J. Visvader, Keeping abreast of the mammary epithelial hierarchy and breast tumorigenesis, Genes & Development, vol.23, issue.22, pp.2563-2577, 2009.
DOI : 10.1101/gad.1849509

M. Asselin-labat, M. Shackleton, J. Stingl, F. Vaillant, and N. Forrest, Steroid Hormone Receptor Status of Mouse Mammary Stem Cells, JNCI Journal of the National Cancer Institute, vol.98, issue.14, pp.1011-1014, 2006.
DOI : 10.1093/jnci/djj267

M. Shackleton, F. Vaillant, K. Simpson, J. Stingl, and G. Smyth, Generation of a functional mammary gland from a single stem cell, Nature, vol.30, issue.7072, pp.84-88, 2006.
DOI : 10.1038/nature04372

K. Sleeman, H. Kendrick, A. Ashworth, C. Isacke, and M. Smalley, CD24 staining of mouse mammary gland cells defines luminal epithelial, myoepithelial/basal and non-epithelial cells, Breast Cancer Research, vol.34, issue.1, p.7, 2006.
DOI : 10.1007/s11626-998-0067-0

K. Sleeman, H. Kendrick, D. Robertson, C. Isacke, and A. Ashworth, Dissociation of estrogen receptor expression and in vivo stem cell activity in the mammary gland, The Journal of Cell Biology, vol.439, issue.1, pp.19-26, 2007.
DOI : 10.1177/002215549904701012

J. Stingl, P. Eirew, I. Ricketson, M. Shackleton, and F. Vaillant, Purification and unique properties of mammary epithelial stem cells, Nature, vol.96, pp.993-997, 2006.
DOI : 10.1038/nature04496

I. Taddei, M. Deugnier, M. Faraldo, V. Petit, and D. Bouvard, ??1 Integrin deletion from the basal compartment of the mammary epithelium affects stem cells, Nature Cell Biology, vol.19, issue.6, pp.716-722, 2008.
DOI : 10.1016/S1074-7613(00)80216-2

J. Stingl, A. Raouf, J. Emerman, and C. Eaves, Epithelial Progenitors in the Normal Human Mammary Gland, Journal of Mammary Gland Biology and Neoplasia, vol.98, issue.1, pp.49-59, 2005.
DOI : 10.1007/s10911-005-2540-7

W. Meng, Y. Mushika, T. Ichii, and M. Takeichi, Anchorage of Microtubule Minus Ends to Adherens Junctions Regulates Epithelial Cell-Cell Contacts, Cell, vol.135, issue.5, pp.948-959, 2008.
DOI : 10.1016/j.cell.2008.09.040

G. Bellett, J. Carter, J. Keynton, D. Goldspink, and C. James, Microtubule plus-end and minus-end capture at adherens junctions is involved in the assembly of apico-basal arrays in polarised epithelial cells, Cell Motility and the Cytoskeleton, vol.117, issue.4, pp.893-908, 2009.
DOI : 10.1002/cm.20393

N. Turner and J. Reis-filho, Basal-like breast cancer and the BRCA1 phenotype, Oncogene, vol.62, issue.43, pp.5846-5853, 2006.
DOI : 10.1073/pnas.171174298

M. Tischkowitz and W. Foulkes, The Basal Phenotype of BRCA1-Related Breast Cancer: Past, Present and Future, Cell Cycle, vol.5, issue.9, pp.963-967, 2006.
DOI : 10.4161/cc.5.9.2713

W. Foulkes, BRCA1 functions as a breast stem cell regulator, Journal of Medical Genetics, vol.41, issue.1, pp.1-5, 2004.
DOI : 10.1136/jmg.2003.013805

S. Furuta, X. Jiang, B. Gu, E. Cheng, and P. Chen, Depletion of BRCA1 impairs differentiation but enhances proliferation of mammary epithelial cells, Proceedings of the National Academy of Sciences, vol.102, issue.26, pp.9176-9181, 2005.
DOI : 10.1073/pnas.0503793102

L. Burga, N. Tung, S. Troyan, M. Bostina, and P. Konstantinopoulos, Altered Proliferation and Differentiation Properties of Primary Mammary Epithelial Cells from BRCA1 Mutation Carriers, Cancer Research, vol.69, issue.4, pp.1273-1278, 2009.
DOI : 10.1158/0008-5472.CAN-08-2954

S. Liu, C. Ginestier, E. Charafe-jauffret, H. Foco, and C. Kleer, BRCA1 regulates human mammary stem/progenitor cell fate, Proceedings of the National Academy of Sciences, vol.105, issue.5, pp.1680-1685, 2008.
DOI : 10.1073/pnas.0711613105

URL : https://hal.archives-ouvertes.fr/hal-01431959

E. Lim, F. Vaillant, D. Wu, N. Forrest, and B. Pal, Aberrant luminal progenitors as the candidate target population for basal tumor development in BRCA1 mutation carriers, Nature Medicine, vol.5, issue.8, pp.907-913, 2009.
DOI : 10.1038/nm.2000

G. Molyneux, F. Geyer, F. Magnay, A. Mccarthy, and H. Kendrick, BRCA1 Basal-like Breast Cancers Originate from Luminal Epithelial Progenitors and Not from Basal Stem Cells, Cell Stem Cell, vol.7, issue.3, pp.403-417, 2010.
DOI : 10.1016/j.stem.2010.07.010

T. Proia, P. Keller, P. Gupta, I. Klebba, and A. Jones, Genetic Predisposition Directs Breast Cancer Phenotype by Dictating Progenitor Cell Fate, Cell Stem Cell, vol.8, issue.2, pp.149-163, 2011.
DOI : 10.1016/j.stem.2010.12.007

S. Sankaran, L. Starita, A. Groen, M. Ko, and J. Parvin, Centrosomal Microtubule Nucleation Activity Is Inhibited by BRCA1-Dependent Ubiquitination, Molecular and Cellular Biology, vol.25, issue.19, pp.8656-8668, 2005.
DOI : 10.1128/MCB.25.19.8656-8668.2005

L. Starita, Y. Machida, S. Sankaran, J. Elias, and K. Griffin, BRCA1-Dependent Ubiquitination of ??-Tubulin Regulates Centrosome Number, Molecular and Cellular Biology, vol.24, issue.19, pp.8457-8466, 2004.
DOI : 10.1128/MCB.24.19.8457-8466.2004

V. Joukov, A. Groen, T. Prokhorova, R. Gerson, and E. White, The BRCA1/BARD1 Heterodimer Modulates Ran-Dependent Mitotic Spindle Assembly, Cell, vol.127, issue.3, pp.539-552, 2006.
DOI : 10.1016/j.cell.2006.08.053

M. Pujana, J. Han, L. Starita, K. Stevens, and M. Tewari, Network modeling links breast cancer susceptibility and centrosome dysfunction, Nature Genetics, vol.22, issue.11, pp.1338-1349, 2007.
DOI : 10.1038/ng.2007.2

V. Assmann, C. Gillett, R. Poulsom, K. Ryder, and I. Hart, The pattern of expression of the microtubule-binding protein RHAMM/IHABP in mammary carcinoma suggests a role in the invasive behaviour of tumour cells, The Journal of Pathology, vol.82, issue.2, pp.191-196, 2001.
DOI : 10.1002/path.941

I. Bieche, S. Tozlu, I. Girault, and R. Lidereau, Identification of a three-gene expression signature of poor-prognosis breast carcinoma, Molecular Cancer, vol.3, issue.1, p.37, 2004.
DOI : 10.1186/1476-4598-3-37

URL : https://hal.archives-ouvertes.fr/inserm-00094845

A. Groen, L. Cameron, M. Coughlin, D. Miyamoto, and T. Mitchison, XRHAMM Functions in Ran-Dependent Microtubule Nucleation and Pole Formation during Anastral Spindle Assembly, Current Biology, vol.14, issue.20, pp.1801-1811, 2004.
DOI : 10.1016/j.cub.2004.10.002

C. Maxwell, J. Keats, A. Belch, L. Pilarski, and T. Reiman, Receptor for hyaluronan-mediated motility correlates with centrosome abnormalities in multiple myeloma and maintains mitotic integrity, Cancer Res, vol.65, pp.850-860, 2005.

C. Tolg, S. Hamilton, L. Morningsar, J. Zhang, and K. Esguerra, RHAMM Promotes Interphase Microtubule Instability and Mitotic Spindle Integrity through MEK1/ERK1/2 Activity, Journal of Biological Chemistry, vol.285, issue.34, pp.26461-26474, 2010.
DOI : 10.1074/jbc.M110.121491

A. Stefansson, O. , G. Jonasson, J. , T. Johannsson et al., Genomic profiling of breast tumours in relation to BRCAabnormalities and phenotypes, Breast Cancer Research, vol.21, issue.4, p.47, 2009.
DOI : 10.1016/j.clon.2009.04.004

K. Nathanson, Y. Shugart, R. Omaruddin, C. Szabo, and D. Goldgar, CGH-targeted linkage analysis reveals a possible BRCA1 modifier locus on chromosome 5q, Human Molecular Genetics, vol.11, issue.11, pp.1327-1332, 2002.
DOI : 10.1093/hmg/11.11.1327

A. Antoniou, X. Wang, Z. Fredericksen, L. Mcguffog, and R. Tarrell, A locus on 19p13 modifies risk of breast cancer in BRCA1 mutation carriers and is associated with hormone receptor???negative breast cancer in the general population, Nature Genetics, vol.447, issue.10, pp.885-892, 2010.
DOI : 10.1002/gepi.1370050611

URL : https://hal.archives-ouvertes.fr/hal-00698376

A. Antoniou, O. Sinilnikova, L. Mcguffog, S. Healey, and H. Nevanlinna, Common variants in LSP1, 2q35 and 8q24 and breast cancer risk for BRCA1 and BRCA2 mutation carriers, Human Molecular Genetics, vol.18, issue.22, pp.4442-4456, 2009.
DOI : 10.1093/hmg/ddp372

A. Antoniou, A. Spurdle, O. Sinilnikova, S. Healey, and K. Pooley, Common Breast Cancer-Predisposition Alleles Are Associated with Breast Cancer Risk in BRCA1 and BRCA2 Mutation Carriers, The American Journal of Human Genetics, vol.82, issue.4, pp.937-948, 2008.
DOI : 10.1016/j.ajhg.2008.02.008

T. Rebbeck, H. Lynch, S. Neuhausen, S. Narod, and L. Van-'t-veer, Prophylactic oophorectomy in BRCA1 and BRCA2 mutation carriers, European Journal of Cancer, vol.38, pp.1616-1622, 2002.
DOI : 10.1016/S0959-8049(02)00269-1

A. Antoniou, D. Goldgar, N. Andrieu, J. Chang-claude, and R. Brohet, A weighted cohort approach for analysing factors modifying disease risks in carriers of high-risk susceptibility genes, Genetic Epidemiology, vol.10, issue.1, pp.1-11, 2005.
DOI : 10.1002/gepi.20074

M. Buisson, O. Anczukow, A. Zetoune, M. Ware, and S. Mazoyer, The 185delAG mutation (c.68_69delAG) in theBRCA1 gene triggers translation reinitiation at a downstream AUG codon, Human Mutation, vol.16, issue.10, pp.1024-1029, 2006.
DOI : 10.1002/humu.20384

H. Liu, L. Cartegni, M. Zhang, and A. Krainer, A mechanism for exon skipping caused by nonsense or missense mutations in BRCA1 and other genes, Nat Genet, vol.27, pp.55-58, 2001.

S. Mazoyer, N. Puget, L. Perrin-vidoz, H. Lynch, and O. Serova-sinilnikova, A BRCA1 Nonsense Mutation Causes Exon Skipping, The American Journal of Human Genetics, vol.62, issue.3, pp.713-715, 1998.
DOI : 10.1086/301768

L. Perrin-vidoz, O. Sinilnikova, D. Stoppa-lyonnet, G. Lenoir, and S. Mazoyer, The nonsense-mediated mRNA decay pathway triggers degradation of most BRCA1 mRNAs bearing premature termination codons, Human Molecular Genetics, vol.11, issue.23, pp.2805-2814, 2002.
DOI : 10.1093/hmg/11.23.2805

A. Antoniou, O. Sinilnikova, J. Simard, M. Leone, and M. Dumont, RAD51 135G???C Modifies Breast Cancer Risk among BRCA2 Mutation Carriers: Results from a Combined Analysis of 19 Studies, The American Journal of Human Genetics, vol.81, issue.6, pp.1186-1200, 2007.
DOI : 10.1086/522611

I. Adzhubei, S. Schmidt, L. Peshkin, V. Ramensky, and A. Gerasimova, A method and server for predicting damaging missense mutations, Nature Methods, vol.7, issue.4, pp.248-249, 2010.
DOI : 10.1038/nmeth0410-248

P. Kumar, S. Henikoff, and P. Ng, Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm, Nature Protocols, vol.4, issue.8, pp.1073-1081, 2009.
DOI : 10.1101/gr.772403

C. Maxwell, E. Rasmussen, F. Zhan, J. Keats, and S. Adamia, RHAMM expression and isoform balance predict aggressive disease and poor survival in multiple myeloma, Blood, vol.104, issue.4, pp.1151-1158, 2004.
DOI : 10.1182/blood-2003-11-4079

J. Venables, R. Klinck, A. Bramard, L. Inkel, and G. Dufresne-martin, Identification of Alternative Splicing Markers for Breast Cancer, Cancer Research, vol.68, issue.22, pp.9525-9531, 2008.
DOI : 10.1158/0008-5472.CAN-08-1769

J. Debnath, S. Muthuswamy, and J. Brugge, Morphogenesis and oncogenesis of MCF-10A mammary epithelial acini grown in three-dimensional basement membrane cultures, Methods, vol.30, issue.3, pp.256-268, 2003.
DOI : 10.1016/S1046-2023(03)00032-X

O. Petersen, L. Ronnov-jessen, A. Howlett, and M. Bissell, Interaction with basement membrane serves to rapidly distinguish growth and differentiation pattern of normal and malignant human breast epithelial cells., Proceedings of the National Academy of Sciences, vol.89, issue.19, pp.9064-9068, 1992.
DOI : 10.1073/pnas.89.19.9064

B. Weigelt and M. Bissell, Unraveling the microenvironmental influences on the normal mammary gland and breast cancer, Seminars in Cancer Biology, vol.18, issue.5, pp.311-321, 2008.
DOI : 10.1016/j.semcancer.2008.03.013

C. Plachot, L. Chaboub, H. Adissu, L. Wang, and A. Urazaev, Factors necessary to produce basoapical polarity in human glandular epithelium formed in conventional and high-throughput three-dimensional culture: example of the breast epithelium, BMC Biology, vol.7, issue.1, p.77, 2009.
DOI : 10.1186/1741-7007-7-77

S. Rodríguez-pinilla, D. Sarrio, E. Honrado, G. Moreno-bueno, and D. Hardisson, Vimentin and laminin expression is associated with basal-like phenotype in both sporadic and BRCA1-associated breast carcinomas, Journal of Clinical Pathology, vol.60, issue.9, pp.1006-1012, 2007.
DOI : 10.1136/jcp.2006.042143

W. Lingle and J. Salisbury, Altered Centrosome Structure Is Associated with Abnormal Mitoses in Human Breast Tumors, The American Journal of Pathology, vol.155, issue.6, pp.1941-1951, 1999.
DOI : 10.1016/S0002-9440(10)65513-7

L. Littlepage and J. Ruderman, Identification of a new APC/C recognition domain, the A box, which is required for the Cdh1-dependent destruction of the kinase Aurora-A during mitotic exit, Genes & Development, vol.16, issue.17, pp.2274-2285, 2002.
DOI : 10.1101/gad.1007302

S. Sankaran, D. Crone, R. Palazzo, and J. Parvin, Aurora-A Kinase Regulates Breast Cancer Associated Gene 1 Inhibition of Centrosome-Dependent Microtubule Nucleation, Cancer Research, vol.67, issue.23, pp.11186-11194, 2007.
DOI : 10.1158/0008-5472.CAN-07-2578

R. Bayliss, T. Sardon, I. Vernos, and E. Conti, Structural Basis of Aurora-A Activation by TPX2 at the Mitotic Spindle, Molecular Cell, vol.12, issue.4, pp.851-862, 2003.
DOI : 10.1016/S1097-2765(03)00392-7

E. Harrington, D. Bebbington, J. Moore, R. Rasmussen, and A. Ajose-adeogun, VX-680, a potent and selective small-molecule inhibitor of the Aurora kinases, suppresses tumor growth in vivo, Nature Medicine, vol.10, issue.3, pp.262-267, 2004.
DOI : 10.1038/nm1003

G. Carter, D. Galas, and T. Galitski, Maximal Extraction of Biological Information from Genetic Interaction Data, PLoS Computational Biology, vol.25, issue.4, 2009.
DOI : 10.1371/journal.pcbi.1000347.s002

Y. Shi, T. Reiman, W. Li, C. Maxwell, and S. Sen, Targeting aurora kinases as therapy in multiple myeloma, Blood, vol.109, issue.9, pp.3915-3921, 2007.
DOI : 10.1182/blood-2006-07-037671

M. Nousiainen, H. Sillje, G. Sauer, E. Nigg, and R. Korner, Phosphoproteome analysis of the human mitotic spindle, Proceedings of the National Academy of Sciences, vol.103, issue.14, pp.5391-5396, 2006.
DOI : 10.1073/pnas.0507066103

I. Cheeseman, S. Anderson, M. Jwa, E. Green, and J. Kang, Phospho-Regulation of Kinetochore-Microtubule Attachments by the Aurora Kinase Ipl1p, Cell, vol.111, issue.2, pp.163-172, 2002.
DOI : 10.1016/S0092-8674(02)00973-X

M. Fournier, K. Martin, P. Kenny, K. Xhaja, and I. Bosch, Gene Expression Signature in Organized and Growth-Arrested Mammary Acini Predicts Good Outcome in Breast Cancer, Cancer Research, vol.66, issue.14, pp.7095-7102, 2006.
DOI : 10.1158/0008-5472.CAN-06-0515

L. Emery, A. Tripathi, C. King, M. Kavanah, and J. Mendez, Early Dysregulation of Cell Adhesion and Extracellular Matrix Pathways in Breast Cancer Progression, The American Journal of Pathology, vol.175, issue.3, pp.1292-1302, 2009.
DOI : 10.2353/ajpath.2009.090115

B. Kalmyrzaev, P. Pharoah, D. Easton, B. Ponder, and A. Dunning, Hyaluronan-Mediated Motility Receptor Gene Single Nucleotide Polymorphisms and Risk of Breast Cancer, Cancer Epidemiology Biomarkers & Prevention, vol.17, issue.12, pp.3618-3620, 2008.
DOI : 10.1158/1055-9965.EPI-08-0216

O. Fletcher, N. Johnson, C. Palles, S. Silva, I. Mccormack et al., Inconsistent Association Between the STK15 F31I Genetic Polymorphism and Breast Cancer Risk, JNCI Journal of the National Cancer Institute, vol.98, issue.14, pp.1014-1018, 2006.
DOI : 10.1093/jnci/djj268

C. Maxwell, J. Keats, M. Crainie, X. Sun, and T. Yen, RHAMM Is a Centrosomal Protein That Interacts with Dynein and Maintains Spindle Pole Stability, Molecular Biology of the Cell, vol.14, issue.6, pp.2262-2276, 2003.
DOI : 10.1091/mbc.E02-07-0377

J. Olson, X. Wang, V. Pankratz, Z. Fredericksen, and C. Vachon, Centrosome-related genes, genetic variation, and risk of breast cancer, Breast Cancer Research and Treatment, vol.41, issue.5, pp.221-228, 2011.
DOI : 10.1007/s10549-010-0950-8

C. Maxwell, V. Moreno, X. Solé, L. Gómez, and P. Hernández, Genetic interactions: the missing links for a better understanding of cancer susceptibility, progression and treatment, Molecular Cancer, vol.7, issue.1, 2008.
DOI : 10.1186/1476-4598-7-4

M. Labarge, C. Nelson, R. Villadsen, A. Fridriksdottir, and J. Ruth, Human mammary progenitor cell fate decisions are products of interactions with combinatorial microenvironments, Integr. Biol., vol.10, issue.Pt 7, pp.70-79, 2009.
DOI : 10.1039/B816472J

X. Fant, V. Srsen, A. Espigat-georger, and A. Merdes, Nuclei of Non-Muscle Cells Bind Centrosome Proteins upon Fusion with Differentiating Myoblasts, PLoS ONE, vol.4, issue.12, 2009.
DOI : 10.1371/journal.pone.0008303.g007

M. Stiess, N. Maghelli, L. Kapitein, S. Gomis-ruth, and M. Wilsch-brauninger, Axon Extension Occurs Independently of Centrosomal Microtubule Nucleation, Science, vol.327, issue.5966, pp.704-707, 2010.
DOI : 10.1126/science.1182179

C. Oikonomou and F. Cross, Frequency control of cell cycle oscillators, Current Opinion in Genetics & Development, vol.20, issue.6, pp.605-612, 2010.
DOI : 10.1016/j.gde.2010.08.006

M. Ouchi, N. Fujiuchi, K. Sasai, H. Katayama, and Y. Minamishima, to M Transition, Journal of Biological Chemistry, vol.279, issue.19, pp.19643-19648, 2004.
DOI : 10.1074/jbc.M311780200

S. Chang, K. Biswas, B. Martin, S. Stauffer, and S. Sharan, Expression of human BRCA1 variants in mouse ES cells allows functional analysis of BRCA1 mutations, Journal of Clinical Investigation, vol.119, issue.10, pp.3160-3171, 2009.
DOI : 10.1172/JCI39836DS1

T. Huang, P. Cheng, Y. Chan, T. Yeh, and Y. Young, Regulation of ciliary differentiation of human respiratory epithelial cells by the receptor for hyaluronan-mediated motility on hyaluronan-based biomaterials, Biomaterials, vol.31, issue.26, pp.6701-6709, 2010.
DOI : 10.1016/j.biomaterials.2010.05.054

V. Calvo and M. Beato, BRCA1 Counteracts Progesterone Action by Ubiquitination Leading to Progesterone Receptor Degradation and Epigenetic Silencing of Target Promoters, Cancer Research, vol.71, issue.9, pp.3422-3431, 2011.
DOI : 10.1158/0008-5472.CAN-10-3670

C. Ginestier, S. Liu, and M. Wicha, Getting to the Root of BRCA1-Deficient Breast Cancer, Cell Stem Cell, vol.5, issue.3, pp.229-230, 2009.
DOI : 10.1016/j.stem.2009.08.007

URL : https://hal.archives-ouvertes.fr/hal-01431956

K. Markianos, M. Daly, and L. Kruglyak, Efficient Multipoint Linkage Analysis through Reduction of Inheritance Space, The American Journal of Human Genetics, vol.68, issue.4, pp.963-977, 2001.
DOI : 10.1086/319507

V. Assmann, D. Jenkinson, J. Marshall, and I. Hart, The intracellular hyaluronan receptor RHAMM/IHABP interacts with microtubules and actin filaments, J Cell Sci, vol.112, pp.3943-3954, 1999.