H. Ginisty, H. Sicard, B. Roger, and P. Bouvet, Structure and functions of nucleolin, J Cell Sci, vol.112, pp.761-772, 1999.
URL : https://hal.archives-ouvertes.fr/hal-00023721

L. Ghisolfi-nieto, G. Joseph, F. Puvion-dutilleul, F. Amalric, and P. Bouvet, Nucleolin is a sequence-specific RNA-binding protein: characterization of targets on pre-ribosomal RNA, J Mol Biol, vol.260, pp.34-53, 1996.

H. Ginisty, F. Amalric, and P. Bouvet, Two different combinations of RNA-binding domains determine the RNA binding specificity of nucleolin, J Biol Chem, vol.276, pp.14338-14343, 2001.
URL : https://hal.archives-ouvertes.fr/hal-00023731

C. Cong, S. Das, and P. Bouvet, The Multiple Properties and Functions of Nucleolin. Protein Reviews series, Nucleolus, Springer Editorin-Chief is M. Zouhair Atassi, p.185, 2011.

C. M. Berger, X. Gaume, and P. Bouvet, The roles of nucleolin subcellular localization in cancer, Biochimie, vol.113, pp.78-85, 2015.

F. Mongelard and P. Bouvet, AS-1411, a guanosine-rich oligonucleotide aptamer targeting nucleolin for the potential treatment of cancer, including acute myeloid leukemia, Curr Opin Mol Ther, vol.12, pp.107-114, 2010.

A. Hammoudi, Proteomic profiling of a mouse model of acute intestinal Apc deletion leads to identification of potential novel biomarkers of human colorectal cancer (CRC), Biochem Biophys Res Commun, vol.440, pp.364-370, 2013.

W. Qiu, Overexpression of nucleolin and different expression sites both related to the prognosis of gastric cancer, APMIS, vol.121, pp.919-925, 2013.

M. E. Gilles, Nucleolin Targeting Impairs the Progression of Pancreatic Cancer and Promotes the Normalization of Tumor Vasculature, Cancer Res, vol.76, pp.7181-7193, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02262283

V. Marcel, Expression Profiling of Ribosome Biogenesis Factors Reveals Nucleolin as a Novel Potential Marker to Predict Outcome in AML Patients, PLoS One, vol.12, p.170160, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01567808

R. Galzio, Glycosilated nucleolin as marker for human gliomas, J Cell Biochem, vol.113, pp.571-579, 2012.

E. Benedetti, Nucleolin antagonist triggers autophagic cell death in human glioblastoma primary cells and decreased in vivo tumor growth in orthotopic brain tumor model, Oncotarget, vol.6, pp.42091-42104, 2015.

L. A. Hanakahi, L. A. Dempsey, M. J. Li, and N. Maizels, Nucleolin is one component of the B cell-specific transcription factor and switch region binding protein, LR1, Proc Natl Acad Sci, vol.94, pp.3605-3610, 1997.

J. Allinne, Perinucleolar relocalization and nucleolin as crucial events in the transcriptional activation of key genes in mantle cell lymphoma, Blood, vol.123, pp.2044-2053, 2014.

E. Grinstein, Nucleolin regulates gene expression in CD34-positive hematopoietic cells, J Biol Chem, vol.282, pp.12439-12449, 2007.

T. H. Yang, Purification and characterization of nucleolin and its identification as a transcription repressor, Mol Cell Biol, vol.14, pp.6068-6074, 1994.

D. Angelov, Nucleolin is a histone chaperone with FACT-like activity and assists remodeling of nucleosomes, EMBO J, vol.25, pp.1669-1679, 2006.
URL : https://hal.archives-ouvertes.fr/inserm-00091623

M. Goldstein, F. A. Derheimer, J. Tait-mulder, and M. B. Kastan, Nucleolin mediates nucleosome disruption critical for DNA doublestrand break repair, Proc Natl Acad Sci, vol.110, pp.16874-16879, 2013.

, SCIENtIfIC REPORTS |, vol.7

X. Gaume, K. Monier, F. Argoul, F. Mongelard, and P. Bouvet, In vivo Study of the Histone Chaperone Activity of Nucleolin by FRAP, Biochem Res Int, p.187624, 2011.

K. Abdelmohsen, Enhanced translation by Nucleolin via G-rich elements in coding and non-coding regions of target mRNAs, Nucleic Acids Res, vol.39, pp.8513-8530, 2011.

S. Das, Characterization of nucleolin K88 acetylation defines a new pool of nucleolin colocalizing with pre-mRNA splicing factors, FEBS Lett, vol.587, pp.417-424, 2013.

V. Marchand, Identification of protein partners of the human immunodeficiency virus 1 tat/rev exon 3 leads to the discovery of a new HIV-1 splicing regulator, protein hnRNP K, RNA Biol, vol.8, pp.325-342, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01866938

B. F. Pickering, D. Yu, and M. W. Van-dyke, Nucleolin protein interacts with microprocessor complex to affect biogenesis of microRNAs 15a and 16, J Biol Chem, vol.286, pp.44095-44103, 2011.

F. Pichiorri, In vivo NCL targeting affects breast cancer aggressiveness through miRNA regulation, J Exp Med, vol.210, pp.951-968, 2013.

A. Salvetti, Nuclear functions of nucleolin through global proteomics and interactomic approaches, J Proteome Res, vol.15, pp.1659-1669, 2016.
URL : https://hal.archives-ouvertes.fr/hal-02191488

S. Storck, M. Thiry, and P. Bouvet, Conditional knockout of nucleolin in DT40 cells reveals the functional redundancy of its RNAbinding domains, Biol Cell, vol.101, pp.153-167, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00371314

I. Ugrinova, Inactivation of nucleolin leads to nucleolar disruption, cell cycle arrest and defects in centrosome duplication, BMC Mol Biol, vol.8, p.66, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00337707

X. Gaume, Centrosomal nucleolin is required for microtubule network organization, Cell Cycle, vol.14, pp.902-919, 2015.

X. Gaume, Expression of Nucleolin Affects Microtubule Dynamics, PLoS One, vol.11, p.157534, 2016.

S. M. Murphy, GCP5 and GCP6: two new members of the human gamma-tubulin complex, Mol Biol Cell, vol.12, pp.3340-3352, 2001.

A. E. Leonard, S. L. Pereira, H. Sprecher, and Y. S. Huang, Elongation of long-chain fatty acids, Prog Lipid Res, vol.43, pp.36-54, 2004.

A. C. Kimmelman, Metabolic Dependencies in RAS-Driven Cancers, Clin Cancer Res, vol.21, pp.1828-1834, 2015.

N. Subramanian, Nucleolin-aptamer therapy in retinoblastoma: molecular changes and mass spectrometry-based imaging, Mol Ther Nucleic Acids, vol.5, p.358, 2016.

J. Bi, Identification of nucleolin as a lipid-raft-dependent beta1-integrin-interacting protein in A375 cell migration, Mol Cells, vol.36, pp.507-517, 2013.

H. Jin, Divergent behaviors and underlying mechanisms of cell migration and invasion in non-metastatic T24 and its metastatic derivative T24T bladder cancer cell lines, Oncotarget, vol.6, pp.522-536, 2015.

W. Qiu, The involvement of cell surface nucleolin in the initiation of CCR6 signaling in human hepatocellular carcinoma, Med Oncol, vol.32, p.75, 2015.

T. Watanabe, Epithelial-mesenchymal transition in human gastric cancer cell lines induced by TNF-alpha-inducing protein of Helicobacter pylori, Int J Cancer, vol.134, pp.2373-2382, 2014.

M. Hollander and D. Wolfe, Nonparametric Statistical Methods, 1999.

P. Sethupathy, B. Corda, and A. G. Hatzigeorgiou, TarBase: A comprehensive database of experimentally supported animal microRNA targets, RNA, vol.12, pp.192-197, 2006.

T. I. Jeon, An SREBP-responsive microRNA operon contributes to a regulatory loop for intracellular lipid homeostasis, Cell Metab, vol.18, pp.51-61, 2013.

W. L. Wang, J. Welsh, and M. Tenniswood, 1,25-Dihydroxyvitamin D3 modulates lipid metabolism in prostate cancer cells through miRNA mediated regulation of PPARA, J Steroid Biochem Mol Biol, vol.136, pp.247-251, 2013.

Q. Zhang, miR-17 is involved in the regulation of LC-PUFA biosynthesis in vertebrates: effects on liver expression of a fatty acyl desaturase in the marine teleost Siganus canaliculatus, Biochim Biophys Acta, vol.1841, pp.934-943, 2014.

D. Destouches, A simple approach to cancer therapy afforded by multivalent pseudopeptides that target cell-surface nucleoproteins, Cancer Res, vol.71, pp.3296-3305, 2011.
URL : https://hal.archives-ouvertes.fr/hal-00578863

M. Koutsioumpa and E. Papadimitriou, Cell surface nucleolin as a target for anti-cancer therapies, Recent Pat Anticancer Drug Discov, vol.9, pp.137-152, 2014.

J. W. Lee, H. J. Kim, and K. Heo, Therapeutic aptamers: developmental potential as anticancer drugs, BMB Rep, vol.48, pp.234-237, 2015.

J. Lefils, A. Geloen, H. Vidal, M. Lagarde, and N. Bernoud-hubac, Dietary DHA: time course of tissue uptake and effects on cytokine secretion in mice, Br J Nutr, vol.104, pp.1304-1312, 2010.

R. A. Irizarry, Exploration, normalization, and summaries of high density oligonucleotide array probe level data, Biostatistics, vol.4, pp.249-264, 2003.

Y. Benjamini and Y. Hochberg, Controlling the False Discovery Rate: A Practical and Powerful Approach to Multiple Testing, Journal of the Royal Statistical Society, Series B, vol.57, pp.289-300, 1995.

P. Shannon, Cytoscape: a software environment for integrated models of biomolecular interaction networks, Genome Res, vol.13, pp.2498-2504, 2003.

A. Subramanian, Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles, Proc Natl Acad Sci, vol.102, pp.15545-15550, 2005.