M. Hengartner, The biochemistry of apoptosis, Nature, vol.407, issue.6805, pp.770-776, 2000.
DOI : 10.1038/35037710

C. Thompson, Apoptosis in the pathogenesis and treatment of disease, Science, vol.267, issue.5203, pp.1456-1462, 1995.
DOI : 10.1126/science.7878464

J. Reed, APOPTOSIS-BASED THERAPIES, Nature Reviews Drug Discovery, vol.1, issue.2, pp.111-121, 2002.
DOI : 10.1038/nrd726

B. Bell, E. Scheer, and T. L. , Identification of hTAFII80?? Links Apoptotic Signaling Pathways to Transcription Factor TFIID Function, Molecular Cell, vol.8, issue.3, pp.591-600, 2001.
DOI : 10.1016/S1097-2765(01)00325-2

E. Wilhelm, F. Pellay, A. Benecke, and B. Bell, TAF6?? Controls Apoptosis and Gene Expression in the Absence of p53, PLoS ONE, vol.12, issue.7, p.2721, 2008.
DOI : 10.1371/journal.pone.0002721.s005

B. Bell and T. L. , Regulation of Gene Expression by Multiple Forms of TFIID and Other Novel TAFII-Containing Complexes, Experimental Cell Research, vol.246, issue.1, pp.11-19, 1999.
DOI : 10.1006/excr.1998.4294

M. Green, TBP-associated factors (TAFIIs): multiple, selective transcriptional mediators in common complexes, Trends in Biochemical Sciences, vol.25, issue.2, pp.59-63, 2000.
DOI : 10.1016/S0968-0004(99)01527-3

F. Muller, M. Demeny, and T. L. , New Problems in RNA Polymerase II Transcription Initiation: Matching the Diversity of Core Promoters with a Variety of Promoter Recognition Factors, Journal of Biological Chemistry, vol.282, issue.20, pp.14685-14689, 2007.
DOI : 10.1074/jbc.R700012200

URL : https://hal.archives-ouvertes.fr/hal-00189974

R. Weinzierl, S. Ruppert, B. Dynlacht, N. Tanese, and R. Tjian, Cloning and expression of Drosophila TAFII60 and human TAFII70 reveal conserved interactions with other subunits of TFIID, Embo J, vol.12, issue.13, pp.5303-5309, 1993.

K. Wright, M. Marr, and R. Tjian, TAF4 nucleates a core subcomplex of TFIID and mediates activated transcription from a TATA-less promoter, Proceedings of the National Academy of Sciences, vol.103, issue.33, pp.12347-12352, 2006.
DOI : 10.1073/pnas.0605499103

W. Selleck, R. Howley, Q. Fang, V. Podolny, M. Fried et al., A histone fold TAF octamer within the yeast TFIID transcriptional coactivator, Nature Structural Biology, vol.8, issue.8, pp.695-700, 2001.
DOI : 10.1038/90408

B. Michel, P. Komarnitsky, and S. Buratowski, Histone-like TAFs Are Essential for Transcription In Vivo, Molecular Cell, vol.2, issue.5, pp.663-673, 1998.
DOI : 10.1016/S1097-2765(00)80164-1

K. Hisatake, T. Ohta, R. Takada, M. Guermah, M. Horikoshi et al., Evolutionary conservation of human TATA-bindingpolypeptide-associated factors TAFII31 and TAFII80 and interactions of TAFII80 with other TAFs and with general transcription factors, Proc Natl Acad Sci, issue.18, pp.928195-8199, 1995.

X. Xie, T. Kokubo, S. Cohen, U. Mirza, A. Hoffmann et al., Structural similarity between TAFs and the heterotetrameric core of the histone octamer, Nature, vol.380, issue.6572, pp.380316-322, 1996.
DOI : 10.1038/380316a0

Z. Chen and J. Manley, In Vivo Functional Analysis of the Histone 3-like TAF9 and a TAF9-related Factor, TAF9L, Journal of Biological Chemistry, vol.278, issue.37, pp.27835172-83, 2003.
DOI : 10.1074/jbc.M304241200

M. Frontini, E. Soutoglou, M. Argentini, C. Bole-feysot, B. Jost et al., TAF9b (Formerly TAF9L) Is a Bona Fide TAF That Has Unique and Overlapping Roles with TAF9, Molecular and Cellular Biology, vol.25, issue.11, pp.254638-4649, 2005.
DOI : 10.1128/MCB.25.11.4638-4649.2005

URL : https://hal.archives-ouvertes.fr/hal-00187538

C. Thut, J. Chen, R. Klemm, and R. Tjian, p53 transcriptional activation mediated by coactivators TAFII40 and TAFII60, Science, vol.267, issue.5194, pp.53100-104, 1995.
DOI : 10.1126/science.7809597

W. Liu, R. Coleman, E. Ma, P. Grob, J. Yang et al., Structures of three distinct activator-TFIID complexes, Genes & Development, vol.23, issue.13, pp.1510-1521, 2009.
DOI : 10.1101/gad.1790709

G. Farmer, J. Colgan, Y. Nakatani, J. Manley, and C. Prives, Functional interaction between p53, the TATA-binding protein (TBP), andTBP-associated factors in vivo., Molecular and Cellular Biology, vol.16, issue.8, pp.164295-4304, 1996.
DOI : 10.1128/MCB.16.8.4295

G. Jimenez, M. Nister, J. Stommel, M. Beeche, E. Barcarse et al., A transactivation-deficient mouse model provides insights into Trp53 regulation and function, Nat Genet, vol.26, issue.1, pp.37-43, 2000.

G. Gill, Death Signals Changes in TFIID, Molecular Cell, vol.8, issue.3, pp.482-484, 2001.
DOI : 10.1016/S1097-2765(01)00338-0

S. Aviel-ronen, B. Coe, S. Lau, G. Da-cunha-santos, C. Zhu et al., Genomic markers for malignant progression in pulmonary adenocarcinoma with bronchioloalveolar features, Proceedings of the National Academy of Sciences, vol.105, issue.29, pp.10510155-10160, 2008.
DOI : 10.1073/pnas.0709618105

J. Campbell, W. Lockwood, T. Buys, R. Chari, B. Coe et al., Integrative genomic and gene expression analysis of chromosome 7 identified novel oncogene loci in non-small cell lung cancer, Genome, issue.12, pp.511032-1039, 2008.

H. Dressman, C. Hans, A. Bild, J. Olson, E. Rosen et al., Gene Expression Profiles of Multiple Breast Cancer Phenotypes and Response to Neoadjuvant Chemotherapy, Clinical Cancer Research, vol.12, issue.3, pp.819-826, 2006.
DOI : 10.1158/1078-0432.CCR-05-1447

W. Wang, R. Nahta, G. Huper, and J. Marks, TAFII70 isoform-specific growth suppression correlates with its ability to complex with the GADD45a protein, Mol Cancer Res, vol.2, issue.8, pp.442-452, 2004.

E. Wilhelm, F. Pellay, A. Benecke, and B. Bell, Determining the impact of alternative splicing events on transcriptome dynamics, BMC Research Notes, vol.1, issue.1, p.94, 2008.
DOI : 10.1186/1756-0500-1-94

V. Batagelj, A. Mrvar, M. Jünger, and S. Leipert, Pajek??? Analysis and Visualization of Large Networks, Graph Drawing: 9th International Symposium, pp.477-478, 2001.
DOI : 10.1007/3-540-45848-4_54

URL : http://citeseerx.ist.psu.edu/viewdoc/summary?doi=10.1.1.108.5239

E. Oda, R. Ohki, H. Murasawa, J. Nemoto, T. Shibue et al., Noxa, a BH3-Only Member of the Bcl-2 Family and Candidate Mediator of p53-Induced Apoptosis, Science, vol.288, issue.5468, pp.2881053-1058, 2000.
DOI : 10.1126/science.288.5468.1053

S. Jarriault, C. Brou, F. Logeat, E. Schroeter, R. Kopan et al., Signalling downstream of activated mammalian Notch, Nature, vol.377, issue.6547, pp.355-358, 1995.
DOI : 10.1038/377355a0

L. Nikitenko, S. Fox, S. Kehoe, M. Rees, and R. Bicknell, Adrenomedullin and tumour angiogenesis, British Journal of Cancer, vol.282, issue.1, pp.1-7, 2006.
DOI : 10.1016/S0167-0115(03)00037-5

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2361077

J. Zhou, Y. Liu, and G. Wu, The Role of Mitogen-Activated Protein Kinase Phosphatase-1 in Oxidative Damage-Induced Cell Death, Cancer Research, vol.66, issue.9, pp.4888-4894, 2006.
DOI : 10.1158/0008-5472.CAN-05-4229

Y. Liu, J. Wang, J. Guo, J. Wu, and H. Lieberman, DUSP1 Is Controlled by p53 during the Cellular Response to Oxidative Stress, Molecular Cancer Research, vol.6, issue.4, pp.624-633, 2008.
DOI : 10.1158/1541-7786.MCR-07-2019

M. Li, J. Zhou, Y. Ge, L. Matherly, and G. Wu, The Phosphatase MKP1 Is a Transcriptional Target of p53 Involved in Cell Cycle Regulation, Journal of Biological Chemistry, vol.278, issue.42, pp.27841059-41068, 2003.
DOI : 10.1074/jbc.M307149200

T. Miyashita and J. Reed, Tumor suppressor p53 is a direct transcriptional activator of the human bax gene, Cell, vol.80, issue.2, pp.293-299, 1995.

P. Hwang, F. Bunz, J. Yu, C. Rago, T. Chan et al., Ferredoxin reductase affects p53-dependent, 5- fluorouracil-induced apoptosis in colorectal cancer cells, Nature Medicine, vol.7, issue.10, pp.1111-1117, 2001.
DOI : 10.1038/nm1001-1111

G. Liu and X. Chen, The ferredoxin reductase gene is regulated by the p53 family and sensitizes cells to oxidative stress-induced apoptosis, Oncogene, vol.21, issue.47, pp.7195-7204, 2002.
DOI : 10.1038/sj.onc.1205862

J. Yu, L. Zhang, P. Hwang, and K. Kinzler, PUMA Induces the Rapid Apoptosis of Colorectal Cancer Cells, Molecular Cell, vol.7, issue.3, pp.673-682, 2001.
DOI : 10.1016/S1097-2765(01)00213-1

L. Phng and H. Gerhardt, Angiogenesis: A Team Effort Coordinated by Notch, Developmental Cell, vol.16, issue.2, pp.196-208, 2009.
DOI : 10.1016/j.devcel.2009.01.015

G. Serini, D. Valdembri, and F. Bussolino, Integrins and angiogenesis: A sticky business, Experimental Cell Research, vol.312, issue.5, pp.651-658, 2006.
DOI : 10.1016/j.yexcr.2005.10.020

R. Kerbel, Tumor Angiogenesis, New England Journal of Medicine, vol.358, issue.19, pp.2039-2049, 2008.
DOI : 10.1056/NEJMra0706596

T. Burke and J. Kadonaga, The downstream core promoter element, DPE, is conserved from Drosophila to humans and is recognized by TAFII60 of??Drosophila, Genes & Development, vol.11, issue.22, pp.3020-3031, 1997.
DOI : 10.1101/gad.11.22.3020

H. Shao, M. Revach, S. Moshonov, Y. Tzuman, K. Gazit et al., Core Promoter Binding by Histone-Like TAF Complexes, Molecular and Cellular Biology, vol.25, issue.1, pp.206-219, 2005.
DOI : 10.1128/MCB.25.1.206-219.2005

D. Mercatante, C. Bortner, J. Cidlowski, and R. Kole, Modification of alternative splicing of Bcl-x pre-mRNA in prostate and breast cancer Wilhelm et al analysis of apoptosis and cell death, BMC Molecular Biology J Biol Chem, vol.11, issue.1019, pp.27616411-16417, 2001.

C. Gaiddon, N. Moorthy, and C. Prives, Ref-1 regulates the transactivation and pro-apoptotic functions of p53 in vivo, The EMBO Journal, vol.18, issue.20, pp.5609-5621, 1999.
DOI : 10.1093/emboj/18.20.5609

T. Ishimitsu, A. Miyata, H. Matsuoka, and K. Kangawa, Transcriptional Regulation of Human Adrenomedullin Gene in Vascular Endothelial Cells, Biochemical and Biophysical Research Communications, vol.243, issue.2, pp.463-470, 1998.
DOI : 10.1006/bbrc.1998.8110

P. Hinds, C. Finlay, R. Quartin, S. Baker, E. Fearon et al., Mutant p53 DNA clones from human colon carcinomas cooperate with ras in transforming primary rat cells: a comparison of the " hot spot " mutant phenotypes, Cell Growth Differ, vol.1, issue.12, pp.571-580, 1990.

M. Thomas, A. Kalita, S. Labrecque, D. Pim, L. Banks et al., Two Polymorphic Variants of Wild-Type p53 Differ Biochemically and Biologically, Molecular and Cellular Biology, vol.19, issue.2, pp.1092-1100, 1999.
DOI : 10.1128/MCB.19.2.1092

C. Brou, J. Wu, S. Ali, E. Scheer, C. Lang et al., by the acidic transactivator GAL-VP16 and the two nonacidic activation functions of the estrogen receptor, Nucleic Acids Research, vol.21, issue.1, pp.5-12, 1993.
DOI : 10.1093/nar/21.1.5

S. Noth, G. Brysbaert, and A. Benecke, Normalization Using Weighted Negative Second Order Exponential Error Functions (NeONORM) Provides Robustness Against Asymmetries in Comparative Transcriptome Profiles and Avoids False Calls, Genomics, Proteomics & Bioinformatics, vol.4, issue.2, pp.90-109, 2006.
DOI : 10.1016/S1672-0229(06)60021-1

S. Noth and A. Benecke, Avoiding inconsistencies over time and tracking difficulties in Applied Biosystems AB1700/Panther probe-to-gene annotations, BMC Bioinformatics, vol.6, issue.1, p.307, 2005.
DOI : 10.1186/1471-2105-6-307

. Wilhelm, TAF6?? orchestrates an apoptotic transcriptome profile and interacts functionally with p53, BMC Molecular Biology, vol.11, issue.1, p.10, 2010.
DOI : 10.1186/1471-2199-11-10

URL : https://hal.archives-ouvertes.fr/inserm-00663537