. Cellules, expression d'oncogène et conditions de culture cellulaire

.. .. Traitements,

. Microscopie-haut-débit and .. .. Et-immunofluorescence,

. , ARN : extraction, RT, qPCR et séquençages hauts débits

. .. Sirna,

.. .. Western-blot,

.. .. Crispr,

. .. Bibliographie,

B. Acosta, J. C. Banito, A. Wuestefeld, T. Georgilis, A. Janich et al., A complex secretory program orchestrated by the inflammasome controls paracrine senescence, Nature Cell Biology, vol.15, issue.8, pp.978-990, 2013.

J. C. Acosta, A. O'loghlen, A. Banito, M. V. Guijarro, A. Augert et al., Chemokine signaling via the CXCR2 receptor reinforces senescence, Cell, vol.133, issue.6, pp.1006-1018, 2008.

M. M. Ahmed, D. Chendil, S. Lele, K. Venkatasubbarao, S. Dey et al., Early growth response-1 gene: potential radiation response gene marker in prostate cancer, American Journal of Clinical Oncology, vol.24, issue.5, pp.500-505, 2001.

O. Aksoy, A. Chicas, T. Zeng, Z. Zhao, M. Mccurrach et al., The atypical E2F family member E2F7 couples the p53 and RB pathways during cellular senescence, Genes and Development, vol.26, issue.14, pp.1546-1557, 2012.

G. Alexandre, V. Céline, M. Patrice, C. Carole, and H. Philippe, Effects of topical corticosteroids on cell proliferation, cell cycle progression and apoptosis: In vitro comparison on HaCaT, International Journal of Pharmaceutics, 2014.

L. G. Aoude, K. A. Wadt, A. L. Pritchard, and N. K. Hayward, Genetics of familial melanoma: 20 years after CDKN2A. Pigment Cell and Melanoma Research, 2015.

P. W. Atadja, K. F. Stringer, and K. T. Riabowol, Loss of serum response element-binding activity and hyperphosphorylation of serum response factor during cellular aging, Molecular and Cellular Biology, vol.14, issue.7, pp.4991-4999, 1994.

E. Ayroldi, L. Cannarile, G. Migliorati, G. Nocentini, D. V. Delfino et al., Mechanisms of the anti-inflammatory effects of glucocorticoids: Genomic and nongenomic interference with MAPK signaling pathways, FASEB Journal, vol.26, issue.12, pp.4805-4820, 2012.

E. Ayroldi, G. Migliorati, S. Bruscoli, C. Marchetti, O. Zollo et al., Modulation of T-cell activation by the glucocorticoid-induced leucine zipper factor via inhibition of nuclear factor ?B, Blood, vol.98, issue.3, pp.743-753, 2001.

E. Ayroldi, M. G. Petrillo, A. Bastianelli, M. C. Marchetti, S. Ronchetti et al., L-GILZ binds p53 and MDM2 and suppresses tumor growth through p53 activation in human, 2014.

M. Benhamed, U. Herbig, T. Ye, A. Dejean, and O. Bischof, Senescence is an endogenous trigger for microRNA-directed transcriptional gene silencing in human cells, Nature Cell Biology, vol.14, issue.3, pp.266-275, 2012.

D. C. Bennett, Human melanocyte senescence and melanoma susceptibility genes. Oncogene, 2003.

D. C. Berry, Y. Jiang, R. W. Arpke, E. D. Berglund, M. Kyba et al., Cellular Aging Contributes to Failure of Cold-Induced Beige Adipocyte Formation in Old Mice and Humans, Cell Metabolism, vol.25, issue.1, pp.1-16, 2017.

C. Bevona, W. Goggins, T. Quinn, J. Fullerton, and H. Tsao, Cutaneous Melanomas Associated With Nevi, Archives of Dermatology, vol.139, issue.12, p.1620, 2003.

S. Bhattacharyya, M. Wu, F. Fang, W. G. Tourtellotte, C. Feghali-bostwick et al., Early growth response transcription factors: Key mediators of fibrosis and novel targets for anti-fibrotic therapy, Matrix Biology, vol.30, issue.4, pp.235-242, 2011.

S. C. Biddie, B. L. Conway-campbell, and S. L. Lightman, Dynamic regulation of glucocorticoid signalling in health and disease, Rheumatology, vol.51, issue.3, pp.403-412, 2012.

,

O. Bischof and R. I. Martínez-zamudio, MicroRNAs and lncRNAs in senescence: A re-view, IUBMB Life, vol.67, issue.4, pp.255-267, 2015.

M. Bittner, P. Meltzer, Y. Chen, Y. Jiang, E. Seftor et al., Molecular classification of cutaneous malignant melanoma by gene expression profiling, Nature, vol.406, issue.6795, pp.536-540, 2000.

A. M. Bode and Z. Dong, The functional contrariety of JNK, Molecular Carcinogenesis, vol.46, issue.8, pp.591-598, 2007.

J. S. Boehm, J. J. Zhao, J. Yao, S. Y. Kim, R. Firestein et al., Integrative Genomic Approaches Identify IKBKE as a Breast Cancer Oncogene, Cell, vol.129, issue.6, pp.1065-1079, 2007.

I. Bogdan, J. Smolle, H. Kerl, G. Burg, and R. Böni, Melanoma ex naevo: a study of the associated naevus, Melanoma Research, vol.13, issue.2, pp.213-217, 2003.

,

N. A. Bonito, J. Drechsler, S. Stoecker, C. R. Carmo, M. J. Seckl et al., Control of gp130 expression by the mitogen-activated protein kinase ERK2, Oncogene, vol.33, issue.17, pp.2255-2263, 2014.

K. Boon, N. W. Bailey, J. Yang, M. P. Steel, S. Groshong et al., Molecular Phenotypes Distinguish Patients with Relatively Stable from Progressive Idiopathic Pulmonary Fibrosis (IPF), PLoS ONE, vol.4, issue.4, 2009.
DOI : 10.1371/journal.pone.0005134

URL : https://journals.plos.org/plosone/article/file?id=10.1371/journal.pone.0005134&type=printable

P. T. Bot, I. E. Hoefer, J. P. Sluijter, P. Van-vliet, A. M. Smits et al., Increased expression of the transforming growth factor-beta signaling pathway, endoglin, and early growth response-1 in stable plaques, Stroke, vol.40, issue.2, pp.439-447, 2009.

,

M. Braig and C. A. Schmitt, Oncogene-induced senescence: Putting the brakes on tumor development, Cancer Research, vol.66, issue.6, pp.2881-2884, 2006.
DOI : 10.1158/0008-5472.can-05-4006

URL : http://cancerres.aacrjournals.org/content/66/6/2881.full.pdf

K. Brown, S. Gerstberger, L. Carlson, G. Franzoso, and U. Siebenlist, Control of I kappa B-alpha proteolysis by site-specific, signal-induced phosphorylation, Science, issue.5203, pp.1485-1488, 1995.

I. Brusuker, J. M. Rhodes, and R. Goldman, ?-Galactosidase-An indicator of the maturational stage of mouse and human mononuclear phagocytes, Journal of Cellular Physiology, vol.112, issue.3, pp.385-390, 1982.

G. Buchwalter, C. Gross, and B. Wasylyk, Ets ternary complex transcription factors, 2004.
DOI : 10.1016/j.gene.2003.09.028

J. C. Buckingham, Glucocorticoids: exemplars of multi-tasking, British Journal of Pharmacology, p.147, 2006.
DOI : 10.1038/sj.bjp.0706456

URL : http://europepmc.org/articles/pmc1760726?pdf=render

,

B. Buehring, R. Viswanathan, N. Binkley, and W. Busse, Glucocorticoid-induced osteoporosis: An update on effects and management, Journal of Allergy and Clinical Immunology, vol.132, issue.5, pp.1019-1030, 2013.
DOI : 10.1016/j.jaci.2013.08.040

D. V. Bulavin, C. Phillips, B. Nannenga, O. Timofeev, L. A. Donehower et al., Inactivation of the Wip1 phosphatase inhibits mammary tumorigenesis through p38 MAPK-mediated activation of the p16Ink4a-p19 Arf pathway, Nature Genetics, vol.36, issue.4, pp.343-350, 2004.

I. E. Bultink, M. Baden, and W. F. Lems, Glucocorticoid-induced osteoporosis: an update on current pharmacotherapy and future directions, Expert Opinion on Pharmacotherapy, vol.14, issue.2, pp.185-197, 2013.
DOI : 10.1517/14656566.2013.761975

T. Buschmann, O. Potapova, A. Bar-shira, V. N. Ivanov, S. Y. Fuchs et al., Z, 2001.

, Jun NH2-terminal kinase phosphorylation of p53 on Thr-81 is important for p53 stabilization and transcriptional activities in response to stress, vol.21, pp.2743-2754

J. M. Busillo, K. M. Azzams, and J. A. Cidlowski, Glucocorticoids sensitize the innate immune system through regulation of the NLRP3 inflammasome, Journal of Biological Chemistry, vol.286, issue.44, pp.38703-38713, 2011.

J. M. Busillo and J. A. Cidlowski, The five Rs of glucocorticoid action during inflammation: Ready, reinforce, repress, resolve, and restore, Trends in Endocrinology and Metabolism, vol.24, issue.3, pp.109-119, 2013.

F. Buttgereit, A fresh look at glucocorticoids: How to use an old ally more effectively, Bulletin of the NYU Hospital for Joint Diseases, vol.70, pp.26-29, 2012.

A. Calipel, G. Lefevre, C. Pouponnot, F. Mouriaux, A. Eychène et al., Mutation of B-Raf in Human Choroidal Melanoma Cells Mediates Cell Proliferation and Transformation through the MEK/ERK Pathway, Journal of Biological Chemistry, vol.278, issue.43, pp.42409-42418, 2003.

A. Calogero, L. Cuomo, M. Onofrio, U. De-grazia, P. Spinsanti et al., Expression of Egr-1 correlates with the transformed phenotype and the type of viral latency in EBV genome positive lymphoid cell lines, Oncogene, vol.13, issue.10, pp.2105-2112, 1996.

J. Campisi, J. Andersen, P. Kapahi, and S. Melov, Cellular senescence: a link between cancer and age related degenerative disease? Seminars in Cancer Biology, vol.21, pp.354-359, 2011.

J. Campisi and F. Adda-di-fagagna, Cellular senescence: when bad things happen to good cells, Nature Reviews. Molecular Cell Biology, vol.8, issue.9, pp.729-740, 2007.

, Genomic Classification of Cutaneous Melanoma, Cancer Genome Atlas Network, vol.161, issue.7, pp.1681-1696, 2015.

X. Cao, R. A. Koski, A. Gashler, M. Mckiernan, C. F. Morris et al., Identification and characterization of the egr-1 gene product, a DNA-binding zinc finger protein by differentiation and growth signals, Molecular and Celluar Biology, vol.10, issue.5, pp.1931-1939, 1990.

X. Cao, R. Mahendran, G. R. Guy, and Y. H. Tan, Detection and characterization of cellular EGR1 binding to its recognition site, Journal of Biological Chemistry, vol.268, issue.23, pp.16949-16957, 1993.

G. Caratti, L. Matthews, T. Poolman, S. Kershaw, M. Baxter et al., Glucocorticoid receptor function in health and disease, Clinical Endocrinology, vol.34, issue.9, pp.518-530, 2015.

C. Carrasco-serrano and M. Criado, Glucocorticoid activation of the neuronal nicotinic acetylcholine receptor ?7 subunit gene: Involvement of transcription factor Egr-1, FEBS Letters, vol.566, issue.1-3, pp.247-250, 2004.

A. Carrel, On the Permanent Life of Tissues Outside of the Organism. The Journal of p53 tumor suppressor, PLoS Biology, vol.6, issue.12, pp.2853-2868, 1912.

,

S. Courtois-cox, S. M. Genther-williams, E. E. Reczek, B. W. Johnson, L. T. Mcgillicuddy et al., A negative feedback signaling network underlies oncogene-induced senescence, Cancer Cell, vol.10, issue.6, pp.459-472, 2006.

,

V. J. Craig, L. Zhang, J. S. Hagood, and C. A. Owen, Matrix metalloproteinases as therapeutic targets for idiopathic pulmonary fibrosis, American Journal of Respiratory Cell and Molecular Biology, 2015.

E. Crescenzi, G. Palumbo, and H. J. Brady, Bcl-2 activates a programme of premature senescence in human carcinoma cells, The Biochemical Journal, vol.375, pp.263-274, 2003.

J. D. Croxtall, P. T. Van-hal, Q. Choudhury, D. W. Gilroy, and R. J. Flower, Different glucocorticoids vary in their genomic and non-genomic mechanism of action in A549 cells, British Journal of Pharmacology, vol.135, issue.2, pp.511-519, 2002.

R. Culerrier, M. Carraz, C. Mann, and M. Djabali, MSK1 triggers the expression of the INK4AB/ARF locus in oncogene-induced senescence, Molecular Biology of the Cell, vol.27, issue.17, pp.2726-2734, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01412354

O. E. Dadzie, S. Yang, A. Emley, M. Keady, J. Bhawan et al., RAS and RAF mutations in banal melanocytic aggregates contiguous with primary cutaneous melanoma: clues to melanomagenesis, British Journal of Dermatology, vol.160, issue.2, pp.368-375, 2009.

,

P. Dalle-pezze, G. Nelson, E. G. Otten, V. I. Korolchuk, T. B. Kirkwood et al., Dynamic modelling of pathways to cellular senescence reveals strategies for targeted interventions, PLoS Computational Biology, vol.10, issue.8, 2014.

W. E. Damsky and M. Bosenberg, Melanocytic nevi and melanoma: Unraveling a complex relationship, Oncogene, vol.36, issue.42, pp.5771-5792, 2017.

W. E. Damsky, G. Micevic, K. Meeth, V. Muthusamy, D. P. Curley et al., mTORC1 activation blocks brafV600E-induced growth arrest but is insufficient for melanoma formation, Cancer Cell, vol.27, issue.1, pp.41-56, 2015.

D. Dankort, D. P. Curley, R. A. Cartlidge, B. Nelson, A. N. Karnezis et al., BrafV600E cooperates with Pten loss to induce metastatic melanoma, Nature Genetics, issue.5, pp.544-552, 2009.

D. Dankort, E. Filenova, M. Collado, M. Serrano, K. Jones et al., A new mouse model to explore the initiation , progression , and therapy tumors, Genes & Development, vol.21, issue.415, pp.379-384, 2007.

A. Das, D. Chendil, S. Dey, M. Mohiuddin, M. Mohiuddin et al., Ionizing Radiation Down-regulates p53 Protein in Primary Egr-1-/-Mouse Embryonic Fibroblast Cells Causing Enhanced Resistance to Apoptosis, Journal of Biological Chemistry, vol.276, issue.5, pp.3279-3286, 2001.

M. Das, F. Jiang, H. K. Sluss, C. Zhang, K. M. Shokat et al., Suppression of p53-dependent senescence by the JNK signal transduction pathway, Proceedings of the National Academy of Sciences of the United States of America, vol.104, pp.15759-15764, 2007.

H. Davies, G. R. Bignell, C. Cox, P. Stephens, S. Edkins et al., Mutations of the BRAF gene in human cancer, Nature, issue.6892, pp.949-954, 2002.

,

K. De-bosscher, W. Vanden-berghe, I. M. Beck, W. Van-molle, N. Hennuyer et al., A fully dissociated compound of plant origin for inflammatory gene repression, Proceedings of the National Academy of Sciences of the United States of America, vol.102, pp.15827-15832, 2005.

,

D. Eyman, M. Damodarasamy, S. R. Plymate, and M. J. Reed, CCL5 secreted by senescent aged fibroblasts induces proliferation of prostate epithelial cells and expression of genes that modulate angiogenesis, Journal of Cellular Physiology, vol.220, issue.2, pp.376-381, 2009.

,

M. Fang, S. A. Wee, K. Ronski, H. Fan, S. Tao et al., Evidence of EGR1 as a differentially expressed gene among proliferative skin diseases, Genomic Medicine, vol.1, issue.1-2, pp.75-85, 2007.

L. Fardet, I. Petersen, and I. Nazareth, Risk of cardiovascular events in people prescribed glucocorticoids with iatrogenic Cushing's syndrome: Cohort study, BMJ (Online), vol.345, issue.7871, 2012.

S. R. Florell, L. J. Meyer, K. M. Boucher, P. A. Porter-gill, M. Hart et al., Longitudinal assessment of the nevus phenotype in a melanoma kindred, Journal of Investigative Dermatology, vol.123, issue.3, pp.576-582, 2004.

D. Franchimont, Overview of the actions of glucocorticoids on the immune response: A good model to characterize new pathways of immunosuppression for new treatment strategies, Annals of the New York Academy of Sciences, vol.1024, issue.1, pp.124-137, 2004.

,

C. Frémin, M. K. Saba-el-leil, K. Lévesque, S. L. Ang, and S. Meloche, Functional redundancy of ERK1 and ERK2 MAP kinases during development, Cell Reports, vol.12, issue.6, pp.913-921, 2015.

A. Freund, C. K. Patil, and J. Campisi, p38MAPK is a novel DNA damage response-independent regulator of the senescence-associated secretory phenotype, The EMBO Journal, vol.30, issue.8, pp.1536-1548, 2011.

R. Fukunaga and T. Hunter, MNK1, a new MAP kinase-activated protein kinase, isolated by a novel expression screening method for identifying protein kinase substrates, EMBO Journal, vol.16, issue.8, pp.1921-1933, 1997.

C. Gaggioli, M. Deckert, G. Robert, P. Abbe, M. Batoz et al., HGF induces fibronectin matrix synthesis in melanoma cells through MAP kinasedependent signaling pathway and induction of Egr-1, Oncogene, vol.24, issue.8, pp.1423-1433, 2005.

S. Gansauge, F. Gansauge, H. Gause, B. Poch, M. H. Schoenberg et al., The induction of apoptosis in proliferating human fibroblasts by oxygen radicals is associated with a p53-and p21(WAF1CIP1) induction, FEBS Letters, vol.404, issue.1, pp.59-67, 1997.

M. J. Garnett and R. Marais, Guilty as charged: B-RAF is a human oncogene, Cancer Cell, vol.6, issue.4, pp.313-319, 2004.

A. Gashler and V. P. Sukhatme, Early Growth Response Protein 1 (Egr-1): Prototype of a Zincfinger Family of Transcription Factors, Progress in Nucleic Acid Research, vol.50, pp.191-224, 1995.

A. C. Gavin and A. R. Nebreda, A MAP kinase docking site is required for phosphorylation and activation of p90(rsk)/MAPKAP kinase-1, Current Biology, vol.9, issue.5, pp.281-284, 1999.

, , pp.80120-80121

C. Gennari, Differential effect of glucocorticoids on calcium absorption and bone mass, British Journal of Rheumatology, vol.32, issue.2, pp.11-14, 1993.

,

G. Gerlini, P. Romagnoli, and N. Pimpinelli, Skin cancer and immunosuppression, Critical Reviews in Oncology/Hematology, issue.1, pp.127-136, 2005.

,

S. Glück, B. Guey, M. F. Gulen, K. Wolter, T. W. Kang et al., Innate immune sensing of cytosolic chromatin fragments through cGAS promotes senescence, Nature Cell Biology, vol.19, issue.9, pp.1061-1070, 2017.

V. K. Goel, N. Ibrahim, G. Jiang, M. Singhal, S. Fee et al., Melanocytic nevus-like hyperplasia and melanoma in transgenic BRAFV600E mice, Oncogene, vol.28, issue.23, pp.2289-2298, 2009.

A. M. Goldminz, S. C. Au, N. Kim, A. B. Gottlieb, and P. F. Lizzul, NF-??B: An essential transcription factor in psoriasis, Journal of Dermatological Science, 2013.

V. C. Gray-schopfer, S. C. Cheong, H. Chong, J. Chow, T. Moss et al., Cellular senescence in naevi and immortalisation in melanoma: A role for p16?, British Journal of Cancer, vol.95, issue.4, pp.496-505, 2006.

S. Grotegut, D. Von-schweinitz, G. Christofori, and F. Lehembre, Hepatocyte growth factor induces cell scattering through MAPK/Egr-1-mediated upregulation of Snail, The EMBO Journal, vol.25, issue.15, pp.3534-3545, 2006.

G. L. Grove and V. J. Cristofalo, Characterization of the cell cycle of cultured human diploid cells: effects of aging and hydrocortisone, Journal of Cellular Physiology, vol.90, issue.3, pp.415-422, 1977.

M. Guha, M. A. O'connell, R. Pawlinski, A. Hollis, P. Mcgovern et al., Lipopolysaccharide activation of the MEK-ERK1/2 pathway in human monocytic cells mediates tissue factor and tumor necrosis factor ? expression by inducing Elk-1 phosphorylation and Egr-1 expression, Blood, vol.98, issue.5, pp.1429-1439, 2001.

V. Gupta, N. Awasthi, and B. J. Wagner, Specific activation of the glucocorticoid receptor and modulation of signal transduction pathways in human lens epithelial cells, Investigative Ophthalmology & Visual Science, vol.48, issue.4, pp.1724-1734, 2007.

J. Gustafsson, Historical overview of nuclear receptors, The Journal of Steroid Biochemistry and Molecular Biology, 2015.

M. Gutierrez-mecinas, A. F. Trollope, A. Collins, H. Morfett, S. A. Hesketh et al., Long-lasting behavioral responses to stress involve a direct interaction of glucocorticoid receptors with ERK1/2-MSK1-Elk-1 signaling, Proceedings of the National Academy of Sciences, vol.108, issue.33, pp.13806-13811, 2011.

H. A. Haenssle, N. Mograby, A. Ngassa, T. Buhl, S. Emmert et al., Association of Patient Risk Factors and Frequency of Nevus-Associated Cutaneous Melanomas, JAMA Dermatology, vol.152, issue.3, p.291, 2016.

S. Haferkamp, L. L. Scurr, T. M. Becker, M. Frausto, R. F. Kefford et al., Oncogeneinduced senescence does not require the p16 INK4a or p14ARF melanoma tumor suppressors, Journal of Investigative Dermatology, vol.129, issue.8, pp.1983-1991, 2009.

S. Haferkamp, S. L. Tran, T. M. Becker, L. L. Scurr, R. F. Kefford et al., The relative contributions of the p53 and pRb pathways in oncogene-induced melanocyte senescence, Aging, vol.1, issue.6, pp.542-556, 2009.

B. M. Hall, V. Balan, A. S. Gleiberman, E. Strom, P. Krasnov et al., Aging of mice is associated with p16(Ink4a)-and ?-galactosidasepositive macrophage accumulation that can be induced in young mice by senescent cells, Aging, vol.8, issue.7, pp.1294-1315, 2016.

J. Han and P. Sun, The pathways to tumor suppression via route p38, Trends in Biochemical Sciences, vol.32, issue.8, pp.364-371, 2007.

B. Y. Hanaoka, C. A. Peterson, and L. J. Crofford, Glucocorticoid effects on skeletal muscle: Benefit and risk in patients with autoimmune inflammatory rheumatoid diseases, Expert Review of Clinical Immunology, vol.8, issue.8, pp.695-697, 2012.

B. Y. Hanaoka, C. A. Peterson, C. Horbinski, and L. J. Crofford, Implications of glucocorticoid therapy in idiopathic inflammatory myopathies, Nature Reviews Rheumatology, 2012.

K. E. Hansen, B. Kleker, N. Safdar, and C. M. Bartels, A systematic review and metaanalysis of glucocorticoid-induced osteoporosis in children, Seminars in Arthritis and Rheumatism, 2014.

M. Harada, M. Suzuki, T. Ikeda, T. Kaneko, S. Harada et al., Clonality in Nevocellular and Melanoma: An Expression-Based Clonality Analysis at the X-Linked Genes by Polymerase Chain Reaction, Journal of Investigative Dermatology, vol.109, issue.5, pp.656-660, 1997.

T. Hardy, F. Oakley, Q. M. Anstee, and C. P. Day, Nonalcoholic Fatty Liver Disease: Pathogenesis and Disease Spectrum, Annual Review of Pathology: Mechanisms of Disease, vol.11, issue.1, pp.451-496, 2016.

A. Harris and J. Seckl, Glucocorticoids, prenatal stress and the programming of disease, Hormones and Behavior, vol.59, issue.3, pp.279-289, 2011.

R. N. Hasan and A. I. Schafer, Hemin upregulates Egr-1 expression in vascular smooth muscle cells via reactive oxygen species ERK-1/2-Elk-1 and NF-?B, Circulation Research, vol.102, issue.1, pp.42-50, 2008.

R. Hass, M. Brach, H. Gunji, S. Kharbanda, and D. Kufe, Inhibition of EGR-1 and NF-?B gene expression by dexamethasone during phorbol ester-induced human monocytic differentiation, Biochemical Pharmacology, vol.44, issue.8, pp.1569-1576, 1992.

R. Hayashi, H. Wada, K. Ito, and I. M. Adcock, Effects of glucocorticoids on gene transcription, European Journal of Pharmacology, vol.500, issue.1-3, pp.51-62, 2004.

,

L. Hayflick and P. S. Moorhead, The serial cultivation of human diploid cell strains, Experimental Cell Research, vol.25, issue.3, pp.90192-90198, 1961.

L. E. Heasley and S. Han, JNK Regulation of Oncogenesis, Molecules and Cells, vol.21, issue.2, pp.167-173, 2006.

L. Hecker, N. J. Logsdon, D. Kurundkar, A. Kurundkar, K. Bernard et al., Reversal of persistent fibrosis in aging by targeting Nox4-Nrf2 redox imbalance, Science Translational Medicine, vol.6, issue.231, 2014.

I. C. Hellstrom, S. K. Dhir, J. C. Diorio, and M. J. Meaney, Maternal licking regulates hippocampal glucocorticoid receptor transcription through a thyroid hormone-serotonin-NGFI-A signalling cascade, Philosophical Transactions of the Royal Society B: Biological Sciences, vol.367, pp.2495-2510, 1601.

A. Helman, A. Klochendler, N. Azazmeh, Y. Gabai, E. Horwitz et al., p16(Ink4a)-induced senescence of pancreatic beta cells enhances insulin secretion, vol.22, 2016.

M. T. Hemann and M. Narita, Oncogenes and senescence: Breaking down in the fast lane, Genes and Development, vol.21, issue.1, pp.1-5, 2007.

H. Henneicke, M. Herrmann, R. Kalak, T. C. Brennan-speranza, U. Heinevetter et al., Corticosterone selectively targets endo-cortical surfaces by an osteoblast-dependent mechanism, Bone, vol.49, issue.4, pp.733-742, 2011.

U. Herbig, W. Jobling, B. P. Chen, D. J. Chen, and J. M. Sedivy, Telomere shortening triggers senescence of human cells through a pathway involving ATM, p53, and p21CIP1, but not p16INK4a, Molecular Cell, vol.14, issue.4, pp.501-513, 2004.

M. A. Hermoso, T. Matsuguchi, K. Smoak, and J. A. Cidlowski, Glucocorticoids and Tumor Necrosis Factor Alpha Cooperatively Regulate Toll-Like Receptor 2 Gene Expression, Molecular and Cellular Biology, vol.24, issue.11, pp.4743-4756, 2004.

J. Hjoberg, L. Le, A. Imrich, V. Subramaniam, S. I. Mathew et al., Induction of early growth-response factor 1 by platelet-derived growth factor in human airway smooth muscle, American Journal of Physiology. Lung Cellular and Molecular Physiology, vol.286, issue.4, pp.817-842, 2004.

T. G. Hofmann, S. P. Hehner, S. Bacher, W. Dröge, and M. L. Schmitz, Various glucocorticoids differ in their ability to induce gene expression, apoptosis and to repress NF-?B-dependent transcription, FEBS Letters, vol.441, issue.3, pp.441-446, 1998.

D. J. Holt and D. W. Grainger, Senescence and quiescence induced compromised function in cultured macrophages, Biomaterials, vol.33, issue.30, pp.7497-7507, 2012.

,

J. Honkaniemi, J. S. Zhang, F. M. Longo, and F. R. Sharp, Stress induces zinc finger immediate early genes in the rat adrenal gland, Brain Research, vol.877, issue.2, pp.2673-2674, 2000.

R. Houben, S. Ortmann, A. Drasche, J. Troppmair, M. J. Herold et al., Proliferation arrest in B-Raf mutant melanoma cell lines upon MAPK pathway activation, Journal of Investigative Dermatology, vol.129, issue.2, pp.406-414, 2009.

T. C. Hsu and P. S. Moorhead, Mammalian chromosomes in vitro. VII. heteroploidy in human cell strains, Journal of the National Cancer Institute, vol.18, issue.3, pp.463-471, 1957.

,

C. Hu, T. Chang, C. Cheng, C. Liu, J. Wu et al., Snail associates with EGR-1 and SP-1 to upregulate transcriptional activation of p15INK4b, The FEBS Journal, vol.277, issue.5, pp.1202-1218, 2010.

R. Huang, Y. Fan, I. De-belle, C. Niemeyer, M. M. Gottardis et al., Decreased Egr-1 expression in human, mouse and rat mammary cells and tissues correlates with tumor formation, International Journal of Cancer, vol.72, issue.1, pp.102-109, 1997.

, , vol.72, 19970703.

W. H. Hudson, C. Youn, and E. A. Ortlund, The structural basis of direct glucocorticoid-mediated transrepression, Nature Structural and Molecular Biology, vol.20, issue.1, pp.53-58, 2013.

,

L. Hui, L. Bakiri, A. Mairhorfer, N. Schweifer, C. Haslinger et al., p38? suppresses normal and cancer cell proliferation by antagonizing the JNK-c-Jun pathway, Nature Genetics, vol.39, issue.6, pp.741-749, 2007.

P. Hui, A. Perkins, and E. Glusac, Assessment of clonality in melanocytic nevi, Journal of Cutaneous Pathology, vol.28, issue.3, pp.140-144, 2001.

A. Iannetti, A. C. Ledoux, S. J. Tudhope, H. Sellier, B. Zhao et al., Regulation of p53 and Rb links the alternative NF-?B pathway to EZH2 expression and cell senescence, PLoS Genetics, vol.10, issue.9, 2014.

F. Ishikawa, Cellular Senescence as a Stress Response, Cornea, vol.25, 2006.

O. Itani, K. Z. Liu, K. L. Cornish, J. R. Campbell, and C. P. Thomas, Glucocorticoids stimulate human sgk1 gene expression by activation of a GRE in its 5'-flanking region, American Journal of Physiology. Endocrinology and Metabolism, vol.283, issue.5, pp.971-980, 2002.

,

H. Iwasa, J. Han, and F. Ishikawa, Mitogen-activated protein kinase p38 defines the common senescence-signalling pathway, Genes to Cells, vol.8, issue.2, pp.131-144, 2003.

A. Jalili, C. Wagner, M. Pashenkov, G. Pathria, K. D. Mertz et al., Dual Suppression of the Cyclin-Dependent Kinase Inhibitors CDKN2C and CDKN1A in Human Melanoma, JNCI: Journal of the National Cancer Institute, vol.104, issue.21, pp.1673-1679, 2012.

M. Jeanblanc, S. Ragu, C. Gey, K. Contrepois, R. Courbeyrette et al., Parallel pathways in RAF-induced senescence and conditions for its reversion, Oncogene, issue.25, pp.3072-3085, 2012.

W. R. Jeck, A. P. Siebold, and N. E. Sharpless, Review: A meta-analysis of GWAS and ageassociated diseases, Aging Cell, vol.11, issue.5, pp.727-731, 2012.

A. Ø. Jensen, H. F. Thomsen, M. C. Engebjerg, A. B. Olesen, S. Friis et al., Use of oral glucocorticoids and risk of skin cancer and non-Hodgkin's lymphoma: a population-based case-control study, British Journal of Cancer, vol.100, issue.1, pp.200-205, 2009.

P. B. Joel, J. Smith, T. W. Sturgill, T. L. Fisher, J. Blenis et al., , 1998.

, Estrogen Receptor-Mediated Transcription through Phosphorylation of Ser-167, Molecular and Cellular Biology, vol.18, issue.4, pp.1978-1984

A. Joselow, D. Lynn, T. Terzian, and N. F. Box, Senescence-Like Phenotypes in Human Nevi, Methods in Molecular Biology, issue.1, pp.175-184, 2017.

J. Jun and L. F. Lau, Cellular senescence controls fibrosis in wound healing, Aging, vol.2, issue.9, pp.627-631, 2010.

J. Jun and L. F. Lau, The matricellular protein CCN1 induces fibroblast senescence and restricts fibrosis in cutaneous wound healing, Nature Cell Biology, vol.12, issue.7, pp.676-685, 2010.

M. Kadmiel and J. A. Cidlowski, Glucocorticoid receptor signaling in health and disease, Trends in Pharmacological Sciences, 2013.

H. Kakavand, O. Crainic, T. Lum, S. A. O'toole, R. F. Kefford et al., , 2014.

, Concordant BRAFV600E mutation status in primary melanomas and associated naevi: implications for mutation testing of primary melanomas, Pathology, vol.46, issue.3, pp.193-198

S. Kang, M. Jung, C. W. Kim, and D. Y. Shin, Inactivation of p38 kinase delays the onset of senescence in rabbit articuilar chondrocytes, Mechanisms of Ageing and Development, vol.126, issue.5, pp.591-597, 2005.

T. Kang, T. Yevsa, N. Woller, L. Hoenicke, T. Wuestefeld et al., Senescence surveillance of pre-malignant hepatocytes limits liver cancer development, Nature, vol.479, issue.7374, pp.547-551, 2011.

M. R. Karagas, G. L. Cushing, E. R. Greenberg, L. A. Mott, S. K. Spencer et al., Non-melanoma skin cancers and glucocorticoid therapy, British Journal of Cancer, vol.85, issue.5, pp.683-686, 2001.

R. Z. Karim, W. Li, A. Sanki, M. H. Colman, Y. H. Yang et al., Reduced p16 and Increased Cyclin D1 and pRb Expression Are Correlated with Progression in Cutaneous Melanocytic Tumors, International Journal of Surgical Pathology, vol.17, issue.5, pp.361-367, 2009.

O. Kassel and P. Herrlich, Crosstalk between the glucocorticoid receptor and other transcription factors: Molecular aspects, Molecular and Cellular Endocrinology, vol.275, issue.1-2, pp.13-29, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00531941

O. Kassel, A. Sancono, J. Krätzschmar, B. Kreft, M. Stassen et al., Glucocorticoids inhibit MAP kinase via increased expression and decreased degradation of MKP-1, The EMBO Journal, vol.20, issue.24, pp.7108-7116, 2001.

K. Kaufmann and G. Thiel, Epidermal growth factor and thrombin induced proliferation of immortalized human keratinocytes is coupled to the synthesis of Egr-1, a zinc finger transcriptional regulator, Journal of Cellular Biochemistry, vol.85, issue.2, pp.381-391, 2002.

,

P. A. Keenan, M. W. Jacobson, R. M. Soleymani, M. D. Mayes, M. E. Stress et al., The effect on memory of chronic prednisone treatment in patients with systemic disease, Neurology, vol.47, issue.6, pp.1396-1402, 1996.

L. M. Khachigian, Early growth response-1 in cardiovascular pathobiology, Circulation Research, vol.98, 2006.

S. Kharbanda, T. Nakamura, R. Stone, R. Hass, S. Bernstein et al., Expression of the early growth response 1 and 2 zinc finger genes during induction of monocytic differentiation, The Journal of Clinical Investigation, vol.88, issue.2, pp.571-577, 1991.

,

A. M. Kidger, L. K. Rushworth, J. Stellzig, J. Davidson, C. J. Bryant et al., Dual-specificity phosphatase 5 controls the localized inhibition, propagation, and transforming potential of ERK signaling, Proceedings of the National Academy of Sciences, vol.114, issue.3, pp.317-326, 2017.

K. Kim, C. Chen, R. I. Monzon, and L. F. Lau, Matricellular Protein CCN1 Promotes Regression of Liver Fibrosis through Induction of Cellular Senescence in Hepatic Myofibroblasts, Molecular and Cellular Biology, vol.33, issue.10, pp.2078-2090, 2013.

S. Kim, J. M. Kim, S. H. Shim, and H. I. Chang, Shikonin induces cell cycle arrest in human gastric cancer (AGS) by early growth response 1 (Egr1)-mediated p21 gene expression, Journal of Ethnopharmacology, vol.151, issue.3, pp.1064-1071, 2014.

K. H. Kirsch, Y. Korradi, and J. P. Johnson, Mader: a novel nuclear protein over expressed in human melanomas, Oncogene, vol.12, issue.5, pp.963-971, 1996.

H. Kittler, M. Seltenheim, M. Dawid, H. Pehamberger, K. Wolff et al., Frequency and Characteristics of Enlarging Common Melanocytic Nevi, Archives of Dermatology, vol.136, issue.3, pp.316-320, 2000.

N. C. Klein, C. H. Go, and B. A. Cunha, Infections associated with steroid use, Infectious Disease Clinics of North America, vol.15, issue.2, pp.423-455, 2001.

C. A. Koch, J. L. Doppman, N. J. Patronas, L. K. Nieman, and G. P. Chrousos, Do glucocorticoids cause spinal epidural lipomatosis? When endocrinology and spinal surgery meet, Trends in Endocrinology and Metabolism: TEM, vol.11, issue.3, pp.86-90, 2000.

G. Kollarovic, M. Studencka, L. Ivanova, C. Lauenstein, K. Heinze et al., To senesce or not to senesce: how primary human fibroblasts decide their cell fate after DNA damage, Aging, vol.8, issue.1, 2016.

P. Krimpenfort, K. C. Quon, W. J. Mooi, A. Loonstra, and A. Berns, Loss of p16Ink4a confers susceptibility to metastatic melanoma in mice, Nature, issue.6851, pp.83-86, 2001.

J. Krishnamurthy, M. R. Ramsey, K. L. Ligon, C. Torrice, A. Koh et al., p16INK4a induces an age-dependent decline in islet regenerative potential, Nature, vol.443, issue.7110, pp.453-457, 2006.

K. Krishnaraju, H. Q. Nguyen, D. A. Liebermann, and B. Hoffman, The zinc finger transcription factor Egr-1 potentiates macrophage differentiation of hematopoietic cells, Mol Cell Biol, vol.15, issue.10, pp.5499-5507, 1995.

V. Krizhanovsky, M. Yon, R. A. Dickins, S. Hearn, J. Simon et al., Senescence of Activated Stellate Cells Limits Liver Fibrosis, Cell, vol.134, issue.4, pp.657-667, 2008.

A. Krones-herzig, E. D. Adamson, and D. Mercola, Early growth response 1 protein, an upstream gatekeeper of the p53 tumor suppressor, controls replicative senescence, Proceedings of the National Academy of Sciences of the United States of America, vol.100, pp.3233-3238, 2003.

A. Krones-herzig, S. Mittal, K. Yule, H. Liang, C. English et al., Early growth response 1 acts as a tumor suppressor in vivo and in vitro via regulation of p53, Cancer Research, vol.65, issue.12, pp.5133-5143, 2005.

A. Krtolica, S. Parrinello, S. Lockett, P. Desprez, and J. Campisi, Senescent fibroblasts promote epithelial cell growth and tumorigenesis: A link between cancer and aging, Proceedings of the National Academy of Sciences, vol.98, issue.21, pp.12072-12077, 2001.

,

B. Krynitz, G. Edgren, B. Lindelöf, E. Baecklund, C. Brattström et al., Risk of skin cancer and other malignancies in kidney, liver, heart and lung transplant recipients 1970 to 2008-A Swedish population-based study, International Journal of Cancer, vol.132, issue.6, pp.1429-1438, 2013.

A. Kubosaki, Y. Tomaru, M. Tagami, E. Arner, H. Miura et al., Genomewide investigation of in vivo EGR-1 binding sites in monocytic differentiation, Genome Biology, vol.10, issue.4, 2009.

T. Kuilman, C. Michaloglou, W. J. Mooi, and D. S. Peeper, The essence of senescence, Genes & Development, vol.24, issue.22, pp.2463-2479, 2010.

T. Kuilman, C. Michaloglou, L. C. Vredeveld, S. Douma, R. Van-doorn et al., Oncogene-Induced Senescence Relayed by an Interleukin-Dependent Inflammatory Network, Cell, vol.133, issue.6, pp.1019-1031, 2008.
DOI : 10.1016/j.cell.2008.03.039

URL : https://doi.org/10.1016/j.cell.2008.03.039

T. Kuilman and D. S. Peeper, Senescence-messaging secretome: SMS-ing cellular stress, Nature Reviews Cancer, vol.9, issue.2, pp.81-94, 2009.
DOI : 10.1038/nrc2560

J. Kumbrink, K. H. Kirsch, and J. P. Johnson, EGR1, EGR2, and EGR3 activate the expression of their coregulator NAB2 establishing a negative feedback loop in cells of neuroectodermal and epithelial origin, Journal of Cellular Biochemistry, vol.111, issue.1, pp.207-217, 2010.

,

D. J. Kurz, S. Decary, Y. Hong, and J. D. Erusalimsky, Senescence-associated (beta)-galactosidase reflects an increase in lysosomal mass during replicative ageing of human endothelial cells, Journal of Cell Science, vol.113, pp.3613-3622, 2000.

T. Kuwata, M. Kitagawa, and T. Kasuga, Proliferative activity of primary cutaneous melanocytic tumours, Virchows Archiv. A, Pathological Anatomy and Histopathology, vol.423, issue.5, pp.359-364, 1993.

R. M. Laberge, Y. Sun, A. V. Orjalo, C. K. Patil, A. Freund et al., MTOR regulates the pro-tumorigenic senescence-associated secretory phenotype by promoting IL1A translation, Nature Cell Biology, vol.17, issue.8, pp.1049-1061, 2015.
DOI : 10.1038/ncb3195

URL : http://europepmc.org/articles/pmc4691706?pdf=render

R. M. Laberge, L. Zhou, M. R. Sarantos, F. Rodier, A. Freund et al., Glucocorticoids suppress selected components of the senescence-associated secretory phenotype, International Journal of Cancer, vol.11, issue.4, pp.302-303, 2001.

M. Lazarus and H. Kaufman, An association between corticosteroid use and melanoma recurrence: A case report and review of the literature, Medical Oncology, vol.29, issue.3, pp.2018-2020, 2012.

L. Beau, M. M. Albain, K. S. Larson, R. A. Vardiman, J. W. Davis et al., Clinical and cytogenetic correlations in 63 patients with therapy-related myelodysplastic syndromes and acute nonlymphocytic leukemia: further evidence for characteristic abnormalities of chromosomes no. 5 and 7, Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology, vol.4, issue.3, pp.325-345, 1986.

J. Leclerc, R. Ballotti, and C. Bertolotto, Pathways from senescence to melanoma: focus on MITF sumoylation, 2017.
DOI : 10.1038/onc.2017.292

N. Leclerc, C. A. Luppen, V. V. Ho, S. Nagpal, J. G. Hacia et al., Gene expression profiling of glucocorticoid-inhibited osteoblasts, Journal of Molecular Endocrinology, vol.33, issue.1, pp.175-193, 2004.

N. Leclerc, T. Noh, J. Cogan, D. B. Samarawickrama, E. Smith et al., Opposing effects of glucocorticoids and Wnt signaling on Krox20 and mineral deposition in osteoblast cultures, Journal of Cellular Biochemistry, vol.103, issue.6, pp.1938-1951, 2008.

N. Leclerc, T. Noh, A. Khokhar, E. Smith, and B. Frenkel, Glucocorticoids inhibit osteocalcin transcription in osteoblasts by suppressing Egr2/Krox20-binding enhancer, Arthritis and Rheumatism, vol.52, issue.3, pp.929-939, 2005.
DOI : 10.1002/art.20872

K. H. Lee and J. Kim, Hepatocyte growth factor induced up-regulations of VEGF through Egr1 in hepatocellular carcinoma cells, Clinical & Experimental Metastasis, vol.26, issue.7, pp.685-692, 2009.

S. L. Lee, Y. Sadovsky, A. H. Swirnoff, J. A. Polish, P. Goda et al., Luteinizing hormone deficiency and female infertility in mice lacking the transcription factor NGFI-A (Egr-1), Science, issue.5279, pp.1219-1221, 1996.

,

S. R. Lee, H. K. Kim, J. B. Youm, L. A. Dizon, I. S. Song et al., Non-genomic effect of glucocorticoids on cardiovascular system, Pflugers Archiv-European Journal of Physiology, vol.464, issue.6, pp.1-11, 2012.

R. Lefloch, J. Pouysségur, and P. Lenormand, Single and Combined Silencing of ERK1 and ERK2 Reveals Their Positive Contribution to Growth Signaling Depending on Their Expression Levels, Molecular and Cellular Biology, vol.28, issue.1, pp.511-527, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00319825

M. Lehmann, M. Korfei, K. Mutze, S. Klee, W. Skronska-wasek et al., Senolytic drugs target alveolar epithelial cell function and attenuate experimental lung fibrosis ex vivo, European Respiratory Journal, vol.50, issue.2, p.1602367, 2017.

O. V. Leontieva and M. V. Blagosklonny, DNA damaging agents and p53 do not cause senescence in quiescent cells, while consecutive re-activation of mTOR is associated with conversion to senescence, Aging, vol.2, issue.12, pp.924-935, 2010.

D. A. Lewis, Q. Yi, J. B. Travers, and D. F. Spandau, UVB-induced senescence in human keratinocytes requires a functional insulin-like growth factor-1 receptor and p53, Molecular Biology of the Cell, vol.19, issue.4, pp.1346-1353, 2008.

C. Li, W. Ning, M. A. Matthay, C. Feghali-bostwick, and A. M. Choi, MAPK pathway mediates EGR-1-HSP70-dependent cigarette smoke-induced chemokine production, American Journal of Physiology. Lung Cellular and Molecular Physiology, vol.292, issue.5, pp.1297-303, 2007.

C. Li, D. Yang, X. Cao, F. Wang, D. Hong et al., Fibronectin induces epithelial-mesenchymal transition in human breast cancer MCF-7 cells via activation of calpain, Oncology Letters, vol.13, issue.5, pp.3889-3895, 2017.

H. Li, W. Qian, X. Weng, Z. Wu, H. Li et al., Glucocorticoid receptor and sequential P53 activation by Dexamethasone mediates apoptosis and cell cycle arrest of Osteoblastic MC3T3-E1 cells, PLoS ONE, vol.7, issue.6, 2012.

J. Li, S. Han, W. Cousin, and I. M. Conboy, Age-specific functional epigenetic changes in p21 and p16 in injury-activated satellite cells, Stem Cells, pp.1-16, 2014.

S. Li, M. Mawal-dewan, V. J. Cristofalo, and C. Sell, Enhanced proliferation of human fibroblasts, in the presence of dexamethasone, is accompanied by changes in p21(Waf1/Cip1/Sdi1) and the insulin-like growth factor type 1 receptor, Journal of Cellular Physiology, vol.177, pp.396-401, 1998.

A. W. Lin, M. Barradas, J. C. Stone, L. Van-aelst, M. Serrano et al., Premature senescence involving p53 and p16 is activated in response to constitutive MEK/MAPK mitogenic signaling, Genes and Development, vol.12, issue.19, pp.3008-3019, 1998.

W. M. Lin, S. Luo, A. Muzikansky, A. Z. Lobo, K. K. Tanabe et al., Outcome of patients with de novo versus nevus-associated melanoma, Journal of the American Academy of Dermatology, vol.72, issue.1, pp.54-58, 2015.

C. Liu, V. M. Rangnekar, E. D. Adamson, and D. Mercola, Suppression of growth and transformation and induction of apoptosis by EGR-1, Cancer Gene Therapy, vol.5, issue.1, pp.3-28, 1998.

C. Liu, J. Yao, D. Mercola, and E. D. Adamson, The transcription factor EGR-1 directly transactivates the fibronectin gene and enhances attachment of human glioblastoma cell line U251, Journal of Biological Chemistry, vol.275, issue.27, pp.20315-20323, 2000.

,

J. Liu, L. Grogan, M. M. Nau, C. J. Allegra, E. Chu et al., Physical interaction between p53 and primary response gene Egr-1, International Journal of Oncology, vol.18, issue.4, pp.863-870, 2001.

M. Lohoff, M. Giaisi, R. Köhler, B. Casper, P. H. Krammer et al., Early growth response protein-1 (Egr-1) is preferentially expressed in T helper type 2 (Th2) cells and is involved 148 in acute transcription of the Th2 cytokine interleukin-4, Journal of Biological Chemistry, vol.285, issue.3, pp.1643-1652, 2010.

D. M. Lonard and B. W. Malley, Nuclear Receptor Coregulators: Judges, Juries, and Executioners of Cellular Regulation, Molecular Cell, vol.27, issue.5, pp.691-700, 2007.

,

R. Louw-du-toit, J. P. Hapgood, and D. Africander, Medroxyprogesterone Acetate Differentially Regulates Interleukin (IL)-12 and IL-10 in a Human Ectocervical Epithelial Cell Line in a Glucocorticoid Receptor (GR)-dependent Manner, The Journal of Biological Chemistry, vol.289, issue.45, pp.31136-31149, 2014.

J. Ma, Z. Ren, Y. Ma, L. Xu, Y. Zhao et al., Targeted knockdown of EGR-1 inhibits IL-8 production and IL-8-mediated invasion of prostate cancer cells throughsuppressing EGR-1/NF-?B synergy, Journal of Biological Chemistry, vol.284, issue.50, pp.34600-34606, 2009.

R. M. Mackie, J. English, T. C. Aitchison, C. P. Fitzsimons, and P. Wilson, The number and distribution of benign pigmented moles (melanocytic naevi) in a healthy British population, British Journal of Dermatology, vol.113, issue.2, pp.167-174, 1985.

G. M. Maelandsmo, R. Holm, O. Fodstad, R. S. Kerbel, and V. A. Flørenes, Cyclin kinase inhibitor p21 in malignant melanoma: Reduced expression in metastatic lesions, American Journal of Pathology, vol.149, issue.6, pp.1813-1822, 1996.

N. Malaquin, A. Martinez, and F. Rodier, Keeping the senescence secretome under control: Molecular reins on the senescence-associated secretory phenotype, Experimental Gerontology, vol.82, pp.39-49, 2016.

S. P. Malkoski and R. I. Dorin, Composite Glucocorticoid Regulation at a Functionally Defined Negative Glucocorticoid Response Element of the Human Corticotropin-Releasing Hormone Gene, Molecular Endocrinology, vol.13, issue.10, pp.1629-1644, 1999.

F. A. Mallette, M. F. Gaumont-leclerc, and G. Ferbeyre, The DNA damage signaling pathway is a critical mediator of oncogene-induced senescence, Genes and Development, vol.21, issue.1, pp.43-48, 2007.

D. J. Mangelsdorf, C. Thummel, M. Beato, P. Herrlich, G. Schütz et al., The nuclear receptor superfamily: the second decade, Cell, vol.83, pp.835-839, 1995.

N. Manwani, S. Gagnon, M. Post, S. Joza, L. Muglia et al., Reduced viability of mice with lung epithelial-specific knockout of glucocorticoid receptor, American Journal of Respiratory Cell and Molecular Biology, vol.43, issue.5, pp.599-606, 2010.

,

R. Marais, Y. Light, H. Fpaterson, and C. J. Marshall, Ras recruits Raf-1 to the plasma membrane for activation by tyrosine phosphorylation, The EMBO Journal, issue.13, pp.3136-3145, 1995.

R. Marcotte, C. Lacelle, and E. Wang, Senescent fibroblasts resist apoptosis by downregulating caspase-3, Mechanisms of Ageing and Development, vol.125, pp.777-783, 2004.

C. J. Marshall, MAP kinase kinase kinase, MAP kinase kinase and MAP kinase, Current Opinion in Genetics & Development, vol.4, issue.1, pp.90095-90102, 1994.

D. X. Mason, T. J. Jackson, and A. W. Lin, Molecular signature of oncogenic ras-induced senescence, Oncogene, vol.23, issue.57, pp.9238-9246, 2004.

M. Mawal-dewan, L. Frisoni, V. J. Cristofalo, and C. Sell, Extension of replicative lifespan in WI38 human fibroblasts by dexamethasone treatment is accompanied by suppression of p21Waf1/Cip1/Sdi1 levels, Experimental Cell Research, vol.285, issue.1, pp.91-98, 2003.

, , pp.13-15

M. Mazzantini and O. Di-munno, Glucocorticoid-induced osteoporosis: 2013 update, Reumatismo, vol.66, issue.2, p.144, 2014.

A. S. Mcneal, K. Liu, V. Nakhate, C. Natale, E. K. Duperret et al., CDKN2B loss promotes progression from benign melanocytic nevus to melanoma, Cancer Discovery, vol.5, issue.10, pp.1072-1085, 2015.

S. H. Meijsing, M. A. Pufall, A. Y. So, D. L. Bates, L. Chen et al., DNA binding site sequence directs glucocorticoid receptor structure and activity, Science, vol.324, issue.5925, pp.407-410, 2009.

A. Melk, W. Kittikowit, I. Sandhu, K. M. Halloran, P. Grimm et al., Cell senescence in rat kidneys in vivo increases with growth and age despite lack of telomere shortening, Kidney International, vol.63, issue.6, pp.2134-2143, 2003.

A. Melk, B. M. Schmidt, O. Takeuchi, B. Sawitzki, D. C. Rayner et al., Expression of p16INK4a and other cell cycle regulator and senescence associated genes in aging human kidney, Kidney International, vol.65, issue.2, pp.510-520, 2004.

S. Meloche and J. Pouysségur, The ERK1/2 mitogen-activated protein kinase pathway as a master regulator of the G1-to S-phase transition. Oncogene, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00319874

S. W. Menzies, M. L. Stevenson, D. Altamura, and K. Byth, Variables Predicting Change in Benign Melanocytic Nevi Undergoing Short-term Dermoscopic Imaging, Archives of Dermatology, vol.147, issue.6, 2011.

M. Meyyappan, K. Wheaton, and K. T. Riabowol, Decreased expression and activity of the immediate-early growth response (Egr-1) gene product during cellular senescence, Journal of Cellular Physiology, vol.179, issue.1, pp.29-39, 1999.

C. Michaloglou, L. C. Vredeveld, M. S. Soengas, C. Denoyelle, T. Kuilman et al., BRAFE600-associated senescence-like cell cycle arrest of human naevi, Nature, vol.436, issue.7051, pp.720-724, 2005.

V. C. Midgley and L. M. Khachigian, Fibroblast growth factor-2 induction of platelet-derived growth factor-C chain transcription in vascular smooth muscle cells is ERK-dependent but not JNK-dependent and mediated by Egr-1, Journal of Biological Chemistry, vol.279, issue.39, pp.40289-40295, 2004.

J. Milbrandt, A nerve growth factor-induced gene encodes a possible transcriptional regulatory factor, Science, issue.4828, pp.797-799, 1987.

A. J. Miller and M. C. Mihm, Melanoma. New England Journal of Medicine, vol.355, issue.1, pp.51-65, 2006.

D. M. Miller and K. T. Flaherty, Cyclin-dependent kinases as therapeutic targets in melanoma, Pigment Cell and Melanoma Research, vol.27, issue.3, pp.351-365, 2014.

L. Mire, K. Hollowood, D. Gray, C. Bordea, and F. Wojnarowska, Melanomas in renal transplant recipients, British Journal of Dermatology, vol.154, issue.3, pp.472-477, 2006.

A. Mitchell, C. R. Dass, L. Sun, and L. M. Khachigian, Inhibition of human breast carcinoma proliferation, migration, chemoinvasion and solid tumour growth by DNAzymes targeting the zinc finger transcription factor EGR-1, Nucleic Acids Research, vol.32, issue.10, pp.3065-3069, 2004.

P. R. Mittelstadt and J. D. Ashwell, Inhibition of AP-1 by the Glucocorticoid-inducible Protein GILZ, Journal of Biological Chemistry, vol.276, issue.31, pp.29603-29610, 2001.

,

S. Moghadam-kia and V. P. Werth, Prevention and treatment of systemic glucocorticoid side effects, International Journal of Dermatology, vol.49, issue.3, pp.239-248, 2010.

O. Moiseeva, F. A. Mallette, U. K. Mukhopadhyay, A. Moores, and G. Ferbeyre, DNA damage signaling and p53-dependent senescence after prolonged beta-interferon stimulation, Molecular Biology of the Cell, vol.17, issue.4, pp.1583-1592, 2006.

W. J. Mooi and T. Krausz, Common acquired melanocytic naevi, Biopsy Pathology of Melanocytic Disorders, pp.56-105, 1992.

W. J. Mooi and D. S. Peeper, Pathogenesis of melanocytic naevi: growth arrest linked with cellular senescence?, Histopathology. Histopathology, 2002.

G. R. Mora, K. R. Olivier, J. C. Cheville, R. F. Mitchell, W. L. Lingle et al., The cytoskeleton differentially localizes the early growth response gene-1 protein in cancer and benign cells of the prostate, Molecular Cancer Research, vol.2, issue.2, pp.115-128, 2004.

D. K. Morrison, MAP kinase pathways, Cold Spring Harbor Perspectives in Biology, issue.11, p.4, 2012.

D. Muñoz-espín, M. Cañamero, A. Maraver, G. Gómez-lópez, J. Contreras et al., Programmed cell senescence during mammalian embryonic development, Cell, vol.155, issue.5, pp.1104-1118, 2013.

S. Muthukkumar, P. Nair, S. F. Sells, N. G. Maddiwar, R. J. Jacob et al., Role of EGR-1 in thapsigargin-inducible apoptosis in the melanoma cell line A375-C6, Mol.Cell Biol, vol.15, pp.6262-6272, 1995.

A. S. Muthusamy, L. Mumford, A. Hudson, S. V. Fuggle, P. J. Friend et al., Pancreas Transplantation From Donors After Circulatory Death From the United Kingdom, American Journal of Transplantation, vol.12, issue.8, pp.20131-20138, 1997.

H. Nakhoul, J. Ke, X. Zhou, W. Liao, S. X. Zeng et al., Ribosomopathies: Mechanisms of Disease, Clinical Medicine Insights: Blood Disorders, vol.7, 2014.

M. Narita, S. N?nez, E. Heard, M. Narita, A. W. Lin et al., Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence, Journal of Investigative Dermatology, vol.113, issue.6, pp.28-33, 2000.

R. ;. Newton, K. Abdelmohsen, M. Gorospe, N. Ejiogu, A. B. Zonderman et al., microRNA expression patterns reveal differential expression of target genes with age, PLoS ONE, vol.5, issue.5, pp.603-613, 2000.

R. H. Oakley and J. A. Cidlowski, Cellular processing of the glucocorticoid receptor gene and protein: New mechanisms for generating tissue-specific actions of glucocorticoids, Journal of Biological Chemistry, vol.286, issue.5, pp.3177-3184, 2011.

R. H. Oakley and J. A. Cidlowski, The biology of the glucocorticoid receptor: new signaling mechanisms in health and disease, The Journal of Allergy and Clinical Immunology, vol.132, issue.5, pp.1033-1044, 2013.

F. Olivieri, M. R. Rippo, V. Monsurrò, S. Salvioli, M. Capri et al., MicroRNAs linking inflamm-aging, cellular senescence and cancer, Ageing Research Reviews, vol.12, issue.4, pp.1056-1068, 2013.

M. Oubaha, K. Miloudi, A. Dejda, V. Guber, G. Mawambo et al., , 2016.

, Senescence-associated secretory phenotype contributes to pathological angiogenesis in retinopathy, Science Translational Medicine, vol.8, issue.362, pp.362-144

,

J. Pagel and E. Deindl, Early growth response 1-a transcription factor in the crossfire of signal transduction cascades, Indian Journal of Biochemistry & Biophysics, vol.48, issue.4, pp.226-235, 2011.

E. Parra, L. Gutiérrez, and J. Ferreira, Increased expression of p21Waf1/Cip1 and JNK with costimulation of prostate cancer cell activation by an siRNA Egr-1 inhibitor, Oncology Reports, vol.30, issue.2, pp.911-916, 2013.

S. Parrinello, E. Samper, A. Krtolica, J. Goldstein, S. Melov et al., Oxygen sensitivity severely limits the replicative lifespan of murine fibroblasts, Nature Cell Biology, vol.5, issue.8, pp.741-747, 2003.

E. E. Patton, H. R. Widlund, J. L. Kutok, K. R. Kopani, J. F. Amatruda et al., BRAF mutations are sufficient to promote nevi formation and cooperate with p53 in the genesis of melanoma, Current Biology, vol.15, issue.3, pp.249-254, 2005.

,

E. Pazolli, X. Luo, S. Brehm, K. Carbery, J. J. Chung et al., Senescent stromal-derived osteopontin promotes preneoplastic cell growth, Cancer Research, vol.69, issue.3, pp.1230-1239, 2009.

M. Pedersen, A. Viros, M. Cook, and R. Marais, G12DNRAS and kinase-dead BRAF cooperate to drive naevogenesis and melanomagenesis, Pigment Cell and Melanoma Research, 2014.

A. Pellicoro, P. Ramachandran, J. P. Iredale, and J. A. Fallowfield, Liver fibrosis and repair: Immune regulation of wound healing in a solid organ, Nature Reviews Immunology, vol.14, issue.3, pp.181-194, 2014.

M. Pende, T. L. Fisher, P. B. Simpson, J. T. Russell, J. Blenis et al., Neurotransmitterand growth factor-induced cAMP response element binding protein phosphorylation in glial cell progenitors: role of calcium ions, protein kinase C, and mitogen-activated protein kinase/ribosomal S6 kinase pathway, The Journal of Neuroscience : The Official Journal of the Society for Neuroscience, vol.17, issue.4, pp.1291-1301, 1997.

C. Peyssonnaux, A. Eychène, E. Pimenta, M. Wolley, and M. Stowasser, The Raf / MEK / ERK pathway : new concepts of activation, Biology of the Cell, vol.93, issue.1-2, pp.5137-5142, 2001.

A. Piris, M. C. Mihm, and M. P. Hoang, BAP1 and BRAFV600E expression in benign and malignant melanocytic proliferations, Human Pathology, vol.46, issue.2, pp.239-245, 2015.

,

R. F. Place, E. J. Noonan, and C. Giardina, HDAC inhibition prevents NF-??B activation by suppressing proteasome activity: Down-regulation of proteasome subunit expression stabilizes I??B??, Biochemical Pharmacology, vol.70, issue.3, pp.394-406, 2005.

R. Poirier, H. Cheval, C. Mailhes, S. Garel, P. Charnay et al., Distinct functions of egr gene family members in cognitive processes, Frontiers in Neuroscience, vol.2, issue.1, pp.47-55, 2008.
URL : https://hal.archives-ouvertes.fr/hal-00320617

P. M. Pollock, U. L. Harper, K. S. Hansen, L. M. Yudt, M. Stark et al., High frequency of BRAF mutations in nevi, Nature Genetics, vol.33, issue.1, pp.19-20, 2002.

Y. Pommier, E. Leo, H. Zhang, and C. Marchand, DNA topoisomerases and their poisoning by anticancer and antibacterial drugs. Chemistry and Biology, 2010.
DOI : 10.1016/j.chembiol.2010.04.012

URL : https://doi.org/10.1016/j.chembiol.2010.04.012

N. Popov and J. Gil, Epigenetic regulation of the INK4B-ARF-INK4a locus: In sickness and in 152 health, Epigenetics, vol.5, issue.8, pp.685-690, 2010.

R. C. Poulsen, A. C. Watts, R. J. Murphy, S. J. Snelling, A. J. Carr et al., Glucocorticoids induce senescence in primary human tenocytes by inhibition of sirtuin 1 and activation of the p53/p21 pathway: In vivo and in vitro evidence, Annals of the Rheumatic Diseases, vol.73, issue.7, pp.1405-1413, 2014.

J. N. Poynter, J. T. Elder, D. R. Fullen, R. P. Nair, M. S. Soengas et al., BRAF and NRAS mutations in melanoma and melanocytic nevi, Melanoma Research, vol.16, issue.4, pp.267-273, 2006.
DOI : 10.1097/01.cmr.0000222600.73179.f3

C. M. Price, Telomere flip-flop: An unfolding passage to senescence, EMBO Reports, vol.13, issue.1, pp.5-6, 2012.
DOI : 10.1038/embor.2011.237

URL : http://embor.embopress.org/content/13/1/5.full.pdf

A. Ramalingam, A. Hirai, and E. A. Thompson, Glucocorticoid inhibition of fibroblast proliferation and regulation of the cyclin kinase inhibitor p21Cip1, Molecular Endocrinology, issue.5, pp.577-586, 1997.

M. Raman, W. Chen, and M. H. Cobb, Differential regulation and properties of MAPKs. Oncogene, 2007.
DOI : 10.1038/sj.onc.1210392

URL : https://www.nature.com/articles/1210392.pdf

H. Rayess, M. B. Wang, and E. S. Srivatsan, Cellular senescence and tumor suppressor gene p16, International Journal of Cancer. Journal International Du Cancer, vol.130, issue.8, pp.1715-1725, 2012.
DOI : 10.1002/ijc.27316

URL : http://europepmc.org/articles/pmc3288293?pdf=render

A. Rebbaa, X. Zheng, P. M. Chou, and B. L. Mirkin, Caspase inhibition switches doxorubicininduced apoptosis to senescence, Oncogene, vol.22, issue.18, pp.2805-2811, 2003.
DOI : 10.1038/sj.onc.1206366

URL : https://www.nature.com/articles/1206366.pdf

,

J. P. Reddy and Y. Li, Oncogene-induced senescence and its role in tumor suppression, Journal of Mammary Gland Biology and Neoplasia, vol.16, issue.3, pp.247-256, 2011.
DOI : 10.1007/s10911-011-9221-5

M. Reimann, S. Lee, C. Loddenkemper, J. R. Dörr, V. Tabor et al., Tumor Stroma-Derived TGF-? Limits Myc-Driven Lymphomagenesis via Suv39h1-Dependent Senescence, Cancer Cell, vol.17, issue.3, pp.262-272, 2010.
DOI : 10.1016/j.ccr.2009.12.043

URL : https://doi.org/10.1016/j.ccr.2009.12.043

J. Revest, F. Di-blasi, P. Kitchener, F. Rougé-pont, A. Desmedt et al., The MAPK pathway and Egr-1 mediate stress-related behavioral effects of glucocorticoids, Nature Neuroscience, vol.8, issue.5, pp.664-672, 2005.

J. Revest, N. Kaouane, M. Mondin, A. Le-roux, F. Rougé-pont et al., Glucocorticoid treatment induces expression of Egr-1 and synapsin-I proteins in primary culture of hippocampal neurons, Molecular Psychiatry, vol.15, issue.12, p.1125, 2010.

J. Revollo and J. A. Cidlowski, Mechanisms generating diversity in glucocorticoid receptor signaling, Annals of the New York Academy of ?, 1179, pp.167-178, 2009.

T. Rhen and J. A. Cidlowski, Antiinflammatory action of glucocorticoids-new mechanisms for old drugs, New England Journal of Medicine, pp.1711-1723, 2005.

B. Ritschka, M. Storer, A. Mas, F. Heinzmann, M. C. Ortells et al., The senescence-associated secretory phenotype induces cellular plasticity and tissue regeneration, Genes and Development, vol.31, issue.2, pp.172-183, 2017.

R. Rizzoli and E. Biver, Glucocorticoid-induced osteoporosis: Who to treat with what agent?, Nature Reviews Rheumatology, 2015.

D. Roberts, J. Brown, N. Medley, and S. R. Dalziel, Antenatal corticosteroids for accelerating fetal lung maturation for women at risk of preterm birth, Cochrane Database of Systematic Reviews. Chichester, 2017.

M. J. Robinson and M. H. Cobb, Mitogen-activated protein kinase pathways. Current Opinion in Cell Biology, 1997.

W. A. Robinson, M. Lemon, A. Elefanty, M. Harrison-smith, N. Markham et al., Human acquired naevi are clonal, Melanoma Research, vol.8, issue.6, pp.499-503, 1998.

F. Rodier and J. Campisi, Four faces of cellular senescence, Journal of Cell Biology, 2011.

F. Rodier, J. Coppe, C. K. Patil, W. A. Hoeijmakers, D. P. Muñoz et al., Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion, Nature Cell Biology, vol.11, issue.8, pp.973-979, 2009.

F. Rodier, D. P. Munoz, R. Teachenor, V. Chu, O. Le et al., DNA-SCARS: distinct nuclear structures that sustain damage-induced senescence growth arrest and inflammatory cytokine secretion, Journal of Cell Science, vol.124, issue.1, pp.68-81, 2011.

M. R. Roh, P. Eliades, S. Gupta, and H. Tsao, Genetics of melanocytic nevi, Pigment Cell & Melanoma Research, vol.28, issue.6, pp.661-672, 2015.

I. B. Roninson, Tumor cell senescence in cancer treatment, Cancer Research, vol.63, issue.11, pp.2705-2715, 2003.

A. J. Rose, A. Vegiopoulos, and S. Herzig, Role of glucocorticoids and the glucocorticoid receptor in metabolism: Insights from genetic manipulations, Journal of Steroid Biochemistry and Molecular Biology, vol.122, issue.1-3, pp.10-20, 2010.

B. Rosner and V. J. Cristofalo, Hydrocortisone: a specific modulator of in vitro proliferation and aging, Mechanisms of Ageing and Development, vol.9, pp.485-496, 1979.

O. G. Rössler and G. Thiel, Thrombin induces Egr-1 expression in fibroblasts involving elevation of the intracellular Ca2+ concentration, phosphorylation of ERK and activation of ternary complex factor, BMC Molecular Biology, vol.10, issue.1, 2009.

M. W. Russo, B. R. Sevetson, and J. Milbrandt, Identification of NAB1, a repressor of NGFI-Aand Krox20-mediated transcription, Proceedings of the National Academy of Sciences of the United States of America, vol.92, pp.6873-6877, 1995.

S. J. Ryu, Y. S. Oh, and S. C. Park, Failure of stress-induced downregulation of Bcl-2 contributes to apoptosis resistance in senescent human diploid fibroblasts, Cell Death and Differentiation, vol.14, issue.5, p.1020, 2007.

J. Sage, G. J. Mulligan, L. D. Attardi, A. Miller, S. Chen et al., Targeted disruption of the three Rb-related genes leads to loss of G1control and immortalization, Genes and Development, vol.14, issue.23, pp.3037-3050, 2000.

Z. Salah, M. Maoz, G. Pizov, and R. Bar-shavit, Transcriptional regulation of human proteaseactivated receptor 1: a role for the early growth response-1 protein in prostate cancer, Cancer Research, vol.67, issue.20, pp.9835-9843, 2007.

J. Salotti, K. Sakchaisri, W. G. Tourtellotte, and P. F. Johnson, An Arf-Egr-C/EBP? Pathway Linked to Ras-Induced Senescence and Cancer, Molecular and Cellular Biology, vol.35, issue.5, pp.866-883, 2015.

A. Sanchis, L. Alba, V. Latorre, L. M. Sevilla, and P. Pérez, Keratinocyte-targeted overexpression of the glucocorticoid receptor delays cutaneous wound healing, PLoS ONE, vol.7, issue.1, 2012.

A. Sanchis, P. Bayo, L. M. Sevilla, and P. Pérez, Glucocorticoid receptor antagonizes EGFR function to regulate eyelid development, The International Journal of Developmental Biology, vol.54, issue.10, pp.1473-1480, 2010.
DOI : 10.1387/ijdb.103071as

URL : http://www.ijdb.ehu.es/web/descarga/paper/103071as

L. Sauer, D. Gitenay, C. Vo, and V. Baron, Mutant p53 initiates a feedback loop that involves Egr-1/EGF receptor/ERK in prostate cancer cells, Oncogene, vol.29, issue.18, pp.2628-2637, 2010.

M. J. Schafer, T. A. White, K. Iijima, A. J. Haak, G. Ligresti et al., Cellular senescence mediates fibrotic pulmonary disease, Nature Communications, vol.8, p.14532, 2017.
DOI : 10.1038/ncomms14532

URL : https://www.nature.com/articles/ncomms14532.pdf

J. V. Schaffer, Update on melanocytic nevi in children, Clinics in Dermatology, vol.33, issue.3, pp.368-386, 2015.
DOI : 10.1016/j.clindermatol.2014.12.015

M. Schreiber, A. Kolbus, F. Piu, A. Szabowski, U. Möhle-steinlein et al., Control of cell cycle progression by c-Jun is p53 dependent, Genes and Development, vol.13, issue.5, pp.607-619, 1999.

M. J. Seibel, M. S. Cooper, and H. Zhou, Glucocorticoid-induced osteoporosis: Mechanisms, management, and future perspectives. The Lancet Diabetes and Endocrinology, pp.70045-70052, 2013.
DOI : 10.1016/s2213-8587(13)70045-7

S. F. Sells, S. Muthukkumar, V. P. Sukhatme, S. Crist, V. M. Rangnekar et al., Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16(INK4a), Molecular and Cellular Biology, vol.15, issue.2, pp.81902-81911, 1995.

B. R. Sevetson, J. Svaren, and J. Milbrandt, A novel activation function for NAB proteins in EGRdependent transcription of the luteinizing hormone beta gene, The Journal of Biological Chemistry, vol.275, issue.13, pp.9749-9757, 2000.

L. M. Sevilla, V. Latorre, A. Sanchis, and P. Pérez, Epidermal inactivation of the glucocorticoid receptor triggers skin barrier defects and cutaneous inflammation, Journal of Investigative Dermatology, vol.133, issue.2, pp.361-370, 2013.
DOI : 10.1038/jid.2012.281

URL : https://doi.org/10.1038/jid.2012.281

V. L. Seyfert, S. B. Mcmahon, W. D. Glenn, A. J. Yellen, V. P. Sukhatme et al., Methylation of an immediate-early inducible gene as a mechanism for B cell tolerance induction, Science, issue.4982, pp.797-800, 1990.

,

V. L. Seyfert, V. P. Sukhatme, and J. G. Monroe, Differential expression of a zinc finger-encoding gene in response to positive versus negative signaling through receptor immunoglobulin in murine B lymphocytes, Molecular and Cellular Biology, vol.9, issue.5, pp.2083-2088, 1989.

S. Shah, E. M. King, A. Chandrasekhar, and R. Newton, Roles for the mitogen-Activated protein kinase (MAPK) phosphatase, DUSP1, in feedback control of inflammatory gene expression and repression by dexamethasone, Journal of Biological Chemistry, vol.289, issue.19, pp.13667-13679, 2014.

A. H. Shain and B. C. Bastian, From melanocytes to melanomas, Nature Reviews Cancer, vol.16, issue.6, pp.345-358, 2016.
DOI : 10.1038/nrc.2016.37

A. H. Shain, I. Yeh, I. Kovalyshyn, A. Sriharan, E. Talevich et al., The Genetic Evolution of Melanoma from Precursor Lesions, New England Journal of Medicine, vol.373, issue.20, pp.1926-1936, 2015.

N. E. Sharpless and C. J. Sherr, Forging a signature of in vivo senescence, Nature Reviews Cancer, vol.15, issue.7, pp.397-408, 2015.

K. E. Sheppard, R. B. Pearson, and R. D. Hannan, Unexpected role of CDK4 in a G2/M checkpoint, Cell Cycle, vol.14, issue.9, pp.1351-1352, 2015.

C. J. Sherr and R. A. Depinho, Cellular senescence: mitotic clock or culture shock?, Cell, vol.102, issue.4, pp.46-51, 2000.

J. Shin, J. Yang, J. C. Lee, and K. H. Baek, Depletion of ERK2 but not ERK1 abrogates oncogenic Ras-induced senescence, Cellular Signalling, vol.25, issue.12, pp.2540-2547, 2013.
DOI : 10.1016/j.cellsig.2013.08.014

,

S. Shin, C. A. Dimitri, S. O. Yoon, W. Dowdle, and J. Blenis, ERK2 but Not ERK1 Induces Epithelialto-Mesenchymal Transformation via DEF Motif-Dependent Signaling Events, Molecular Cell, vol.38, issue.1, pp.114-127, 2010.

S. Y. Shin, Y. Y. Bahk, J. Ko, I. Chung, Y. S. Lee et al., Suppression of Egr-1 transcription through targeting of the serum response factor by oncogenic H-Ras, The EMBO Journal, vol.25, issue.5, pp.1093-1103, 2006.

S. Y. Shin, C. G. Kim, S. H. Kim, Y. S. Kim, Y. Lim et al., Chlorpromazine activates p21Waf1/Cip1 gene transcription via early growth response-1 (Egr-1) in C6 glioma cells, Experimental & Molecular Medicine, vol.42, issue.5, pp.395-405, 2010.

,

D. Shitara, M. M. Nascimento, S. Puig, S. Yamada, M. M. Enokihara et al., Nevus-Associated Melanomas: Clinicopathologic Features, American Journal of Clinical Pathology, vol.142, issue.4, pp.485-491, 2014.

D. Shitara, G. Tell-martí, C. Badenas, M. M. Enokihara, L. Alós et al., Mutational status of naevus-associated melanomas, British Journal of Dermatology, vol.173, issue.3, pp.671-680, 2015.

L. K. Smith and J. A. Cidlowski, Glucocorticoid-induced apoptosis of healthy and malignant lymphocytes, Progress in brain research, vol.182, issue.10, pp.82001-82002, 2010.

K. Smoak and J. A. Cidlowski, Glucocorticoids regulate tristetraprolin synthesis and posttranscriptionally regulate tumor necrosis factor alpha inflammatory signaling, Molecular and Cellular Biology, vol.26, issue.23, pp.9126-9135, 2006.

N. J. Solan, H. Miyoshi, E. M. Carmona, G. D. Bren, and C. V. Paya, RelB cellular regulation and transcriptional activity are regulated by p100, Journal of Biological Chemistry, vol.277, issue.2, pp.1405-1418, 2002.

H. Sone and Y. Kagawa, Pancreatic beta cell senescence contributes to the pathogenesis of type 2 diabetes in high-fat diet-induced diabetic mice, Diabetologia, vol.48, issue.1, pp.58-67, 2005.

F. Spaapen, G. G. Van-den-akker, M. M. Caron, P. Prickaerts, C. Rofel et al., The Immediate Early Gene Product EGR1 and Polycomb Group Proteins Interact in Epigenetic Programming during Chondrogenesis, PLoS ONE, vol.8, issue.3, 2013.

R. Srinivasan, G. M. Mager, R. M. Ward, J. Mayer, and J. Svaren, NAB2 represses transcription by interacting with the CHD4 subunit of the nucleosome remodeling and deacetylase (NuRD) complex, The Journal of Biological Chemistry, vol.281, issue.22, pp.15129-15137, 2006.

,

O. C. Stegmaier, Natural regression of the melanocytic nevus, The Journal of Investigative Dermatology, vol.32, issue.3, pp.413-421, 1959.

C. ;. Stellato, A. Mas, A. Robert-moreno, M. Pecoraro, M. C. Ortells et al., Senescence is a developmental mechanism that contributes to embryonic growth and patterning, Posttranscriptional Gene Regulation: Novel Pathways for Glucocorticoids' Antiinflammatory Action, vol.3, pp.1119-1130, 2012.

T. W. Sturgill, L. B. Ray, E. Erikson, and J. L. Maller, Insulin-stimulated MAP-2 kinase phosphorylates and activates ribosomal protein S6 kinase II, Nature, vol.334, issue.6184, pp.715-718, 1988.

I. Sturmlechner, M. Durik, C. J. Sieben, D. J. Baker, and J. M. Van-deursen, Cellular senescence in renal ageing and disease, Nature Reviews Nephrology, vol.13, issue.2, pp.77-89, 2017.

V. P. Sukhatme, Early transcriptional events in cell growth: the Egr family, Journal of the American Society of Nephrology : JASN, vol.1, issue.6, pp.859-866, 1990.

V. P. Sukhatme, X. Cao, L. C. Chang, C. H. Tsai-morris, D. Stamenkovich et al., A zinc finger-encoding gene coregulated with c-fos during growth and differentiation, and after cellular depolarization, Cell, vol.53, issue.1, pp.37-43, 1988.

, , pp.90485-90485

N. Sundahl, J. Bridelance, C. Libert, K. De-bosscher, and I. M. Beck, Selective glucocorticoid receptor modulation: New directions with non-steroidal scaffolds, Pharmacology & Therapeutics, 2015.

M. Surjit, K. P. Ganti, A. Mukherji, T. Ye, G. Hua et al., Widespread negative response elements mediate direct repression by agonist-liganded glucocorticoid receptor, Cell, vol.145, issue.2, pp.224-241, 2011.

L. J. Suva, M. Ernst, and G. A. Rodan, Retinoic Acid Increases zij268 Early Gene Expression in RatPreosteoblastic Cells, Mol Cell Biol, vol.11, issue.5, pp.2503-2510, 1991.

J. Svaren, B. R. Sevetson, E. D. Apel, D. B. Zimonjic, N. C. Popescu et al., NAB2, a corepressor of NGFI-A (Egr-1) and Krox20, is induced by proliferative and differentiative stimuli, Molecular and Cellular Biology, vol.16, issue.7, pp.3545-3553, 1996.

J. Svaren, B. R. Sevetson, T. Golda, J. J. Stanton, A. H. Swirnoff et al., Novel mutants of NAB corepressors enhance activation by Egr transactivators, The EMBO Journal, vol.17, issue.20, pp.6010-6019, 1998.

M. Takekawa, M. Adachi, A. Nakahata, I. Nakayama, F. Itoh et al., p53inducible wip1 phosphatase mediates a negative feedback regulation of p38 MAPK-p53 signaling in response to UV radiation, The EMBO Journal, vol.19, issue.23, pp.6517-6526, 2000.

,

L. Talve, I. Sauroja, Y. Collan, K. Punnonen, and T. Ekfors, Loss of expression of the p16(INK4)/CDKN2 gene in cutaneous malignant melanoma correlates with tumor cell proliferation and invasive stage, International Journal of Cancer, vol.74, issue.3, pp.255-259, 1997.

G. Thiel and G. Cibelli, Regulation of life and death by the zinc finger transcription factor Egr-1, Journal of Cellular Physiology, vol.193, issue.3, pp.287-292, 2002.

A. E. Thigpen, K. M. Cala, J. M. Guileyardo, K. H. Molberg, J. D. Mcconnell et al., Increased Expression of Early Growth Response-1 Messenger Ribonucleic Acid in Prostatic Adenocarcinoma, The Journal of Urology, vol.155, issue.3, pp.66361-66365, 1996.

P. Topilko, S. Schneider-maunoury, G. Levi, A. Trembleau, D. Gourdji et al., Multiple Pituitary and Ovarian Defects in Krox-24 ( NGFI-A, Egr-1 )-Targeted Mice, Molecular Endocrinology, vol.12, issue.1, pp.107-122, 1998.

R. Treisman, Regulation of transcription by MAP kinase cascades, Current Opinion in Cell Biology, vol.8, issue.2, pp.80067-80073, 1996.

J. R. Troche, L. M. Ferrucci, B. Cartmel, D. J. Leffell, A. E. Bale et al., Systemic glucocorticoid use and early-onset basal cell carcinoma, Annals of Epidemiology, vol.24, issue.8, pp.625-627, 2014.

F. Tronche, C. Kellendonk, O. Kretz, P. Gass, K. Anlag et al., Disruption of the glucocorticoid receptor gene in the nervous system results in reduced anxiety, Nature Genetics, vol.23, issue.1, pp.99-103, 1999.

I. P. Trougakos, A. Saridaki, G. Panayotou, and E. S. Gonos, Identification of differentially expressed proteins in senescent human embryonic fibroblasts, Mechanisms of Ageing and Development, vol.127, issue.1, pp.88-92, 2006.

P. Tschandl, A. S. Berghoff, M. Preusser, S. Burgstaller-muehlbacher, H. Pehamberger et al., NRAS and BRAF Mutations in Melanoma-Associated Nevi and Uninvolved Nevi, vol.8, 2013.

S. W. Tyan, M. C. Tsai, C. L. Lin, Y. L. Ma, and E. H. Lee, Serum-and glucocorticoid-inducible kinase 1 enhances zif268 expression through the mediation of SRF and CREB1 associated with spatial memory formation, Journal of Neurochemistry, vol.105, issue.3, pp.820-832, 2008.

,

A. K. Uryga and M. R. Bennett, Ageing induced vascular smooth muscle cell senescence in atherosclerosis, The Journal of Physiology, vol.594, issue.8, pp.2115-2124, 2016.

J. M. Van-deursen, The role of senescent cells in ageing, Nature, vol.509, issue.7501, pp.439-446, 2014.

B. C. Varnum, R. W. Lim, D. A. Kujubu, S. J. Luner, S. E. Kaufman et al., Granulocyte-macrophage colony-stimulating factor and tetradecanoyl phorbol acetate induce a distinct, restricted subset of primary-response TIS genes in both proliferating and terminally differentiated myeloid cells, Molecular and Cellular Biology, vol.9, issue.8, pp.3580-3583, 1989.

M. Vilasco, L. Communal, N. Mourra, A. Courtin, P. Forgez et al., Glucocorticoid receptor and breast cancer, Breast Cancer Research and Treatment, vol.130, issue.1, pp.1-10, 2011.

L. Voisin, M. K. Saba-el-leil, C. Julien, C. Fremin, and S. Meloche, Genetic Demonstration of a Redundant Role of Extracellular Signal-Regulated Kinase 1 (ERK1) and ERK2 Mitogen-Activated Protein Kinases in Promoting Fibroblast Proliferation, Molecular and Cellular Biology, vol.30, issue.12, pp.2918-2932, 2010.

H. Von-der-kammer, M. Mayhaus, C. Albrecht, J. Enderich, M. Wegner et al., Muscarinic acetylcholine receptors activate expression of the EGR gene family of transcription factors, The Journal of Biological Chemistry, vol.273, issue.23, pp.14538-14544, 1998.

,

E. Von-scheven, K. J. Corbin, S. Stefano, and R. Cimaz, Glucocorticoid-associated osteoporosis in chronic inflammatory diseases: Epidemiology, mechanisms, diagnosis, and treatment, Current Osteoporosis Reports, 2014.

I. A. Voutsadakis and A. Cairoli, A critical review of the molecular pathophysiology of lenalidomide sensitivity in 5q ? myelodysplastic syndromes, Leukemia & Lymphoma, vol.53, issue.5, pp.779-788, 2012.

L. C. Vredeveld, P. A. Possik, M. A. Smit, K. Meissl, C. Michaloglou et al., Abrogation of BRAFV600E-induced senescence by PI3K pathway activation contributes to melanomagenesis, Genes & Development, vol.26, issue.10, pp.1055-1069, 2012.

,

T. Wada, N. Joza, H. M. Cheng, T. Sasaki, I. Kozieradzki et al., MKK7 couples stress signalling to G2/M cell-cycle progression and cellular senescence, Nature Cell Biology, vol.6, issue.3, pp.12-226, 2004.

T. Wada and J. M. Penninger, Stress kinase MKK7: Savior of cell cycle arrest and cellular senescence, Cell Cycle, vol.3, issue.5, pp.577-579, 2004.

T. Wada, E. Stepniak, L. Hui, A. Leibbrandt, T. Katada et al., Antagonistic control of cell fates by JNK and p38-MAPK signaling, Cell Death and Differentiation, vol.15, issue.1, pp.89-93, 2008.

N. Wajapeyee, R. W. Serra, X. Zhu, M. Mahalingam, and M. R. Green, Oncogenic BRAF Induces Senescence and Apoptosis through Pathways Mediated by the Secreted Protein IGFBP7, Cell, vol.132, issue.3, pp.363-374, 2008.

E. Wang, Senescent Human Fibroblasts Resist Programmed Cell Death, and Failure to Suppress bell Is Involved, Cancer Research, vol.55, issue.11, pp.2284-2292, 1995.

J. Wang, M. K. Derynck, D. F. Nonaka, D. B. Khodabakhsh, C. Haqq et al., From The Cover: Chromatin immunoprecipitation (ChIP) scanning identifies primary glucocorticoid receptor target genes, Proceedings of the National Academy of Sciences, vol.101, issue.44, pp.15603-15608, 2004.

W. Wang, J. X. Chen, R. Liao, Q. Deng, J. J. Zhou et al., Sequential activation of the MEK-extracellular signal-regulated kinase and MKK3/6-p38 mitogen-activated protein kinase pathways mediates oncogenic ras-induced premature senescence, Molecular and Cellular Biology, vol.22, issue.10, pp.3389-3403, 2002.

I. C. Weaver, I. C. Hellstrom, S. E. Brown, S. D. Andrews, S. Dymov et al., The methylated-DNA binding protein MBD2 enhances NGFI-A (egr-1)-mediated 158 transcriptional activation of the glucocorticoid receptor, Philosophical Transactions of the Royal Society B: Biological Sciences, vol.369, pp.20130513-20130513, 1652.

,

L. L. Wei, X. J. Wu, C. C. Gong, and D. S. Pei, Egr-1 suppresses breast cancer cells proliferation by arresting cell cycle progression via down-regulating CyclinDs, International Journal of Clinical and Experimental Pathology, vol.10, issue.10, pp.10212-10222, 2017.

N. Wei, Y. Yu, V. Joshi, and T. Schmidt, Glucocorticoid receptor antagonist and siRNA prevent senescence of human bone marrow mesenchymal stromal cells in vitro, Cell and Tissue ?, vol.354, issue.2, pp.461-470, 2013.

S. U. Wiemann, A. Satyanarayana, M. Tsahuridu, H. L. Tillmann, L. Zender et al., Hepatocyte telomere shortening and senescence are general markers of human liver cirrhosis, The FASEB Journal : Official Publication of the Federation of American Societies for Experimental Biology, vol.16, issue.9, pp.935-942, 2002.

A. C. Wikström, Glucocorticoid action and novel mechanisms of steroid resistance: Role of glucocorticoid receptor-interacting proteins for glucocorticoid responsiveness, Journal of Endocrinology, vol.178, issue.3, pp.331-337, 2003.

G. S. Wu, The functional interactions between the p53 and MAPK signaling pathways, Cancer Biology & Therapy, vol.3, issue.2, pp.156-161, 2004.

J. Wu and E. H. Bresnick, Bare rudiments of notch signaling: how receptor levels are regulated, Trends in Biochemical Sciences, 2007.
DOI : 10.1016/j.tibs.2007.09.002

M. Wu, D. S. Melichian, M. De-la-garza, K. Gruner, S. Bhattacharyya et al., Essential roles for early growth response transcription factor Egr-1 in tissue fibrosis and wound healing, The American Journal of Pathology, vol.175, issue.3, pp.1041-1055, 2009.

,

M. Xing, BRAF mutation in thyroid cancer, Endocrine-Related Cancer, vol.12, issue.2, pp.245-262, 2005.

M. Xu, A. K. Palmer, H. Ding, M. M. Weivoda, T. Pirtskhalava et al., Targeting senescent cells enhances adipogenesis and metabolic function in old age, 2015.
DOI : 10.7554/elife.12997

URL : https://doi.org/10.7554/elife.12997

W. Xue, L. Zender, C. Miething, R. A. Dickins, E. Hernando et al., Senescence and tumour clearance is triggered by p53 restoration in murine liver carcinomas, Nature, vol.445, issue.7128, pp.656-660, 2007.

S. F. Yan, T. Fujita, J. Lu, K. Okada, Y. Shan-zou et al., Egr-1, a master switch coordinating upregulation of divergent gene families underlying ischemic stress, Nature Medicine, vol.6, issue.12, pp.1355-1361, 2000.

H. Yanai, A. Shteinberg, Z. Porat, A. Budovsky, A. Braiman et al., Cellular senescence-like features of lung fibroblasts derived from idiopathic pulmonary fibrosis patients, Aging, vol.7, issue.9, pp.664-672, 2015.
DOI : 10.18632/aging.100807

URL : http://europepmc.org/articles/pmc4600624?pdf=render

Y. E. Yegorov, S. S. Akimov, R. Hass, A. V. Zelenin, and I. A. Prudovsky, Endogenous ?galactosidase activity in continuously nonproliferating cells, Experimental Cell Research, vol.243, issue.1, pp.207-211, 1998.
DOI : 10.1006/excr.1998.4169

I. Yeh, A. Von-deimling, and B. C. Bastian, Clonal BRAF mutations in melanocytic nevi and initiating role of BRAF in melanocytic neoplasia, Journal of the National Cancer Institute, vol.105, issue.12, pp.917-919, 2013.

J. Yu, S. S. Zhang, K. Saito, S. Williams, Y. Arimura et al., PTEN regulation by Akt-EGR1-ARF-PTEN axis, The EMBO Journal, vol.28, issue.1, pp.21-33, 2009.
DOI : 10.1038/emboj.2008.238

URL : http://emboj.embopress.org/content/28/1/21.full.pdf

,

E. Zeggini, M. N. Weedon, C. M. Lindgren, T. M. Frayling, K. S. Elliott et al., Replication of genome-wide association signals in UK samples reveals risk loci for type 2 diabetes, Science, issue.5829, pp.1336-1341, 2007.

M. Zen, M. Canova, C. Campana, S. Bettio, L. Nalotto et al., The kaleidoscope of glucorticoid effects on immune system, Autoimmunity Reviews, vol.10, issue.6, pp.305-310, 2011.

C. Zhang, T. Wenger, J. Mattern, S. Ilea, C. Frey et al., Clinical and mechanistic aspects of glucocorticoid-induced chemotherapy resistance in the majority of solid tumors, Cancer Biology & Therapy, vol.6, issue.2, pp.278-287, 2007.

H. H. Zhang, X. Chen, J. Wang, W. Guang, W. Han et al., EGR1 decreases the malignancy of human non-small cell lung carcinoma by regulating KRT18 expression, Scientific Reports, vol.4, p.5416, 2014.

H. Zhang, H. Zheng, W. Mu, Z. He, B. Yang et al., DUSP16 ablation arrests the cell cycle and induces cellular senescence, FEBS Journal, vol.282, issue.23, pp.4580-4594, 2015.

L. Zhang, W. Zhou, V. E. Velculescu, S. E. Kern, R. H. Hruban et al., Gene expression profiles in normal and cancer cells, Science, vol.276, issue.5316, pp.1268-1272, 1997.

J. Zhu, D. Woods, M. Mcmahon, and J. M. Bishop, Senescence of human fibroblasts induced by oncogenic Raf, Genes and Development, vol.12, issue.19, pp.2997-3007, 1998.

,

Y. Zwang, M. Oren, and Y. Yarden, Consistency test of the cell cycle: Roles for p53 and EGR1. Cancer Research, 2012.