R. B. Goldschmidt, In and Out of the Ivory Tower, p.311, 1960.

B. G. Childs, M. Durik, D. J. Baker, and J. M. Van-deursen, Cellular senescence in aging and age-related disease: from mechanisms to therapy, Nature medicine, vol.21, pp.1424-1435, 2015.

A. Cournil and T. B. Kirkwood, If you would live long, choose your parents well, Trends in genetics: TIG, vol.17, pp.233-235, 2001.

P. Hasty, The impact of DNA damage, genetic mutation and cellular responses on cancer prevention, longevity and aging: observations in humans and mice, Mechanisms of ageing and development, vol.126, pp.71-77, 2005.

J. H. Hoeijmakers, DNA damage, aging, and cancer, The New England journal of medicine, vol.361, pp.1475-1485, 2009.

S. I. Liochev, Which Is the Most Significant Cause of Aging?, Antioxidants, vol.4, pp.793-810, 2015.

M. T. Mc-auley, Modelling the molecular mechanisms of aging, Bioscience reports, vol.37, 2017.

S. J. Mitchell, M. Scheibye-knudsen, D. L. Longo, and R. De-cabo, Animal Models of Aging Research: Implications for Human Aging and Age-Related Diseases, Annual Review of Animal Biosciences, vol.3, pp.283-303, 2015.

P. S. Azevedo, B. F. Polegato, M. F. Minicucci, S. A. Paiva, and L. A. Zornoff, Cardiac Remodeling: Concepts, Clinical Impact, Pathophysiological Mechanisms and Pharmacologic Treatment, Arquivos brasileiros de cardiologia, vol.106, pp.62-69, 2016.

E. Dirkx, Nfat and miR-25 cooperate to reactivate the transcription factor Hand2 in heart failure, Nature Cell Biology, vol.15, 2013.

E. E. Creemers, A. A. Wilde, and Y. M. Pinto, Heart failure: advances through genomics, Nature reviews. Genetics, vol.12, pp.357-362, 2011.

, An integrated encyclopedia of DNA elements in the human genome, Nature, vol.489, pp.57-74, 2012.

C. Lopez-otin, M. A. Blasco, L. Partridge, M. Serrano, and G. Kroemer, The hallmarks of aging, Cell, vol.153, pp.1194-1217, 2013.

G. C. Van-almen, MicroRNA-18 and microRNA-19 regulate CTGF and TSP-1 expression in age-related heart failure, Aging cell, vol.10, pp.769-779, 2011.

J. A. Klein, The harlequin mouse mutation downregulates apoptosis-inducing factor, Nature, vol.419, pp.367-374, 2002.

V. P. Van-empel, Downregulation of apoptosis-inducing factor in harlequin mutant mice sensitizes the myocardium to oxidative stress-related cell death and pressure overload-induced decompensation, Circulation research, vol.96, pp.92-101, 2005.

D. T. Lucas and L. I. Szweda, Cardiac reperfusion injury: aging, lipid peroxidation, and mitochondrial dysfunction, Proc Natl Acad Sci, vol.95, pp.510-514, 1998.

D. T. Lucas and L. I. Szweda, Cardiac reperfusion injury: Aging, lipid peroxidation, and mitochondrial dysfunction, Proceedings of the National Academy of Sciences 95, 510, 1998.

C. Heymes, Increased myocardial NADPH oxidase activity in human heart failure, Journal of the American College of Cardiology, vol.41, pp.471-477, 2003.

G. Lenaz, Mitochondrial bioenergetics in aging, Biochimica et Biophysica Acta (BBA) -Bioenergetics, vol.1459, pp.397-404, 2000.

H. Ryu, ERCC1 expression status predicts the response and survival of patients with metastatic or recurrent cervical cancer treated via platinum-based chemotherapy, Medicine, vol.96, 2017.

J. Mcwhir, J. Selfridge, D. J. Harrison, S. Squires, and D. W. Melton, Mice with DNA repair gene (ERCC-1) deficiency have elevated levels of p53, liver nuclear abnormalities and die before weaning, Nature genetics, vol.5, pp.217-224, 1993.

G. Weeda, Disruption of mouse ERCC1 results in a novel repair syndrome with growth failure, nuclear abnormalities and senescence, Current biology: CB, vol.7, pp.427-439, 1997.

Y. Liu, The telomerase reverse transcriptase is limiting and necessary for telomerase function in vivo, Current biology: CB, vol.10, pp.1459-1462, 2000.

Y. Liu, H. Kha, M. Ungrin, M. O. Robinson, and L. Harrington, Preferential maintenance of critically short telomeres in mammalian cells heterozygous for mTert, Proceedings of the National Academy of Sciences of the United States of America, vol.99, pp.3597-3602, 2002.

A. Canela, E. Vera, P. Klatt, and M. A. Blasco, High-throughput telomere length quantification by FISH and its application to human population studies, Proceedings of the National Academy of Sciences of the United States of America, vol.104, pp.5300-5305, 2007.

D. Meyer and T. , Telomere Length as Cardiovascular Aging Biomarker: JACC Review Topic of the Week, Journal of the American College of Cardiology, vol.72, pp.805-813, 2018.

E. Vera, The rate of increase of short telomeres predicts longevity in mammals, Cell reports, vol.2, pp.732-737, 2012.

E. Sahin, Telomere dysfunction induces metabolic and mitochondrial compromise, Nature, vol.470, pp.359-365, 2011.

S. Andrews and . Fastqc, A Quality Control tool for High Throughput Sequence Data, 2014.

A. M. Bolger, M. Lohse, and B. Usadel, Trimmomatic: a flexible trimmer for Illumina sequence data, Bioinformatics, vol.30, pp.2114-2120, 2014.

D. Kim, B. Langmead, and S. L. Salzberg, HISAT: a fast spliced aligner with low memory requirements, Nature methods, vol.12, pp.357-360, 2015.

M. Pertea, StringTie enables improved reconstruction of a transcriptome from RNA-seq reads, Nature biotechnology, vol.33, pp.290-295, 2015.

M. I. Love, W. Huber, and S. Anders, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq. 2, Genome biology, vol.15, 2014.

G. Yu, L. G. Wang, Y. Han, and Q. Y. He, clusterProfiler: an R package for comparing biological themes among gene clusters, Omics: a journal of integrative biology, vol.16, pp.284-287, 2012.

M. Pinti, Aging of the immune system: Focus on inflammation and vaccination, European journal of immunology, vol.46, pp.2286-2301, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02452484

N. A. Duggal, G. Niemiro, S. D. Harridge, R. J. Simpson, and J. M. Lord, Can physical activity ameliorate immunosenescence and thereby reduce age-related multi-morbidity?, Nature reviews. Immunology, vol.19, pp.563-572, 2019.

M. Dizdaroglu and P. Jaruga, Mechanisms of free radical-induced damage to DNA, Free radical research, vol.46, pp.382-419, 2012.

Z. A. Zhao, Lack of Cardiac Improvement After Cardiosphere-Derived Cell Transplantation in Aging Mouse Hearts, Circulation research, vol.123, pp.21-31, 2018.

A. B. Gevaert, Endothelial Senescence Contributes to Heart Failure With Preserved Ejection Fraction in an Aging Mouse Model, Circulation. Heart failure, vol.10, 2017.

J. M. Snyder, J. M. Ward, and P. M. Treuting, Cause-of-Death Analysis in Rodent Aging Studies, Veterinary pathology, vol.53, pp.233-243, 2016.

, With Chartbook on Long-term Trends in Health, 2016.

R. Anderson, Length-independent telomere damage drives post-mitotic cardiomyocyte senescence, The EMBO journal, vol.38, 2019.
URL : https://hal.archives-ouvertes.fr/inserm-02445949

C. M. Smith, The mouse Gene Expression Database (GXD): 2019 update, Nucleic Acids Res, vol.47, pp.774-779, 2019.

C. Parks, Comparison and Functional Genetic Analysis of Striatal Protein Expression Among Diverse Inbred Mouse Strains, Frontiers in molecular neuroscience, vol.12, 2019.

D. Bottomly, Evaluating gene expression in C57BL/6J and DBA/2J mouse striatum using RNA-Seq and microarrays, PLoS One, vol.6, 2011.