C. Chad and D. Botsteid, Isolation and Characterizationof Chromosome-Gain and Increase-in-Ploidy Mutants in Yeast, Genetics, vol.135, pp.677-691, 1993.

J. Paris and M. Philippe, Poly(A) metabolism and polysomal recruitment of maternal mRNAs during early Xenopus development, Dev Biol, vol.140, pp.221-224, 1990.

T. Andrésson and J. V. Ruderman, The kinase Eg2 is a component of the Xenopus oocyte progesterone-activated signaling pathway, EMBO J, vol.17, pp.5627-5637, 1998.

,

C. Roghi, R. Giet, and R. Uzbekov, The Xenopus protein kinase pEg2 associates with the centrosome in a cell cycle-dependent manner, binds to the spindle microtubules and is involved in bipolar mitotic spindle assembly, J Cell Sci, vol.111, pp.557-572, 1998.
URL : https://hal.archives-ouvertes.fr/inserm-00966036

D. M. Glover, M. H. Leibowitz, D. A. Mclean, and H. Parry, Mutations in aurora prevent centrosome separation leading to the formation of monopolar spindles, Cell, vol.81, pp.90374-90381, 1995.

F. Wirtz-peitz, T. Nishimura, and J. A. Knoblich, Linking cell cycle to asymmetric division: Aurora-A phosphorylates the Par complex to regulate Numb localization, Cell, vol.135, pp.161-173, 2008.

R. Giet, D. Mclean, and S. Descamps, Drosophila Aurora A kinase is required to localize D-TACC to centrosomes and to regulate astral microtubules, J Cell Biol, vol.156, pp.437-451, 2002.
URL : https://hal.archives-ouvertes.fr/inserm-00966242

C. Lee, R. O. Andersen, and C. Cabernard, Drosophila Aurora-A kinase inhibits neuroblast self-renewal by regulating aPKC/Numb cortical polarity and spindle orientation, Genes Dev, vol.20, pp.3464-3474, 2006.

H. Wang, G. W. Somers, and A. Bashirullah, Aurora-A acts as a tumor suppressor and regulates self-renewal of Drosophila neuroblasts, Genes Dev, vol.20, pp.3453-3463, 2006.

,

P. Meraldi, R. Honda, and E. A. Nigg, Aurora-A overexpression reveals tetraploidization as a major route to centrosome amplification in p53-/-cells, EMBO J, vol.21, pp.483-492, 2002.

D. Zhang, T. Hirota, and T. Marumoto, Cre-loxP-controlled periodic Aurora-A overexpression induces mitotic abnormalities and hyperplasia in mammary glands of mouse models, Oncogene, vol.23, pp.8720-8730, 2004.

G. Vader and S. Lens, The Aurora kinase family in cell division and cancer, Biochimica et Biophysica Acta (BBA) -Reviews on Cancer, vol.1786, pp.60-72, 2008.

,

A. S. Nikonova, I. Astsaturov, and I. G. Serebriiskii, Aurora A kinase (AURKA) in normal and pathological cell division, Cellular and Molecular Life Sciences, vol.70, pp.661-687, 2013.

J. J. Isola, S. Fuqua, S. G. Hilsenbeck, and F. M. Waldman, Genetic Aberrations Detected by Comparative Genomic Hybridization Predict Outcome in Node-Negative Breast Cancer, vol.147, p.7, 1995.

S. S. Farag, The potential role of Aurora kinase inhibitors in haematological malignancies, Review. British Journal of Haematology, vol.155, pp.561-579, 2011.

H. Zhou, J. Kuang, and L. Zhong, Tumour amplified kinase STK15/BTAK induces centrosome amplification, aneuploidy and transformation, Nat Genet, vol.20, pp.189-193, 1998.

Y. Miyoshi, K. Iwao, C. Egawa, and S. Noguchi, Association of centrosomal kinaseSTK15/BTAK mRNA expression with chromosomal instability in human breast cancers, Int J Cancer, vol.92, pp.370-373, 2001.

Y. Jeng, S. Peng, C. Lin, and H. Hsu, Overexpression and Amplification of Aurora-A in Hepatocellular Carcinoma, Clin Cancer Res, vol.10, pp.2065-2071, 2004.

V. Bavetsias and S. Linardopoulos, Aurora Kinase Inhibitors: Current Status and Outlook, Front Oncol, vol.5, 2015.

B. A. Haubrich and D. C. Swinney, Enzyme Activity Assays for Protein Kinases: Strategies to Identify Active Substrates, Curr Drug Discov Technol, vol.13, pp.2-15, 2016.

J. Knight, T. Pawson, and A. Gingras, Profiling the kinome: Current capabilities and future challenges, Journal of Proteomics, vol.81, pp.43-55, 2013.

G. Bertolin, A. Bulteau, and M. Alves-guerra, Aurora kinase A localises to mitochondria to control organelle dynamics and energy production, vol.7, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01862597

M. Kamran, Z. Long, and D. Xu, Aurora kinase A regulates Survivin stability through targeting FBXL7 in gastric cancer drug resistance and prognosis. Oncogenesis 6:e298, 2017.

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, pp.851-862, 2003.

Y. Zhang, J. Ni, and Q. Huang, Identification of the auto-inhibitory domains of Aurora-A kinase, Biochemical and Biophysical Research Communications, vol.357, pp.347-352, 2007.

,

L. E. Littlepage, H. Wu, and T. Andresson, Identification of phosphorylated residues that affect the activity of the mitotic kinase Aurora-A, Proc Natl Acad Sci, vol.99, pp.15440-15445, 2002.

C. A. Dodson, S. Yeoh, T. Haq, and R. Bayliss, A Kinetic Test Characterizes Kinase Intramolecular and Intermolecular Autophosphorylation Mechanisms, Sci Signal, vol.6, pp.54-54, 2013.

A. Zorba, V. Buosi, and S. Kutter, Molecular mechanism of Aurora A kinase autophosphorylation and its allosteric activation by TPX2, vol.3, p.2667, 2014.

T. A. Kufer, H. Silljé, and R. Körner, Human TPX2 is required for targeting Aurora-A kinase to the spindle, J Cell Biol, vol.158, pp.617-623, 2002.

P. A. Eyers, E. Erikson, L. G. Chen, and J. L. Maller, A novel mechanism for activation of the protein kinase Aurora A, Current Biology, vol.13, pp.691-697, 2003.

S. Brunet, T. Sardon, and T. Zimmerman, Characterization of the TPX2 Domains Involved in Microtubule Nucleation and Spindle Assembly in Xenopus Egg Extracts, Mol Biol Cell, vol.15, pp.5318-5328, 2004.

S. G. Burgess and R. Bayliss, The structure of C290A:C393A Aurora A provides structural insights into kinase regulation, Acta Crystallogr F Struct Biol Commun, vol.71, pp.315-319, 2015.

C. A. Dodson and R. Bayliss, Activation of Aurora-A kinase by protein partner binding and phosphorylation are independent and synergistic, J Biol Chem, vol.287, pp.1150-1157, 2012.

M. Toya, M. Terasawa, and K. Nagata, A kinase-independent role for Aurora A in the assembly of mitotic spindle microtubules in Caenorhabditis elegans embryos, Nature Cell Biology, vol.13, pp.708-714, 2011.

H. Katayama, K. Sasai, and M. Kloc, Aurora kinase-A regulates kinetochore/chromatin associated microtubule assembly in human cells, Cell Cycle, vol.7, pp.2691-2704, 2008.

D. L. Satinover, C. A. Leach, P. T. Stukenberg, and D. L. Brautigan, Activation of Aurora-A kinase by protein phosphatase inhibitor-2, a bifunctional signaling protein, PNAS, vol.101, pp.8625-8630, 2004.

K. Zeng, R. N. Bastos, F. A. Barr, and U. Gruneberg, Protein phosphatase 6 regulates mitotic spindle formation by controlling the T-loop phosphorylation state of Aurora A bound to its activator TPX2, J Cell Biol, vol.191, pp.1315-1332, 2010.

A. N. Kettenbach, K. A. Schlosser, and S. P. Lyons, Global assessment of its network dynamics reveals that the kinase Plk1 inhibits the phosphatase PP6 to promote Aurora A activity, Sci Signal, vol.11, 2018.

V. Krenn and A. Musacchio, The Aurora B Kinase in Chromosome Bi-Orientation and Spindle Checkpoint Signaling, Front Oncol, vol.5, 2015.

E. Willems, M. Dedobbeleer, and M. Digregorio, The functional diversity of Aurora kinases: a comprehensive review, Cell Div, vol.13, 2018.

E. C. Greenwald, S. Mehta, and J. Zhang, Genetically Encoded Fluorescent Biosensors Illuminate the Spatiotemporal Regulation of Signaling Networks, Chem Rev, vol.118, pp.11707-11794, 2018.

B. G. Fuller, M. A. Lampson, and E. A. Foley, Midzone activation of aurora B in anaphase produces an intracellular phosphorylation gradient, Nature, vol.453, pp.1132-1136, 2008.

,

P. D. Andrews, Y. Ovechkina, and N. Morrice, Aurora B Regulates MCAK at the Mitotic Centromere. Developmental Cell, vol.6, pp.253-268, 2004.

J. D. Violin, J. Zhang, R. Y. Tsien, and A. C. Newton, A genetically encoded fluorescent reporter reveals oscillatory phosphorylation by protein kinase C, The Journal of Cell Biology, vol.161, pp.899-909, 2003.

L. Mac?rek, A. Lindqvist, and D. Lim, Polo-like kinase-1 is activated by aurora A to promote checkpoint recovery, Nature, vol.455, pp.119-123, 2008.

I. M. Cheeseman, S. Anderson, and M. Jwa, Phospho-Regulation of Kinetochore-Microtubule Attachments by the Aurora Kinase Ipl1p, Cell, vol.111, issue.02, p.973, 2002.

S. Lens, E. E. Voest, and R. H. Medema, Shared and separate functions of polo-like kinases and aurora kinases in cancer, Nature Reviews Cancer, vol.10, pp.825-841, 2010.

A. A. Ye, J. Deretic, and C. M. Hoel, Aurora A Kinase Contributes to a Pole-Based Error Correction Pathway, Curr Biol, vol.25, pp.1842-1851, 2015.

A. A. Ye, J. Torabi, and T. J. Maresca, Aurora A Kinase Amplifies a Midzone Phosphorylation Gradient to Promote High-Fidelity Cytokinesis, Biol Bull, vol.231, pp.61-72, 2016.

T. Courtheoux, A. Diallo, and A. P. Damodaran, Aurora A kinase activity is required to maintain an active spindle assembly checkpoint during prometaphase, Journal of Cell Science, vol.131, 2018.
URL : https://hal.archives-ouvertes.fr/hal-01780367

G. Bertolin, F. Sizaire, and G. Herbomel, A FRET biosensor reveals spatiotemporal activation and functions of aurora kinase A in living cells, Nature Communications, vol.7, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01394773

C. De-la-cova, R. Townley, S. Regot, and I. Greenwald, A Real-Time Biosensor for ERK Activity Reveals Signaling Dynamics during C. elegans Cell Fate Specification, Developmental Cell, vol.42, pp.542-553, 2017.

V. Mayr, C. Sturtzel, and M. Stadler, Fast Dynamic in vivo Monitoring of Erk Activity at Single Cell Resolution in DREKA Zebrafish, Front Cell Dev Biol, vol.6, 2018.

Y. Konishi, K. Terai, and Y. Furuta, Live-Cell FRET Imaging Reveals a Role of Extracellular Signal-Regulated Kinase Activity Dynamics in Thymocyte Motility. iScience, vol.10, pp.98-113, 2018.

S. Floyd, J. Pines, and C. Lindon, APC/CCdh1 Targets Aurora Kinase to Control Reorganization of the Mitotic Spindle at Anaphase, Current Biology, vol.18, pp.1649-1658, 2008.

,

C. Alfieri, S. Zhang, and D. Barford, Visualizing the complex functions and mechanisms of the anaphase promoting complex/cyclosome (APC/C), Open Biol, vol.7, 2017.

A. Castro, Y. Arlot-bonnemains, and S. Vigneron, APC/Fizzy-Related targets Aurora-A kinase for proteolysis, EMBO reports, vol.3, pp.457-462, 2002.
URL : https://hal.archives-ouvertes.fr/inserm-00966246

X. Yu, K. Minter-dykhouse, and L. Malureanu, Chfr is required for tumor suppression and Aurora A regulation, Nat Genet, vol.37, pp.401-406, 2005.

S. R. Horn, M. J. Thomenius, and E. S. Johnson, Regulation of mitochondrial morphology by APC/CCdh1-mediated control of Drp1 stability, Molecular biology of the cell, vol.22, pp.1207-1216, 2011.

L. S. Kiat, K. M. Hui, and G. Gopalan, Aurora-A Kinase Interacting Protein (AIP), a Novel Negative Regulator of Human Aurora-A Kinase, J Biol Chem, vol.277, pp.45558-45565, 2002.

,

S. K. Lim and G. Gopalan, Antizyme1 mediates AURKAIP1-dependent degradation of Aurora-A, Oncogene, vol.26, pp.6593-6603, 2007.

S. K. Lim and G. Gopalan, Aurora-A kinase interacting protein 1 (AURKAIP1) promotes Aurora-A degradation through an alternative ubiquitin-independent pathway, Biochem J, vol.403, pp.119-127, 2007.

S. Qiu, J. Liu, and F. Xing, Antizyme inhibitor 1: a potential carcinogenic molecule, Cancer Sci, vol.108, pp.163-169, 2017.

O. V. Plotnikova, E. N. Pugacheva, R. L. Dunbrack, and E. A. Golemis, Rapid calcium-dependent activation of Aurora-A kinase, Nat Commun, vol.1, pp.1-8, 2010.

D. Reboutier, M. Troadec, and J. Cremet, Nucleophosmin/B23 activates Aurora A at the centrosome through phosphorylation of serine 89, The Journal of Cell Biology, vol.197, pp.19-26, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00679670

E. N. Pugacheva, S. A. Jablonski, and T. R. Hartman, HEF1-Dependent Aurora A Activation Induces Disassembly of the Primary Cilium, Cell, vol.129, pp.1351-1363, 2007.

,

G. Wheway, L. Nazlamova, and J. T. Hancock, Signaling through the Primary Cilium, Front Cell Dev Biol, vol.6, 2018.

J. Pan, T. Seeger-nukpezah, and E. A. Golemis, The role of the cilium in normal and abnormal cell cycles: emphasis on renal cystic pathologies, Cell Mol Life Sci, vol.70, pp.1849-1874, 2013.

E. N. Pugacheva and E. A. Golemis, The focal adhesion scaffolding protein HEF1 regulates activation of the Aurora-A and Nek2 kinases at the centrosome, Nature Cell Biology, vol.7, pp.937-946, 2005.

D. Kinzel, K. Boldt, and E. E. Davis, Pitchfork Regulates Primary Cilia Disassembly and Left-Right Asymmetry, Developmental Cell, vol.19, pp.66-77, 2010.

K. H. Lee, Y. Johmura, and L. Yu, Identification of a novel Wnt5a-CK1?-Dvl2-Plk1-mediated primary cilia disassembly pathway: Primary cilia disassembly by the Dvl2-Plk1 complex, The EMBO Journal, vol.31, pp.3104-3117, 2012.

M. Mergen, C. Engel, and B. Muller, The nephronophthisis gene product NPHP2/Inversin interacts with Aurora A and interferes with HDAC6-mediated cilia disassembly, Nephrology Dialysis Transplantation, vol.28, pp.2744-2753, 2013.

O. V. Plotnikova, A. S. Nikonova, and Y. V. Loskutov, Calmodulin activation of Aurora-A kinase (AURKA) is required during ciliary disassembly and in mitosis, Molecular Biology of the Cell, vol.23, pp.2658-2670, 2012.

J. M. Frade and O. Mc, Neuronal cell cycle: the neuron itself and its circumstances, Cell Cycle, vol.14, pp.712-720, 2015.

D. Mori, M. Yamada, and Y. Mimori-kiyosue, An essential role of the aPKC-Aurora A-NDEL1 pathway in neurite elongation by modulation of microtubule dynamics, Nature Cell Biology, vol.11, pp.1057-1068, 2009.

F. Bleichert, M. R. Botchan, and J. M. Berger, Mechanisms for initiating cellular DNA replication, Science, vol.355, p.6317, 2017.

T. Tsunematsu, Y. Takihara, and N. Ishimaru, Aurora-A controls pre-replicative complex assembly and DNA replication by stabilizing geminin in mitosis, Nature Communications, vol.4, p.1885, 2013.

T. Tsunematsu, R. Arakaki, and A. Yamada, The Non-Canonical Role of Aurora-A in DNA Replication, Front Oncol, vol.5, 2015.

A. Veerakumarasivam, L. D. Goldstein, and K. Saeb-parsy, AURKA overexpression accompanies dysregulation of DNA-damage response genes in invasive urothelial cell carcinoma, Cell Cycle, vol.7, pp.3525-3533, 2008.

G. Yang, B. Chang, and F. Yang, Aurora Kinase A Promotes Ovarian Tumorigenesis through Dysregulation of the Cell Cycle and Suppression of BRCA2, Clin Cancer Res, vol.16, pp.3171-3181, 2010.

T. Marumoto, T. Hirota, and T. Morisaki, Roles of aurora-A kinase in mitotic entry and G2 checkpoint in mammalian cells, Genes to Cells, vol.7, pp.1173-1182, 2002.

T. Do, J. Hirst, and S. Hyter, Aurora A kinase regulates non-homologous end-joining and poly(ADP-ribose) polymerase function in ovarian carcinoma cells, Oncotarget, vol.8, pp.50376-50392, 2017.

R. Chaudhuri, A. Nussenzweig, and A. , The multifaceted roles of PARP1 in DNA repair and chromatin remodelling, Nat Rev Mol Cell Biol, vol.18, pp.610-621, 2017.

J. P. Vaughn, P. L. Davis, and M. D. Jarboe, BRCA1 expression is induced before DNA synthesis in both normal and tumor-derived breast cells, Cell Growth & Differentiation, vol.7, p.711, 1996.

H. Ruffner and I. M. Verma, BRCA1 is a cell cycle-regulated nuclear phosphoprotein, Proc Natl Acad Sci U S A, vol.94, pp.7138-7143, 1997.

F. Zheng, C. Yue, and G. Li, Nuclear AURKA acquires kinase-independent transactivating function to enhance breast cancer stem cell phenotype, Nature Communications, vol.7, 2016.

P. Cammareri, A. Scopelliti, and M. Todaro, Aurora-A Is Essential for the Tumorigenic Capacity and Chemoresistance of Colorectal Cancer Stem Cells, Cancer Res, vol.70, pp.4655-4665, 2010.

I. Chefetz, J. C. Holmberg, and A. B. Alvero, Inhibition of Aurora-A kinase induces cell cycle arrest in epithelial ovarian cancer stem cells by affecting NF?B pathway, Cell Cycle, vol.10, pp.2206-2214, 2011.

F. Zheng, Z. Long, and Z. Hou, A novel small molecule aurora kinase inhibitor attenuates breast tumor-initiating cells and overcomes drug resistance, Mol Cancer Ther, vol.13, 1991.

M. Mannino, N. Gomez-roman, H. Hochegger, and A. J. Chalmers, Differential sensitivity of Glioma stem cells to Aurora kinase A inhibitors: implications for stem cell mitosis and centrosome dynamics, Stem Cell Res, vol.13, pp.135-143, 2014.

J. Yang, T. Ikezoe, and C. Nishioka, CD34 + /CD38 ? acute myelogenous leukemia cells aberrantly express Aurora kinase A, Int J Cancer, vol.133, pp.2706-2719, 2013.

H. Clevers, The cancer stem cell: premises, promises and challenges, Nature Medicine, vol.17, pp.313-319, 2011.

A. Katsha, M. Soutto, and V. Sehdev, Aurora Kinase A Promotes Inflammation and Tumorigenesis in Mice and Human Gastric Neoplasia, Gastroenterology, vol.145, 2013.

T. Wei, P. Wu, and T. Wu, Aurora A and NF-?B Survival Pathway Drive Chemoresistance in Acute Myeloid Leukemia via the TRAF-Interacting Protein TIFA, Cancer Res, vol.77, pp.494-508, 2017.

K. Neben, A. Korshunov, and A. Benner, Microarray-based screening for molecular markers in medulloblastoma revealed STK15 as independent predictor for survival, Cancer Res, vol.64, pp.3103-3111, 2004.

I. Dimova, S. Raitcheva, and R. Dimitrov, Correlations between c-myc gene copy-number and clinicopathological parameters of ovarian tumours, European Journal of Cancer, vol.42, pp.674-679, 2006.

S. Lassmann, Y. Shen, and U. Jütting, Predictive value of Aurora-A/STK15 expression for late stage epithelial ovarian cancer patients treated by adjuvant chemotherapy, Clin Cancer Res, vol.13, pp.4083-4091, 2007.

T. Otto, S. Horn, and M. Brockmann, Stabilization of N-Myc is a critical function of Aurora A in human neuroblastoma, Cancer Cell, vol.15, pp.67-78, 2009.

H. Beltran, D. S. Rickman, and K. Park, Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets, Cancer Discov, vol.1, pp.487-495, 2011.

H. Yang, C. C. Ou, and R. I. Feldman, Aurora-A kinase regulates telomerase activity through c-Myc in human ovarian and breast epithelial cells, Cancer Res, vol.64, pp.463-467, 2004.

S. Yang, S. He, and X. Zhou, Suppression of Aurora-A oncogenic potential by c-Myc downregulation, Exp Mol Med, vol.42, pp.759-767, 2010.

A. A. Dar, A. Belkhiri, and W. El-rifai, The aurora kinase A regulates GSK-3beta in gastric cancer cells, Oncogene, vol.28, pp.866-875, 2009.

J. Hollander, S. Rimpi, and J. R. Doherty, Aurora kinases A and B are up-regulated by Myc and are essential for maintenance of the malignant state, Blood, vol.116, pp.1498-1505, 2010.

J. Wu, T. Chen, and C. Yu, Identification of V23RalA-Ser194 as a Critical Mediator for Aurora-A-induced Cellular Motility and Transformation by Small Pool Expression Screening, J Biol Chem, vol.280, pp.9013-9022, 2005.

K. Lim, D. C. Brady, and D. F. Kashatus, Aurora-A Phosphorylates, Activates, and Relocalizes the Small GTPase RalA, Molecular and Cellular Biology, vol.30, pp.508-523, 2010.

,

D. F. Kashatus, K. Lim, and D. C. Brady, RALA and RALBP1 regulate mitochondrial fission at mitosis, Nature Cell Biology, vol.13, pp.1108-1115, 2011.

L. Wang, L. De-oliveira, R. Wang, and C. , High-Throughput Functional Genetic and Compound Screens Identify Targets for Senescence Induction in, Cancer. Cell Rep, vol.21, pp.773-783, 2017.

L. Wang, J. Wang, and J. Chen, Aurora A Kinase Inhibitor AKI603 Induces Cellular Senescence in Chronic Myeloid Leukemia Cells Harboring T315I Mutation, Sci Rep, vol.6, p.35533, 2016.

F. Zheng, Z. Long, and Z. Hou, A Novel Small Molecule Aurora Kinase Inhibitor Attenuates Breast Tumor-Initiating Cells and Overcomes Drug Resistance, Mol Cancer Ther, vol.13, 1991.

Y. Liu, O. E. Hawkins, and A. E. Vilgelm, Combining an Aurora Kinase Inhibitor and a Death Receptor Ligand/Agonist Antibody Triggers Apoptosis in Melanoma Cells and Prevents Tumor Growth in Preclinical Mouse Models, Clin Cancer Res, vol.21, pp.5338-5348, 2015.

N. Tikhmyanova, J. L. Little, and E. A. Golemis, CAS proteins in normal and pathological cell growth control, Cellular and Molecular Life Sciences, vol.67, pp.1025-1048, 2010.

T. Do, X. F. Bickel, and L. E. , Aurora kinase A mediates epithelial ovarian cancer cell migration and adhesion, Oncogene, vol.33, pp.539-549, 2014.

M. Mahankali, K. M. Henkels, F. Speranza, and J. Gomez-cambronero, A non-mitotic role for Aurora kinase A as a direct activator of cell migration upon interaction with PLD, FAK and Src, J Cell Sci, vol.128, pp.516-526, 2015.

J. Wu and Y. L. Shan-y, AURKA promotes cell migration and invasion of head and neck squamous cell carcinoma through regulation of the AURKA/Akt/FAK signaling pathway, Oncol Lett, vol.11, pp.1889-1894, 2016.

L. Wang, J. Xiang, and M. Yan, The Mitotic Kinase Aurora-A Induces Mammary Cell Migration and Breast Cancer Metastasis by Activating the Cofilin-F-actin Pathway, Cancer Research, vol.70, pp.9118-9128, 2010.

G. Kanellos and M. C. Frame, Cellular functions of the ADF/cofilin family at a glance, Journal of Cell Science, vol.129, pp.3211-3218, 2016.

A. B. D'assoro, T. Liu, and C. Quatraro, The mitotic kinase Aurora-A promotes distant metastases by inducing epithelial-to-mesenchymal transition in ER?+ breast cancer cells, Oncogene, vol.33, pp.599-610, 2014.

J. Wang, K. Nikhil, and K. Viccaro, Phosphorylation-dependent regulation of ALDH1A1 by Aurora kinase A: insights on their synergistic relationship in pancreatic cancer, BMC Biol, vol.15, issue.10, 2017.

J. Wang, K. Nikhil, and K. Viccaro, The Aurora-A-Twist1 axis promotes highly aggressive phenotypes in pancreatic carcinoma, J Cell Sci, vol.130, pp.1078-1093, 2017.

A. Dongre and R. A. Weinberg, New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer, Nat Rev Mol Cell Biol, vol.20, pp.69-84, 2019.

F. Wang, H. Li, and X. Yan, Alisertib induces cell cycle arrest and autophagy and suppresses epithelial-to-mesenchymal transition involving PI3K/Akt/mTOR and sirtuin 1-mediated signaling pathways in human pancreatic cancer cells, Drug Des Devel Ther, vol.9, pp.575-601, 2015.

R. Grant, A. Abdelbaki, and A. Bertoldi, Constitutive regulation of mitochondrial morphology by Aurora A kinase depends on a predicted cryptic targeting sequence at the N-terminus, Open Biology, vol.8, p.170272, 2018.

C. Chang and C. Blackstone, Dynamic regulation of mitochondrial fission through modification of the dynamin-related protein Drp1: Chang & Blackstone, Annals of the New York Academy of Sciences, vol.1201, pp.34-39, 2010.

P. Kaestner, A. Stolz, and H. Bastians, Determinants for the efficiency of anticancer drugs targeting either Aurora-A or Aurora-B kinases in human colon carcinoma cells, Mol Cancer Ther, vol.8, pp.2046-2056, 2009.

C. T. Durlacher, Z. Li, and X. Chen, An update on the pharmacokinetics and pharmacodynamics of alisertib, a selective Aurora kinase A inhibitor, Clinical and Experimental Pharmacology and Physiology, vol.43, pp.585-601, 2016.

M. G. Manfredi, J. A. Ecsedy, and A. Chakravarty, Characterization of Alisertib (MLN8237), an investigational small-molecule inhibitor of aurora A kinase using novel in vivo pharmacodynamic assays, Clin Cancer Res, vol.17, pp.7614-7624, 2011.

K. R. Kelly, J. Ecsedy, and E. Medina, The novel Aurora A kinase inhibitor MLN8237 is active in resistant chronic myeloid leukaemia and significantly increases the efficacy of nilotinib, J Cell Mol Med, vol.15, pp.2057-2070, 2011.

N. Zhou, K. Singh, and M. C. Mir, The investigational Aurora kinase A inhibitor MLN8237 induces defects in cell viability and cell-cycle progression in malignant bladder cancer cells in vitro and in vivo, Clin Cancer Res, vol.19, pp.1717-1728, 2013.

T. Shimomura, S. Hasako, and Y. Nakatsuru, MK-5108, a Highly Selective Aurora-A Kinase Inhibitor, Shows Antitumor Activity Alone and in Combination with Docetaxel, Molecular Cancer Therapeutics, vol.9, pp.157-166, 2010.

W. Shan, P. Y. Akinfenwa, and K. B. Savannah, A Small-Molecule Inhibitor Targeting the Mitotic Spindle Checkpoint Impairs the Growth of Uterine Leiomyosarcoma, Clinical Cancer Research, vol.18, pp.3352-3365, 2012.

V. Tagal, S. Wei, and W. Zhang, SMARCA4-inactivating mutations increase sensitivity to Aurora kinase A inhibitor VX-680 in non-small cell lung cancers, Nat Commun, vol.8, 2017.

M. Amin, S. E. Minton, and P. M. Lorusso, A phase I study of MK-5108, an oral aurora a kinase inhibitor, administered both as monotherapy and in combination with docetaxel, in patients with advanced or refractory solid tumors, Invest New Drugs, vol.34, pp.84-95, 2016.

G. C. Fletcher, R. D. Brokx, and T. A. Denny, ENMD-2076 Is an Orally Active Kinase Inhibitor with Antiangiogenic and Antiproliferative Mechanisms of Action, Molecular Cancer Therapeutics, vol.10, pp.126-137, 2011.

K. Shiomitsu, E. Sajo, C. Rubin, and I. Sehgal, The radiosensitizing effect of the aurora kinase inhibitors, ENMD-2076, on canine mast cell tumours in vitro: Radiosensitizing effect of ENMD-2076, Vet Comp Oncol, vol.14, pp.13-27, 2016.

K. Yee, H. Chen, and D. W. Hedley, A phase I trial of the aurora kinase inhibitor, ENMD-2076, in patients with relapsed or refractory acute myeloid leukemia or chronic myelomonocytic leukemia, Invest New Drugs, vol.34, pp.614-624, 2016.

A. Mazumdar, Y. C. Henderson, and A. K. El-naggar, Aurora kinase A inhibition and paclitaxel as targeted combination therapy for head and neck squamous cell carcinoma, Head Neck, vol.31, pp.625-634, 2009.

Y. Lin, F. M. Richards, and B. Krippendorff, Paclitaxel and CYC3, an aurora kinase A inhibitor, synergise in pancreatic cancer cells but not bone marrow precursor cells, Br J Cancer, vol.107, pp.1692-1701, 2012.

V. Sehdev, A. Katsha, and J. Ecsedy, The combination of alisertib, an investigational Aurora kinase A inhibitor, and docetaxel promotes cell death and reduces tumor growth in preclinical cell models of upper gastrointestinal adenocarcinomas: Alisertib & Docetaxel Inhibit Tumor Growth, Cancer, vol.119, pp.904-914, 2013.

W. Qi, L. S. Cooke, and X. Liu, Aurora inhibitor MLN8237 in combination with docetaxel enhances apoptosis and anti-tumor activity in mantle cell lymphoma, Biochemical Pharmacology, vol.81, pp.881-890, 2011.

E. Caputo, R. Miceli, and M. L. Motti, AurkA inhibitors enhance the effects of B-RAF and MEK inhibitors in melanoma treatment, J Transl Med, vol.12, p.216, 2014.

G. Pathria, B. Garg, and V. Borgdorff, Overcoming MITF-conferred drug resistance through dual AURKA/MAPK targeting in human melanoma cells, Cell Death Dis, vol.7, 2016.

K. N. Shah, R. Bhatt, and J. Rotow, Aurora kinase A drives the evolution of resistance to thirdgeneration EGFR inhibitors in lung cancer, Nat Med, vol.25, pp.111-118, 2019.

K. Sumi, K. Tago, T. Kasahara, and M. Funakoshi-tago, Aurora kinase A critically contributes to the resistance to anti-cancer drug cisplatin in JAK2 V617F mutant-induced transformed cells, FEBS Lett, vol.585, pp.1884-1890, 2011.

A. P. Damodaran, L. Vaufrey, O. Gavard, and C. Prigent, Aurora A Kinase Is a Priority Pharmaceutical Target for the Treatment of Cancers, Trends in Pharmacological Sciences, vol.38, pp.687-700, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01578074

I. A. Asteriti, F. Daidone, and G. Colotti, Identification of small molecule inhibitors of the Aurora-A/TPX2 complex, Oncotarget, vol.8, 2017.

Y. Kong, A. Bender, and A. Yan, Identification of Novel Aurora Kinase A (AURKA) Inhibitors via Hierarchical Ligand-Based Virtual Screening, J Chem Inf Model, vol.58, pp.36-47, 2018.

L. Schermelleh, A. Ferrand, and T. Huser, Super-resolution microscopy demystified, Nature Cell Biology, vol.21, p.72, 2019.

Y. Wu and H. Shroff, Faster, sharper, and deeper: structured illumination microscopy for biological imaging, Nature Methods, vol.15, p.1011, 2018.

R. Heintzmann and T. Huser, Super-Resolution Structured Illumination Microscopy, vol.25, 2017.

P. Dedecker, G. Mo, T. Dertinger, and J. Zhang, Widely accessible method for superresolution fluorescence imaging of living systems, PNAS, vol.109, pp.10909-10914, 2012.

,

T. Dertinger, R. Colyer, and G. Iyer, Fast, background-free, 3D super-resolution optical fluctuation imaging (SOFI), PNAS, vol.106, pp.22287-22292, 2009.

G. Mo, B. Ross, and F. Hertel, Genetically encoded biosensors for visualizing live-cell biochemical activity at super-resolution, Nature Methods, vol.14, pp.427-434, 2017.

,

I. Pérez-de-castro, M. Carmena, C. Prigent, and D. M. Glover, Aurora Kinases: Classical Mitotic Roles, Non-canonical Functions and Translational Views, Frontiers Media SA, 2017.

A. Piljic and C. Schultz, Simultaneous Recording of Multiple Cellular Events by FRET, ACS Chem Biol, vol.3, pp.156-160, 2008.

D. M. Grant, W. Zhang, and E. J. Mcghee, Multiplexed FRET to Image Multiple Signaling Events in Live Cells, Biophys J, vol.95, pp.69-71, 2008.

C. Demeautis, F. Sipieter, and J. Roul, Multiplexing PKA and ERK1&2 kinases FRET biosensors in living cells using single excitation wavelength dual colour FLIM, Scientific Reports, vol.7, p.41026, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01478382

B. L. Ross, B. Tenner, and M. L. Markwardt, Single-color, ratiometric biosensors for detecting signaling activities in live cells, vol.7, 2018.