W. Arancio, A bioinformatics analysis of Lamin-A regulatory network: A perspective on epigenetic involvement in Hutchinson-Gilford progeria syndrome, Rejuvenation Research, vol.15, issue.2, pp.123-127, 2012.

W. Arancio, C. Giordano, and G. Pizzolanti, A ceRNA analysis on LMNA gene focusing on the Hutchinson-Gilford progeria syndrome, Journal of Clinical Bioinformatics, vol.3, issue.1, 2013.

D. Ara-ujo-vilar, G. Lattanzi, B. Gonz-alez-m-endez, A. T. Costa-freitas, D. Prieto et al., Site-dependent differences in both prelamin A and adipogenic genes in subcutaneous adipose tissue of patients with type 2 familial partial lipodystrophy, Journal of Medical Genetics, vol.46, issue.1, pp.40-48, 2009.

S. Bandiera, E. Hatem, S. Lyonnet, and A. Henrion-caude, microRNAs in diseases: From candidate to modifier genes, Clinical Genetics, vol.77, issue.4, pp.306-313, 2010.

D. P. Bartel, MicroRNA target recognition and regulatory functions, Cell, vol.136, issue.2, pp.215-233, 2009.

F. Barth-el-emy, C. Navarro, R. Fayek, N. Da-silva, P. Roll et al., Truncated prelamin A expression in HGPS-like patients: A transcriptional study, European Journal of Human Genetics: EJHG, vol.23, issue.8, pp.1051-1061, 2015.

R. Ben-yaou, C. Navarro, S. Quijano-roy, A. T. Bertrand, C. Massart et al., Type B mandibuloacral dysplasia with congenital myopathy due to homozygous ZMPSTE24 missense mutation, European Journal of Human Genetics, vol.19, issue.6, pp.647-654, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00611256

B. Er-eziat, V. Cervera, P. Le-dour, C. Verpont, M. Dumont et al., Lipodystrophy Study Group (2011) LMNA mutations induce a non-inflammatory fibrosis and a brown fat-like dystrophy of enlarged cervical adipose tissue, The American Journal of Pathology, vol.179, issue.5, pp.2443-2453

G. Bidault, M. Garcia, M. Vantyghem, P. Ducluzeau, R. Morichon et al., Lipodystrophy-Linked LMNA p.R482W Mutation Induces Clinical Early Atherosclerosis and In Vitro Endothelial DysfunctionSignificance. Arteriosclerosis, Thrombosis, and Vascular Biology, vol.33, pp.2162-2171, 2013.

M. T. Bohnsack, K. Czaplinski, and D. Gorlich, Exportin 5 is a RanGTP-dependent dsRNA-binding protein that mediates nuclear export of pre-miRNAs, RNA, vol.10, issue.2, pp.185-191, 2004.

G. Bonne, M. R. Di-barletta, S. Varnous, H. M. B-ecane, E. H. Hammouda et al., Mutations in the gene encoding lamin A/C cause autosomal dominant Emery-Dreifuss muscular dystrophy, Nature Genetics, vol.21, issue.3, pp.285-288, 1999.

C. P. Bracken, H. S. Scott, and G. J. Goodall, A network-biology perspective of microRNA function and dysfunction in cancer, Nature Reviews Genetics, vol.17, issue.12, pp.719-732, 2016.

M. Caron, M. Auclair, B. Donadille, V. Guerci, B. Laville et al., Human lipodystrophies linked to mutations in A-type lamins and to HIV protease inhibitor therapy are both associated with prelamin A accumulation, oxidative stress and premature cellular senescence, Cell Death and Differentiation, vol.14, issue.10, pp.1759-1767, 2007.

P. Cau, C. Navarro, K. Harhouri, P. Roll, S. Sigaudy et al., Nuclear matrix, nuclear envelope and premature aging syndromes in a translational research perspective, Seminars in Cell & Developmental Biology, vol.29, pp.125-147, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01646524

F. Caux, E. Dubosclard, O. Lascols, B. Buendia, O. Chazouill-eres et al., A new clinical condition linked to a novel mutation in lamins A and C with generalized lipoatrophy, insulin-resistant diabetes, disseminated leukomelanodermic papules, liver steatosis, and cardiomyopathy, The Journal of Clinical Endocrinology and Metabolism, vol.88, issue.3, pp.1006-1013, 2003.

L. Chen, L. Lee, B. A. Kudlow, H. G. Santos, O. Sletvold et al., LMNA mutations in atypical Werner's syndrome, Lancet, issue.9382, pp.440-445, 2003.

J. Chen, E. P. Murchison, R. Tang, T. E. Callis, M. Tatsuguchi et al., Targeted deletion of Dicer in the heart leads to dilated cardiomyopathy and heart failure, Proceedings of the National Academy of Sciences of the United States of America, vol.105, pp.2111-2116, 2008.

C. Chou, N. Chang, S. Shrestha, S. Hsu, Y. Lin et al., Updates to the experimentally validated miRNA-target interactions database, Nucleic Acids Research, vol.44, issue.D1, pp.239-247, 2016.

C. Coffinier, S. E. Hudon, R. Lee, E. A. Farber, C. Nobumori et al., A potent HIV protease inhibitor, darunavir, does not inhibit ZMPSTE24 or lead to an accumulation of farnesyl-prelamin A in cells, The Journal of Biological Chemistry, vol.283, issue.15, pp.9797-9804, 2008.

A. Dallaire, C. Garand, E. R. Paquel, S. J. Mitchell, R. De-cabo et al., Down regulation of miR-124 in both Werner syndrome DNA helicase mutant mice and mutant Caenorhabditis elegans wrn-1 reveals the importance of this microRNA in accelerated aging, Aging, vol.4, issue.9, pp.636-647, 2012.

D. Sandre-giovannoli, A. Bernard, R. Cau, P. Navarro, C. Amiel et al., Lamin a truncation in Hutchinson-Gilford progeria, Science, issue.5628, p.2055, 2003.

D. Sandre-giovannoli, A. Chaouch, M. Kozlov, S. Vallat, J. Tazir et al., Homozygous defects in LMNA, encoding lamin A/C nuclear-envelope proteins, cause autosomal recessive axonal neuropathy in human (Charcot-Marie-Tooth disorder type 2) and mouse, American Journal of Human Genetics, vol.70, issue.3, pp.726-736, 2002.
URL : https://hal.archives-ouvertes.fr/hal-01724642

E. Deboy, M. Puttaraju, P. Jailwala, M. Kasoji, M. Cam et al., Identification of novel RNA isoforms of LMNA, Nucleus, vol.8, issue.5, pp.1-10, 2017.

A. M. Denli, B. B. Tops, R. H. Plasterk, R. F. Ketting, and G. J. Hannon, Processing of primary microRNAs by the Microprocessor complex, Nature, vol.432, issue.7014, pp.231-235, 2004.

Y. Doubaj, A. De-sandre-giovannoli, E. Vera, C. L. Navarro, S. C. Elalaoui et al., An inherited LMNA gene mutation in atypical Progeria syndrome, American Journal of Medical Genetics Part A, vol.158, issue.11, pp.2881-2887, 2012.

O. Dreesen, A. Chojnowski, P. F. Ong, T. Y. Zhao, J. E. Common et al., Lamin B1 fluctuations have differential effects on cellular proliferation and senescence, The Journal of Cell Biology, vol.200, issue.5, pp.605-617, 2013.

I. Eisenberg, A. Eran, I. Nishino, M. Moggio, C. Lamperti et al., Distinctive patterns of microRNA expression in primary muscular disorders, Proceedings of the National Academy of Sciences of the United States of America, vol.104, pp.17016-17021, 2007.

R. Elkhatib, G. Longepied, M. Paci, V. Achard, J. Grillo et al., Nuclear envelope remodelling during human spermiogenesis involves somatic B-type lamins and a spermatid-specific B3 lamin isoform. MHR: Basic Science of Reproductive, Medicine, vol.21, issue.3, pp.225-236, 2015.

M. Eriksson, W. T. Brown, L. B. Gordon, M. W. Glynn, J. Singer et al., Recurrent de novo point mutations in lamin A cause Hutchinson-Gilford progeria syndrome, Nature, vol.423, issue.6937, pp.293-298, 2003.

D. Fatkin, C. Macrae, T. Sasaki, M. R. Wolff, M. Porcu et al., Missense mutations in the rod domain of the lamin A/C gene as causes of dilated cardiomyopathy and conduction-system disease, The New England Journal of Medicine, vol.341, issue.23, pp.1715-1724, 1999.

K. Furukawa, H. Inagaki, and Y. Hotta, Identification and cloning of an mRNA coding for a germ cell-specific A-type lamin in mice. Experimental Cell Research, vol.212, pp.426-430, 1994.

V. A. Gennarino, G. Angelo, G. Dharmalingam, S. Fernandez, G. Russolillo et al.,

S. Banfi, Identification of microRNA-regulated gene networks by expression analysis of target genes, Genome Research, vol.22, issue.6, pp.1163-1172, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01126203

R. Gernapudi, B. Wolfson, Y. Zhang, Y. Yao, P. Yang et al., MicroRNA 140 Promotes Expression of Long Noncoding RNA NEAT1 in Adipogenesis, Molecular and Cellular Biology, vol.36, issue.1, pp.30-38, 2016.

M. M. Ghodgaonkar, R. G. Shah, F. Kandan-kulangara, E. Affar, H. H. Qi et al., Abrogation of DNA vectorbased RNAi during apoptosis in mammalian cells due to caspasemediated cleavage and inactivation of Dicer-1, Cell Death and Differentiation, vol.16, issue.6, pp.858-868, 2009.

S. Griffiths-jones, R. J. Grocock, S. Van-dongen, A. Bateman, and A. J. Enright, miRBase: microRNA sequences, targets and gene nomenclature, Nucleic Acids Research, vol.34, pp.140-144, 2006.

Y. Gruenbaum and R. Foisner, Lamins: Nuclear intermediate filament proteins with fundamental functions in nuclear mechanics and genome regulation, Annual Review of Biochemistry, vol.84, pp.131-164, 2015.

H. Guo, N. T. Ingolia, J. S. Weissman, and D. P. Bartel, Mammalian microRNAs predominantly act to decrease target mRNA levels, Nature, vol.466, issue.7308, pp.835-840, 2010.

S. M. Hammond, An overview of microRNAs, Advanced Drug Delivery Reviews, vol.87, pp.3-14, 2015.

J. Han, Y. Lee, K. Yeom, Y. Kim, H. Jin et al., The Drosha-DGCR8 complex in primary microRNA processing, Genes & Development, vol.18, issue.24, pp.3016-3027, 2004.

K. Harhouri, C. Navarro, C. Baquerre, N. Da-silva, C. Bartoli et al., Antisense-based progerin downregulation in HGPS-like patients, cells. Cells, vol.5, issue.3, p.31, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01769404

K. Harhouri, D. Frankel, C. Bartoli, P. Roll, A. De-sandre-giovannoli et al., An overview of treatment strategies for HutchinsonGilford Progeria Syndrome, Nucleus, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01774301

J. Hausser and M. Zavolan, Identification and consequences of miRNA-target interactions-beyond repression of gene expression, Nature Reviews Genetics, vol.15, issue.9, pp.599-612, 2014.

J. Hayes, P. P. Peruzzi, and S. Lawler, MicroRNAs in cancer: Biomarkers, functions and therapy, Trends in Molecular Medicine, vol.20, issue.8, pp.460-469, 2014.

R. A. Hegele, H. Cao, D. M. Liu, G. A. Costain, V. Charlton-menys et al., Sequencing of the reannotated LMNB2 gene reveals novel mutations in patients with acquired partial lipodystrophy, American Journal of Human Genetics, vol.79, issue.2, pp.383-389, 2006.

A. Helwak, G. Kudla, T. Dudnakova, and D. Tollervey, Mapping the human miRNA interactome by CLASH reveals frequent noncanonical binding, Cell, vol.153, issue.3, pp.654-665, 2013.

D. G. Hendrickson, D. J. Hogan, H. L. Mccullough, J. W. Myers, D. Herschlag et al., Concordant regulation of translation and mRNA abundance for hundreds of targets of a human microRNA, PLoS Biology, vol.7, issue.11, 2009.

S. C. Hoelscher, S. A. Doppler, M. Dreßen, H. Lahm, R. Lange et al., MicroRNAs: Pleiotropic players in congenital heart disease and regeneration, Journal of Thoracic Disease, vol.9, issue.1, pp.64-81, 2017.

H. Houlden, M. Laura, . Wavrant-de, F. Vri-eze, J. Blake et al., Mutations in the HSP27 (HSPB1) gene cause dominant, recessive, and sporadic distal HMN/CMT type 2, Neurology, vol.71, issue.21, pp.1660-1668, 2008.

P. W. Hsu, H. Huang, S. Hsu, L. Lin, A. Tsou et al., A cellular function for the RNA-interference enzyme Dicer in the maturation of the let-7 small temporal RNA, Nucleic Acids Research, vol.34, issue.5531, pp.834-838, 2001.

H. L. Janssen, H. W. Reesink, E. J. Lawitz, S. Zeuzem, M. Rodrigueztorres et al., Treatment of HCV infection by targeting microRNA, The New England Journal of Medicine, vol.368, issue.18, pp.1685-1694, 2013.

C. D. Johnson, A. Esquela-kerscher, G. Stefani, M. Byrom, K. Kelnar et al., The let-7 microRNA represses cell proliferation pathways in human cells, Cancer Research, vol.67, issue.16, pp.7713-7722, 2007.

S. Jonas and E. Izaurralde, Towards a molecular understanding of microRNA-mediated gene silencing, Nature Reviews Genetics, vol.16, issue.7, pp.421-433, 2015.

H. Jung, C. Coffinier, Y. Choe, A. P. Beigneux, B. S. Davies et al., Regulation of prelamin A but not lamin C by miR-9, a brain-specific microRNA, Proceedings of the National Academy of Sciences of the United States of America, vol.109, pp.423-431, 2012.

H. Jung, Y. Tu, S. H. Yang, A. Tatar, C. Nobumori et al., New Lmna knock-in mice provide a molecular mechanism for the "segmental aging" in Hutchinson-Gilford progeria syndrome, Human Molecular Genetics, vol.23, issue.6, pp.1506-1515, 2014.

H. Kobayashi and Y. Tomari, RISC assembly: Coordination between small RNAs and Argonaute proteins, Biochimica et Biophysica ActaGene Regulatory Mechanisms, vol.1859, issue.1, pp.71-81, 2016.

A. J. Koch and J. M. Holaska, Loss of emerin alters myogenic signaling and miRNA expression in mouse myogenic progenitors, PLoS One, vol.7, issue.5, 2012.

A. Kozomara and S. Griffiths-jones, miRBase: Integrating microRNA annotation and deep-sequencing data, Nucleic Acids Research, vol.39, pp.152-157, 2011.

A. Krek, D. Gr?-un, E. J. Epstein, I. Da-piedade, K. C. Gunsalus et al., Combinatorial microRNA target predictions, Nature Genetics, vol.37, issue.5, p.495, 2005.

M. Lagos-quintana, R. Rauhut, A. Yalcin, J. Meyer, W. Lendeckel et al., Identification of tissue-specific microRNAs from mouse, Current Biology, vol.12, issue.9, pp.735-739, 2002.

F. Al, , vol.15, p.17

Y. Lee, C. Ahn, J. Han, H. Choi, J. Kim et al.,

V. N. Kim, The nuclear RNase III Drosha initiates microRNA processing, Nature, vol.425, issue.6956, pp.415-419, 2003.

R. C. Lee, R. L. Feinbaum, and V. Ambros, The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14, Cell, vol.75, issue.5, pp.843-854, 1993.
URL : https://hal.archives-ouvertes.fr/in2p3-00597159

S. Lin and Y. Fu, miR-23 regulation of lamin B1 is crucial for oligodendrocyte development and myelination, Disease Models & Mechanisms, vol.2, issue.3-4, pp.178-188, 2009.

S. Lin, M. Y. Heng, L. J. Cek, and Y. Fu, Regulation of myelination in the central nervous system by nuclear lamin B1 and non-coding RNAs, Translational Neurodegeneration, vol.3, issue.1, 2014.

S. Lin, Y. Huang, L. Zhang, M. Y. Heng, L. J. Pt-acek et al., MicroRNA-23a promotes myelination in the central nervous system, Proceedings of the National Academy of Sciences of the United States of America, vol.110, pp.17468-17473, 2013.

F. Lin and H. J. Worman, Structural organization of the human gene encoding nuclear lamin A and nuclear lamin C, The Journal of Biological Chemistry, vol.268, issue.22, pp.16321-16326, 1993.

F. Lin and H. J. Worman, Structural organization of the human gene (LMNB1) encoding nuclear lamin B1, Genomics, vol.27, issue.2, pp.230-236, 1995.

B. M. Machiels, A. H. Zorenc, J. M. Endert, H. J. Kuijpers, G. J. Van-eys et al., An alternative splicing product of the lamin A/C gene lacks exon 10, The Journal of Biological Chemistry, vol.271, issue.16, pp.9249-9253, 1996.

A. Malhas, N. J. Saunders, and D. J. Vaux, The nuclear envelope can control gene expression and cell cycle progression via miRNA regulation, Cell Cycle, vol.9, issue.3, pp.531-539, 2010.

G. Mari~-no, A. P. Ugalde, A. F. Osorio, F. G. Fueyo, A. Freije et al., Insulin-like growth factor 1 treatment extends longevity in a mouse model of human premature aging by restoring somatotroph axis function, Proceedings of the National Academy of Sciences of the United States of America, vol.107, pp.16268-16273, 2010.

M. Miranda, M. R. Chac-on, F. Vidal, A. Megia, C. Richart et al.,

J. Vendrell, LMNA messenger RNA expression in highly active antiretroviral therapy-treated HIV-positive patients, Journal of Acquired Immune Deficiency Syndromes, vol.46, issue.4, pp.384-389, 2007.

G. F. Mok, E. Lozano-velasco, and A. Unsterberg, microRNAs in skeletal muscle development, Seminars in Cell & Developmental Biology, vol.72, pp.67-76, 2017.

R. D. Mortensen, M. Serra, J. A. Steitz, and S. Vasudevan, Posttranscriptional activation of gene expression in Xenopus laevis oocytes by microRNA-protein complexes (microRNPs), Proceedings of the National Academy of Sciences of the United States of America, vol.108, pp.8281-8286, 2011.

C. L. Moulson, L. G. Fong, J. M. Gardner, E. A. Farber, G. Go et al., Increased progerin expression associated with unusual LMNA mutations causes severe progeroid syndromes, Human Mutation, vol.28, issue.9, pp.882-889, 2007.

A. Muchir, G. Bonne, A. J. Van-der-kooi, M. Van-meegen, F. Baas et al.,

K. Schwartz, Identification of mutations in the gene encoding lamins A/C in autosomal dominant limb girdle muscular dystrophy with atrioventricular conduction disturbances (LGMD1B), Human Molecular Genetics, vol.9, issue.9, pp.1453-1459, 2000.

N. Naetar, S. Ferraioli, and R. Foisner, Lamins in the nuclear interior-life outside the lamina, Journal of Cell Science, vol.130, issue.13, pp.2087-2096, 2017.

C. L. Navarro, J. Cadi~-nanos, A. De-sandre-giovannoli, R. Bernard, S. Courrier et al., Loss of ZMPSTE24 (FACE1) causes autosomal recessive restrictive dermopathy and accumulation of Lamin A precursors, Human Molecular Genetics, vol.14, issue.11, pp.1503-1513, 2005.
URL : https://hal.archives-ouvertes.fr/hal-01669073

C. L. Navarro, D. Sandre-giovannoli, A. Bernard, R. Boccaccio, I. Boyer et al., Lamin A and ZMPSTE24 (FACE-1) defects cause nuclear disorganization and identify restrictive dermopathy as a lethal neonatal laminopathy, Human Molecular Genetics, vol.13, issue.20, pp.2493-2503, 2004.
URL : https://hal.archives-ouvertes.fr/hal-01668977

F. Navarro and J. Lieberman, Small RNAs guide hematopoietic cell differentiation and function, The Journal of Immunology, vol.184, issue.11, pp.5939-5947, 2010.

X. Nissan, S. Blondel, C. Navarro, Y. Maury, C. Denis et al.,

M. Peschanski, Unique preservation of neural cells in Hutchinson-Gilford progeria syndrome is due to the expression of the neural-specific miR-9 MicroRNA, Cell Reports, vol.2, issue.1, pp.1-9, 2012.

G. Novelli, A. Muchir, F. Sangiuolo, A. Helbling-leclerc, M. R. D'apice et al., Mandibuloacral dysplasia is caused by a mutation in LMNA-encoding lamin A/C, American Journal of Human Genetics, vol.71, issue.2, pp.426-431, 2002.

A. Oldenburg, N. Briand, A. L. Sørensen, I. Cahyani, A. Shah et al., A lipodystrophy-causing lamin A mutant alters conformation and epigenetic regulation of the anti-adipogenic MIR335 locus, The Journal of Cell Biology, vol.216, issue.9, pp.2731-2743, 2017.

Q. S. Padiath, K. Saigoh, R. Schiffmann, H. Asahara, T. Yamada et al., Lamin B1 duplications cause autosomal dominant leukodystrophy, Nature Genetics, vol.38, issue.10, pp.1114-1123, 2006.

A. E. Pasquinelli, MicroRNAs and their targets: Recognition, regulation and an emerging reciprocal relationship, Nature Reviews Genetics, vol.13, issue.4, pp.271-282, 2012.

A. M. Pend-as, Z. Zhou, J. Cadi~-nanos, J. M. Freije, J. Wang et al., Defective prelamin A processing and muscular and adipocyte alterations in Zmpste24 metalloproteinasedeficient mice, Nature Genetics, vol.31, issue.1, pp.94-99, 2002.

R. F. Place, L. Li, D. Pookot, E. J. Noonan, and R. Dahiya, MicroRNA-373 induces expression of genes with complementary promoter sequences, Proceedings of the National Academy of Sciences of the United States of America, vol.105, pp.1608-1613, 2008.

E. R. Porrello, 2013) microRNAs in cardiac development and regeneration, Clinical Science, vol.125, issue.4, pp.151-166, 1979.

S. Quijano-roy, B. Mbieleu, C. G. B?-onnemann, P. Jeannet, J. Colomer et al., De novo LMNA mutations cause a new form of congenital muscular dystrophy, Annals of Neurology, vol.64, issue.2, pp.177-186, 2008.

R. Di-barletta, M. Ricci, E. Galluzzi, G. Tonali, P. Mora et al., Different mutations in the LMNA gene cause autosomal dominant and autosomal recessive Emery-Dreifuss muscular dystrophy, American Journal of Human Genetics, vol.66, issue.4, pp.1407-1412, 2000.

B. J. Reinhart, F. J. Slack, M. Basson, A. E. Pasquinelli, J. C. Bettinger et al., The 21-nucleotide let-7 RNA regulates developmental timing in Caenorhabditis elegans, Nature, issue.6772, pp.901-906, 2000.

D. Renard, G. Fourcade, D. Milhaud, D. Bessis, V. Esteves-vieira et al., Novel LMNA mutation in atypical Werner syndrome presenting with ischemic disease, Stroke, vol.40, issue.2, pp.11-14, 2009.

L. Renou, S. Stora, R. B. Yaou, M. Volk, M. Sinkovec et al.,

G. Bonne, Heart-hand syndrome of Slovenian type: A new kind of laminopathy, Journal of Medical Genetics, vol.45, issue.10, pp.666-671, 2008.

A. Rodriguez, S. Griffiths-jones, J. L. Ashurst, and A. Bradley, Identification of mammalian microRNA host genes and transcription units, Genome Research, vol.14, issue.10A, pp.1902-1910, 2004.

A. Sato, T. Omi, A. Yamamoto, A. Satake, A. Hiramoto et al., MicroRNA-351 regulates two-types of cell death, necrosis and apoptosis, induced by 5-fluoro-2 0-deoxyuridine, PLoS One, vol.11, issue.4, 2016.

K. H. Schreiber and B. K. Kennedy, When lamins go bad: Nuclear structure and disease, Cell, vol.152, issue.6, pp.1365-1375, 2013.

S. Shackleton, D. J. Lloyd, S. N. Jackson, R. Evans, M. F. Niermeijer et al., LMNA, encoding lamin A/C, is mutated in partial lipodystrophy, Nature Genetics, vol.24, issue.2, pp.153-156, 2000.

X. Sun, Z. Zhou, D. J. Fink, and M. Mata, HspB1 silences translation of PDZ-RhoGEF by enhancing miR-20a and miR-128 expression to promote neurite extension, Molecular and Cellular Neurosciences, vol.57, pp.111-119, 2013.

N. Sylvius, G. Bonne, K. Straatman, T. Reddy, T. W. Gant et al., MicroRNA expression profiling in patients with lamin A/C-associated muscular dystrophy, FASEB Journal, vol.25, issue.11, pp.3966-3978, 2011.

W. Tang, A. I. Robles, R. P. Beyer, L. T. Gray, G. H. Nguyen et al., The Werner syndrome RECQ helicase targets G4 DNA in human cells to modulate transcription, Human Molecular Genetics, vol.25, issue.10, pp.2060-2069, 2016.

J. S. Tsang, M. S. Ebert, and A. Van-oudenaarden, , 2010.

, Genome-wide dissection of microRNA functions and cotargeting networks using gene set signatures, Molecular Cell, vol.38, issue.1, pp.140-153

Y. Turgay, M. Eibauer, A. E. Goldman, T. Shimi, M. Khayat et al., The molecular architecture of lamins in somatic cells, Nature, vol.543, issue.7644, pp.261-264, 2017.

A. P. Ugalde, Y. Espa~-nol, and C. L-opez-ot-in, Micromanaging aging with miRNAs: New messages from the nuclear envelope, Nucleus, vol.2, issue.6, pp.549-555, 2011.

A. P. Ugalde, A. J. Ramsay, J. De-la-rosa, I. Varela, G. Mari~-no et al., Aging and chronic DNA damage response activate a regulatory pathway involving miR-29 and p53, The EMBO Journal, vol.30, issue.11, pp.2219-2232, 2011.

N. J. Ullrich and L. B. Gordon, Hutchinson-Gilford progeria syndrome, Handbook of Clinical Neurology, vol.132, pp.249-264, 2015.

R. Ungricht, U. ;. Kutay, A. J. Van-der-meer, A. C. Van-nuenen, J. De-bruijne et al., Miravirsen dosing in chronic hepatitis C patients results in decreased microRNA122 levels without affecting other microRNAs in plasma, Alimentary Pharmacology & Therapeutics, vol.18, issue.4, pp.851-864, 2014.

E. Van-rooij, N. Liu, and E. N. Olson, Lamina-associated domains: Links with chromosome architecture, heterochromatin, and gene repression, Trends in Genetics, vol.24, issue.4, pp.780-791, 2008.

S. Vasudevan, Y. Tong, and J. A. Steitz, Switching from repression to activation: microRNAs can up-regulate translation, Science, vol.318, issue.5858, pp.1931-1934, 2007.

V. L. Verstraeten, J. L. Broers, M. A. Van-steensel, S. Zinn-justin, F. C. Ramaekers et al., Compound heterozygosity for mutations in LMNA causes a progeria syndrome without prelamin A accumulation, Human Molecular Genetics, vol.15, issue.16, pp.2509-2522, 2006.

N. Vignier, F. Amor, P. Fogel, A. Duvallet, J. Poupiot et al.,

D. Israeli, Distinctive serum miRNA profile in mouse models of striated muscular pathologies, PLoS One, vol.8, issue.2, 2013.

N. Wong and X. Wang, miRDB: An online resource for microRNA target prediction and functional annotations, Nucleic Acids Research, vol.43, issue.D1, pp.146-152, 2015.

H. J. Worman, Nuclear lamins and laminopathies, The Journal of Pathology, vol.226, issue.2, pp.316-325, 2012.

H. J. Worman and G. Bonne, Laminopathies": A wide spectrum of human diseases, Experimental Cell Research, vol.313, issue.10, pp.2121-2133, 2007.

X. Xiong, H. J. Jung, S. Gombar, J. Y. Park, C. Zhang et al.,

Y. Suh, MicroRNA transcriptome analysis identifies miR-365 as a novel negative regulator of cell proliferation in Zmpste24-deficient mouse embryonic fibroblasts, Mutation Research, vol.777, pp.69-78, 2015.

S. H. Yang, S. Procaccia, H. Jung, C. Nobumori, A. Tatar et al.,

S. G. Young, Mice that express farnesylated versions of prelamin A in neurons develop achalasia, Human Molecular Genetics, vol.24, issue.10, pp.2826-2840, 2015.

R. Yi, Y. Qin, I. G. Macara, and B. R. Cullen, Exportin-5 mediates the nuclear export of pre-microRNAs and short hairpin RNAs, Genes & Development, vol.17, issue.24, pp.3011-3016, 2003.

S. G. Young, H. Jung, C. Coffinier, and L. G. Fong, Understanding the roles of nuclear A-and B-type lamins in brain development, Journal of Biological Chemistry, vol.287, issue.20, pp.16103-16110, 2012.

K. Yu, S. Lee, J. Jung, I. Hong, H. Kim et al., MicroRNA-141-3p plays a role in human mesenchymal stem cell aging by directly targeting ZMPSTE24, Journal of Cell Science, vol.126, pp.5422-5431, 2013.

C. Zhang, X. Liu, Q. He, H. Zheng, S. Xu et al.,

S. Deng, miR-342-5p promotes Zmpste24-deficient mouse embryonic fibroblasts proliferation by suppressing GAS2, Molecular Medicine Reports, vol.16, pp.8944-8952, 2017.

S. Zhu, W. Pan, Y. Qian, D. Frankel, V. Delecourt et al., MicroRNAs in hereditary and sporadic premature aging syndromes and other laminopathies, Journal of Molecular Medicine, vol.17, issue.9, p.12766, 2013.

F. Al, , vol.17, p.17