1
|
farrukh S, Baig S, Hussain R, Imad R, kulsoom O, Yousaf Rana M. Identification of polymorphic alleles in TERC and TERT gene reprogramming the telomeres of newborn and legacy with parental health. Saudi J Biol Sci 2024; 31:103897. [PMID: 38192544 PMCID: PMC10772381 DOI: 10.1016/j.sjbs.2023.103897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/02/2023] [Accepted: 12/08/2023] [Indexed: 01/10/2024] Open
Abstract
Telomere and telomerase genes (TERC and TERT) highlighted many novel genetic polymorphisms related to common diseases. This study explored the polymorphic alleles of TERC and TERT gene in parents-newborn (triad) and its association with telomere length (TL) and parental diseases (mother: Gestational Diabetes Mellitus (GDM), Preeclampsia, fathers: Diabetes, Hypertension). In this cross-sectional study, the blood samples (n = 612) were collected from parents-newborn triad (204 each) for TL (T/S ratio) quantification by using qPCR, and gene (TERC and TERT) polymorphism was detected by Sanger sequencing. The correlation analysis was used to find an association between paternal TL (T/S ratio) and newborn TL. The multivariate linear regression was applied to determine the effect of parents genes and diseases on newborn TL. A positive association (r = 0.42,0.39) (p < 0.0001) among parents and newborn TL was observed. In the diseased group, both TERC (rs10936599) and TERT (rs2736100) genes had a high frequency of allele C in newborns (OR = 0.94, P = 0.90, OR = 4.24, P = 0.012). However, among parents, TERT gene [Mother CC (B = 0.575; P = 0.196), Father CC (B = -0.739; P = 0.071)] was found significant contributing factor for Newborn TL. Diseased parents with T/T and A/C genotypes had longer newborn TL (2.82 ± 2.43, p < 0.022; 1.80 ± 1.20, p < 0.00) than the C/C genotype. Therefore, the study, confirmed that major allele C of TERC and TERT genes is associated with smaller TL in diseased parents-newborns of the targeted population.
Collapse
Affiliation(s)
- Sadia farrukh
- Department of Biochemistry, Ziauddin University, Karachi, Pakistan
- Department of Community Health Sciences, The Agha Khan University, Karachi, Pakistan
| | - Saeeda Baig
- Department of Biochemistry, Ziauddin University, Karachi, Pakistan
| | - Rubina Hussain
- Department Gynecology and obstetrician, Ziauddin university and hospitals, Karachi, Pakistan
| | - Rehan Imad
- Department of Molecular medicine, Ziauddin University Karachi, Pakistan
| | - Ome kulsoom
- Department Gynecology and obstetrician, Ziauddin hospital, Karachi, Pakistan
| | - Mehreen Yousaf Rana
- Department Gynecology and obstetrician, Ziauddin hospital, Karachi, Pakistan
| |
Collapse
|
2
|
Novau-Ferré N, Rojas M, Gutierrez-Tordera L, Arcelin P, Folch J, Papandreou C, Bulló M. Lipoprotein Particle Profiles Associated with Telomere Length and Telomerase Complex Components. Nutrients 2023; 15:nu15112624. [PMID: 37299586 DOI: 10.3390/nu15112624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/29/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Telomere length (TL) is a well-known marker of age-related diseases. Oxidative stress and inflammation increase the rate of telomere shortening, triggering cellular senescence. Although lipoproteins could have anti-inflammatory and proinflammatory functional properties, the relationship between lipoprotein particles with TL and telomerase activity-related genes has not been investigated much. In this study, we assessed the associations of lipoprotein subfractions with telomere length, TERT, and WRAP53 expression in a total of 54 pre-diabetic subjects from the EPIRDEM study. We regressed TL, TERT, and WRAP53 on 12 lipoprotein subclasses, employing a Gaussian linear regression method with Lasso penalty to determine a lipoprotein profile associated with telomere-related parameters. The covariates included age, sex, body mass index (BMI), dyslipidemia, statin consumption, and physical activity leisure time. We identified a lipoprotein profile composed of four lipoprotein subfractions associated with TL (Pearson r = 0.347, p-value = 0.010), two lipoprotein subfractions associated with TERT expression (Pearson r = 0.316, p-value = 0.020), and five lipoprotein subfractions associated with WRAP53 expression (Pearson r = 0.379, p-value =0.005). After adjusting for known confounding factors, most lipoprotein profiles maintained the association with TL, TERT, and WRAP53. Overall, medium and small-sized HDL particles were associated with shorter telomeres and lower expression of TERT and WRAP53. Large HDL particles were associated with longer telomere and lower expression of WRAP53, but not with TERT. Our results suggest that the lipoprotein profiles are associated with telomere length, TERT, and WRAP53 expression and should be considered when assessing the risk of chronic diseases.
Collapse
Affiliation(s)
- Nil Novau-Ferré
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology(TecnATox), Rovira i Virgili University, 43201 Reus, Spain
| | - Melina Rojas
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology(TecnATox), Rovira i Virgili University, 43201 Reus, Spain
| | - Laia Gutierrez-Tordera
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology(TecnATox), Rovira i Virgili University, 43201 Reus, Spain
| | - Pierre Arcelin
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Atención Básica de Salud (ABS) Reus V. Centro de Atención Primaria Marià Fortuny, SAGESSA, 43204 Reus, Spain
| | - Jaume Folch
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology(TecnATox), Rovira i Virgili University, 43201 Reus, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Carlos III Health Institute, 28031 Madrid, Spain
| | - Christopher Papandreou
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology(TecnATox), Rovira i Virgili University, 43201 Reus, Spain
- Department of Nutrition and Dietetics Sciences, School of Health Sciences, Hellenic Mediterranean University (HMU), 72300 Siteia, Greece
| | - Mònica Bulló
- Nutrition and Metabolic Health Research Group, Department of Biochemistry and Biotechnology, Rovira i Virgili University (URV), 43201 Reus, Spain
- Institute of Health Pere Virgili (IISPV), 43204 Reus, Spain
- Center of Environmental, Food and Toxicological Technology(TecnATox), Rovira i Virgili University, 43201 Reus, Spain
- Atención Básica de Salud (ABS) Reus V. Centro de Atención Primaria Marià Fortuny, SAGESSA, 43204 Reus, Spain
- CIBER Physiology of Obesity and Nutrition (CIBEROBN), Carlos III Health Institute, 28029 Madrid, Spain
| |
Collapse
|
3
|
Banerjee P, Rosales JE, Chau K, Nguyen MTH, Kotla S, Lin SH, Deswal A, Dantzer R, Olmsted-Davis EA, Nguyen H, Wang G, Cooke JP, Abe JI, Le NT. Possible molecular mechanisms underlying the development of atherosclerosis in cancer survivors. Front Cardiovasc Med 2023; 10:1186679. [PMID: 37332576 PMCID: PMC10272458 DOI: 10.3389/fcvm.2023.1186679] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Cancer survivors undergone treatment face an increased risk of developing atherosclerotic cardiovascular disease (CVD), yet the underlying mechanisms remain elusive. Recent studies have revealed that chemotherapy can drive senescent cancer cells to acquire a proliferative phenotype known as senescence-associated stemness (SAS). These SAS cells exhibit enhanced growth and resistance to cancer treatment, thereby contributing to disease progression. Endothelial cell (EC) senescence has been implicated in atherosclerosis and cancer, including among cancer survivors. Treatment modalities for cancer can induce EC senescence, leading to the development of SAS phenotype and subsequent atherosclerosis in cancer survivors. Consequently, targeting senescent ECs displaying the SAS phenotype hold promise as a therapeutic approach for managing atherosclerotic CVD in this population. This review aims to provide a mechanistic understanding of SAS induction in ECs and its contribution to atherosclerosis among cancer survivors. We delve into the mechanisms underlying EC senescence in response to disturbed flow and ionizing radiation, which play pivotal role in atherosclerosis and cancer. Key pathways, including p90RSK/TERF2IP, TGFβR1/SMAD, and BH4 signaling are explored as potential targets for cancer treatment. By comprehending the similarities and distinctions between different types of senescence and the associated pathways, we can pave the way for targeted interventions aim at enhancing the cardiovascular health of this vulnerable population. The insights gained from this review may facilitate the development of novel therapeutic strategies for managing atherosclerotic CVD in cancer survivors.
Collapse
Affiliation(s)
- Priyanka Banerjee
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Julia Enterría Rosales
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- School of Medicine, Instituto Tecnológico de Monterrey, Guadalajara, Mexico
| | - Khanh Chau
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Minh T. H. Nguyen
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
- Department of Life Science, University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven H. Lin
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elizabeth A. Olmsted-Davis
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Hung Nguyen
- Cancer Division, Burnett School of Biomedical Science, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Guangyu Wang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - John P. Cooke
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
4
|
Booth LK, Redgrave RE, Tual-Chalot S, Spyridopoulos I, Phillips HM, Richardson GD. Heart Disease and Ageing: The Roles of Senescence, Mitochondria, and Telomerase in Cardiovascular Disease. Subcell Biochem 2023; 103:45-78. [PMID: 37120464 DOI: 10.1007/978-3-031-26576-1_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
During ageing molecular damage leads to the accumulation of several hallmarks of ageing including mitochondrial dysfunction, cellular senescence, genetic instability and chronic inflammation, which contribute to the development and progression of ageing-associated diseases including cardiovascular disease. Consequently, understanding how these hallmarks of biological ageing interact with the cardiovascular system and each other is fundamental to the pursuit of improving cardiovascular health globally. This review provides an overview of our current understanding of how candidate hallmarks contribute to cardiovascular diseases such as atherosclerosis, coronary artery disease and subsequent myocardial infarction, and age-related heart failure. Further, we consider the evidence that, even in the absence of chronological age, acute cellular stress leading to accelerated biological ageing expedites cardiovascular dysfunction and impacts on cardiovascular health. Finally, we consider the opportunities that modulating hallmarks of ageing offer for the development of novel cardiovascular therapeutics.
Collapse
Affiliation(s)
- Laura K Booth
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Rachael E Redgrave
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Helen M Phillips
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
5
|
Brandt M, Dörschmann H, Khraisat S, Knopp T, Ringen J, Kalinovic S, Garlapati V, Siemer S, Molitor M, Göbel S, Stauber R, Karbach SH, Münzel T, Daiber A, Wenzel P. Telomere Shortening in Hypertensive Heart Disease Depends on Oxidative DNA Damage and Predicts Impaired Recovery of Cardiac Function in Heart Failure. Hypertension 2022; 79:2173-2184. [PMID: 35862118 DOI: 10.1161/hypertensionaha.121.18935] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Heart failure (HF) coincides with cardiomyocyte telomere shortening. Arterial hypertension is the most prominent risk factor for HF. Both HF and arterial hypertension are associated with dysregulation of the neurohormonal axis. How neurohormonal activation is linked to telomere shortening in the pathogenesis of HF is incompletely understood. METHODS Cardiomyocyte telomere length was assessed in a mouse model of hypertensive HF induced by excess neurohormonal activation (AngII [angiotensin II] infusion, high salt diet, and uninephrectomy), in AngII-stimulated cardiomyocytes and in endomyocardial biopsies from patients with HF. Superoxide production, expression of NOX2 (NADPH oxidase 2) and PRDX1 (peroxiredoxin 1) and HDAC6 (histone deacetylase 6) activity were assessed. RESULTS Telomere shortening occurred in vitro and in vivo, correlating with both left ventricular (LV) dilatation and LV systolic function impairment. Telomere shortening coincided with increased superoxide production, increased NOX2 expression, increased HDAC6 activity, loss of the telomere-specific antioxidant PRDX1, and increased oxidative DNA-damage. NOX2 knockout prevented PRDX1 depletion, DNA-damage and telomere shortening confirming this enzyme as a critical source of reactive oxygen species. Cotreatment with the NOX inhibitor apocynin ameliorated hypertensive HF and telomere shortening. Similarly, treatment with the HDAC6 inhibitor tubastatin A, which increases PRDX1 bioavailability, prevented telomere shortening in adult cardiomyocytes. To explore the clinical relevance of our findings, we examined endomyocardial biopsies from an all-comer population of patients with HF with reduced ejection fraction. Here, cardiomyocyte telomere length predicted the recovery of cardiac function. CONCLUSIONS Cardiomyocyte telomere shortening and oxidative damage in heart failure with reduced ejection fraction induced by excess neurohormonal activation depends on NOX2-derived superoxide and may help to stratify HF therapy.
Collapse
Affiliation(s)
- Moritz Brandt
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Hendrik Dörschmann
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Sana'a Khraisat
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Tanja Knopp
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Julia Ringen
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Sanela Kalinovic
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Venkata Garlapati
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Svenja Siemer
- Department of Otolaryngology, Head and Neck Surgery, University Medical Center Mainz' Mainz' Germany (S.S., R.S.)
| | - Michael Molitor
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Sebastian Göbel
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Roland Stauber
- Department of Otolaryngology, Head and Neck Surgery, University Medical Center Mainz' Mainz' Germany (S.S., R.S.)
| | - Susanne Helena Karbach
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Thomas Münzel
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Andreas Daiber
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Philip Wenzel
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Department of Biochemistry, Cardiovascular Research Institute Maastricht School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands (P.W.)
| |
Collapse
|
6
|
Yilmaz S, Bedir E, Ballar Kirmizibayrak P. The role of cycloastragenol at the intersection of NRF2/ARE, telomerase, and proteasome activity. Free Radic Biol Med 2022; 188:105-116. [PMID: 35718303 DOI: 10.1016/j.freeradbiomed.2022.06.230] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 10/18/2022]
Abstract
Aging is well-characterized by the gradual decline of cellular functionality. As redox balance, proteostasis, and telomerase systems have been found to be associated with aging and age-related diseases, targeting these systems with small compounds has been considered a promising therapeutic approach. Cycloastragenol (CA), a small molecule telomerase activator obtained from Astragalus species, has been reported to positively affect several age-related pathophysiologies, but the mechanisms underlying CA activity have yet to be reported. Here, we presented that CA increased NRF2 nuclear localization and activity leading to upregulation of cytoprotective enzymes and attenuation of oxidative stress-induced ROS levels. Furthermore, CA-mediated induction of telomerase activity was found to be regulated by NRF2. CA not only increased the expression of hTERT but also its nuclear localization via upregulating the Hsp90-chaperon complex. In addition to modulating nuclear hTERT levels at unstressed conditions, CA alleviated oxidative stress-induced mitochondrial hTERT levels while increasing nuclear hTERT levels. Concomitantly, H2O2-induced mitochondrial ROS level was found to be significantly decreased by CA administration. Our data also revealed that CA strongly enhanced proteasome activity and assembly. More importantly, the proteasome activator effect of CA is dependent on the induction of telomerase activity, which is mediated by NRF2 system. In conclusion, our results not only revealed the cross-talk among NRF2, telomerase, and proteasome systems but also that CA functions at the intersection of these three major aging-related cellular pathways.
Collapse
Affiliation(s)
- Sinem Yilmaz
- Department of Biotechnology, Graduate School of Natural and Applied Sciences, Ege University, Bornova, Izmir, Turkey; Department of Bioengineering, Faculty of Engineering, University of Alanya Aladdin Keykubat, Antalya, Turkey
| | - Erdal Bedir
- Department of Bioengineering, Izmir Institute of Technology, 35430, Urla, Izmir, Turkey.
| | | |
Collapse
|
7
|
Accelerated Growth, Differentiation, and Ploidy with Reduced Proliferation of Right Ventricular Cardiomyocytes in Children with Congenital Heart Defect Tetralogy of Fallot. Cells 2022; 11:cells11010175. [PMID: 35011735 PMCID: PMC8750260 DOI: 10.3390/cells11010175] [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: 11/27/2021] [Revised: 12/29/2021] [Accepted: 01/01/2022] [Indexed: 02/06/2023] Open
Abstract
The myocardium of children with tetralogy of Fallot (TF) undergoes hemodynamic overload and hypoxemia immediately after birth. Comparative analysis of changes in the ploidy and morphology of the right ventricular cardiomyocytes in children with TF in the first years of life demonstrated their significant increase compared with the control group. In children with TF, there was a predominantly diffuse distribution of Connexin43-containing gap junctions over the cardiomyocytes sarcolemma, which redistributed into the intercalated discs as cardiomyocytes differentiation increased. The number of Ki67-positive cardiomyocytes varied greatly and amounted to 7.0–1025.5/106 cardiomyocytes and also were decreased with increased myocytes differentiation. Ultrastructural signs of immaturity and proliferative activity of cardiomyocytes in children with TF were demonstrated. The proportion of interstitial tissue did not differ significantly from the control group. The myocardium of children with TF under six months of age was most sensitive to hypoxemia, it was manifested by a delay in the intercalated discs and myofibril assembly and the appearance of ultrastructural signs of dystrophic changes in the cardiomyocytes. Thus, the acceleration of ontogenetic growth and differentiation of the cardiomyocytes, but not the reactivation of their proliferation, was an adaptation of the immature myocardium of children with TF to hemodynamic overload and hypoxemia.
Collapse
|
8
|
Bogorodskiy A, Okhrimenko I, Burkatovskii D, Jakobs P, Maslov I, Gordeliy V, Dencher NA, Gensch T, Voos W, Altschmied J, Haendeler J, Borshchevskiy V. Role of Mitochondrial Protein Import in Age-Related Neurodegenerative and Cardiovascular Diseases. Cells 2021; 10:3528. [PMID: 34944035 PMCID: PMC8699856 DOI: 10.3390/cells10123528] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/10/2021] [Accepted: 12/12/2021] [Indexed: 11/17/2022] Open
Abstract
Mitochondria play a critical role in providing energy, maintaining cellular metabolism, and regulating cell survival and death. To carry out these crucial functions, mitochondria employ more than 1500 proteins, distributed between two membranes and two aqueous compartments. An extensive network of dedicated proteins is engaged in importing and sorting these nuclear-encoded proteins into their designated mitochondrial compartments. Defects in this fundamental system are related to a variety of pathologies, particularly engaging the most energy-demanding tissues. In this review, we summarize the state-of-the-art knowledge about the mitochondrial protein import machinery and describe the known interrelation of its failure with age-related neurodegenerative and cardiovascular diseases.
Collapse
Affiliation(s)
- Andrey Bogorodskiy
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.B.); (I.O.); (D.B.); (I.M.); (V.G.); (N.A.D.)
| | - Ivan Okhrimenko
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.B.); (I.O.); (D.B.); (I.M.); (V.G.); (N.A.D.)
| | - Dmitrii Burkatovskii
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.B.); (I.O.); (D.B.); (I.M.); (V.G.); (N.A.D.)
| | - Philipp Jakobs
- Environmentally-Induced Cardiovascular Degeneration, Central Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, University Hospital and Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; (P.J.); (J.A.); (J.H.)
| | - Ivan Maslov
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.B.); (I.O.); (D.B.); (I.M.); (V.G.); (N.A.D.)
| | - Valentin Gordeliy
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.B.); (I.O.); (D.B.); (I.M.); (V.G.); (N.A.D.)
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52428 Jülich, Germany
- JuStruct: Jülich Center for Structural Biology, Forschungszentrum Jülich, 52428 Jülich, Germany
- Institut de Biologie Structurale (IBS), Université Grenoble Alpes, CEA, CNRS, 38400 Grenoble, France
| | - Norbert A. Dencher
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.B.); (I.O.); (D.B.); (I.M.); (V.G.); (N.A.D.)
- Physical Biochemistry, Chemistry Department, Technical University of Darmstadt, 64289 Darmstadt, Germany
| | - Thomas Gensch
- Institute of Biological Information Processing (IBI-1: Molecular and Cellular Physiology), Forschungszentrum Jülich, 52428 Jülich, Germany;
| | - Wolfgang Voos
- Institute of Biochemistry and Molecular Biology (IBMB), Faculty of Medicine, University of Bonn, 53113 Bonn, Germany;
| | - Joachim Altschmied
- Environmentally-Induced Cardiovascular Degeneration, Central Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, University Hospital and Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; (P.J.); (J.A.); (J.H.)
- IUF—Leibniz Research Institute for Environmental Medicine, 40225 Düsseldorf, Germany
| | - Judith Haendeler
- Environmentally-Induced Cardiovascular Degeneration, Central Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, University Hospital and Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany; (P.J.); (J.A.); (J.H.)
| | - Valentin Borshchevskiy
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia; (A.B.); (I.O.); (D.B.); (I.M.); (V.G.); (N.A.D.)
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52428 Jülich, Germany
- JuStruct: Jülich Center for Structural Biology, Forschungszentrum Jülich, 52428 Jülich, Germany
| |
Collapse
|
9
|
Pietri P, Stefanadis C. Cardiovascular Aging and Longevity: JACC State-of-the-Art Review. J Am Coll Cardiol 2021; 77:189-204. [PMID: 33446313 DOI: 10.1016/j.jacc.2020.11.023] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/25/2022]
Abstract
Cardiovascular aging and longevity are interrelated through many pathophysiological mechanisms. Many factors that promote atherosclerotic cardiovascular disease are also implicated in the aging process and vice versa. Indeed, cardiometabolic disorders such as hyperglycemia, insulin resistance, dyslipidemia, and arterial hypertension share common pathophysiological mechanisms with aging and longevity. Moreover, genetic modulators of longevity have a significant impact on cardiovascular aging. The current knowledge of genetic, molecular, and biochemical pathways of aging may serve as a substrate to introduce interventions that might delay cardiovascular aging, thus approaching the goal of longevity. In the present review, the authors describe pathophysiological links between cardiovascular aging and longevity and translate these mechanisms into clinical data by reporting genetic, dietary, and environmental characteristics from long-living populations.
Collapse
Affiliation(s)
- Panagiota Pietri
- Athens Medical School, University of Athens, Athens, Greece; Research Institute for Longevity and Aging-related diseases, Athens, Greece
| | - Christodoulos Stefanadis
- Athens Medical School, University of Athens, Athens, Greece; Research Institute for Longevity and Aging-related diseases, Athens, Greece.
| |
Collapse
|
10
|
Senescence and senolytics in cardiovascular disease: Promise and potential pitfalls. Mech Ageing Dev 2021; 198:111540. [PMID: 34237321 PMCID: PMC8387860 DOI: 10.1016/j.mad.2021.111540] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/28/2021] [Accepted: 07/04/2021] [Indexed: 02/08/2023]
Abstract
Ageing is the biggest risk factor for impaired cardiovascular health, with cardiovascular disease being the cause of death in 40 % of individuals over 65 years old. Ageing is associated with an increased prevalence of atherosclerosis, coronary artery stenosis and subsequent myocardial infarction, thoracic aortic aneurysm, valvular heart disease and heart failure. An accumulation of senescence and increased inflammation, caused by the senescence-associated secretory phenotype, have been implicated in the aetiology and progression of these age-associated diseases. Recently it has been demonstrated that compounds targeting components of anti-apoptotic pathways expressed by senescent cells can preferentially induce senescence cells to apoptosis and have been termed senolytics. In this review, we discuss the evidence demonstrating that senescence contributes to cardiovascular disease, with a particular focus on studies that indicate the promise of senotherapy. Based on these data we suggest novel indications for senolytics as a treatment of cardiovascular diseases which have yet to be studied in the context of senotherapy. Finally, while the potential benefits are encouraging, several complications may result from senolytic treatment. We, therefore, consider these challenges in the context of the cardiovascular system.
Collapse
|
11
|
Erdem HB, Bahsi T, Ergün MA. Function of telomere in aging and age related diseases. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 85:103641. [PMID: 33774188 DOI: 10.1016/j.etap.2021.103641] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/08/2021] [Accepted: 03/22/2021] [Indexed: 06/12/2023]
Abstract
Telomeres consist of specialized non-coding DNA repeat sequences. They are essential for preserving the integrity of the genome during cancer development, senescence. Mammalian telomeres might have 1-50 kb of telomeric DNA, which becomes 40-200 base pairs shorter after per cell cycle, and becomes 5-8 kilobase shorter during senescence. There are many studies on the correlation of telomere length and aging rate. However, as the differences in the methods used in the studies and the scarcity of prospective studies, factors affecting telomere length are not really well understood. Some of the age related diseases may develop due to telomere dysfunction and telomere shortness. The short telomere structure detected in both peripheral blood leukocytes and cells of the disease-related tissue has the feature of being a predictive marker for many age-related diseases. It is expected that with future research, telomere length analysis is expected to enter clinical practice.
Collapse
Affiliation(s)
- Haktan Bağış Erdem
- Department of Medical Genetics, University of Health Sciences, Dr. Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, Yenimahalle, Ankara, Turkey.
| | - Taha Bahsi
- Department of Medical Genetics, University of Health Sciences, Dr. Abdurrahman Yurtaslan Ankara Oncology Training and Research Hospital, Yenimahalle, Ankara, Turkey.
| | - Mehmet Ali Ergün
- Department of Medical Genetics, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey.
| |
Collapse
|
12
|
Pillai VB, Samant S, Hund S, Gupta M, Gupta MP. The nuclear sirtuin SIRT6 protects the heart from developing aging-associated myocyte senescence and cardiac hypertrophy. Aging (Albany NY) 2021; 13:12334-12358. [PMID: 33934090 PMCID: PMC8148452 DOI: 10.18632/aging.203027] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 02/01/2021] [Indexed: 12/23/2022]
Abstract
Sirtuins have been shown to regulate the aging process. We have previously demonstrated that Sirt6 blocks the pressure overload-induced cardiac hypertrophy in mice. Here, we show that Sirt6 can also mitigate aging-induced cardiomyocyte senescence and cardiac hypertrophy. We found that aging is associated with altered Sirt6 activity along with development of cardiac hypertrophy and fibrosis. Compared to young mice (4-months), the hearts of aged mice (24-months) showed increased levels of mitochondrial DNA damage, shortened telomere length, and increased accumulation of 8-oxo-dG adducts, which are hallmarks of aging. The aged hearts also showed reduced levels of NAD+ and altered levels of mitochondrial fusion-fission proteins. Similar characteristics were observed in the hearts of Sirt6 deficient mice. Additionally, we found that doxorubicin (Dox) induced cardiomyocyte senescence, as measured by expression of p16INK4a, p53, and β-galactosidase, was associated with loss of Sirt6. However, Sirt6 overexpression protected cardiomyocytes from developing Dox-induced senescence. Further, compared to wild-type mice, the hearts of Sirt6.Tg mice showed reduced expression of aging markers, and the development of aging-associated cardiac hypertrophy and fibrosis. Our data suggest that Sirt6 is a critical anti-aging molecule that regulates various cellular processes associated with aging and protects the heart from developing aging-induced cardiac hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Vinodkumar B Pillai
- Department of Surgery, Basic Science Division, The Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Sadhana Samant
- Department of Surgery, Basic Science Division, The Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Samantha Hund
- Department of Surgery, Basic Science Division, The Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Madhu Gupta
- Department of Surgery, Basic Science Division, The Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Mahesh P Gupta
- Department of Surgery, Basic Science Division, The Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
13
|
Santos F, Correia M, Nóbrega-Pereira S, Bernardes de Jesus B. Age-Related Pathways in Cardiac Regeneration: A Role for lncRNAs? Front Physiol 2021; 11:583191. [PMID: 33551829 PMCID: PMC7855957 DOI: 10.3389/fphys.2020.583191] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/16/2020] [Indexed: 12/13/2022] Open
Abstract
Aging imposes a barrier for tissue regeneration. In the heart, aging leads to a severe rearrangement of the cardiac structure and function and to a subsequent increased risk of heart failure. An intricate network of distinct pathways contributes to age-related alterations during healthy heart aging and account for a higher susceptibility of heart disease. Our understanding of the systemic aging process has already led to the design of anti-aging strategies or to the adoption of protective interventions. Nevertheless, our understanding of the molecular determinants operating during cardiac aging or repair remains limited. Here, we will summarize the molecular and physiological alterations that occur during aging of the heart, highlighting the potential role for long non-coding RNAs (lncRNAs) as novel and valuable targets in cardiac regeneration/repair.
Collapse
Affiliation(s)
- Francisco Santos
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Magda Correia
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - Sandrina Nóbrega-Pereira
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
- Faculdade de Medicina, Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
| | - Bruno Bernardes de Jesus
- Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
14
|
Carlone DL, Riba-Wolman RD, Deary LT, Tovaglieri A, Jiang L, Ambruzs DM, Mead BE, Shah MS, Lengner CJ, Jaenisch R, Breault DT. Telomerase expression marks transitional growth-associated skeletal progenitor/stem cells. Stem Cells 2021; 39:296-305. [PMID: 33438789 PMCID: PMC7986156 DOI: 10.1002/stem.3318] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/20/2020] [Indexed: 12/28/2022]
Abstract
Skeletal progenitor/stem cells (SSCs) play a critical role in postnatal bone growth and maintenance. Telomerase (Tert) activity prevents cellular senescence and is required for maintenance of stem cells in self‐renewing tissues. Here we investigated the role of mTert‐expressing cells in postnatal mouse long bone and found that mTert expression is enriched at the time of adolescent bone growth. mTert‐GFP+ cells were identified in regions known to house SSCs, including the metaphyseal stroma, growth plate, and the bone marrow. We also show that mTert‐expressing cells are a distinct SSC population with enriched colony‐forming capacity and contribute to multiple mesenchymal lineages, in vitro. In contrast, in vivo lineage‐tracing studies identified mTert+ cells as osteochondral progenitors and contribute to the bone‐forming cell pool during endochondral bone growth with a subset persisting into adulthood. Taken together, our results show that mTert expression is temporally regulated and marks SSCs during a discrete phase of transitional growth between rapid bone growth and maintenance.
Collapse
Affiliation(s)
- Diana L Carlone
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Rebecca D Riba-Wolman
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Luke T Deary
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Alessio Tovaglieri
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Lijie Jiang
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Dana M Ambruzs
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Benjamin E Mead
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Manasvi S Shah
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher J Lengner
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA.,Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
15
|
Telomeres and telomerase in risk assessment of cardiovascular diseases. Exp Cell Res 2020; 397:112361. [PMID: 33171154 DOI: 10.1016/j.yexcr.2020.112361] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/02/2020] [Indexed: 01/14/2023]
Abstract
Telomeres are repetitive nucleoprotein structures located at the ends of chromosomes. Reduction in the number of repetitions causes cell senescence. Cells with high proliferative potential age with each replication cycle. Post-mitotic cells (e.g. cardiovascular cells) have a different aging mechanism. During the aging of cardiovascular system cells, permanent DNA damage occurs in the telomeric regions caused by mitochondrial dysfunction, which is a phenomenon independent of cell proliferation and telomere length. Mitochondrial dysfunction is accompanied by increased production of reactive oxygen species and development of inflammation. This phenomenon in the cells of blood vessels can lead to atherosclerosis development. Telomere damage in cardiomyocytes leads to the activation of the DNA damage response system, histone H2A.X phosphorylation, p53 activation and p21 and p16 protein synthesis, resulting in the SASP phenotype (senescence-associated secretory phenotype), increased inflammation and cardiac dysfunction. Cardiovascular cells show the activity of the TERT subunit of telomerase, an enzyme that prevents telomere shortening. It turns out that disrupting the activity of this enzyme can also contribute to the formation of cardiovascular diseases. Measurements of telomere length according to the "blood-muscle" model may help in the future to assess the risk of cardiovascular complications in people undergoing cardiological procedures, as well as to assess the effectiveness of some drugs.
Collapse
|
16
|
Dookun E, Walaszczyk A, Redgrave R, Palmowski P, Tual‐Chalot S, Suwana A, Chapman J, Jirkovsky E, Donastorg Sosa L, Gill E, Yausep OE, Santin Y, Mialet‐Perez J, Andrew Owens W, Grieve D, Spyridopoulos I, Taggart M, Arthur HM, Passos JF, Richardson GD. Clearance of senescent cells during cardiac ischemia-reperfusion injury improves recovery. Aging Cell 2020; 19:e13249. [PMID: 32996233 PMCID: PMC7576252 DOI: 10.1111/acel.13249] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/02/2020] [Accepted: 09/13/2020] [Indexed: 12/16/2022] Open
Abstract
A key component of cardiac ischemia-reperfusion injury (IRI) is the increased generation of reactive oxygen species, leading to enhanced inflammation and tissue dysfunction in patients following intervention for myocardial infarction. In this study, we hypothesized that oxidative stress, due to ischemia-reperfusion, induces senescence which contributes to the pathophysiology of cardiac IRI. We demonstrate that IRI induces cellular senescence in both cardiomyocytes and interstitial cell populations and treatment with the senolytic drug navitoclax after ischemia-reperfusion improves left ventricular function, increases myocardial vascularization, and decreases scar size. SWATH-MS-based proteomics revealed that biological processes associated with fibrosis and inflammation that were increased following ischemia-reperfusion were attenuated upon senescent cell clearance. Furthermore, navitoclax treatment reduced the expression of pro-inflammatory, profibrotic, and anti-angiogenic cytokines, including interferon gamma-induced protein-10, TGF-β3, interleukin-11, interleukin-16, and fractalkine. Our study provides proof-of-concept evidence that cellular senescence contributes to impaired heart function and adverse remodeling following cardiac ischemia-reperfusion. We also establish that post-IRI the SASP plays a considerable role in the inflammatory response. Subsequently, senolytic treatment, at a clinically feasible time-point, attenuates multiple components of this response and improves clinically important parameters. Thus, cellular senescence represents a potential novel therapeutic avenue to improve patient outcomes following cardiac ischemia-reperfusion.
Collapse
Affiliation(s)
- Emily Dookun
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Anna Walaszczyk
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | | | - Pawel Palmowski
- School of Environmental SciencesFaculty of ScienceAgriculture & EngineeringNewcastle UniversityNewcastle upon TyneUK
| | | | - Averina Suwana
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - James Chapman
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | | | | | - Eleanor Gill
- School of MedicineDentistry and Biomedical SciencesCentre for Experimental MedicineInstitute for Health SciencesQueen`s University BelfastBelfastUK
| | - Oliver E Yausep
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | | | | | - W Andrew Owens
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - David Grieve
- School of MedicineDentistry and Biomedical SciencesCentre for Experimental MedicineInstitute for Health SciencesQueen`s University BelfastBelfastUK
| | | | - Michael Taggart
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - Helen M. Arthur
- Biosciences InstituteNewcastle UniversityNewcastle upon TyneUK
| | - João F. Passos
- Department of Physiology and Biomedical EngineeringMayo ClinicRochesterMNUSA
| | | |
Collapse
|
17
|
Farhangi MA, Najafi M. The association between dietary quality indices and serum telomerase activity in patient candidates for CABG. Eat Weight Disord 2020; 25:1461-1468. [PMID: 32056109 DOI: 10.1007/s40519-020-00861-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 01/28/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Diet and dietary habits are major determinants of human telomere length. Telomerase activity is affected mostly by oxidative stress and inflammation. However, the association of telomerase activity with dietary quality indices has not been evaluated before. In the current work, we aimed to test the association of telomerase activity with dietary antioxidant quality score (DAQ), dietary inflammatory index and dietary patterns in patients who were candidate for coronary artery bypass grafting surgery (CABG). METHODS AND MATERIALS In the current cross-sectional study, 454 candidates for the CABG were enrolled from Tehran Heart Center-Coronary Outcome Measurement (THC-COM) cohort. Laboratory measurements included Hb-A1C, serum lipid profile, creatinine, blood urea nitrogen, hematocrit, lipoprotein (LP)-a, telomerase activity, serum vitamin D and C-reactive protein. Dietary status was measured by semiquantitative food frequency questionnaire, and dietary indices were calculated. Dietary patterns were extracted by factor analysis method. RESULTS High telomerase activity was associated with lower prevalence of myocardial infarction (MI) (P = 0.04), high dietary vitamin E and high total dietary antioxidant quality scores. Telomerase activity in top quartile of neo-traditional dietary pattern was higher than other quartiles (P = 0.021). No significant association between telomerase activity and other dietary patterns was obtained. Higher telomerase activity was also associated with higher serum creatinine and lower LP-(a) concentrations (P < 0.05). CONCLUSION To our findings, higher telomerase activity was associated with higher DAQ and lower MI prevalence. It seems that adherence to healthy diet increases serum telomerase activity and reduced telomerase concentration is associated with increased cardio-metabolic risk factors. Moreover, adherence to neo-traditional pattern with higher intake of low-fat dairy products was associated with higher telomerase activity. LEVEL OF EVIDENCE Level V: A well-designed observational cross-sectional study.
Collapse
Affiliation(s)
| | - Mahdi Najafi
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada. .,Department of Research, Tehran Heart Center, Tehran University of Medical Sciences, North Karegar Street, Tehran, 1411713138, Iran.
| |
Collapse
|
18
|
Pańczyszyn A, Boniewska-Bernacka E, Goc A. The role of telomeres and telomerase in the senescence of postmitotic cells. DNA Repair (Amst) 2020; 95:102956. [PMID: 32937289 DOI: 10.1016/j.dnarep.2020.102956] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022]
Abstract
Senescence is a process related to the stopping of divisions and changes leading the cell to the SASP phenotype. Permanent senescence of many SASP cells contributes to faster aging of the body and development of age-related diseases due to the release of pro-inflammatory factors. Both mitotically active and non-dividing cells can undergo senescence as a result of activation of different molecular pathways. Telomeres, referred to as the molecular clock, direct the dividing cell into the aging pathway when reaching a critical length. In turn, the senescence of postmitotic cells depends not on the length of telomeres, but their functionality. Dysfunctional telomeres are responsible for triggering the signaling of DNA damage response (DDR). Telomerase subunits in post-mitotic cells translocate between the nucleus, cytoplasm and mitochondria, participating in the regulation of their activity. Among other things, they contribute to the reduction of reactive oxygen species generation, which leads to telomere dysfunction and, consequently, senescence. Some proteins of the shelterin complex also play a protective role by inhibiting senescence-initiating kinases and limiting ROS production by mitochondria.
Collapse
Affiliation(s)
- Anna Pańczyszyn
- University of Opole, Institute of Medical Sciences, Department of Biology and Genetics, Opole 45-040, Pl.Kopernika 11a, Poland.
| | - Ewa Boniewska-Bernacka
- University of Opole, Institute of Medical Sciences, Department of Biology and Genetics, Opole 45-040, Pl.Kopernika 11a, Poland.
| | - Anna Goc
- University of Opole, Institute of Medical Sciences, Department of Biology and Genetics, Opole 45-040, Pl.Kopernika 11a, Poland.
| |
Collapse
|
19
|
Zarei M, Najafi M, Movahedi E, Javanbakht MH, Choi YH, Yaseri M, Shirvani A, Sellke FW, Stranges S. The predictive role of circulating telomerase and vitamin D for long-term survival in patients undergoing coronary artery bypass grafting surgery (CABG). PLoS One 2020; 15:e0237477. [PMID: 32790742 PMCID: PMC7425905 DOI: 10.1371/journal.pone.0237477] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/27/2020] [Indexed: 11/21/2022] Open
Abstract
Backgrounds Cardiovascular disease (CVD) is the leading cause of mortality all over the globe. Inflammation is believed to play a pivotal role in the pathophysiology of CVD. While there are studies on the interrelationship of telomerase and vitamin D and their involvement in CVD, their independent contributions to long-term outcomes in patients with CVD are not well-defined. This study aimed to investigate the association of both telomerase and vitamin D concentrations with 10-year survival among candidates of coronary artery bypass grafting (CABG) surgery. Methods Participants were 404 patients from Tehran Heart Center-Coronary Outcome Measurement (THC-COM) cohort who were recruited from CABG surgery candidates in 2006. In addition to demographic and clinical data including risk factors for coronary artery disease, laboratory parameters such as markers of inflammation as well as baseline serum 25-hydroxy vitamin D [25(OH)D] and telomerase concentrations were measured. Cardiac function indexes alongside outcome measures such as mortality and survival days were recorded for every patient up to 10 years after CABG. Cox-proportional hazard model was used to study the association between all-cause mortality and research parameters. Results The mean serum telomerase enzyme level was 24.92 ±21.4 nmol/L and the mean serum 25(OH)D was 27.27±10.3 ng/mL. 10-year mortality was reported in 64 (15.8%) patients. 25(OH)D was categorized into three groups (<20, 20–30, and >30) and the cut-point for telomerase was set at 25.0 nmol/L. In Cox regression analysis, higher levels of telomerase (>25 nmol/L) were significantly associated with longer survival (p = 0.041), whereas 25(OH)D concentrations were not associated with survival time. Further analysis showed that telomerase concentrations significantly predicted survival only in the presence of insufficient levels of 25(OH)D (20–30 ng/mL) (p = 0.037). Conclusions Telomerase can be regarded as a potential predictor of long-term outcomes in patients who underwent CABG. However, the association of telomerase with the mortality may be modified by vitamin D concentrations.
Collapse
Affiliation(s)
- Mahtab Zarei
- Department of Cellular and Molecular Nutrition, Faculty of Nutritional Sciences & Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Najafi
- Department of Medical Biophysics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anesthesiology, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
- * E-mail: ,
| | - Elnaz Movahedi
- Department of Cellular and Molecular Nutrition, Faculty of Nutritional Sciences & Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamad Hassan Javanbakht
- Department of Cellular and Molecular Nutrition, Faculty of Nutritional Sciences & Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Yun-Hee Choi
- Department of Epidemiology and Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Mehdi Yaseri
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Arash Shirvani
- Department of Medicine, Section of Endocrinology, Nutrition, and Diabetes, Vitamin D, Skin and Bone Research Laboratory, Boston University Medical Campus, Boston, Massachusetts, United States of America
| | - Frank W. Sellke
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island, United States of America
| | - Saverio Stranges
- Department of Epidemiology and Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Family Medicine, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Population Health, Luxembourg Institute of Health, Strassen, Luxembourg
| |
Collapse
|
20
|
Martynowicz H, Gać P, Kornafel-Flak O, Filipów S, Łaczmański Ł, Sobieszczańska M, Mazur G, Porȩba R. The Relationship Between the Effectiveness of Blood Pressure Control and Telomerase Reverse Transcriptase Concentration, Adipose Tissue Hormone Concentration and Endothelium Function in Hypertensives. Heart Lung Circ 2020; 29:e200-e209. [DOI: 10.1016/j.hlc.2019.12.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/12/2019] [Accepted: 12/01/2019] [Indexed: 12/23/2022]
|
21
|
Rosen J, Jakobs P, Ale-Agha N, Altschmied J, Haendeler J. Non-canonical functions of Telomerase Reverse Transcriptase - Impact on redox homeostasis. Redox Biol 2020; 34:101543. [PMID: 32502898 PMCID: PMC7267725 DOI: 10.1016/j.redox.2020.101543] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/11/2020] [Accepted: 04/13/2020] [Indexed: 02/08/2023] Open
Abstract
Telomerase consists of the catalytic subunit Telomerase Reverse Transcriptase (TERT) and the Telomerase RNA Component. Its canonical function is the prevention of telomere erosion. Over the last years it became evident that TERT is also present in tissues with low replicative potential. Important non-canonical functions of TERT are protection against apoptosis and maintenance of the cellular redox homeostasis in cancer as well as in somatic tissues. Intriguingly, TERT and reactive oxygen species (ROS) are interdependent on each other, with TERT being regulated by changes in the redox balance and itself controlling ROS levels in the cytosol and in the mitochondria. The latter is achieved because TERT is present in the mitochondria, where it protects mitochondrial DNA and maintains levels of anti-oxidative enzymes. Since numerous diseases are associated with oxidative stress, increasing the mitochondrial TERT level could be of therapeutic value.
Collapse
Affiliation(s)
- Julia Rosen
- Environmentally-induced Cardiovascular Degeneration, Institute of Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Philipp Jakobs
- Environmentally-induced Cardiovascular Degeneration, Institute of Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Niloofar Ale-Agha
- Environmentally-induced Cardiovascular Degeneration, Institute of Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Joachim Altschmied
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| | - Judith Haendeler
- Environmentally-induced Cardiovascular Degeneration, Institute of Clinical Chemistry and Laboratory Diagnostics, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany; IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| |
Collapse
|
22
|
Biohorology and biomarkers of aging: Current state-of-the-art, challenges and opportunities. Ageing Res Rev 2020; 60:101050. [PMID: 32272169 DOI: 10.1016/j.arr.2020.101050] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 02/06/2020] [Accepted: 03/22/2020] [Indexed: 02/08/2023]
Abstract
The aging process results in multiple traceable footprints, which can be quantified and used to estimate an organism's age. Examples of such aging biomarkers include epigenetic changes, telomere attrition, and alterations in gene expression and metabolite concentrations. More than a dozen aging clocks use molecular features to predict an organism's age, each of them utilizing different data types and training procedures. Here, we offer a detailed comparison of existing mouse and human aging clocks, discuss their technological limitations and the underlying machine learning algorithms. We also discuss promising future directions of research in biohorology - the science of measuring the passage of time in living systems. Overall, we expect deep learning, deep neural networks and generative approaches to be the next power tools in this timely and actively developing field.
Collapse
|
23
|
Sadeghi-Tabas S, Saghebjoo M, Sarir H, Hedayati M. Effects of work/rest interval manipulation of high-intensity interval training and detraining on telomerase activity and p53 levels in cardiac muscle. Sci Sports 2020. [DOI: 10.1016/j.scispo.2019.06.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
24
|
Vecoli C, Borghini A, Andreassi MG. The molecular biomarkers of vascular aging and atherosclerosis: telomere length and mitochondrial DNA 4977 common deletion. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2020; 784:108309. [PMID: 32430098 DOI: 10.1016/j.mrrev.2020.108309] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022]
Abstract
Age is the dominant risk factor for the most prevalent atherosclerotic vascular diseases, including coronary artery disease, myocardial infarction, cerebrovascular disease and stroke. In human, telomere erosion and mitochondrial DNA (mtDNA) damage play a central role in the mechanisms leading to cellular aging decline. This review summarizes the most relevant findings on the role of telomere shortening and the common mtDNA4977 deletion in the progression and evolution of atherosclerosis by combining insight from experimental models and human clinical studies. The current evidence shows a link between telomere erosion and compromised mitochondrial function and provides a new perspective regarding their potential role as clinical biomarkers and therapeutic targets.
Collapse
|
25
|
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: 1.6] [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
|
26
|
Chan D, Martin-Ruiz C, Saretzki G, Neely D, Qiu W, Kunadian V. The association of telomere length and telomerase activity with adverse outcomes in older patients with non-ST-elevation acute coronary syndrome. PLoS One 2020; 15:e0227616. [PMID: 31923255 PMCID: PMC6953865 DOI: 10.1371/journal.pone.0227616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/22/2019] [Indexed: 12/24/2022] Open
Abstract
Background Non-ST elevation acute coronary syndrome (NSTEACS) occurs more frequently in older patients with an increased occurrence of recurrent cardiac events following the index presentation. Telomeres are structures consisting of repeated DNA sequences as associated shelterin proteins at the ends of chromosomes. We aim to determine whether telomere length (TL) and telomerase activity (TA) predicted poor outcomes in older patients presenting with NSTEACS undergoing invasive care. Method Older patients undergoing invasive management for NSTEACS were recruited to the ICON-1 biomarker study (NCT01933581). Peripheral blood mononuclear cells (PBMC) were recovered on 153 patients. DNA was isolated and mean TL was measured by quantitative PCR expressed as relative T (telomere repeat copy number) to S (single copy gene number) ratio (T/S ratio), and a telomere repeat amplification assay was used to assess TA during index presentation with NSTEACS. Primary clinical outcomes consisted of death, myocardial infarction (MI), unplanned revascularisation, stroke and significant bleeding recorded at 1 year. TL and TA were divided into tertile groups for analysis. Cox proportional hazards regression was performed. Ordinal regression was performed to evaluate the relationship between TL and TA and traditional cardiovascular risk factors at baseline. Results 298 patients were recruited in the ICON-1 study of which 153 had PBMC recovered. The mean age was 81.0 ± 4.0 years (64% male). Mean telomere length T/S ratio was 0.47 ± 0.25 and mean TA was 1.52 ± 0.61 units. The primary composite outcome occurred in 44 (28.8%) patients. There was no association between short TL or low TA and incidence of the primary composite outcome (Hazard Ratio [HR] 1.50, 95% Confidence Interval [CI] 0.68–3.34, p = 0.32 and HR 1.33, 95% CI 0.52–3.36, p = 0.51 respectively). Conclusion TL and TA are not found to be associated with the incidence of adverse outcomes in older patients presenting with NSTEACS undergoing invasive care. Clinical trial registration URL: https://www.clinicaltrials.gov Unique identifier: NCT01933581
Collapse
Affiliation(s)
- Danny Chan
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- Cardiothoracic Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Carmen Martin-Ruiz
- BioScreening Facility, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Gabriele Saretzki
- Ageing Biology Centre and Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Dermot Neely
- Department of Biochemistry, Newcastle upon Tyne Hospitals NHS Foundations Trust, United Kingdom
| | - Weiliang Qiu
- Sanofi Genzyme, Framingham, MA, United States of America
| | - Vijay Kunadian
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
- Cardiothoracic Centre, Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
- * E-mail:
| |
Collapse
|
27
|
Lundberg M, Millischer V, Backlund L, Martinsson L, Stenvinkel P, Sellgren CM, Lavebratt C, Schalling M. Lithium and the Interplay Between Telomeres and Mitochondria in Bipolar Disorder. Front Psychiatry 2020; 11:586083. [PMID: 33132941 PMCID: PMC7553080 DOI: 10.3389/fpsyt.2020.586083] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/31/2020] [Indexed: 01/06/2023] Open
Abstract
Bipolar disorder is a severe psychiatric disorder which affects more than 1% of the world's population and is a leading cause of disability among young people. For the past 50 years, lithium has been the drug of choice for maintenance treatment of bipolar disorder due to its potent ability to prevent both manic and depressive episodes as well as suicide. However, though lithium has been associated with a multitude of effects within different cellular pathways and biological systems, its specific mechanism of action in stabilizing mood remains largely elusive. Mitochondrial dysfunction and telomere shortening have been implicated in both the pathophysiology of bipolar disorder and as targets of lithium treatment. Interestingly, it has in recent years become clear that these phenomena are intimately linked, partly through reactive oxygen species signaling and the subcellular translocation and non-canonical actions of telomerase reverse transcriptase. In this review, we integrate the current understanding of mitochondrial dysfunction, oxidative stress and telomere shortening in bipolar disorder with documented effects of lithium. Moreover, we propose that lithium's mechanism of action is intimately connected with the interdependent regulation of mitochondrial bioenergetics and telomere maintenance.
Collapse
Affiliation(s)
- Martin Lundberg
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Vincent Millischer
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Lena Backlund
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Lina Martinsson
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Healthcare Services, Region Stockholm, Stockholm, Sweden
| | - Peter Stenvinkel
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Carl M Sellgren
- Centre for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Healthcare Services, Region Stockholm, Stockholm, Sweden.,Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Catharina Lavebratt
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Martin Schalling
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
28
|
Zheng Q, Huang J, Wang G. Mitochondria, Telomeres and Telomerase Subunits. Front Cell Dev Biol 2019; 7:274. [PMID: 31781563 PMCID: PMC6851022 DOI: 10.3389/fcell.2019.00274] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 10/24/2019] [Indexed: 12/11/2022] Open
Abstract
Mitochondrial functions and telomere functions have mostly been studied independently. In recent years, it, however, has become clear that there are intimate links between mitochondria, telomeres, and telomerase subunits. Mitochondrial dysfunctions cause telomere attrition, while telomere damage leads to reprogramming of mitochondrial biosynthesis and mitochondrial dysfunctions, which has important implications in aging and diseases. In addition, evidence has accumulated that telomere-independent functions of telomerase also exist and that the protein component of telomerase TERT shuttles between the nucleus and mitochondria under oxidative stress. Our previously published data show that the RNA component of telomerase TERC is also imported into mitochondria, processed, and exported back to the cytosol. These data show a complex regulation network where telomeres, nuclear genome, and mitochondria are co-regulated by multi-localization and multi-function proteins and RNAs. This review summarizes the connections between mitochondria and telomeres, the mitochondrion-related functions of telomerase subunits, and how they play a role in crosstalk between mitochondria and the nucleus.
Collapse
Affiliation(s)
- Qian Zheng
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Jinliang Huang
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Geng Wang
- School of Life Sciences, Tsinghua University, Beijing, China.,School of Life Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
29
|
Denham J. Telomere regulation: lessons learnt from mice and men, potential opportunities in horses. Anim Genet 2019; 51:3-13. [PMID: 31637754 DOI: 10.1111/age.12870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2019] [Indexed: 11/26/2022]
Abstract
Telomeres are genetically conserved nucleoprotein complexes located at the ends of chromosomes that preserve genomic stability. In large mammals, somatic cell telomeres shorten with age, owing to the end replication problem and lack of telomere-lengthening events (e.g. telomerase and ALT activity). Therefore, telomere length reflects cellular replicative reserve and mitotic potential. Environmental insults can accelerate telomere attrition in response to cell division and DNA damage. As such, telomere shortening is considered one of the major hallmarks of ageing. Much effort has been dedicated to understanding the environmental perturbations that accelerate telomere attrition and therapeutic strategies to preserve or extend telomeres. As telomere dynamics seem to reflect cumulative cellular stress, telomere length could serve as a biomarker of animal welfare. The assessment of telomere dynamics (i.e. rate of shortening) in conjunction with telomere-regulating genes and telomerase activity in racehorses could monitor long-term animal health, yet it could also provide some unique opportunities to address particular limitations with the use of other animal models in telomere research. Considering the ongoing efforts to optimise the health and welfare of equine athletes, the purpose of this review is to discuss the potential utility of assessing telomere length in Thoroughbred racehorses. A brief review of telomere biology in large and small mammals will be provided, followed by discussion on the biological implications of telomere length and environmental (e.g. lifestyle) factors that accelerate or attenuate telomere attrition. Finally, the utility of quantifying telomere dynamics in horses will be offered with directions for future research.
Collapse
Affiliation(s)
- J Denham
- School of Health and Biomedical Sciences, Bundoora West Campus, RMIT University, Room 53, Level 4, Building 202, Bundoora, VIC, 3083, Australia
| |
Collapse
|
30
|
Herrmann M, Pusceddu I, März W, Herrmann W. Telomere biology and age-related diseases. Clin Chem Lab Med 2019; 56:1210-1222. [PMID: 29494336 DOI: 10.1515/cclm-2017-0870] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 01/30/2018] [Indexed: 11/15/2022]
Abstract
Telomeres are the protective end caps of chromosomes and shorten with every cell division. Telomere length has been proposed as a biomarker of biological age and a risk factor for age-related diseases. Epidemiologic studies show an association between leukocyte telomere length (LTL) and mortality. There is solid evidence that links LTL with cardiovascular disease. Short telomeres promote atherosclerosis and impair the repair of vascular lesions. Alzheimer's disease patients have also a reduced LTL. Telomeres measured in tumor tissue from breast, colon and prostate are shorter than in healthy tissue from the same organ and the same patient. In healthy tissue directly adjacent to these tumors, telomeres are also shorter than in cells that are more distant from the cancerous lesion. A reduced telomere length in cancer tissue from breast, colon and prostate is associated with an advanced disease state at diagnosis, faster disease progression and poorer survival. By contrast, results regarding LTL and cancer are inconsistent. Furthermore, the majority of studies did not find significant associations between LTL, bone mineral density (BMD) and osteoporosis. The present manuscript gives an overview about our current understanding of telomere biology and reviews existing knowledge regarding the relationship between telomere length and age-related diseases.
Collapse
Affiliation(s)
- Markus Herrmann
- Department of Clinical Pathology, Bolzano Hospital, Lorenz-Boehler-Str. 5, 39100 Bolzano, Italy.,Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
| | - Irene Pusceddu
- Laboratory of Clinical Pathology, Hospital of Bolzano, Bolzano, Italy
| | - Winfried März
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria.,Medical Clinic V (Nephrology, Hypertensiology, Rheumatology, Endocrinology, Diabetology), Medical Faculty of Mannheim, University of Heidelberg, Mannheim, Germany.,Synlab Academy, Synlab Holding Deutschland GmbH, Mannheim, Germany
| | - Wolfgang Herrmann
- Department of Clinical Chemistry, University of Saarland, Homburg, Germany
| |
Collapse
|
31
|
Telomere Gene Therapy: Polarizing Therapeutic Goals for Treatment of Various Diseases. Cells 2019; 8:cells8050392. [PMID: 31035374 PMCID: PMC6563133 DOI: 10.3390/cells8050392] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 04/22/2019] [Accepted: 04/24/2019] [Indexed: 02/07/2023] Open
Abstract
Modulation of telomerase maintenance by gene therapy must meet two polarizing requirements to achieve different therapeutic outcomes: Anti-aging/regenerative applications require upregulation, while anticancer applications necessitate suppression of various genes integral to telomere maintenance (e.g., telomerase, telomerase RNA components, and shelterin complex). Patients suffering from aging-associated illnesses often exhibit telomere attrition, which promotes chromosomal instability and cellular senescence, thus requiring the transfer of telomere maintenance-related genes to improve patient outcomes. However, reactivation and overexpression of telomerase are observed in 85% of cancer patients; this process is integral to cancer immortality. Thus, telomere-associated genes in the scope of cancer gene therapy must be inactivated or inhibited to induce anticancer effects. These contradicting requirements for achieving different therapeutic outcomes mean that any vector-mediated upregulation of telomere-associated genes must be accompanied by rigorous evaluation of potential oncogenesis. Thus, this review aims to discuss how telomere-associated genes are being targeted or utilized in various gene therapy applications and provides some insight into currently available safety hazard assessments.
Collapse
|
32
|
Saghebjoo M, Sadeghi-Tabas S, Saffari I, Ghane A, Dimauro I. Sex Differences in antiaging response to short- and long-term high-intensity interval exercise in rat cardiac muscle: Telomerase activity, total antioxidant/oxidant status. CHINESE J PHYSIOL 2019; 62:261-266. [DOI: 10.4103/cjp.cjp_52_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
33
|
Affiliation(s)
- Chao-Yung Wang
- Department of Cardiology, Chang Gung Memorial Hospital, and Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
34
|
Richardson GD, Sage A, Bennaceur K, Al Zhrany N, Coelho-Lima J, Dookun E, Draganova L, Saretzki G, Breault DT, Mallat Z, Spyridopoulos I. Telomerase Mediates Lymphocyte Proliferation but Not the Atherosclerosis-Suppressive Potential of Regulatory T-Cells. Arterioscler Thromb Vasc Biol 2018; 38:1283-1296. [PMID: 29599138 PMCID: PMC5965929 DOI: 10.1161/atvbaha.117.309940] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 03/05/2018] [Indexed: 01/05/2023]
Abstract
Supplemental Digital Content is available in the text. Objective— Atherosclerosis is an age-related disease characterized by systemic oxidative stress and low-grade inflammation. The role of telomerase and telomere length in atherogenesis remains contentious. Short telomeres of peripheral leukocytes are predictive for coronary artery disease. Conversely, attenuated telomerase has been demonstrated to be protective for atherosclerosis. Hence, a potential causative role of telomerase in atherogenesis is critically debated. Approach and Results— In this study, we used multiple mouse models to investigate the regulation of telomerase under oxidative stress as well as its impact on atherogenesis in vitro and in vivo. Using primary lymphocytes and myeloid cell cultures, we demonstrate that cultivation under hyperoxic conditions induced oxidative stress resulting in chronic activation of CD4+ cells and significantly reduced CD4+ T-cell proliferation. The latter was telomerase dependent because oxidative stress had no effect on the proliferation of primary lymphocytes isolated from telomerase knockout mice. In contrast, myeloid cell proliferation was unaffected by oxidative stress nor reliant on telomerase. Telomerase reverse transcriptase deficiency had no effect on regulatory T-cell (Treg) numbers in vivo or suppressive function ex vivo. Adoptive transfer of telomerase reverse transcriptase–/– Tregs into Rag2–/– ApoE–/– (recombination activating gene 2/apolipoprotein E) double knockout mice demonstrated that telomerase function was not required for the ability of Tregs to protect against atherosclerosis. However, telomere length was critical for Treg function. Conclusions— Telomerase contributes to lymphocyte proliferation but plays no major role in Treg function, provided that telomere length is not critically short. We suggest that oxidative stress may contribute to atherosclerosis via suppression of telomerase and acceleration of telomere attrition in Tregs.
Collapse
Affiliation(s)
- Gavin David Richardson
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| | - Andrew Sage
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.S., Z.M.)
| | - Karim Bennaceur
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| | - Nayef Al Zhrany
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| | - Jose Coelho-Lima
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| | - Emily Dookun
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| | - Lilia Draganova
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| | - Gabriele Saretzki
- Institute for Cell and Molecular Biosciences, The Ageing Biology Centre, Newcastle University Institute for Ageing, Campus for Ageing and Vitality (G.S.), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - David T Breault
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, MA (D.T.B.).,Harvard Stem Cell Institute, Cambridge, MA (D.T.B.)
| | - Ziad Mallat
- Division of Cardiovascular Medicine, Department of Medicine, University of Cambridge, United Kingdom (A.S., Z.M.).,INSERM U970, Paris Cardiovascular Research Center, France (Z.M.).,Université Paris Descartes, Sorbonne Paris Cité, France (Z.M.)
| | - Ioakim Spyridopoulos
- From the Cardiovascular Research Centre, Institute of Genetic Medicine, International Centre for Life (G.D.R., K.B., N.A.Z., J.C.-L., E.D., L.D., I.S.)
| |
Collapse
|
35
|
Aix E, Gallinat A, Flores I. Telomeres and telomerase in heart regeneration. Differentiation 2018; 100:26-30. [PMID: 29453108 DOI: 10.1016/j.diff.2018.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 01/19/2018] [Accepted: 01/23/2018] [Indexed: 01/08/2023]
Abstract
Although recent advances have overturned the old view of the human heart as an inert postmitotic organ, it is clear that the adult heart´s capacity to regenerate after an ischemic episode is very limited. Unlike humans, zebrafish and other lower vertebrates vigorously regenerate damaged myocardium after cardiac injury. Understanding how the zebrafish is able to conserve life-long cardiac regeneration capacity while mammals lose it soon after birth is crucial for the development of new treatments for myocardial infarction. Mammals and lower vertebrates differ markedly in their rates of cardiomyocyte proliferation and levels of telomerase activity. Here, we review recent discoveries identifying lack of telomerase activity and concomitant telomere dysfunction as natural barriers to cardiomyocyte proliferation and cardiac regeneration.
Collapse
Affiliation(s)
- Esther Aix
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernandez Almagro 3, Madrid E-28029, Spain
| | - Alex Gallinat
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernandez Almagro 3, Madrid E-28029, Spain
| | - Ignacio Flores
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernandez Almagro 3, Madrid E-28029, Spain.
| |
Collapse
|
36
|
Anderson R, Richardson GD, Passos JF. Mechanisms driving the ageing heart. Exp Gerontol 2017; 109:5-15. [PMID: 29054534 DOI: 10.1016/j.exger.2017.10.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 10/16/2017] [Indexed: 01/07/2023]
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally. One of the main risk factors for CVD is age, however the biological processes that occur in the heart during ageing are poorly understood. It is therefore important to understand the fundamental mechanisms driving heart ageing to enable the development of preventions and treatments targeting these processes. Cellular senescence is often described as the irreversible cell-cycle arrest which occurs in somatic cells. Emerging evidence suggests that cellular senescence plays a key role in heart ageing, however the cell-types involved and the underlying mechanisms are not yet elucidated. In this review we discuss the current understanding of how mechanisms known to contribute to senescence impact on heart ageing and CVD. Finally, we evaluate recent data suggesting that targeting senescent cells may be a viable therapy to counteract the ageing of the heart.
Collapse
Affiliation(s)
- Rhys Anderson
- The Randall Division, King's College London, London, UK; Ageing Research Laboratories, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK; Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK.
| | - Gavin D Richardson
- Cardiovascular Research Centre, Institute for Genetic Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - João F Passos
- Ageing Research Laboratories, Newcastle University Institute for Ageing, Newcastle University, Newcastle upon Tyne, UK; Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
37
|
de Almeida AJPO, Ribeiro TP, de Medeiros IA. Aging: Molecular Pathways and Implications on the Cardiovascular System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7941563. [PMID: 28874954 PMCID: PMC5569936 DOI: 10.1155/2017/7941563] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/27/2017] [Indexed: 02/06/2023]
Abstract
The world's population over 60 years is growing rapidly, reaching 22% of the global population in the next decades. Despite the increase in global longevity, individual healthspan needs to follow this growth. Several diseases have their prevalence increased by age, such as cardiovascular diseases, the leading cause of morbidity and mortality worldwide. Understanding the aging biology mechanisms is fundamental to the pursuit of cardiovascular health. In this way, aging is characterized by a gradual decline in physiological functions, involving the increased number in senescent cells into the body. Several pathways lead to senescence, including oxidative stress and persistent inflammation, as well as energy failure such as mitochondrial dysfunction and deregulated autophagy, being ROS, AMPK, SIRTs, mTOR, IGF-1, and p53 key regulators of the metabolic control, connecting aging to the pathways which drive towards diseases. In addition, senescence can be induced by cellular replication, which resulted from telomere shortening. Taken together, it is possible to draw a common pathway unifying aging to cardiovascular diseases, and the central point of this process, senescence, can be the target for new therapies, which may result in the healthspan matching the lifespan.
Collapse
Affiliation(s)
- Arthur José Pontes Oliveira de Almeida
- Departamento de Ciências Farmacêuticas/Centro de Ciências da Saúde, Universidade Federal da Paraíba, Cidade Universitária-Campus I, Caixa Postal 5009, 58.051-970 João Pessoa, PB, Brazil
| | - Thaís Porto Ribeiro
- Departamento de Ciências Farmacêuticas/Centro de Ciências da Saúde, Universidade Federal da Paraíba, Cidade Universitária-Campus I, Caixa Postal 5009, 58.051-970 João Pessoa, PB, Brazil
| | - Isac Almeida de Medeiros
- Departamento de Ciências Farmacêuticas/Centro de Ciências da Saúde, Universidade Federal da Paraíba, Cidade Universitária-Campus I, Caixa Postal 5009, 58.051-970 João Pessoa, PB, Brazil
| |
Collapse
|
38
|
Rudenko TE, Bobkova IN, Kamyshova ES, Gorelova IA. [Role of the mechanisms of replicative cellular senescence in structural and functional changes of the vascular wall in chronic kidney disease]. TERAPEVT ARKH 2017; 89:102-109. [PMID: 28745697 DOI: 10.17116/terarkh2017896102-109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This review considers the mechanisms and risk factors for the development of replicative cellular senescence of the vascular wall in patients with CKD and discusses therapeutic approaches to slowing the accelerated vascular aging.
Collapse
Affiliation(s)
- T E Rudenko
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| | - I N Bobkova
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| | - E S Kamyshova
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| | - I A Gorelova
- I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| |
Collapse
|
39
|
Booth SA, Charchar FJ. Cardiac telomere length in heart development, function, and disease. Physiol Genomics 2017; 49:368-384. [DOI: 10.1152/physiolgenomics.00024.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Telomeres are repetitive nucleoprotein structures at chromosome ends, and a decrease in the number of these repeats, known as a reduction in telomere length (TL), triggers cellular senescence and apoptosis. Heart disease, the worldwide leading cause of death, often results from the loss of cardiac cells, which could be explained by decreases in TL. Due to the cell-specific regulation of TL, this review focuses on studies that have measured telomeres in heart cells and critically assesses the relationship between cardiac TL and heart function. There are several lines of evidence that have identified rapid changes in cardiac TL during the onset and progression of heart disease as well as at critical stages of development. There are also many factors, such as the loss of telomeric proteins, oxidative stress, and hypoxia, that decrease cardiac TL and heart function. In contrast, antioxidants, calorie restriction, and exercise can prevent both cardiac telomere attrition and the progression of heart disease. TL in the heart is also indicative of proliferative potential and could facilitate the identification of cells suitable for cardiac rejuvenation. Although these findings highlight the involvement of TL in heart function, there are important questions regarding the validity of animal models, as well as several confounding factors, that need to be considered when interpreting results and planning future research. With these in mind, elucidating the telomeric mechanisms involved in heart development and the transition to disease holds promise to prevent cardiac dysfunction and potentiate regeneration after injury.
Collapse
Affiliation(s)
- S. A. Booth
- Faculty of Science and Technology, School of Applied and Biomedical Sciences, Federation University Australia, Balllarat, Australia
| | - F. J. Charchar
- Faculty of Science and Technology, School of Applied and Biomedical Sciences, Federation University Australia, Balllarat, Australia
- Department of Physiology, The University of Melbourne, Melbourne, Australia; and
- Department of Cardiovascular Sciences, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
40
|
“Pro-youthful” factors in the “labyrinth” of cardiac rejuvenation. Exp Gerontol 2016; 83:1-5. [DOI: 10.1016/j.exger.2016.07.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/12/2016] [Accepted: 07/13/2016] [Indexed: 12/22/2022]
|
41
|
Abstract
Telomeres are tandem repeat DNA sequences present at the ends of each eukaryotic chromosome to stabilize the genome structure integrity. Telomere lengths progressively shorten with each cell division. Inflammation and oxidative stress, which are implicated as major mechanisms underlying cardiovascular diseases, increase the rate of telomere shortening and lead to cellular senescence. In clinical studies, cardiovascular risk factors such as smoking, obesity, sedentary lifestyle, and hypertension have been associated with short leukocyte telomere length. In addition, low telomerase activity and short leukocyte telomere length have been observed in atherosclerotic plaque and associated with plaque instability, thus stroke or acute myocardial infarction. The aging myocardium with telomere shortening and accumulation of senescent cells limits the tissue regenerative capacity, contributing to systolic or diastolic heart failure. In addition, patients with ion-channel defects might have genetic imbalance caused by oxidative stress-related accelerated telomere shortening, which may subsequently cause sudden cardiac death. Telomere length can serve as a marker for the biological status of previous cell divisions and DNA damage with inflammation and oxidative stress. It can be integrated into current risk prediction and stratification models for cardiovascular diseases and can be used in precise personalized treatments. In this review, we summarize the current understanding of telomeres and telomerase in the aging process and their association with cardiovascular diseases. In addition, we discuss therapeutic interventions targeting the telomere system in cardiovascular disease treatments.
Collapse
|
42
|
Saki B, Bahrami A, Ebrahim K, Abedi-Yekta A, Hedayati M. Effect of concurrent training on telomere length in patients with myocardial infarction: Randomised clinical trial of cardiac rehabilitation. GENE REPORTS 2016. [DOI: 10.1016/j.genrep.2016.07.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
43
|
Yeh JK, Wang CY. Telomeres and Telomerase in Cardiovascular Diseases. Genes (Basel) 2016; 7:genes7090058. [PMID: 27598203 PMCID: PMC5042389 DOI: 10.3390/genes7090058] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 08/25/2016] [Accepted: 08/29/2016] [Indexed: 12/20/2022] Open
Abstract
Telomeres are tandem repeat DNA sequences present at the ends of each eukaryotic chromosome to stabilize the genome structure integrity. Telomere lengths progressively shorten with each cell division. Inflammation and oxidative stress, which are implicated as major mechanisms underlying cardiovascular diseases, increase the rate of telomere shortening and lead to cellular senescence. In clinical studies, cardiovascular risk factors such as smoking, obesity, sedentary lifestyle, and hypertension have been associated with short leukocyte telomere length. In addition, low telomerase activity and short leukocyte telomere length have been observed in atherosclerotic plaque and associated with plaque instability, thus stroke or acute myocardial infarction. The aging myocardium with telomere shortening and accumulation of senescent cells limits the tissue regenerative capacity, contributing to systolic or diastolic heart failure. In addition, patients with ion-channel defects might have genetic imbalance caused by oxidative stress-related accelerated telomere shortening, which may subsequently cause sudden cardiac death. Telomere length can serve as a marker for the biological status of previous cell divisions and DNA damage with inflammation and oxidative stress. It can be integrated into current risk prediction and stratification models for cardiovascular diseases and can be used in precise personalized treatments. In this review, we summarize the current understanding of telomeres and telomerase in the aging process and their association with cardiovascular diseases. In addition, we discuss therapeutic interventions targeting the telomere system in cardiovascular disease treatments.
Collapse
Affiliation(s)
- Jih-Kai Yeh
- Department of Cardiology, Chang Gung Memorial Hospital, 33305 Taoyuan, Taiwan.
| | - Chao-Yung Wang
- Department of Cardiology, Chang Gung Memorial Hospital, 33305 Taoyuan, Taiwan.
- Chang Gung University College of Medicine, 33302 Taoyuan, Taiwan.
| |
Collapse
|
44
|
Bartold PM, Ivanovski S, Darby I. Implants for the aged patient: biological, clinical and sociological considerations. Periodontol 2000 2016; 72:120-34. [DOI: 10.1111/prd.12133] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2015] [Indexed: 02/06/2023]
|
45
|
Zurek M, Altschmied J, Kohlgrüber S, Ale-Agha N, Haendeler J. Role of Telomerase in the Cardiovascular System. Genes (Basel) 2016; 7:genes7060029. [PMID: 27322328 PMCID: PMC4929428 DOI: 10.3390/genes7060029] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/06/2016] [Accepted: 06/14/2016] [Indexed: 12/22/2022] Open
Abstract
Aging is one major risk factor for the incidence of cardiovascular diseases and the development of atherosclerosis. One important enzyme known to be involved in aging processes is Telomerase Reverse Transcriptase (TERT). After the discovery of the enzyme in humans, TERT had initially only been attributed to germ line cells, stem cells and cancer cells. However, over the last few years it has become clear that TERT is also active in cells of the cardiovascular system including cardiac myocytes, endothelial cells, smooth muscle cells and fibroblasts. Interference with the activity of this enzyme greatly contributes to cardiovascular diseases. This review will summarize the findings on the role of TERT in cardiovascular cells. Moreover, recent findings concerning TERT in different mouse models with respect to cardiovascular diseases will be described. Finally, the extranuclear functions of TERT will be covered within this review.
Collapse
Affiliation(s)
- Mark Zurek
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Duesseldorf, Germany.
| | - Joachim Altschmied
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Duesseldorf, Germany.
| | - Stefanie Kohlgrüber
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Duesseldorf, Germany.
| | - Niloofar Ale-Agha
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Duesseldorf, Germany.
| | - Judith Haendeler
- IUF-Leibniz Research Institute for Environmental Medicine, 40225 Duesseldorf, Germany.
- Central Institute of Clinical Chemistry and Laboratory Medicine, Medical Faculty, University of Duesseldorf, 40225 Duesseldorf, Germany.
| |
Collapse
|
46
|
Aix E, Gutiérrez-Gutiérrez Ó, Sánchez-Ferrer C, Aguado T, Flores I. Postnatal telomere dysfunction induces cardiomyocyte cell-cycle arrest through p21 activation. J Cell Biol 2016; 213:571-83. [PMID: 27241915 PMCID: PMC4896054 DOI: 10.1083/jcb.201510091] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 05/06/2016] [Indexed: 12/21/2022] Open
Abstract
The molecular mechanisms that drive mammalian cardiomyocytes out of the cell cycle soon after birth remain largely unknown. Here, we identify telomere dysfunction as a critical physiological signal for cardiomyocyte cell-cycle arrest. We show that telomerase activity and cardiomyocyte telomere length decrease sharply in wild-type mouse hearts after birth, resulting in cardiomyocytes with dysfunctional telomeres and anaphase bridges and positive for the cell-cycle arrest protein p21. We further show that premature telomere dysfunction pushes cardiomyocytes out of the cell cycle. Cardiomyocytes from telomerase-deficient mice with dysfunctional telomeres (G3 Terc(-/-)) show precocious development of anaphase-bridge formation, p21 up-regulation, and binucleation. In line with these findings, the cardiomyocyte proliferative response after cardiac injury was lost in G3 Terc(-/-) newborns but rescued in G3 Terc(-/-)/p21(-/-) mice. These results reveal telomere dysfunction as a crucial signal for cardiomyocyte cell-cycle arrest after birth and suggest interventions to augment the regeneration capacity of mammalian hearts.
Collapse
Affiliation(s)
- Esther Aix
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | | | | | - Tania Aguado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | - Ignacio Flores
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| |
Collapse
|
47
|
Deane JA, Ong YR, Cain JE, Jayasekara WSN, Tiwari A, Carlone DL, Watkins DN, Breault DT, Gargett CE. The mouse endometrium contains epithelial, endothelial and leucocyte populations expressing the stem cell marker telomerase reverse transcriptase. Mol Hum Reprod 2016; 22:272-84. [PMID: 26740067 DOI: 10.1093/molehr/gav076] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 12/22/2015] [Indexed: 12/11/2022] Open
Abstract
STUDY HYPOTHESIS The mouse endometrium harbours stem/progenitor cells that express the stem cell marker mouse telomerase reverse transcriptase (mTert). STUDY FINDING We used a mouse carrying a transgenic reporter for mTert promoter activity to identify rare endometrial populations of epithelial and endothelial cells that express mTert. WHAT IS KNOWN ALREADY Stem/progenitor cells are hypothesized to be responsible for the remarkable regenerative capacity of the endometrium, but the lack of convenient endometrial stem/progenitor markers in the mouse has hampered investigations into the identity of these cells. STUDY DESIGN, SAMPLES/MATERIALS, METHODS A mouse containing a green fluorescent protein (GFP) reporter under the control of the telomerase reverse transcriptase promoter (mTert-GFP) was used to identify potential stem/progenitor cells in the endometrium. mTert promoter activity was determined using fluorescence microscopy and flow cytometry to identify GFP(+) cells. GFP(+) cells were examined for epithelial, stromal, endothelial, leucocyte and proliferation markers and bromodeoxyuridine retention to determine their identity. The endometrium of ovariectomized mice was compared to that of intact cycling mice to establish the role of ovarian hormones in maintaining mTert-expressing cells. MAIN RESULTS AND THE ROLE OF CHANCE We found that mTert-GFP is expressed by rare luminal and glandular epithelial cells (0.3% of epithelial cells by flow cytometry), rare CD45(-) cells in the stromal compartment (0.028 ± 0.010% of stromal cells by microscopy) and many CD45(+) leucocytes. Ovariectomy resulted in significant decrease of mTert-GFP(+) epithelial cells (P = 0.029 for luminal epithelium; P = 0.034 for glandular epithelium) and a decrease in the percentage of mTert-GFP(+) CD45(+) leucocytes in the stromal compartment (P = 0.015). However, CD45(-) mTert-GFP(+) cells in the stromal compartment were maintained in ovariectomized mice. This population is enriched for cells bearing the endothelial marker CD31 (10.3% of CD90(-) CD45(-) and 97.8% CD90(+) CD45(-) by flow cytometry). CD45(-) mTert-GFP(+) cells also immunostained for the endothelial marker von Willebrand factor. These results suggest that the endometrial epithelium and vasculature are foci of stem/progenitor activity and provide a system to investigate molecular mechanisms involved in endometrial regeneration and repair. LIMITATIONS, REASONS FOR CAUTION The stem/progenitor activity of endometrial mTert-GFP(+) cells needs to be experimentally verified. WIDER IMPLICATIONS OF THE FINDINGS The identification and characterization of mTert-expressing progenitor cells in the mouse will facilitate the identification of equivalent populations in the human endometrium that are likely to be involved in endometrial function, fertility and disease. LARGE-SCALE DATA Not applicable. STUDY FUNDING AND COMPETING INTERESTS This study was funded by National Health and Medical Research Council (NHMRC) of Australia grants (1085435, C.E.G., J.A.D.), 1021127 (C.E.G.), NHMRC Senior Research Fellowship (1042298, C.E.G.), the Victorian Infrastructure Support Program, U.S. National Institutes of Health grant R01 DK084056 (D.T.B.) and the Harvard Stem Cell Institute (D.T.B.). The authors have no conflicts of interest to declare.
Collapse
Affiliation(s)
- James A Deane
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St., Clayton, Victoria, Australia Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| | - Y Rue Ong
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St., Clayton, Victoria, Australia
| | - Jason E Cain
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - W Samantha N Jayasekara
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Abhilasha Tiwari
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St., Clayton, Victoria, Australia
| | - Diana L Carlone
- Boston Children's Hospital, Harvard Medical School/Harvard Stem Cell Institute, Boston, MA, USA
| | - D Neil Watkins
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria, Australia Present address: The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia Present address: UNSW Faculty of Medicine, St Vincent's Clinical School, St Vincent's Hospital, Randwick, New South Wales, Australia
| | - David T Breault
- Boston Children's Hospital, Harvard Medical School/Harvard Stem Cell Institute, Boston, MA, USA
| | - Caroline E Gargett
- The Ritchie Centre, Hudson Institute of Medical Research, 27-31 Wright St., Clayton, Victoria, Australia Department of Obstetrics and Gynaecology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
48
|
Liu T, Yu H, Ding L, Wu Z, Gonzalez De Los Santos F, Liu J, Ullenbruch M, Hu B, Martins V, Phan SH. Conditional Knockout of Telomerase Reverse Transcriptase in Mesenchymal Cells Impairs Mouse Pulmonary Fibrosis. PLoS One 2015; 10:e0142547. [PMID: 26555817 PMCID: PMC4640706 DOI: 10.1371/journal.pone.0142547] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/25/2015] [Indexed: 12/31/2022] Open
Abstract
Telomerase is typically expressed in cellular populations capable of extended replication, such as germ cells, tumor cells, and stem cells, but is also induced in tissue injury, repair and fibrosis. Its catalytic component, telomerase reverse transcriptase (TERT) is induced in lung fibroblasts from patients with fibrotic interstitial lung disease and in rodents with bleomycin-induced pulmonary fibrosis. To evaluate the fibroblast specific role of TERT in pulmonary fibrosis, transgenic mice bearing a floxed TERT allele were generated, and then crossed with an inducible collagen α2(I)-Cre mouse line to generate fibroblast specific TERT conditional knockout mice. TERT-specific deficiency in mesenchymal cells caused attenuation of pulmonary fibrosis as manifested by reduced lung hydroxyproline content, type I collagen and α-smooth muscle actin mRNA levels. The TERT-deficient mouse lung fibroblasts displayed decreased cell proliferative capacity and higher susceptibility to induced apoptosis compared with control cells. Additionally TERT deficiency was associated with heightened α-smooth muscle actin expression indicative of myofibroblast differentiation. However the impairment of cell proliferation and increased susceptibility to apoptosis would cause a reduction in the myofibroblast progenitor population necessary to mount a successful myofibroblast-dependent fibrotic response. These findings identified a key role for TERT in fibroblast proliferation and survival essential for pulmonary fibrosis.
Collapse
Affiliation(s)
- Tianju Liu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Hongfeng Yu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Lin Ding
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Zhe Wu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | | | - Jianhua Liu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Matthew Ullenbruch
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Biao Hu
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
| | - Vanessa Martins
- Faculty of Medicine, University of São Paulo, São Paulo, Brazil
| | - Sem H. Phan
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
49
|
Valiente-Alandi I, Albo-Castellanos C, Herrero D, Arza E, Garcia-Gomez M, Segovia JC, Capecchi M, Bernad A. Cardiac Bmi1(+) cells contribute to myocardial renewal in the murine adult heart. Stem Cell Res Ther 2015; 6:205. [PMID: 26503423 PMCID: PMC4620653 DOI: 10.1186/s13287-015-0196-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/15/2015] [Accepted: 10/02/2015] [Indexed: 12/19/2022] Open
Abstract
Introduction The mammalian adult heart maintains a continuous, low cardiomyocyte turnover rate throughout life. Although many cardiac stem cell populations have been studied, the natural source for homeostatic repair has not yet been defined. The Polycomb protein BMI1 is the most representative marker of mouse adult stem cell systems. We have evaluated the relevance and role of cardiac Bmi1+ cells in cardiac physiological homeostasis. Methods Bmi1CreER/+;Rosa26YFP/+ (Bmi1-YFP) mice were used for lineage tracing strategy. After tamoxifen (TM) induction, yellow fluorescent protein (YFP) is expressed under the control of Rosa26 regulatory sequences in Bmi1+ cells. These cells and their progeny were tracked by FACS, immunofluorescence and RT-qPCR techniques from 5 days to 1 year. Results FACS analysis of non-cardiomyocyte compartment from TM-induced Bmi1-YFP mice showed a Bmi1+-expressing cardiac progenitor cell (Bmi1-CPC: B-CPC) population, SCA-1 antigen-positive (95.9 ± 0.4 %) that expresses some stemness-associated genes. B-CPC were also able to differentiate in vitro to the three main cardiac lineages. Pulse-chase analysis showed that B-CPC remained quite stable for extended periods (up to 1 year), which suggests that this Bmi1+ population contains cardiac progenitors with substantial self-maintenance potential. Specific immunostaining of Bmi1-YFP hearts serial sections 5 days post-TM induction indicated broad distribution of B-CPC, which were detected in variably sized clusters, although no YFP+ cardiomyocytes (CM) were detected at this time. Between 2 to 12 months after TM induction, YFP+ CM were clearly identified (3 ± 0.6 % to 6.7 ± 1.3 %) by immunohistochemistry of serial sections and by flow cytometry of total freshly isolated CM. B-CPC also contributed to endothelial and smooth muscle (SM) lineages in vivo. Conclusions High Bmi1 expression identifies a non-cardiomyocyte resident cardiac population (B-CPC) that contributes to the main lineages of the heart in vitro and in vivo. Electronic supplementary material The online version of this article (doi:10.1186/s13287-015-0196-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Iñigo Valiente-Alandi
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Carmen Albo-Castellanos
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,Vivebiotech, San Sebastian, Spain.
| | - Diego Herrero
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain.
| | - Elvira Arza
- Microscopy Unit, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain.
| | - Maria Garcia-Gomez
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)- Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain.
| | - José C Segovia
- Hematopoietic Innovative Therapies Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT)- Centro de Investigaciones Biomédicas en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Advanced Therapies Mixed Unit, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD, UAM), Madrid, Spain.
| | - Mario Capecchi
- Howard Hughes Medical Institute University of Utah, Salt Lake City, UT, USA.
| | - Antonio Bernad
- Cardiovascular Development and Repair Department, Spanish National Cardiovascular Research Center (CNIC), Madrid, Spain. .,Immunology and Oncology Department, Spanish National Center for Biotechnology (CNB-CSIC), Madrid, Spain.
| |
Collapse
|
50
|
Pulp Revascularization on Permanent Teeth with Open Apices in a Middle-aged Patient. J Endod 2015; 41:1571-5. [PMID: 26071100 DOI: 10.1016/j.joen.2015.04.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Revised: 03/04/2015] [Accepted: 04/28/2015] [Indexed: 12/17/2022]
Abstract
Pulp revascularization is a promising procedure for the treatment of adolescents' immature permanent teeth with necrotic pulp and/or apical periodontitis. However, the ability to successfully perform pulp revascularization in a middle-aged patient remains unclear. A 39-year-old woman was referred for treatment of teeth #20 and #29 with necrotic pulp, extensive periapical radiolucencies, and incomplete apices. Pulp revascularization procedures were attempted, including root canal debridement, triple antibiotic paste medication, and platelet-rich plasma transplantation to act as a scaffold. Periapical radiographic and cone-beam computed tomographic examinations were used to review the changes in the apical lesions and root apex configuration. The patient remained asymptomatic throughout the 30-month follow-up. Periapical radiographic examination revealed no change in the apical lesions of either tooth at 8 months. The periapical radiolucency disappeared on tooth #20 and significantly decreased on tooth #29 by the 30-month follow-up, findings that were also confirmed by cone-beam computed tomographic imaging. No evidence of root lengthening or thickening was observed. Successful revascularization was achieved in a middle-aged patient's teeth.
Collapse
|