P < 2.7 × 10-11). Causal modeling indicates TERT-specific and independent effects on LTL and IEAA. Experimental hTERT-expression in primary human fibroblasts engenders a linear increase in DNA methylation age with cell population doubling number. Together, these findings indicate a critical role for hTERT in regulating the epigenetic clock, in addition to its established role of compensating for cell replication-dependent telomere shortening.
Collapse
Affiliation(s)
- Ake T Lu
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Luting Xue
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Elias L Salfati
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Brian H Chen
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
- National Heart, Lung and Blood Institute, Bethesda, MD, 20824-0105, USA
| | - Luigi Ferrucci
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Daniel Levy
- National Heart, Lung and Blood Institute, Bethesda, MD, 20824-0105, USA
| | - Roby Joehanes
- National Heart, Lung and Blood Institute, Bethesda, MD, 20824-0105, USA
| | - Joanne M Murabito
- Department of Medicine, Section of General Medicine, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Douglas P Kiel
- Institute for Aging Research, Hebrew SeniorLife, Beth Israel Deaconess Medical Centre, Harvard Medical School, Boston, MA, 02215, USA
| | - Pei-Chien Tsai
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, SE1 7EH, UK
| | - Idil Yet
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, SE1 7EH, UK
| | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, SE1 7EH, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, SE1 7EH, UK
| | - Toshiko Tanaka
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Allan F McRae
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, QLD, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Riccardo E Marioni
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, QLD, Australia
- Centre for Cognitive Aging and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
- Medical Genetics Section, Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, UK
| | - Peter M Visscher
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, QLD, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Naomi R Wray
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, 4072, QLD, Australia
- Queensland Brain Institute, The University of Queensland, Brisbane, 4072, QLD, Australia
| | - Ian J Deary
- Centre for Cognitive Aging and Cognitive Epidemiology, Department of Psychology, University of Edinburgh, 7 George Square, Edinburgh, EH8 9JZ, UK
| | - Morgan E Levine
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Austin Quach
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Themistocles Assimes
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Philip S Tsao
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- VA Palo Alto Health Care System, Palo Alto, CA, 94304, USA
| | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, 35806, USA
| | - James D Stewart
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Yun Li
- Department of Genetics, School of Medicine, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA
| | - Alex P Reiner
- Fred Hutchinson Cancer Research Center Box 358080, WHI Clinical Coordinating Ctr/Public Health Sciences M3-A4, Seattle, WA, 98109, USA
| | - Lifang Hou
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University Chicago, Evanston, IL, 60611, USA
- Center for Population Epigenetics, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University Chicago, Evanston, IL, 60611, USA
| | - Andrea A Baccarelli
- Laboratory of Environmental Epigenetics, Departments of Environmental Health Sciences Epidemiology, Columbia University Mailman School of Public Health, New York, NY, 10032, USA
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, 27599, USA
- Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, 27516, USA
| | - Abraham Aviv
- The Center for Human Development and Aging, University of Medicine and Dentistry, New Jersey Medical School, Rutgers, Newark, NJ, 07103, USA
| | - Alexia Cardona
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, CB2 0SL, UK
| | - Felix R Day
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, CB2 0SL, UK
| | - Nicholas J Wareham
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, CB2 0SL, UK
| | - John R B Perry
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, CB2 0SL, UK
| | - Ken K Ong
- Medical Research Council (MRC) Epidemiology Unit, University of Cambridge School of Clinical Medicine, Institute of Metabolic Science, Cambridge Biomedical Campus, Cambridge, CB2 0SL, UK
- Department of Paediatrics, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, CB2 0SP, UK
| | - Kenneth Raj
- Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, Oxfordshire, OX11 0RQ, UK
| | - Kathryn L Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, 02118, USA
| | - Steve Horvath
- Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Biostatistics, School of Public Health, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
50
|
Telomeres: Implications for Cancer Development. Int J Mol Sci 2018; 19:ijms19010294. [PMID: 29351238 PMCID: PMC5796239 DOI: 10.3390/ijms19010294] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/12/2018] [Accepted: 01/16/2018] [Indexed: 12/31/2022] Open
Abstract
Telomeres facilitate the protection of natural ends of chromosomes from constitutive exposure to the DNA damage response (DDR). This is most likely achieved by a lariat structure that hides the linear telomeric DNA through protein-protein and protein-DNA interactions. The telomere shortening associated with DNA replication in the absence of a compensatory mechanism culminates in unmasked telomeres. Then, the subsequent activation of the DDR will define the fate of cells according to the functionality of cell cycle checkpoints. Dysfunctional telomeres can suppress cancer development by engaging replicative senescence or apoptotic pathways, but they can also promote tumour initiation. Studies in telomere dynamics and karyotype analysis underpin telomere crisis as a key event driving genomic instability. Significant attainment of telomerase or alternative lengthening of telomeres (ALT)-pathway to maintain telomere length may be permissive and required for clonal evolution of genomically-unstable cells during progression to malignancy. We summarise current knowledge of the role of telomeres in the maintenance of chromosomal stability and carcinogenesis.
Collapse
|