G. Bernard, E. Chouery, M. Putorti, M. Tetreault, A. Takanohashi et al., Mutations of POLR3A Encoding a Catalytic Subunit of RNA Polymerase Pol III Cause a Recessive Hypomyelinating Leukodystrophy, The American Journal of Human Genetics, vol.89, issue.3, pp.415-438, 2011.
DOI : 10.1016/j.ajhg.2011.07.014

M. Tetreault, K. Choquet, S. Orcesi, D. Tonduti, U. Balottin et al., Recessive Mutations in POLR3B, Encoding the Second Largest Subunit of Pol III, Cause a Rare Hypomyelinating Leukodystrophy, The American Journal of Human Genetics, vol.89, issue.5, pp.652-657, 2011.
DOI : 10.1016/j.ajhg.2011.10.006

I. Thiffault, N. Wolf, D. Forget, K. Guerrero, L. Tran et al., Recessive mutations in POLR1C cause a leukodystrophy by impairing biogenesis of RNA polymerase III, Nature Communications, vol.40, p.7623, 2015.
DOI : 10.1038/ncomms8623

H. Saitsu, H. Osaka, M. Sasaki, J. Takanashi, K. Hamada et al., Mutations in POLR3A and POLR3B Encoding RNA Polymerase III Subunits Cause an Autosomal-Recessive Hypomyelinating Leukoencephalopathy, The American Journal of Human Genetics, vol.89, issue.5, pp.644-51, 2011.
DOI : 10.1016/j.ajhg.2011.10.003

URL : http://doi.org/10.1016/j.ajhg.2011.10.003

N. Wolf, A. Vanderver, R. Van-spaendonk, R. Schiffmann, B. Brais et al., Clinical spectrum of 4H leukodystrophy caused by POLR3A and POLR3B mutations, Neurology, vol.83, issue.21, pp.1898-905, 2014.
DOI : 10.1212/WNL.0000000000001002

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4248461

L. Piana, R. Tonduti, D. , G. Dressman, H. Schmidt et al., Brain Magnetic Resonance Imaging (MRI) Pattern Recognition in Pol III-Related Leukodystrophies, Journal of Child Neurology, vol.55, issue.204, pp.214-234, 2014.
DOI : 10.2307/2529310

M. Steenweg, A. Vanderver, S. Blaser, A. Bizzi, T. De-koning et al., Magnetic resonance imaging pattern recognition in hypomyelinating disorders, Brain, vol.133, issue.10, pp.2971-82, 2010.
DOI : 10.1093/brain/awq257

URL : http://doi.org/10.1093/brain/awq257

L. Piana, R. Cayami, F. Tran, L. Guerrero, K. Van-spaendonk et al., Diffuse hypomyelination is not obligate for POLR3-related disorders, Neurology, vol.86, issue.17, pp.1622-1628, 2016.
DOI : 10.1212/WNL.0000000000002612

G. Dieci, G. Fiorino, M. Castelnuovo, M. Teichmann, and A. Pagano, The expanding RNA polymerase III transcriptome, Trends in Genetics, vol.23, issue.12, pp.614-636, 2007.
DOI : 10.1016/j.tig.2007.09.001

URL : http://hdl.handle.net/11381/1706964

H. Daoud, M. Tetreault, W. Gibson, K. Guerrero, A. Cohen et al., are a major cause of hypomyelinating leukodystrophies with or without dental abnormalities and/or hypogonadotropic hypogonadism, Journal of Medical Genetics, vol.50, issue.3, pp.194-201, 2013.
DOI : 10.1136/jmedgenet-2012-101357

M. Gutierrez, I. Thiffault, K. Guerrero, G. Martos-moreno, L. Tran et al., Large exonic deletions in POLRB gene cause POLR3-related leukodystrophy, Orphanet Journal of Rare Diseases, vol.9, issue.18, p.69, 2015.
DOI : 10.1186/s13023-015-0279-9

A. Potic, B. Brais, K. Choquet, R. Schiffmann, and G. Bernard, 4H Syndrome With Late-Onset Growth Hormone Deficiency Caused by POLR3A Mutations, Archives of Neurology, vol.69, issue.7, pp.920-923, 2012.
DOI : 10.1001/archneurol.2011.1963

K. Shimojima, S. Shimada, A. Tamasaki, S. Akaboshi, Y. Komoike et al., Novel compound heterozygous mutations of POLR3A revealed by whole-exome sequencing in a patient with hypomyelination, Brain and Development, vol.36, issue.4, pp.315-336, 2014.
DOI : 10.1016/j.braindev.2013.04.011

Y. Terao, H. Saitsu, M. Segawa, Y. Kondo, K. Sakamoto et al., Diffuse central hypomyelination presenting as 4H syndrome caused by compound heterozygous mutations in POLR3A encoding the catalytic subunit of polymerase III, Journal of the Neurological Sciences, vol.320, issue.1-2, pp.102-107, 2012.
DOI : 10.1016/j.jns.2012.07.005

R. Battini, S. Bertelloni, G. Astrea, M. Casarano, L. Travaglini et al., Longitudinal follow up of a boy affected by Pol III-related leukodystrophy: a detailed phenotype description, BMC Medical Genetics, vol.23, issue.9, p.53, 2015.
DOI : 10.1016/j.ghir.2013.07.002

E. Jurkiewicz, D. Dunin-wasowicz, D. Gieruszczak-bialek, K. Malczyk, K. Guerrero et al., Recessive Mutations in POLR3B Encoding RNA Polymerase III Subunit Causing Diffuse Hypomyelination in Patients with 4H Leukodystrophy with Polymicrogyria and Cataracts, Clinical Neuroradiology, vol.300, issue.1???2
DOI : 10.1007/s00062-015-0472-1

E. Billington, G. Bernard, W. Gibson, and B. Corenblum, Endocrine Aspects of 4H Leukodystrophy: A Case Report and Review of the Literature, Case Reports in Endocrinology, vol.77, issue.1, p.314594, 2015.
DOI : 10.1016/j.jns.2012.07.005

M. Synofzik, G. Bernard, T. Lindig, and J. Gburek-augustat, Teaching NeuroImages: Hypomyelinating leukodystrophy with hypodontia due to POLR3B: Look into a leukodystrophy's mouth, Neurology, vol.81, issue.19, p.145, 2013.
DOI : 10.1212/01.wnl.0000435300.64776.7e

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3812106

M. Richards, L. Plummer, Y. Chan, M. Lippincott, R. Quinton et al., mutations: isolated hypogonadotropic hypogonadism without neurological or dental anomalies, Journal of Medical Genetics, vol.19, issue.1, pp.19-25, 2016.
DOI : 10.1136/jmedgenet-2016-104064

D. Azmanov, S. Siira, T. Chamova, A. Kaprelyan, V. Guergueltcheva et al., gene mutation in patients with an unusual phenotype of striatal involvement, Human Molecular Genetics, vol.25, issue.19, pp.4302-4316, 2016.
DOI : 10.1093/hmg/ddw263

A. Arimbasseri and R. Maraia, RNA Polymerase III Advances: Structural and tRNA Functional Views, Trends in Biochemical Sciences, vol.41, issue.6, pp.546-59, 2016.
DOI : 10.1016/j.tibs.2016.03.003

A. Arimbasseri, N. Blewett, J. Iben, T. Lamichhane, V. Cherkasova et al., RNA Polymerase III Output Is Functionally Linked to tRNA Dimethyl-G26 Modification, PLOS Genetics, vol.4, issue.12, p.1005671, 2015.
DOI : 10.1371/journal.pgen.1005671.s006

URL : http://doi.org/10.1371/journal.pgen.1005671

K. Dittmar, J. Goodenbour, and T. Pan, Tissue-Specific Differences in Human Transfer RNA Expression, PLoS Genetics, vol.95, issue.12, p.221, 2006.
DOI : 10.1371/journal.pgen.0020221.st006

URL : http://doi.org/10.1371/journal.pgen.0020221

J. Castle, C. Armour, M. Lower, D. Haynor, M. Biery et al., Digital Genome-Wide ncRNA Expression, Including SnoRNAs, across 11 Human Tissues Using PolyA-Neutral Amplification, PLoS ONE, vol.31, issue.7, p.11779, 2010.
DOI : 10.1371/journal.pone.0011779.s011

URL : http://doi.org/10.1371/journal.pone.0011779

E. Karaca, S. Weitzer, D. Pehlivan, H. Shiraishi, T. Gogakos et al., Human CLP1 Mutations Alter tRNA Biogenesis, Affecting Both Peripheral and Central Nervous System Function, Cell, vol.157, issue.3, pp.636-50, 2014.
DOI : 10.1016/j.cell.2014.02.058

URL : http://doi.org/10.1016/j.cell.2014.02.058

A. Schaffer, V. Eggens, A. Caglayan, M. Reuter, E. Scott et al., CLP1 Founder Mutation Links tRNA Splicing and Maturation to Cerebellar Development and Neurodegeneration, Cell, vol.157, issue.3, pp.651-63, 2014.
DOI : 10.1016/j.cell.2014.03.049

URL : http://doi.org/10.1016/j.cell.2014.03.049

S. Blanco, S. Dietmann, J. Flores, S. Hussain, C. Kutter et al., Aberrant methylation of tRNAs links cellular stress to neuro-developmental disorders, The EMBO Journal, vol.33, issue.18, pp.2020-2059, 2014.
DOI : 10.15252/embj.201489282

R. Taft, A. Vanderver, R. Leventer, S. Damiani, C. Simons et al., Mutations in DARS Cause Hypomyelination with Brain Stem and Spinal Cord Involvement and Leg Spasticity, The American Journal of Human Genetics, vol.92, issue.5, pp.774-80, 2013.
DOI : 10.1016/j.ajhg.2013.04.006

URL : http://doi.org/10.1016/j.ajhg.2013.04.006

M. Feinstein, M. B. Noyman, I. Shalev, H. Flusser, H. Shelef et al., Pelizaeus-Merzbacher-like Disease Caused by AIMP1/p43 Homozygous Mutation, The American Journal of Human Genetics, vol.87, issue.6, pp.820-828, 2010.
DOI : 10.1016/j.ajhg.2010.10.016

URL : http://doi.org/10.1016/j.ajhg.2010.10.016

N. Wolf, G. Salomons, R. Rodenburg, P. Pouwels, J. Schieving et al., cause hypomyelination, Annals of Neurology, vol.14, issue.1, pp.134-143, 2014.
DOI : 10.1002/ana.24167

A. Vanderver, D. Tonduti, G. Bernard, J. Lai, C. Rossi et al., More Than Hypomyelination in Pol-III Disorder, Journal of Neuropathology & Experimental Neurology, vol.72, issue.1, pp.67-75, 2013.
DOI : 10.1097/NEN.0b013e31827c99d2

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3797528

R. Upadhya, J. Lee, and I. Willis, Maf1 Is an Essential Mediator of Diverse Signals that Repress RNA Polymerase III Transcription, Molecular Cell, vol.10, issue.6, pp.1489-94, 2002.
DOI : 10.1016/S1097-2765(02)00787-6

N. Bonhoure, A. Byrnes, R. Moir, W. Hodroj, F. Preitner et al., Loss of the RNA polymerase III repressor MAF1 confers obesity resistance, Genes & Development, vol.29, issue.9, pp.934-981, 2015.
DOI : 10.1101/gad.258350.115

A. Michels, A. Robitaille, D. Buczynski-ruchonnet, W. Hodroj, J. Reina et al., mTORC1 Directly Phosphorylates and Regulates Human MAF1, mTORC1 directly phosphorylates and regulates human MAF1, pp.3749-57, 2010.
DOI : 10.1128/MCB.00319-10

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2916396

R. Ishimura, G. Nagy, I. Dotu, H. Zhou, X. Yang et al., Ribosome stalling induced by mutation of a CNS-specific tRNA causes neurodegeneration, Science, vol.4, issue.1, pp.455-464, 2014.
DOI : 10.1038/nprot.2008.211

N. Yee, W. Gong, Y. Huang, K. Lorent, A. Dolan et al., Mutation of RNA Pol III Subunit rpc2/polr3b Leads to Deficiency of Subunit Rpc11 and Disrupts Zebrafish Digestive Development, PLoS Biology, vol.370, issue.11, p.312, 2007.
DOI : 10.1371/journal.pbio.0050312.t001

R. White, Transcription by RNA polymerase III: more complex than we thought, Nature Reviews Genetics, vol.21, issue.7, pp.459-63, 2011.
DOI : 10.1038/nrg3001

F. Sievers, A. Wilm, D. Dineen, T. Gibson, K. Karplus et al., Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega, Molecular Systems Biology, vol.7, issue.1, p.539, 2011.
DOI : 10.1093/nar/gkn174

I. Ornelas, L. Mclane, A. Saliu, A. Evangelou, L. Khandker et al., Heterogeneity in oligodendroglia: Is it relevant to mouse models and human disease?, Journal of Neuroscience Research, vol.34, issue.47, pp.1421-1454, 2016.
DOI : 10.1007/s10803-016-2803-8

R. Fields, White matter matters, Sci Am, vol.298, pp.42-51, 2008.

O. Tress, M. Maglione, A. Zlomuzica, D. May, N. Dicke et al., Pathologic and Phenotypic Alterations in a Mouse Expressing a Connexin47 Missense Mutation That Causes Pelizaeus-Merzbacher???Like Disease in Humans, PLoS Genetics, vol.64, issue.7, p.1002146, 2011.
DOI : 10.1371/journal.pgen.1002146.s001

I. Jakovcevski, R. Filipovic, Z. Mo, S. Rakic, and N. Zecevic, Oligodendrocyte development and the onset of myelination in the human fetal brain, Frontiers in Neuroanatomy, vol.3, p.5, 2009.
DOI : 10.3389/neuro.05.005.2009

B. Odermatt, K. Wellershaus, A. Wallraff, G. Seifert, J. Degen et al., Connexin 47 (Cx47)- deficient mice with enhanced green fluorescent protein reporter gene reveal predominant oligodendrocytic expression of Cx47 and display vacuolized myelin in the CNS, J Neurosci, vol.23, pp.4549-59, 2003.

A. Pujol, C. Hindelang, N. Callizot, U. Bartsch, M. Schachner et al., Late onset neurological phenotype of the X-ALD gene inactivation in mice: a mouse model for adrenomyeloneuropathy, Human Molecular Genetics, vol.11, issue.5, pp.499-505, 2002.
DOI : 10.1093/hmg/11.5.499

K. Duning, F. Buck, A. Barnekow, and J. Kremerskothen, SYNCRIP, a component of dendritically localized mRNPs, binds to the translation regulator BC200 RNA, Journal of Neurochemistry, vol.13, issue.2, pp.351-360, 2008.
DOI : 10.1074/jbc.M504286200

H. Tiedge, W. Chen, and J. Brosius, Primary structure, neural-specific expression, and dendritic location of human BC200 RNA, J Neurosci, vol.13, pp.2382-90, 1993.

S. Massone, I. Vassallo, M. Castelnuovo, G. Fiorino, E. Gatta et al., RNA polymerase III drives alternative splicing of the potassium channel???interacting protein contributing to brain complexity and neurodegeneration, The Journal of Cell Biology, vol.9, issue.5, pp.851-66, 2011.
DOI : 10.1038/ng1852

S. Massone, I. Vassallo, G. Fiorino, M. Castelnuovo, F. Barbieri et al., 17A, a novel non-coding RNA, regulates GABA B alternative splicing and signaling in response to inflammatory stimuli and in Alzheimer disease, Neurobiology of Disease, vol.41, issue.2, pp.308-325, 2011.
DOI : 10.1016/j.nbd.2010.09.019

M. Castelnuovo, S. Massone, R. Tasso, G. Fiorino, M. Gatti et al., An Alu-like RNA promotes cell differentiation and reduces malignancy of human neuroblastoma cells, The FASEB Journal, vol.24, issue.10, pp.4033-4079, 2010.
DOI : 10.1096/fj.10-157032

A. Gigoni, D. Costa, M. Gaetani, R. Tasso, F. Villa et al., Down-regulation of 21A Alu RNA as a tool to boost proliferation maintaining the tissue regeneration potential of progenitor cells, Cell Cycle, vol.126, issue.18, pp.2420-2450, 2016.
DOI : 10.1186/1475-2867-14-59

I. Penna, I. Vassallo, M. Nizzari, D. Russo, D. Costa et al., A novel snRNA-like transcript affects amyloidogenesis and cell cycle progression through perturbation of Fe65L1 (APBB2) alternative splicing, Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, vol.1833, issue.6, pp.1511-1537, 1833.
DOI : 10.1016/j.bbamcr.2013.02.020

A. Pagano, M. Castelnuovo, F. Tortelli, R. Ferrari, G. Dieci et al., New Small Nuclear RNA Gene-Like Transcriptional Units as Sources of Regulatory Transcripts, PLoS Genetics, vol.37, issue.2, p.1, 2007.
DOI : 10.1371/journal.pgen.0030001.st005

J. Mack, V. Beljanski, A. Soulika, D. Townsend, C. Brown et al., "Skittish" Abca2 Knockout Mice Display Tremor, Hyperactivity, and Abnormal Myelin Ultrastructure in the Central Nervous System, Molecular and Cellular Biology, vol.27, issue.1, pp.44-53, 2007.
DOI : 10.1128/MCB.01824-06

D. Munter, S. Verheijden, S. Vanderstuyft, E. Malheiro, A. Brites et al., Early-onset Purkinje cell dysfunction underlies cerebellar ataxia in peroxisomal multifunctional protein-2 deficiency, Neurobiology of Disease, vol.94, pp.157-68, 2016.
DOI : 10.1016/j.nbd.2016.06.012

W. Coley, L. Bogdanik, M. Vila, Y. Q. Van-der-meulen, J. Rayavarapu et al., mice, Human Molecular Genetics, vol.25, issue.1, pp.130-175, 2016.
DOI : 10.1093/hmg/ddv460

T. Hatzipetros, L. Bogdanik, V. Tassinari, J. Kidd, A. Moreno et al., C57BL/6J congenic Prp-TDP43A315T mice develop progressive neurodegeneration in the myenteric plexus of the colon without exhibiting key features of ALS, Brain Research, vol.1584, pp.59-72, 2014.
DOI : 10.1016/j.brainres.2013.10.013

W. Frankel, C. Mahaffey, T. Mcgarr, B. Beyer, and V. Letts, Unraveling Genetic Modifiers in the Gria4 Mouse Model of Absence Epilepsy, PLoS Genetics, vol.4, issue.7, p.1004454, 2014.
DOI : 10.1371/journal.pgen.1004454.s001

M. Van-der-knaap, J. Pronk, and G. Scheper, Vanishing white matter disease, The Lancet Neurology, vol.5, issue.5, pp.413-436, 2006.
DOI : 10.1016/S1474-4422(06)70440-9

A. Orioli, V. Praz, P. Lhote, and N. Hernandez, Human MAF1 targets and represses active RNA polymerase III genes by preventing recruitment rather than inducing long-term transcriptional arrest, Genome Research, vol.26, issue.5, pp.624-659, 2016.
DOI : 10.1101/gr.201400.115

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4864463

H. Gingold, D. Tehler, N. Christoffersen, M. Nielsen, F. Asmar et al., A Dual Program for Translation Regulation in Cellular Proliferation and Differentiation, Cell, vol.158, issue.6, pp.1281-92, 2014.
DOI : 10.1016/j.cell.2014.08.011

A. Orioli, tRNA biology in the omics era: Stress signalling dynamics and cancer progression, BioEssays, vol.107, issue.3, 2016.
DOI : 10.1002/bies.201600158

G. Dieci, H. Denmat, S. Lukhtanov, E. Thuriaux, P. Werner et al., A universally conserved region of the largest subunit participates in the active site of RNA polymerase III, EMBO J, vol.14, pp.3766-76, 1995.

V. Thuillier, I. Brun, A. Sentenac, and M. Werner, Mutations in the alpha-amanitin conserved domain of the largest subunit of yeast RNA polymerase III affect pausing, RNA cleavage and transcriptional transitions, EMBO J, vol.15, pp.618-647, 1996.

I. Brun, A. Sentenac, and M. Werner, Dual role of the C34 subunit of RNA polymerase III in transcription initiation, The EMBO Journal, vol.16, issue.18, pp.5730-5771, 1997.
DOI : 10.1093/emboj/16.18.5730

F. Schnutgen and N. Ghyselinck, Adopting the good reFLEXes when generating conditional alterations in the mouse genome, Transgenic Research, vol.30, issue.4, pp.405-418, 2007.
DOI : 10.1007/s11248-007-9089-8

URL : https://hal.archives-ouvertes.fr/hal-00189128

T. Luong, H. Carlisle, A. Southwell, and P. Patterson, Assessment of Motor Balance and Coordination in Mice using the Balance Beam, Journal of Visualized Experiments, issue.49, 2011.
DOI : 10.3791/2376

R. Lariviere, R. Gaudet, B. Gentil, M. Girard, T. Conte et al., Sacs knockout mice present pathophysiological defects underlying autosomal recessive spastic ataxia of Charlevoix-Saguenay, Human Molecular Genetics, vol.24, issue.3, pp.727-766, 2015.
DOI : 10.1093/hmg/ddu491

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4291249

R. Carter, J. Morton, and S. Dunnett, Motor Coordination and Balance in Rodents, Curr Protoc Neurosci, vol.35, pp.8-12, 2001.
DOI : 10.1002/0471142301.ns0812s15

M. Girard, R. Lariviere, D. Parfitt, E. Deane, R. Gaudet et al., Mitochondrial dysfunction and Purkinje cell loss in autosomal recessive spastic ataxia of Charlevoix-Saguenay (ARSACS), Proceedings of the National Academy of Sciences, vol.143, issue.4, pp.1661-1667, 2012.
DOI : 10.1093/jb/mvm245

B. Skryabin, V. Sukonina, U. Jordan, L. Lewejohann, N. Sachser et al., Neuronal Untranslated BC1 RNA: Targeted Gene Elimination in Mice, Molecular and Cellular Biology, vol.23, issue.18, pp.6435-6476, 2003.
DOI : 10.1128/MCB.23.18.6435-6441.2003

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC193692

M. Lavallee-adam, J. Rousseau, C. Domecq, A. Bouchard, D. Forget et al., Discovery of Cell Compartment Specific Protein???Protein Interactions using Affinity Purification Combined with Tandem Mass Spectrometry, Journal of Proteome Research, vol.12, issue.1, pp.272-81, 2013.
DOI : 10.1021/pr300778b