R. Abbotts and S. Madhusudan, Human AP endonuclease 1 (APE1): From mechanistic insights to druggable target in cancer, Cancer Treat. Rev, vol.36, pp.425-435, 2010.

R. Abbotts and D. M. Wilson, Coordination of DNA single strand break repair. Free Radic, Biol. Med, vol.107, pp.228-244, 2017.

A. Adegbola, L. Musante, B. Callewaert, P. Maciel, H. Hu et al., Redefining the MED13L syndrome, Eur. J. Hum. Genet, vol.23, pp.1308-1317, 2015.

K. Adelman and J. Lis, Promoter-proximal pausing of RNA polymerase II: emerging roles on metazoans, Nat. Rev. Genet, vol.13, pp.720-731, 2012.

S. Akoulitchev, S. Chuikov, and D. Reinberg, TFIIH is negatively regulated by cdk8-containing mediator complexes, Nature, vol.407, pp.102-106, 2000.

N. L. Alderson, Y. Wang, M. Blatnik, N. Frizzell, M. D. Walla et al., S-(2-Succinyl)cysteine: A novel chemical modification of tissue proteins by a Krebs cycle intermediate, Arch. Biochem. Biophys, vol.450, pp.1-8, 2006.

B. L. Allen and D. J. Taatjes, The Mediator complex: A central integrator of transcription, Nat. Rev. Mol. Cell Biol, vol.16, pp.155-166, 2015.

R. Amouroux, A. Campalans, B. Epe, and J. P. Radicella, Oxidative stress triggers the preferential assembly of base excision repair complexes on open chromatin regions, Nucleic Acids Res, vol.38, pp.2878-2890, 2010.

G. Antoniali, L. Lirussi, C. Ambrosio, F. Piaz, C. Vascotto et al., SIRT1 gene expression upon genotoxic damage is regulated by APE1 through nCaRE-promoter elements, Mol. Biol. Cell, vol.25, pp.532-547, 2014.

L. Aragon, E. Martinez-perez, and M. Merkenschlager, Condensin, cohesin and the control of chromatin states, Curr. Opin. Genet. Dev, vol.23, pp.204-211, 2013.

T. Arai, V. P. Kelly, O. Minowa, T. Noda, and S. Nishimura, High accumulation of oxidative DNA damage , 8-hydroxyguanine, in Mmh / Ogg1 deficient mice by chronic oxidative stress, Carcinogenesis, vol.23, pp.2005-2010, 2002.

T. Arai, V. P. Kelly, O. Minowa, T. Noda, and S. Nishimura, The study using wild-type and Ogg1 knockout mice exposed to potassium bromate shows no tumor induction despite an extensive accumulation of 8-hydroxyguanine in kidney DNA, Toxicology, vol.221, pp.179-186, 2006.

G. Arents and E. N. Moudrianakis, The histone fold: A ubiquitous architectural motif utilized in DNA compaction and protein dimerization, Proc. Natl. Acad. Sci. U. S. A, vol.92, pp.11170-11174, 1995.

M. Audebert, S. Chevillard, C. Levalois, G. Gyapay, A. Vieillefond et al., Alterations of the DNA repair gene OGG1 in human clear cell carcinomas of the kidney, Cancer Res, vol.60, pp.4740-4744, 2000.

D. Ballmaier and B. Epe, DNA damage by bromate: Mechanism and consequences, Toxicology, vol.221, pp.166-171, 2006.

J. Bancerek, Z. C. Poss, I. Steinparzer, V. Sedlyarov, T. Pfaffenwimmer et al., CDK8 Kinase Phosphorylates Transcription Factor STAT1 to Selectively Regulate the Interferon Response, Immunity, vol.38, pp.250-262, 2013.

C. Barbieri, S. Baca, M. Lawrence, F. Demichelis, M. Blattner et al., Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer, Nat. Genet, vol.44, pp.685-689, 2012.

C. Barrington, R. Finn, and S. Hadjur, Cohesin biology meets the loop extrusion model, Chromosom. Res, vol.25, pp.51-60, 2017.

N. C. Bauer, A. H. Corbett, and P. W. Doetsch, The current state of eukaryotic DNA base damage and repair, Nucleic Acids Res, vol.43, pp.10083-10101, 2015.

M. Baumann, J. Pontiller, and W. Ernst, Structure and basal transcription complex of RNA Polymerase II core promoters in the mammalian genome: An overview, Mol. Biotechnol, vol.45, pp.241-247, 2010.

J. Bednar, R. A. Horowitz, S. A. Grigoryev, L. M. Carruthers, J. C. Hansen et al., Nucleosomes, linker DNA, and linker histone form a unique structural motif that directs the higher-order folding and compaction of chromatin, Proc. Natl. Acad. Sci. U. S. A, vol.95, pp.14173-14178, 1998.

K. K. Bhakat, S. K. Mokkapati, I. Boldogh, T. K. Hazra, and S. Mitra, Acetylation of Human 8-Oxoguanine-DNA Glycosylase by p300 and Its Role in 8-Oxoguanine Repair In Vivo, Mol. Cell. Biol, vol.26, pp.1654-1665, 2006.

E. Birben, U. Murat, S. Md, C. Sackesen, S. Erzurum et al., Oxidative Stress and Antioxidant Defense, WAO J, vol.5, pp.9-19, 2012.

B. Biterge and R. Schneider, Histone variants: Key players of chromatin, Cell Tissue Res, vol.356, pp.457-466, 2014.

M. Bjørås, E. Seeberg, L. Luna, L. H. Pearl, and T. E. Barrett, Reciprocal "flipping" underlies substrate recognition and catalytic activation by the human 8-oxo-guanine DNA glycosylase, J. Mol. Biol, vol.317, pp.171-177, 2002.

K. R. Blahnik, L. Dou, L. Echipare, S. Iyengar, H. O'geen et al., Characterization of the Contradictory Chromatin Signatures at the 3? exons of Zinc finger genes, PLoS One, vol.6, p.17121, 2011.

P. C. Blainey, A. M. Van-oijen, A. Banerjee, G. L. Verdine, and X. S. Xie, A base-excision DNArepair protein finds intrahelical lesion bases by fast sliding in contact with DNA, Proc. Natl. Acad. Sci. U. S. A, vol.103, pp.5752-5757, 2006.

H. Blons, J. P. Radicella, O. Laccourreye, D. Brasnu, P. Beaune et al., Frequent allelic loss at chromosome 3p distinct from genetic alterations of the 8-oxoguanine DNA glycosylase 1 gene in head and neck cancer, Mol. Carcinog, vol.26, pp.254-260, 1999.

A. Bobba, G. Amadoro, D. Valenti, V. Corsetti, R. Lassandro et al., Mitochondrial respiratory chain Complexes I and IV are impaired by ?-amyloid via direct interaction and through Complex I-dependent ROS production, respectively. Mitochondrion, vol.13, pp.298-311, 2013.

S. Boeing, C. Rigault, M. Heidemann, D. Eick, and M. Meisterernst, RNA polymerase II Cterminal heptarepeat domain Ser-7 phosphorylation is established in a mediator-dependent fashion, J. Biol. Chem, vol.285, pp.188-196, 2010.

S. Boiteux, F. Coste, and B. Castaing, Repair of 8-oxo-7 , 8-dihydroguanine in prokaryotic and eukaryotic cells : Properties and biological roles of the Fpg and OGG1 DNA N-glycosylases. Free Radic, Biol. Med, vol.107, pp.179-201, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01618166

S. Boiteux and J. P. Radicella, Base excision repair of 8-hydroxyguanine protects DNA from endogenous oxidative stress, Biochimie, vol.81, pp.59-67, 1999.

H. Bourbon, Comparative genomics supports a deep evolutionary origin for the large, fourmodule transcriptional mediator complex, Nucleic Acids Res, vol.36, pp.3993-4008, 2008.

A. Bravard, M. Vacher, B. Gouget, A. Coutant, F. H. De-boisferon et al., Redox Regulation of Human OGG1 Activity in Response to Cellular Oxidative Stress, Mol. Cell. Biol, vol.26, pp.7430-7436, 2006.

A. Bravard, M. Vacher, E. Moritz, L. Vaslin, J. Hall et al., Oxidation status of human ogg1-s326c polymorphic variant determines cellular DNA repair capacity, Cancer Res, vol.69, pp.3642-3649, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00413809

R. Brem and J. Hall, XRCC1 is required for DNA single-strand break repair in human cells, Nucleic Acids Res, vol.33, pp.2512-2520, 2005.

A. Brooker and K. Berkowitz, The roles of cohesins in mitosis, meiosis, and human health and disease, Methods Mol Biol, vol.1170, pp.229-66, 2014.

S. D. Bruner, D. P. Norman, and G. L. Verdine, Structural basis for recognition and repair of the endogenous mutagen 8-oxoguanine in DNA, Nature, vol.403, pp.859-866, 2000.

H. Budworth, F. R. Harris, P. Williams, D. Y. Lee, A. Holt et al., Suppression of Somatic Expansion Delays the Onset of Pathophysiology in a Mouse Model of Huntington's Disease, PLoS Genet, vol.11, pp.1-22, 2015.

B. Bukowska and B. T. Karwowski, Actual state of knowledge in the field of diseases related with defective nucleotide excision repair, Life Sci, vol.195, pp.6-18, 2018.

S. F. Bunting and A. Nussenzweig, End-joining, translocations and cancer, Nat. Rev. Cancer, vol.13, pp.443-454, 2013.

R. C. Burgess, B. Burman, M. J. Kruhlak, and T. Misteli, Activation of DNA Damage Response Signaling by Condensed Chromatin, Cell Rep, vol.9, pp.1703-1718, 2014.

G. A. Busslinger, R. R. Stocsits, P. Van-der-lelij, E. Axelsson, A. Tedeschi et al., Cohesin is positioned in mammalian genomes by transcription, CTCF and Wapl, Nature, vol.544, pp.503-507, 2017.

J. Cadet, T. Delatour, T. Douki, D. Gasparutto, J. P. Pouget et al., Hydroxyl radicals and DNA base damage, Mutat. Res. -Fundam. Mol. Mech. Mutagen, vol.424, pp.9-21, 1999.

K. W. Caldecott, XRCC1 and DNA strand break repair, DNA Repair (Amst), vol.2, pp.955-969, 2003.

K. W. Caldecott, DNA single-strand break repair and spinocerebellar ataxia, Cell, vol.112, pp.7-10, 2003.

K. W. Caldecott, DNA single-strand break repair, Exp. Cell Res, vol.329, pp.2-8, 2014.

K. W. Caldecott, C. K. Mckeown, J. D. Tucker, S. Ljungquist, and L. H. Thompson, An interaction between the mammalian DNA repair protein XRCC1 and DNA ligase III, Mol. Cell. Biol, vol.14, pp.68-76, 1994.

A. Campalans, R. Amouroux, A. Bravard, B. Epe, and J. P. Radicella, UVA irradiation induces relocalisation of the DNA repair protein hOGG1 to nuclear speckles, J. Cell Sci, vol.120, pp.23-32, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00169632

A. Campalans, T. Kortulewski, R. Amouroux, H. Menoni, W. Vermeulen et al., Distinct spatiotemporal patterns and PARP dependence of XRCC1 recruitment to single-strand break and base excision repair, Nucleic Acids Res, vol.41, pp.3115-3129, 2013.

A. Campalans, S. Marsin, Y. Nakabeppu, T. R. O'connor, S. Boiteux et al., XRCC1 interactions with multiple DNA glycosylases: A model for its recruitment to base excision repair, DNA Repair (Amst), vol.4, pp.826-835, 2005.

A. Campalans, E. Moritz, T. Kortulewski, D. Biard, B. Epe et al., Interaction with OGG1 Is Required for Efficient Recruitment of XRCC1 to Base Excision Repair and Maintenance of Genetic Stability after Exposure to Oxidative Stress, Mol. Cell. Biol, vol.35, pp.1648-1658, 2015.

S. Canudas and S. Smith, Differential regulation of telomere and centromere cohesion by the Scc3 homologues SA1 and SA2, respectively, in human cells, J. Cell Biol, vol.187, pp.165-173, 2009.

M. Cevhler, Y. Shi, D. Li, C. Bt, S. Malik et al., Reconstitution of active human core Mediator complex reveals a pivotal role of the MED14 subunit, Nat Struc Mol Biol, vol.21, pp.1028-1034, 2014.

W. C. Chao, Y. Murayama, S. Muñoz, A. Costa, F. Uhlmann et al., Structural Studies Reveal the Functional Modularity of the Scc2-Scc4 Cohesin Loader, Cell Rep, vol.12, pp.719-725, 2015.

C. Richard, J. L. , M. S. Shukla, H. Menoni, K. Ouararhni et al., FACT Assists Base Excision Repair by Boosting the Remodeling Activity of RSC, PLoS Genet, vol.12, pp.1-21, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-02163206

N. Charlet-berguerand, S. Feuerhahn, S. E. Kong, H. Ziserman, J. W. Conaway et al., RNA polymerase II bypass of oxidative DNA damage is regulated by transcription elongation factors, EMBO J, vol.25, pp.5481-5491, 2006.
URL : https://hal.archives-ouvertes.fr/hal-00187859

L. Chen, A. Elahi, J. Pow-sang, P. Lazarus, and J. Park, Association between polymorphism of human oxoguanine glycosylase 1 and risk of prostate cancer, J Urol, vol.170, pp.2471-2475, 2003.

S. Chevillard, J. P. Radicella, C. Levalois, J. Lebeau, M. F. Poupon et al., Mutations in OGG1, a gene involved in the repair of oxidative DNA damage, are found in human lung and kidney tumours, Oncogene, vol.16, pp.3083-3089, 1998.

T. Chiang, F. Duncan, K. Schindler, R. Schultz, and M. Lampson, Evidence that weakened centromere cohesion is a leading cause of age-related aneuploidy in ovocytes, Curr. Biol, vol.20, pp.1522-1530, 2010.

R. Chien, W. Zeng, S. Kawauchi, M. A. Bender, R. Santos et al., Cohesin mediates chromatin interactions that regulate mammalian ?-globin expression, J. Biol. Chem, vol.286, pp.17870-17878, 2011.

E. S. Chrun, F. Modolo, and F. I. Daniel, Histone modifications: A review about the presence of this epigenetic phenomenon in carcinogenesis, Pathol. Res. Pract, vol.213, pp.1329-1339, 2017.

D. F. Church and W. A. Pryor, Free-radical chemistry of cigarette smoke and its toxicological implications, Environ. Health Perspect, vol.64, pp.111-126, 1985.

A. Ciccia and S. J. Elledge, The DNA Damage Response: Making It Safe to Play with Knives, Mol. Cell, vol.40, pp.179-204, 2010.

A. Clark, M. Oldenbroek, and T. Boyer, Mediator kinase module and human tumorigenesis, Crit. Rev. Biochem. Mol. Biol, vol.50, pp.393-426, 2015.

F. Coin and J. Égly, Formation du complexe d'initiation de la transcription: Des facteurs generaux aux complexes qui destabilisent la chromatine, Medecine/Sciences, vol.16, pp.593-601, 2000.

R. C. Conaway and J. W. Conaway, Function and regulation of the Mediator complex, Curr. Opin. Genet. Dev, vol.21, pp.225-230, 2011.

M. P. Cosma, Ordered recruitment: gene-specific mechanism of transcription activation, Mol. Cell, vol.10, pp.227-263, 2002.

C. Courilleau, C. Chailleux, A. Jauneau, F. Grima, S. Briois et al., The chromatin remodeler p400 atpase facilitates RAD51-mediated repair of DNA double-strand breaks, J. Cell Biol, vol.199, pp.1067-1081, 2012.

J. L. Cracowski, Les isoprostanes: un rôle physiopathologique potentiel en pathologie vasculaire, Rev. Med. Interne, vol.25, pp.459-463, 2004.

J. M. Craig, Heterochromatin -Many flavours, common themes, BioEssays, vol.27, pp.17-28, 2005.

F. Cucco, A. Servadio, V. Gatti, P. Bianchi, L. Mannini et al., Mutant cohesin drives chromosomal instability in early colorectal adenomas, Hum. Mol. Genet, vol.23, pp.6773-6778, 2014.

M. D'errico, E. Parlanti, M. Teson, B. M. De-jesus, P. Degan et al., New functions of XPC in the protection of human skin cells from oxidative damage, EMBO J, vol.25, pp.4305-4315, 2006.

M. E. Dahmus, Reversible phosphorylation of the C-terminal domain of RNA polymerase II, J Biol Chem, vol.271, pp.19009-19012, 1996.

B. Dalhus, M. Forsbring, I. H. Helle, E. S. Vik, R. J. Forstrøm et al., Separation-of-function mutants unravel the dual-reaction mode of human 8-oxoguanine DNA glycosylase, Structure, vol.19, pp.117-127, 2011.

D. Daniels, M. Ford, M. Schwinn, H. Benink, M. Galbraith et al., Mutual Exclusivity of MED12/MED12L, MED13/13L, and CDK8/19 Paralogs Revealed within the CDK-Mediator Kinase Module, J. Proteomics Bioinform, vol.1, pp.2-7, 2013.

F. Dantzer, L. Luna, M. Bjørås, and E. Seeberg, Human OGG1 undergoes serine phosphorylation and associates with the nuclear matrix and mitotic chromatin in vivo, Nucleic Acids Res, vol.30, pp.2349-57, 2002.

S. S. David, V. L. O'shea, and S. Kundu, Base Excision Repair of Oxidative DNA Damage, Nature, vol.447, pp.941-950, 2007.

L. Davidovic, M. Vodenicharov, E. B. Affar, and G. G. Poirier, Importance of poly(ADP-ribose) glycohydrolase in the control of poly(ADP-ribose) metabolism, Exp. Cell Res, vol.268, pp.7-13, 2001.

I. F. Davidson, D. Goetz, M. P. Zaczek, M. I. Molodtsov, P. J. Huis-in-'t-veld et al., Rapid movement and transcriptional re-localization of human cohesin on DNA, EMBO J, vol.35, pp.2671-2685, 2016.

M. A. Davis, E. A. Larimore, B. M. Fissel, J. Swanger, D. J. Taatjes et al., The SCFFbw7 ubiquitin ligase degrades MED13 and MED13L and regulates CDK8 module association with Mediator, Genes Dev, vol.27, pp.151-156, 2013.
DOI : 10.1101/gad.207720.112

URL : http://genesdev.cshlp.org/content/27/2/151.full.pdf

G. Dellaire and D. P. Bazett-jones, PML nuclear bodies: Dynamic sensors of DNA damage and cellular stress, BioEssays, vol.26, pp.963-977, 2004.
DOI : 10.1002/bies.20089

G. Dellaire, R. W. Ching, K. Ahmed, F. Jalali, K. C. Tse et al., Promyelocytic leukemia nuclear bodies behave as DNA damage sensors whose response to DNA double-strand breaks is regulated by NBS1 and the kinases ATM, Chk2, and ATR, J. Cell Biol, vol.175, pp.55-66, 2006.

B. Demple, T. Herman, and D. S. Chen, Cloning and expression of APE, the cDNA encoding the major human apurinic endonuclease: definition of a family of DNA repair enzymes, Proc. Natl. Acad. Sci. U. S. A, vol.88, pp.11450-11454, 1991.

C. Dherin, J. P. Radicella, M. Dizdaroglu, and S. Boiteux, Excision of oxidatively damaged DNA bases by the human ?-hOgg1 protein and the polymorphic ?-hOgg1(Ser326Cys) protein which is frequently found in human populations, Nucleic Acids Res, vol.27, pp.4001-4007, 1999.

I. I. Dianova, K. M. Sleeth, S. L. Allinson, J. L. Parsons, C. Breslin et al., XRCC1-DNA polymerase ? interaction is required for efficient base excision repair, Nucleic Acids Res, vol.32, pp.2550-2555, 2004.

C. Dinant, G. Ampatziadis-michailidis, H. Lans, M. Tresini, A. Lagarou et al., Enhanced chromatin dynamics by fact promotes transcriptional restart after UV-induced DNA damage, Mol. Cell, vol.51, pp.469-479, 2013.

Y. Ding, A. M. Fleming, and C. J. Burrows, Sequencing the Mouse Genome for the Oxidatively Modified Base 8-Oxo-7,8-dihydroguanine by OG-Seq, J. Am. Chem. Soc, vol.139, pp.2569-2572, 2017.

M. F. Dion, S. J. Altschuler, L. F. Wu, and O. J. Rando, Genomic characterization reveals a simple histone H4 acetylation code, Proc. Natl. Acad. Sci. U. S. A, vol.102, pp.5501-5506, 2005.

J. Dixon, S. Selvaraj, F. Yue, A. Kim, Y. Li et al., Topological domains in mammalian genomes identified by analysis of chromatin interactions, Nature, vol.485, pp.376-80, 2012.

M. Dizdaroglu, Oxidatively induced DNA damage: Mechanisms, repair and disease, Cancer Lett, vol.327, pp.26-47, 2012.

A. J. Donner, C. C. Ebmeier, D. J. Taatjes, and J. M. Espinosa, CDK8 is a positive regulator of transcriptional elongation within the serum response network, Nat Struc, vol.17, pp.194-201, 2010.

D. Dorsett and L. Ström, The ancient and evolving roles of cohesin in DNA repair and gene expression, Curr Biol, vol.22, pp.240-250, 2012.

W. Dröge, Free Radicals in the Physiological Control of Cell Function, Physiol. Rev, vol.82, pp.47-95, 2002.

A. Drohat and A. Maiti, Mechanisms for enzymatic cleavage of the N-glycosidic bond in DNA, Org Biomol Chem, vol.12, pp.8367-8378, 2014.

M. F. Dubois, V. T. Nguyen, S. Bellier, and O. Bensaude, Inhibitors of transcription such as 5,6-dichloro-1-?-D-ribofuranosylbenzimidazole and isoquinoline sulfonamide derivatives, 1994.

, *) promote dephosphorylation of the carboxyl-terminal domain of RNA polymerase II largest subunit, J. Biol. Chem, vol.269, pp.13331-13336

G. Dujardin, É. Daguenet, D. G. Bernard, M. Flodrops, S. Durand et al., L'épissage des ARN pré-messagers: quand le splicéosome perd pied, Medecine/Sciences, vol.32, pp.1103-1110, 2016.

A. Dvir, R. C. Conaway, and J. W. Conaway, A role for TFIIH in controlling the activity of early RNA polymerase II elongation complexes, Proc. Natl. Acad. Sci. U. S. A, vol.94, pp.9006-9010, 1997.

C. C. Ebmeier and D. J. Taatjes, Activator-Mediator binding regulates Mediator-cofactor interactions, Proc. Natl. Acad. Sci. U. S. A, vol.107, pp.11283-11288, 2010.

A. Elahi, Z. Zheng, J. Park, K. Eyring, T. Mccaffrey et al., The human OGG1 DNA repair enzyme and its association with orolaryngeal cancer risk, Carcinogenesis, vol.23, pp.1229-1234, 2002.

H. Elmlund, V. Baraznenok, M. Lindahl, C. O. Samuelsen, P. J. Koeck et al., The cyclin-dependent kinase 8 module sterically blocks Mediator interactions with RNA polymerase II, Proc. Natl. Acad. Sci. U. S. A, vol.103, pp.15788-93, 2006.

C. Esnault, Y. Ghavi-helm, S. Brun, J. Soutourina, N. Van-berkum et al., Mediator-dependant recruitment of TFIIH modules in preinitiation complex, Moll Cell, vol.31, pp.337-346, 2008.

F. Eyboulet, C. Cibot, T. Eychenne, H. Neil, O. Alibert et al., Mediator links transcription and DNA repair by facilitating Rad2 / XPG recruitment, Genes Dev, vol.27, pp.2549-62, 2013.

T. Eychenne, M. Werner, and J. Soutourina, Toward understanding of the mechanisms of Mediator function in vivo : Focus on the preinitiation complex assembly, Transcription, vol.8, pp.328-342, 2017.

F. Farinati, R. Cardin, M. Bortolami, D. Nitti, D. Basso et al., Oxidative DNA damage in gastric cancer: CagA status and OGG1 gene polymorphism, Int. J. Cancer, vol.123, pp.51-55, 2008.

A. Favier, Le stress oxydant: Intérêt conceptuel et expérimental dans la compréhension des mécanismes des maladies et potentiel thérapeutique, Actual. Chim, pp.108-115, 2003.

M. Featherstone, Coactivators in transcription initiation: Here are your orders, Curr. Opin. Genet. Dev, vol.12, pp.149-155, 2002.

G. Felsenfeld and M. Groudine, Controlling the double helix, Nature, vol.421, pp.448-453, 2003.

O. Fernandez-capetillo, A. Lee, M. Nussenzweig, and A. Nussenzweig, H2AX: The histone guardian of the genome, DNA Repair (Amst), vol.3, pp.959-967, 2004.

R. Firestein, A. J. Bass, S. Y. Kim, I. F. Dunn, J. Silver et al., CDK8 is a colorectal cancer oncogene that regulates beta-catenin activity, Nature, vol.455, pp.547-551, 2008.

P. M. Flanagan, R. J. Kelleher, M. H. Sayre, H. Tschochner, and R. D. Kornberg, A mediator required for activation of RNA polymerase II transcription in vitro, Nature, vol.350, pp.436-438, 1991.

A. M. Fleming, Y. Ding, and C. J. Burrows, Oxidative DNA damage is epigenetic by regulating gene transcription via base excision repair, Proc. Natl. Acad. Sci. U. S. A, vol.114, pp.2604-2609, 2017.
DOI : 10.1073/pnas.1619809114

URL : https://www.pnas.org/content/pnas/114/10/2604.full.pdf

Y. W. Fong, C. Cattoglio, and R. Tjian, The Intertwined Roles of Transcription and Repair Proteins, Mol. Cell, vol.52, pp.291-302, 2013.

J. R. Friedman, W. J. Fredericks, D. E. Jensen, D. W. Speicher, X. P. Huang et al., KAP-1, a novel corepressor for the highly conserved KRAB repression domain, Genes Dev, vol.10, pp.2067-2078, 1996.

T. Gaarenstroom and C. S. Hill, TGF-? signaling to chromatin: How Smads regulate transcription during self-renewal and differentiation, Semin. Cell Dev. Biol, vol.32, pp.107-118, 2014.

H. Gad, T. Koolmeister, A. S. Jemth, S. Eshtad, S. A. Jacques et al., Nature, vol.508, pp.215-221, 2014.

C. Gaiddon, N. C. Moorthy, and C. Prives, Ref-1 regulates the transactivation and pro-apoptotic functions of p53 in vivo, EMBO J, vol.18, pp.5609-5621, 1999.

M. Galbraith, M. Allen, C. Bensard, X. Wang, M. Schwinn et al., HIF1A employs CDK8-Mediator to stimulate RNAPII elongation, Cell, vol.153, pp.1327-1339, 2013.

R. Garcia-cruz, M. A. Brieño, I. Roig, M. Grossmann, E. Velilla et al., Dynamics of cohesin proteins REC8, STAG3, SMC1? and SMC3 are consistent with a role in sister chromatid cohesion during meiosis in human ovocytes, Hum. Reprod, vol.25, pp.2316-2327, 2010.

N. S. Gavande, P. S. Vandervere-carozza, H. D. Hinshaw, S. I. Jalal, C. R. Sears et al., DNA repair targeted therapy: The past or future of cancer treatment?, Pharmacol. Ther, vol.160, pp.65-83, 2016.

H. P. Gerber, M. Hagmann, K. Seipel, O. Georgiev, M. West et al., RNA polymerase II C-terminal domain required for enhancer-driven transcription, Nature, vol.374, pp.660-662, 1995.

R. Ghirlando and G. Felsenfeld, CTCF: Making the right connections, Genes Dev, vol.30, pp.881-891, 2016.

N. Gilbert, S. Boyle, H. Fiegler, K. Woodfine, N. P. Carter et al., Chromatin Architecture of the Human Genome: Gene-Rich Domains Are Enriched in Open Chromatin Fibers, Cell, vol.118, pp.555-566, 2004.

P. M. Girard, N. Guibourt, and S. Boiteux, The Ogg1 protein of Saccharomyces cerevisiae: A 7,8-dihydro-8-oxoguanine DNA glycosylase/AP lyase whose lysine 241 is a critical residue for catalytic activity, Nucleic Acids Res, vol.25, pp.3204-3211, 1997.

T. G. Gligoris, J. C. Scheinost, F. Bürmann, N. Petela, K. L. Chan et al., Closing the cohesin ring: Structure and function of its Smc3-kleisin interface, Science, vol.346, pp.963-967, 2014.

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

M. G. Goll and T. H. Bestor, Histone modification and replacement in chromatin activation, Genes Dev, vol.16, pp.1739-1742, 2002.

A. Goloborodko, M. V. Imakaev, J. F. Marko, and L. Mirny, Compaction and segregation of sister chromatids via active loop extrusion, Elife, vol.5, pp.1-16, 2016.

A. A. Goodarzi, A. T. Noon, D. Deckbar, Y. Ziv, Y. Shiloh et al., ATM Signaling Facilitates Repair of DNA Double-Strand Breaks Associated with Heterochromatin, Mol. Cell, vol.31, pp.167-177, 2008.

S. A. Gorski, M. Dundr, and T. Misteli, The road much traveled: trafficking in the cell nucleus, Curr. Opin. Cell Biol, vol.18, pp.284-290, 2006.

C. M. Green and G. Almouzni, When repair meets chromatin. First in series on chromatin dynamics, EMBO Rep, vol.3, pp.28-33, 2002.

A. Groth, W. Rocha, A. Verreault, and G. Almouzni, Chromatin Challenges during DNA Replication and Repair, Cell, vol.128, pp.721-733, 2007.

S. Gruber, Shaping chromosomes by DNA capture and release: gating the SMC rings, Curr. Opin. Cell Biol, vol.46, pp.87-93, 2017.

S. Gruber, C. H. Haering, and K. Nasmyth, Chromosomal cohesin forms a ring, Cell, vol.112, pp.765-777, 2003.

L. Guo, A. B.-giasson, M. Glavis-bloom, J. Brewer, A. Shorter et al., A cellular system that degrades misfolded proteins and protects against neurodegeneration, Mol. Cell, vol.55, pp.15-30, 2014.

C. H. Haering, A. M. Farcas, P. Arumugam, J. Metson, and K. Nasmyth, The cohesin ring concatenates sister DNA molecules, Nature, vol.454, pp.297-301, 2008.
DOI : 10.1038/nature07098

URL : http://wrap.warwick.ac.uk/207/1/WRAP_Arumugam_resubmitted_cover_delete.pdf

C. H. Haering, D. Schoffnegger, T. Nishino, W. Helmhart, K. Nasmyth et al., Structure and stability of cohesin's Smc1-kleisin interaction, Mol. Cell, vol.15, pp.951-964, 2004.
DOI : 10.1016/j.molcel.2004.08.030

URL : https://doi.org/10.1016/j.molcel.2004.08.030

J. Haleng, J. Pincemail, J. Defraigne, C. Charlier, and J. Chapelle, Le stress oxydant, Rev Med Liege, vol.62, pp.628-638, 2007.

A. S. Hansen, I. Pustova, C. Cattoglio, R. Tjian, and X. Darzacq, CTCF and cohesin regulate chromatin loop stability with distinct dynamics. Elife, vol.6, pp.1-33, 2017.

U. Hardeland, M. Bentele, J. Jiricny, and P. Schär, The versatile thymine DNA-glycosylase: A comparative characterization of the human, Drosophila and fission yeast orthologs, Nucleic Acids Res, vol.31, pp.2261-2271, 2003.

S. Hauf, E. Roitinger, B. Koch, C. M. Dittrich, K. Mechtler et al., Dissociation of cohesin from chromosome arms and loss of arm cohesion during early mitosis depends on phosphorylation of SA2, PLoS Biol, vol.3, pp.419-0432, 2005.

S. Hauf, I. C. Waizenegger, and J. M. Peters, Cohesin cleavage by separase required for anaphase and cytokinesis in human cells, Science, vol.293, pp.1320-1323, 2001.

K. A. Haushalter, P. T. Stukenberg, M. W. Kirschner, and G. L. Verdine, Identification of a new uracil-DNA glycosylase family by expression cloning using synthetic inhibitors, Curr. Biol, vol.9, pp.80087-80093, 1999.

T. K. Hazra, Y. W. Kow, Z. Hatahet, B. Imhoff, I. Boldogh et al., Identification and characterization of a novel human DNA glycosylase for repair of cytosinederived lesions, J. Biol. Chem, vol.277, pp.30417-30420, 2002.

H. A. Headlam and M. J. Davies, Markers of protein oxidation: Different oxidants give rise to variable yields of bound and released carbonyl products. Free Radic, Biol. Med, vol.36, pp.1175-1184, 2004.

M. L. Hegde, T. K. Hazra, and S. Mitra, Early steps in the DNA base excision/single-strand interruption repair pathway in mammalian cells, Cell Res, vol.18, pp.27-47, 2008.

J. M. Heidinger-pauli, E. Ünal, V. Guacci, and D. Koshland, The Kleisin Subunit of Cohesin Dictates Damage-Induced Cohesion, Mol. Cell, vol.31, pp.47-56, 2008.

E. Heitz, Das Heterochromatin der Moose, Jahrb Wiss Bot, vol.69, pp.762-818, 1928.

T. Helleday, E. Petermann, C. Lundin, B. Hodgson, and R. A. Sharma, DNA repair pathways as targets for cancer therapy, Nat. Rev. Cancer, vol.8, pp.193-204, 2008.

C. J. Hengartner, V. E. Myer, S. Liao, C. J. Wilson, S. S. Koh et al., Temporal Regulation of RNA Polymerase II by Srb10 and Kin28 Cyclin-Dependent Kinases, Mol. Cell, vol.2, pp.43-53, 1998.

W. Heyer, K. T. Ehmsen, and J. Liu, Regulation of Homologous Recombination in Eukaryotes, Annu. Rev. Genet, vol.44, pp.113-139, 2010.

J. W. Hill and M. K. Evans, Dimerization and opposite base-dependent catalytic impairment of polymorphic S326C OGG1 glycosylase, Nucleic Acids Res, vol.34, pp.1620-1632, 2006.

D. Hnisz, B. Abraham, T. Lee, A. Lau, V. Saint-andré et al., Transcriptional super-enhancers connected to cell identity and disease, Cell, vol.155, pp.934-981, 2013.

F. C. Holstege, E. G. Jennings, J. J. Wyrick, T. I. Lee, C. J. Hengartner et al., Dissecting the regulatory circuitry of a eukaryotic genome, Cell, vol.95, pp.717-728, 1998.

P. J. Horn and C. L. Peterson, Chromatin Higher Order Folding Wrapping up Transcription, Science, vol.297, pp.1824-1828, 2002.

J. Hsin and J. L. Manley, The RNA polymerase II CTD coordinates transcription and RNA processing, Genes Dev, vol.26, pp.2119-2137, 2012.

J. Hu, S. Z. Imam, K. Hashiguchi, N. C. Souza-pinto, and V. A. Bohr, Phosphorylation of human oxoguanine DNA glycosylase (?-OGG1) modulates its function, Nucleic Acids Res, vol.33, pp.3271-3282, 2005.

L. E. Huang, Z. Arany, D. M. Livingston, and H. F. Bunn, Activation of hypoxia-inducible transcription factor depends primarily upon redox-sensitive stabilization of its ? subunit, J. Biol. Chem, vol.271, pp.32253-32259, 1996.

S. Huang, Review: Perinucleolar structures, J. Struct. Biol, vol.129, pp.233-240, 2000.

S. Huang, M. Hölzel, T. Knijnenburg, A. Schlicker, U. Mcdermott et al., MED12 Controls the Response to Multiple Cancer Drugs through Regulation of TGF-? Receptor Signaling, Cell, vol.151, pp.937-950, 2012.

R. J. Hung, J. Hall, P. Brennan, and P. Boffetta, Genetic polymorphisms in the base excision repair pathway and cancer risk: A huge review, Am. J. Epidemiol, vol.162, pp.925-942, 2005.

S. Ikeda, T. Biswas, R. Roy, T. Izumi, I. Boldogh et al., Purification and characterization of human NTH1, a homolog of Escherichia coli endonuclease III. Direct identification of Lys-212 as the active nucleophilic residue, J. Biol. Chem, vol.273, pp.21585-93, 1998.

T. Ikura, S. Tashiro, A. Kakino, H. Shima, N. Jacob et al., DNA Damage-Dependent Acetylation and Ubiquitination of H2AX Enhances Chromatin Dynamics, Mol. Cell. Biol, vol.27, pp.7028-7040, 2007.
DOI : 10.1128/mcb.00579-07

URL : https://mcb.asm.org/content/27/20/7028.full.pdf

A. N. Imbalzano, K. S. Zaret, and R. E. Kingston, Transcription factor (TF) IIB and TFIIA can independently increase the affinity of the TATA-binding protein for DNA, J. Biol. Chem, vol.269, pp.8280-8286, 1994.

I. Izeddin, V. Récamier, L. Bosanac, I. I. Cissé, L. Boudarene et al., Single-molecule tracking in live cells reveals distinct target-search strategies of transcription factors in the nucleus, Elife, vol.3, pp.1-27, 2014.

T. Izumi, D. B. Brown, C. V. Naidu, K. K. Bhakat, M. A. Macinnes et al., Two essential but distinct functions of the mammalian abasic endonuclease, Proc. Natl. Acad. Sci. U. S. A, vol.102, pp.5739-5743, 2005.

T. Izumi and S. Mitra, Deletion analysis of human AP-endonuclease: Minimum sequence required for the endonuclease activity, Carcinogenesis, vol.19, pp.525-527, 1998.

D. A. Jackson, Chromatin domains and nuclear compartments: Establishing sites of gene expression in eukaryotic nuclei, Mol. Biol. Rep, vol.24, pp.209-220, 1997.

K. D. Jacob, N. N. Hooten, T. Tadokoro, A. Lohani, J. Barnes et al., Alzheimer's disease-associated polymorphisms in human OGG1 alter catalytic activity and sensitize cells to DNA damage. Free Radic, Biol. Med, vol.63, pp.115-125, 2013.

B. Jakob, J. Splinter, S. Conrad, K. O. Voss, D. Zink et al., DNA double-strand breaks in heterochromatin elicit fast repair protein recruitment, histone H2AX phosphorylation and relocation to euchromatin, Nucleic Acids Res, vol.39, pp.6489-6499, 2011.

C. Jeronimo, M. F. Langelier, A. R. Bataille, J. M. Pascal, B. F. Pugh et al., Tail and Kinase Modules Differently Regulate Core Mediator Recruitment and Function In Vivo, Mol. Cell, vol.64, pp.455-466, 2016.

G. Ji, L. Yan, W. Liu, J. Qu, and A. Gu, OGG1 Ser326Cys polymorphism interacts with cigarette smoking to increase oxidative DNA damage in human sperm and the risk of male infertility, Toxicol. Lett, vol.218, pp.144-149, 2013.

Y. Jiang, X. Wang, S. Bao, R. Guo, D. G. Johnson et al., INO80 chromatin remodeling complex promotes the removal of UV lesions by the nucleotide excision repair pathway, Proc. Natl. Acad. Sci. U. S. A, vol.107, pp.17274-17279, 2010.

M. H. Kagey, J. J. Newman, S. Bilodeau, Y. Zhan, D. A. Orlando et al., Mediator and cohesin connect gene expression and chromatin architecture, Nature, vol.467, pp.430-435, 2010.

M. Kahn, Can we safety target the WNT pathway?, Nat Rev Drug Discov, vol.13, pp.513-532, 2014.

A. Kalousi and E. Soutoglou, Nuclear compartmentalization of DNA repair, Curr. Opin. Genet. Dev, vol.37, pp.148-157, 2016.

L. Kang, W. Zhao, G. Zhang, J. Wu, and H. Guan, Acetylated 8-oxoguanine DNA glycosylase 1 and its relationship with p300 and SIRT1 in lens epithelium cells from age-related cataract, Exp. Eye Res, vol.135, pp.102-108, 2015.

A. Kapoor, M. S. Goldberg, L. K. Cumberland, K. Ratnakumar, M. F. Segura et al., The histone variant macroH2A suppresses melanoma progression through regulation of CDK8, Nature, vol.468, pp.1105-1111, 2010.

Y. Kato, R. Habas, U. Katsutama, A. Näär, and X. He, A component of the ARC/Mediator complex required for TGFb/Nodal signalling, Nature, vol.418, pp.641-646, 2002.

R. Kaufmann, R. Straussberg, H. Mandel, A. Fattal-valevski, B. Ben-zeev et al., Infantile cerebral and cerebellar atrophy is associated with a mutation in the MED17 subunit of the transcription preinitiation mediator complex, Am. J. Hum. Genet, vol.87, pp.667-670, 2010.

M. Kaur, C. Descipio, J. Mccallum, D. Yaeger, M. Devoto et al., Precocious sister chromatid separation (PSCS) in Cornelia de Lange syndrome, Am J Med Genet A, vol.138, pp.27-31, 2005.
DOI : 10.1002/ajmg.a.30919

URL : http://europepmc.org/articles/pmc2766539?pdf=render

B. Kavli, O. Sundheim, M. Akbari, M. Otterlei, H. Nilsen et al., 2002. hUNG2 is the major repair enzyme for removal of uracil from U:A matches, U:G mismatches, and U in single-stranded DNA, with hSMUG1 as a broad specificity backup, J. Biol. Chem, vol.277, pp.39926-39936

P. A. Van-der-kemp, J. B. Charbonnier, M. Audebert, and S. Boiteux, Catalytic and DNA-binding properties of the human Ogg1 DNA N-glycosylase/AP lyase: Biochemical exploration of H270, Q315 and F319, three amino acids of the 8-oxoguanine-binding pocket, Nucleic Acids Res, vol.32, pp.570-578, 2004.

P. Van-der-kemp, D. Thomas, R. Barbey, R. Oliveira, and S. Boiteux, Cloning and expression in Escherichia coli of the OGG1 gene of Saccharomyces cerevisiae, which codes for a DNA glycosylase that excises 7,8-dihydro-8-oxoguanine and 2,6-diamino-4-hydroxy-5-Nmethylformamidopyrimidine, Proc. Natl. Acad. Sci. U. S. A, vol.93, pp.5197-5202, 1996.

N. Khansari, Y. Shakiba, and M. Mahmoudi, Chronic inflammation and oxidative stress as a major cause of age-related diseases and cancer. Recent Pat, Inflamm. Allergy Drug Discov, vol.3, pp.73-80, 2009.

A. Khobta and B. Epe, Interactions between DNA damage, repair, and transcription, Mutat. Res. -Fundam. Mol. Mech. Mutagen, vol.736, pp.5-14, 2012.
DOI : 10.1016/j.mrfmmm.2011.07.014

A. Khobta, N. Kitsera, B. Speckmann, and B. Epe, 8-Oxoguanine DNA glycosylase (Ogg1) causes a transcriptional inactivation of damaged DNA in the absence of functional Cockayne syndrome B (Csb) protein, DNA Repair (Amst), vol.8, pp.309-317, 2009.

Y. Kikuchi, H. Umemura, S. Nishitani, S. Iida, R. Fukasawa et al., Human mediator MED17 subunit plays essential roles in gene regulation by associating with the transcription and DNA repair machineries, Genes to Cells, vol.20, pp.191-202, 2015.

D. I. Kim and K. J. Roux, Filling the Void: Proximity-Based Labeling of Proteins in Living Cells, Trends Cell Biol, vol.26, pp.804-817, 2016.

J. S. Kim, T. B. Krasieva, V. Lamorte, A. Malcolm, R. Taylor et al., Specific recruitment of human cohesin to laser-induced DNA damage, J. Biol. Chem, vol.277, pp.45149-45153, 2002.

S. Kim, X. Xu, A. Hecht, and T. G. Boyer, Mediator is a transducer of Wnt/?-catenin signaling, J. Biol. Chem, vol.281, pp.14066-14075, 2006.

S. T. Kim, B. Xu, and M. B. Kastan, Involvement of the cohesin protein, Smc1, in Atm-dependent and independent responses to DNA damage, Genes Dev, vol.16, pp.560-570, 2002.

R. Kitagawa, C. J. Bakkenist, P. J. Mckinnon, and M. B. Kastan, Phosphorylation of SMC1 is a critical downstream event in the ATM-NBS1-BRCA1 pathway, Genes Dev, vol.18, pp.1423-1438, 2004.

N. Kitsera, D. Stathis, B. Lühnsdorf, H. Müller, T. Carell et al., 8-Oxo-7,8-dihydroguanine in DNA does not constitute a barrier to transcription, but is converted into transcription-blocking damage by OGG1, Nucleic Acids Res, vol.39, pp.5926-5934, 2011.

A. Klungland, I. Rosewell, S. Hollenbach, E. Larsen, G. Daly et al., Accumulation of premutagenic DNA lesions in mice defective in removal of oxidative base damage, Proc. Natl. Acad. Sci. U. S. A, vol.96, pp.13300-13305, 1999.

M. T. Knuesel, K. D. Meyer, C. Bernecky, and D. J. Taatjes, The human CDK8 subcomplex is a molecular switch that controls Mediator coactivator function, Genes Dev, vol.23, pp.439-451, 2009.

M. T. Knuesel, K. D. Meyer, A. J. Donner, J. M. Espinosa, and D. J. Taatjes, The Human CDK8 Subcomplex Is a Histone Kinase That Requires Med12 for Activity and Can Function Independently of Mediator, Mol. Cell. Biol, vol.29, pp.650-661, 2009.

T. Kohno, H. Kunitoh, K. Toyama, S. Yamamoto, A. Kuchiba et al., Association of the OGG1-Ser326Cys polymorphism with lung adenocarcinoma risk, Cancer Sci, vol.97, pp.724-728, 2006.

A. Kon, L. Y. Shih, M. Minamino, M. Sanada, Y. Shiraishi et al., Recurrent mutations in multiple components of the cohesin complex in myeloid neoplasms, Nat. Genet, vol.45, pp.1232-1237, 2013.

R. Kornberg, Chromatin Structure : A Repeating Unit of Histones and DNA, Science, vol.184, pp.868-871, 1974.

I. V. Kovtun, Y. Liu, M. Bjoras, A. Klungland, S. H. Wilson et al., OGG1 initiates age-dependent CAG trinucleotide expansion in somatic cells, Nature, vol.447, pp.447-452, 2007.

H. Krokan, H. Nilsen, F. Skorpen, M. Otterlei, and G. Slupphaug, Base excision repair of DNA in mammalian cells, FEBS Lett, vol.476, pp.73-77, 2000.

M. J. Kruhlak, A. Celeste, G. Dellaire, O. Fernandez-capetillo, W. G. Müller et al., Changes in chromatin structure and mobility in living cells at sites of DNA double-strand breaks, J. Cell Biol, vol.172, pp.823-834, 2006.

M. Kschonsak and C. H. Haering, Shaping mitotic chromosomes: From classical concepts to molecular mechanisms, BioEssays, vol.37, pp.755-766, 2015.

N. A. Kuznetsov, V. V. Koval, D. O. Zharkov, G. A. Nevinsky, K. T. Douglas et al., Kinetics of substrate recognition and cleavage by human 8-oxoguanine-DNA glycosylase, Nucleic Acids Res, vol.33, pp.3919-3931, 2005.

J. Labahn, O. D. Schärer, A. Long, K. Ezaz-nikpay, G. L. Verdine et al., Structural basis for the excision repair of alkylation-damaged DNA, Cell, vol.86, pp.80103-80111, 1996.

M. Lachner, D. O'carroll, S. Rea, K. Mechtler, and T. Jenuwein, Methylation of histone H3 lysine 9 creates a binding site for HP1 proteins, Nature, vol.410, pp.116-136, 2001.

R. Ladurner, E. Kreidl, M. P. Ivanov, H. Ekker, M. H. Idarraga-amado et al., Sororin actively maintains sister chromatid cohesion, EMBO J, vol.35, pp.635-653, 2016.

M. Lafarga, M. Berciano, E. Pena, I. Mayo, J. Castano et al., Clastosome: a subtype of nuclear body enriched in 19S and 20S proteosomes, ubiquitin, and protein substrates of proteasome, Mol. Biol. Cell, vol.13, pp.2771-82, 2002.

F. Lai, U. Orom, M. Cesaroni, M. Beringer, D. Taatjes et al., Activating RNAs associate with Mediator to enhance chromatin architecture and transcription, Nature, vol.494, pp.497-501, 2013.

A. I. Lamond and J. E. Sleeman, Nuclear substructure and dynamics, Curr. Biol, vol.13, pp.825-828, 2003.

L. Lan, S. Nakajima, L. Wei, L. Sun, C. L. Hsieh et al., Novel method for site-specific induction of oxidative DNA damage reveals differences in recruitment of repair proteins to heterochromatin and euchromatin, Nucleic Acids Res, vol.42, pp.2330-2345, 2014.

E. Larsen, K. Kwon, F. Coin, J. M. Egly, and A. Klungland, Transcription activities at 8-oxoG lesions in DNA, DNA Repair (Amst), vol.3, pp.1457-1468, 2004.

T. Lee and R. Young, Transcription of Eukaryotic Protein-Coding Genes, Annu. Rev. Genet, vol.34, pp.77-137, 2000.

T. Lee and R. Young, Transcriptional regulation and its misregulation in disease, Cell, vol.152, pp.1237-1251, 2013.

A. R. Lehmann, DNA polymerases and repair synthesis in NER in human cells, DNA Repair (Amst), vol.10, pp.730-733, 2011.

A. R. Lehmann, M. Walicka, D. J. Griffiths, J. M. Murray, F. Z. Watts et al., The rad18 gene of Schizosaccharomyces pombe defines a new subgroup of the SMC superfamily involved in DNA repair, Mol. Cell. Biol, vol.15, pp.7067-7080, 1995.

I. Liguori, G. Russo, F. Curcio, G. Bulli, L. Aran et al., Oxidative stress, aging, and diseases, Clin Interv Aging, vol.13, pp.757-772, 2018.

T. Lindahl, An N-Glycosidase from Escherichia coli That Releases Free Uracil from DNA Containing Deaminated Cytosine Residues, Proc. Natl. Acad. Sci. U. S. A, vol.71, pp.3649-3653, 1974.

T. Lindahl, Instability and decay of the primary structure od DNA, Nature, vol.362, pp.709-724, 1993.

M. Litt, M. Simpson, M. Gaszner, C. Allis, and G. Felsenfeld, Correlation Between Histone Lysine Methylation and Developmental Changes at the Chicken beta-Globin Locus, Science, vol.293, pp.2453-2455, 2001.

H. Liu, S. Rankin, and H. Yu, Phosphorylation-enabled binding of SGO1-PP2A to cohesin protects sororin and centromeric cohesion during mitosis, Nat. Cell Biol, vol.15, pp.40-49, 2013.

D. Llères, S. Swift, and A. I. Lamond, Detecting Protein-Protein Interactions In Vivo with FRET using Multiphoton Fluorescence Lifetime Imaging Microscopy (FLIM), Curr. Protoc. Cytom, vol.12, pp.1-19, 2007.

B. Van-loon and U. Hübscher, An 8-oxo-guanine repair pathway coordinated by MUTYH glycosylase and DNA polymerase lambda, Proc. Natl. Acad. Sci. U. S. A, vol.106, pp.18201-18206, 2009.

A. Losada, Cohesin in cancer: Chromosome segregation and beyond, Nat. Rev. Cancer, vol.14, pp.389-393, 2014.

A. Losada and T. Hirano, Dynamic molecular linkers of the genome: The first decade of SMC proteins, Genes Dev, vol.19, pp.1269-1287, 2005.

A. Losada, T. Yokochi, R. Kobayashi, and T. Hirano, Identification and characterization of SA/Scc3p subunits in the Xenopus and human cohesin complexes, J. Cell Biol, vol.150, pp.405-416, 2000.

C. De-los-santos, C. Chang, M. Mycek, and R. A. Cardullo, FRAP, FLIM, and FRET: Detection and analysis of cellular dynamics on a molecular scale using fluorescence microscopy, Mol. Reprod. Dev, vol.82, pp.587-604, 2015.

K. Luger, A. W. Mäder, R. K. Richmond, D. F. Sargent, and T. J. Richmond, Crystal structure of the nucleosome core particle at 2.8 Å resolution, Nature, vol.389, pp.251-260, 1997.

M. S. Luijsterburg, M. Lindh, K. Acs, M. G. Vrouwe, A. Pines et al., DDB2 promotes chromatin decondensation at UV-induced DNA damage, J. Cell Biol, vol.197, pp.267-281, 2012.

L. Luna, V. Rolseth, G. A. Hildrestrand, M. Otterlei, F. Dantzer et al., Dynamic relocalization of hOGG1 during the cell cycle is disrupted in cells harbouring the hOGG1-Cys326 polymorphic variant, Nucleic Acids Res, vol.33, pp.1813-1824, 2005.

H. Luo, Y. Li, J. J. Mu, J. Zhang, T. Tonaka et al., Regulation of intra-S phase checkpoint by ionizing radiation (IR)-dependent and IR-independent phosphorylation of SMC3, J. Biol. Chem, vol.283, pp.19176-19183, 2008.

Z. Luo, C. Lin, E. Guest, A. Garrett, N. Mohaghegh et al., The Super Elongation Complex Family of RNA Polymerase II Elongation Factors: Gene Target Specificity and Transcriptional Output, Mol. Cell. Biol, vol.32, pp.2608-2617, 2012.

N. M. Luscombe, S. E. Austin, H. M. Berman, ?. , and J. M. Thornton, An overview of the structures of protein-DNA complexes, Genome Biol, vol.1, pp.1-1, 2000.

G. Maga, G. Villani, E. Crespan, U. Wimmer, E. Ferrari et al., 8-oxoguanine bypass by human DNA polymerases in the presence of auxiliary proteins, Nature, vol.447, pp.606-608, 2007.

N. Mäkinen, M. Mehine, J. Tolvanen, E. Kaasinen, Y. Li et al., MED12, the mediator complex subunit 12 gene, is mutated at high frequency in uterine leiomyomas, Science, vol.334, pp.252-257, 2011.

S. Malik and R. Roeder, The metazoan Mediator co-activator complex as an integrative hub for transcriptional regulation, Nat. Rev. Genet, vol.11, pp.761-772, 2010.

S. Malik and R. G. Roeder, Transcriptional regulation through Mediator-like coactivators in yeast and metazoan cells, Trends Biochem. Sci, vol.25, pp.1596-1602, 2000.

G. Mao, X. Pan, B. Zhu, Y. Zhang, F. Yuan et al., Identification and characterization of OGG1 mutations in patients with Alzheimer's disease, Nucleic Acids Res, vol.35, pp.2759-2766, 2007.

L. Marino-ramirez, M. Kann, B. Shoemaker, and D. Landsman, Histone structure and nucleosome stability, Expert Rev Proteomics, vol.2, pp.719-748, 2005.

A. Marintchev, M. R. Gryk, and G. P. Mullen, Site-directed mutagenesis analysis of the structural interaction of the single-strand-break repair protein, X-ray cross-complementing group 1, with DNA polymerase ?, Nucleic Acids Res, vol.31, pp.580-588, 2003.

E. Markkanen, Not breathing is not an option: How to deal with oxidative DNA damage, DNA Repair (Amst), vol.59, pp.82-105, 2017.

E. Markkanen, J. Dorn, and U. Hübscher, MUTYH DNA glycosylase: The rationale for removing undamaged bases from the DNA, Front. Genet, vol.4, pp.1-20, 2013.

S. Marsin, A. E. Vidal, M. Sossou, J. Ménissier-de-murcia, F. L. Page et al., Role of XRCC1 in the Coordination and Stimulation of Oxidative DNA Damage Repair Initiated by the DNA Glycosylase hOGG1, J. Biol. Chem, vol.278, pp.44068-44074, 2003.

M. Masson, C. Niedergang, V. Schreiber, S. Muller, J. Menissier-de-murcia et al., XRCC1 is specifically associated with poly(ADP-ribose) polymerase and negatively regulates its activity following DNA damage, Mol. Cell. Biol, vol.18, pp.3563-71, 1998.

A. Mccullough, M. Dodson, and R. Lloyd, Initiation of base excision repair: Glycosylase mechanisms and structures, Annu. Rev. Biochem, vol.68, pp.255-285, 1999.

R. Meas, J. J. Wyrick, and M. J. Smerdon, Nucleosomes regulate base excision repair in chromatin, Mutat. Res. -Rev. Mutat. Res, vol.10, pp.1-8, 2017.

L. B. Meira, S. Devaraj, G. E. Kisby, D. K. Burns, R. L. Daniel et al., Heterozygosity for the Mouse Apex Gene Results in Phenotypes Associated with Oxidative Stress, Cancer Res, vol.61, pp.5552-5557, 2001.

H. Menoni, D. Gasparutto, A. Hamiche, J. Cadet, S. Dimitrov et al., ATPDependent Chromatin Remodeling Is Required for Base Excision Repair in Conventional but Not in Variant H2A, Bbd Nucleosomes. Mol. Cell. Biol, vol.27, pp.5949-5956, 2007.
URL : https://hal.archives-ouvertes.fr/inserm-00170196

H. Menoni, J. H. Hoeijmakers, and W. Vermeulen, Nucleotide excision repair-initiating proteins bind to oxidative DNA lesions in vivo, J. Cell Biol, vol.199, pp.1037-1046, 2012.

H. Menoni, M. S. Shukla, V. Gerson, S. Dimitrov, and D. Angelov, Base excision repair of 8-oxoG in dinucleosomes, Nucleic Acids Res, vol.40, pp.692-700, 2012.
URL : https://hal.archives-ouvertes.fr/ensl-00807713

K. D. Meyer, A. J. Donner, M. T. Knuesel, A. G. York, J. M. Espinosa et al., Cooperative activity of cdk8 and GCN5L within Mediator directs tandem phosphoacetylation of histone H3, EMBO J, vol.27, pp.1447-1457, 2008.

M. L. Michaels and J. H. Miller, The GO System Protects Organisms from the Mutagenic Effect of the Spontaneous Lesion 8-Hydroxyguanine (7,8-Dihydro-8-Oxoguanine), J. Bacteriol, vol.174, pp.6321-6325, 1992.

C. Miller, I. Matic, K. C. Maier, B. Schwalb, S. Roether et al., Mediator phosphorylation prevents stress response transcription during non-stress conditions, J. Biol. Chem, vol.287, pp.44017-44026, 2012.

O. Minowa, T. Arai, M. Hirano, Y. Monden, S. Nakai et al., Mmh/Ogg1 gene inactivation results in accumulation of 8-hydroxyguanine in mice, Proc. Natl. Acad. Sci. U. S. A, vol.97, pp.4156-4161, 2000.

T. Misteli, Cell biology of transcription and pre-mRNA splicing: nuclear architecture meets nuclear function, J. Cell Sci, vol.113, pp.1841-1849, 2000.

T. Misteli, J. Cáceres, and D. Spector, The dynamics of a pre-mRNA splicing factor in living cells, Nature, vol.387, pp.523-530, 1997.

C. A. Mizzen, X. J. Yang, T. Kokubo, J. E. Brownell, A. J. Bannister et al., The TAF(II)250 subunit of TFIID has histone acetyltransferase activity, Cell, vol.87, pp.81821-81829, 1996.

R. Mjelle, S. A. Hegre, P. A. Aas, G. Slupphaug, F. Drabløs et al., Cell cycle regulation of human DNA repair and chromatin remodeling genes, DNA Repair (Amst), vol.30, pp.53-67, 2015.

C. Morales and A. Losada, Establishing and dissolving cohesion during the vertebrate cell cycle, Curr. Opin. Cell Biol, vol.52, pp.51-57, 2018.

E. Moritz, K. Pauly, A. Bravard, J. Hall, J. P. Radicella et al., hOGG1-Cys326 variant cells are hypersensitive to DNA repair inhibition by nitric oxide, Carcinogenesis, vol.35, pp.1426-1433, 2014.

I. Morland, V. Rolseth, L. Luna, T. Rognes, M. Bjørås et al., Human DNA glycosylases of the bacterial Fpg/MutM superfamily: an alternative pathway for the repair of 8-oxoguanine and other oxidation products in DNA, Nucleic Acids Res, vol.30, pp.4926-4962, 2002.

E. Morris, J. Ji, F. Yang, L. Stefano, A. Herr et al., E2F1 represses beta-catenin transcription and is antagonized by both pRB and CDK8, Nature, vol.455, pp.552-556, 2008.
DOI : 10.1038/nature07310

URL : http://europepmc.org/articles/pmc3148807?pdf=render

M. Muftuoglu, N. C. Souza-pinto, A. Dogan, M. Aamann, T. Stevnsner et al., Cockayne syndrome group B protein stimulates repair of formamidopyrimidines by NEIL1 DNA glycosylase, J. Biol. Chem, vol.284, pp.9270-9279, 2009.

A. Mukhopadhyay, J. M. Kramer, G. Merkx, D. Lugtenberg, D. F. Smeets et al., CDK19 is disrupted in a female patient with bilateral congenital retinal folds, microcephaly and mild mental retardation, Hum. Genet, vol.128, pp.281-291, 2010.
DOI : 10.1007/s00439-010-0848-x

URL : https://link.springer.com/content/pdf/10.1007%2Fs00439-010-0848-x.pdf

N. Muncke, C. Jung, H. Rüdiger, H. Ulmer, R. Roeth et al., Missense Mutations and Gene Interruption in PROSIT240, a Novel TRAP240-Like Gene, in Patients with Congenital Heart Defect (Transposition of the Great Arteries), Circulation, vol.108, pp.2843-2850, 2003.

Y. Murayama and F. Uhlmann, Biochemical reconstitution of topological DNA binding by the cohesin ring, Nature, vol.505, pp.367-371, 2014.

A. M. Näär, D. J. Taatjes, W. Zhai, E. Nogales, and R. Tjian, Human CRSP interacts with RNA polymerase II CTD and adopts a specific CTD-bound conformation, Genes Dev, vol.16, pp.1339-1344, 2002.

J. Nakamura and J. Swenberg, Endogenous Apurinic/Apyrimidinic Sites in Genomic DNA of Mammalian Tissues, Cancer Res, vol.59, pp.2522-2526, 1999.

R. A. Nash, K. W. Caldecott, D. E. Barnes, and T. Lindahl, XRCC1 protein interacts with one of two distinct forms of DNA ligase III, Biochemistry, vol.36, pp.5207-5211, 1997.

K. Nasmyth, Cohesin: A catenase with separate entry and exit gates?, Nat. Cell Biol, vol.13, pp.1170-1177, 2011.

K. Nasmyth and C. H. Haering, The Structure and Function of Smc and Kleisin Complexes, Annu. Rev. Biochem, vol.74, pp.595-648, 2005.

K. Nasmyth and C. H. Haering, Cohesin: Its Roles and Mechanisms, Annu. Rev. Genet, vol.43, pp.525-558, 2009.

Z. K. Nazarkina, S. N. Khodyreva, S. Marsin, O. I. Lavrik, and J. P. Radicella, XRCC1 interactions with base excision repair DNA intermediates, DNA Repair (Amst), vol.6, pp.254-264, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00169619

A. Németh and I. Grummt, Dynamic regulation of nucleolar architecture, Curr. Opin. Cell Biol, vol.52, pp.105-111, 2018.

H. Niki, R. Imamura, M. Kitaoka, K. Yamanaka, T. Ogura et al., E.coli MukB protein involved in chromosome partition forms a homodimer with a rod-and-hinge structure having DNA binding and ATP/GTP binding activities, EMBO J, vol.11, pp.5101-5110, 1992.

H. Niki, A. Jaffe, R. Imamura, T. Ogura, and S. Hiraga, The new gene mukB codes for a 177 kd protein with coiled-coil domains involved in chromosome partitioning of E.coli, EMBO J, vol.10, pp.183-193, 1991.

T. Nishi, N. Shimizu, M. Hiramoto, I. Sato, Y. Yamaguchi et al., Spatial redox regulation of a critical cysteine residue of NF-kappa B in vivo, J Biol Chem, vol.277, pp.44548-56, 2002.

T. Nishiyama, R. Ladurner, J. Schmitz, E. Kreidl, A. Schleiffer et al., Sororin mediates sister chromatid cohesion by antagonizing Wapl, Cell, vol.143, pp.737-749, 2010.

T. Nishiyama, M. Sykora, P. Huis-in't-veld, K. Mechtler, and J. Peters, Aurora B and Cdk1 mediate Wapl activation and release of acetylated ccohesin from chromosomes by phosphorylating Sororin, Proc. Natl. Acad. Sci. U. S. A, vol.110, pp.13404-13413, 2013.

A. Nitzsche, M. Paszkowski-rogacz, F. Matarese, E. M. Janssen-megens, N. C. Hubner et al., RAD21 cooperates with pluripotency transcription factors in the maintenance of embryonic stem cell identity, PLoS One, vol.6, p.19470, 2011.

N. Noren-hooten, K. Kompaniez, J. Barnes, A. Lohani, and M. K. Evans, Poly(ADP-ribose) polymerase 1 (PARP-1) binds to 8-oxoguanine-DNA glycosylase (OGG1), J. Biol. Chem, vol.286, pp.44679-44690, 2011.

S. Notarnicola, M. Mcintosh, and K. Wise, A Mycoplasma hyorhinis protein with sequence similarities to nucleotide-binding enzymes, Gene, vol.97, pp.77-85, 1991.

I. Novotný, A. Malinová, E. Stejskalová, D. Mat?j?, K. Klime?ová et al., SART3-Dependent Accumulation of Incomplete Spliceosomal snRNPs in Cajal Bodies, Cell Rep, vol.10, pp.429-440, 2015.

V. S. Nunes and N. S. Moretti, Nuclear subcompartments: an overview, Cell Biol. Int, vol.41, pp.2-7, 2017.

J. M. Ordway, D. Eberhart, and T. Curran, Cysteine 64 of Ref-1 is not essential for redox regulation of AP-1 DNA binding, Mol. Cell. Biol, vol.23, pp.4257-66, 2003.

U. A. Ørom, T. Derrien, M. Beringer, K. Gumireddy, A. Gardini et al., Long non-coding RNAs with enhancerlike function in human, Cell, vol.143, pp.46-58, 2010.

S. Palipoch and P. Koomhin, Oxidative stress-associated pathology: A review, Sains Malaysiana, vol.44, pp.1441-1451, 2015.

L. Pan, B. Zhu, W. Hao, X. Zeng, S. A. Vlahopoulos et al., Oxidized guanine base lesions function in 8-oxoguanine DNA glycosylase-1-mediated epigenetic regulation of nuclear factor ?B-driven gene expression, J. Biol. Chem, vol.291, pp.25553-25566, 2016.

M. Pandey and S. Raghavan, DNA Double-strand Break Repair in Mammals, J Radiat Cancer Res, vol.8, pp.93-100, 2017.

V. Parelho, S. Hadjur, M. Spivakov, M. Leleu, S. Sauer et al., Cohesins Functionally Associate with CTCF on Mammalian Chromosome Arms, Cell, vol.132, pp.422-433, 2008.

H. Pelish, B. Liau, I. Nitulescu, A. Tangpeerachaikul, Z. Poss et al., Mediator Kinase Inhibition Further Activates Super-Enhancer Associated Genes in AML, Nature, vol.526, pp.273-276, 2015.

R. Pepperkok, A. Squire, S. Geley, and P. I. Bastiaens, Simultaneous detection of multiple green fluorescent proteins in live cells by fluorescence lifetime imaging microscopy, Curr. Biol, vol.9, pp.80117-80118, 1999.

A. M. Pisoschi and A. Pop, The role of antioxidants in the chemistry of oxidative stress: A review, Eur. J. Med. Chem, vol.97, pp.55-74, 2015.

C. Plaschka, L. Larivière, L. Wenzeck, M. Seizl, M. Hemann et al., Architecture of the RNA polymerase II-Mediator core initiation complex, Nature, vol.518, pp.376-380, 2015.

S. E. Polo and G. Almouzni, Chromatin dynamics after DNA damage: the legacy of the AccessRepair-Restore model, DNA Repair (Amst), vol.36, pp.114-121, 2015.

S. E. Polo, D. Roche, and G. Almouzni, New Histone Incorporation Marks Sites of UV Repair in Human Cells, Cell, vol.127, pp.481-493, 2006.

Y. Pommier, C. Redon, V. A. Rao, J. A. Seiler, O. Sordet et al., Repair of and checkpoint response to topoisomerase I-mediated DNA damage, Mutat. Res. -Fundam. Mol. Mech. Mutagen, vol.532, pp.173-203, 2003.

Z. Poss, C. Ebmeier, A. Odell, A. Tangpeerachaikul, T. Lee et al., Identification of Mediator kinase substrates in human cells using cortistatin A and quantitative phosphoproteomics, Cell, vol.15, pp.436-450, 2016.

Z. C. Poss, C. C. Ebmeier, and D. J. Taatjes, The Mediator complex and transcription regulation, Crit. Rev. Biochem. Mol. Biol, vol.48, pp.575-608, 2013.

H. E. Poulsen, E. Specht, K. Broedbaek, T. Henriksen, C. Ellervik et al., RNA modifications by oxidation: A novel disease mechanism? Free Radic, Biol. Med, vol.52, pp.1353-1361, 2012.

A. Prakash, S. Doublié, and S. Wallace, The Fpg/Nei family of DNA glycosylases: substrates, structures and search for damage, Prg Mol Biol Transl, vol.110, pp.71-91, 2012.

J. L. Pulice and C. Kadoch, Composition and Function of Mammalian SWI/SNF Chromatin Remodeling Complexes in Human Disease, Cold Spring Harb. Symp. Quant. Biol, vol.81, pp.53-60, 2016.

J. P. Radicella, C. Dherin, C. Desmaze, M. S. Fox, and S. Boiteux, Cloning and characterization of hOGG1, a human homolog of the OGG1 gene of Saccharomyces cerevisiae, Proc. Natl. Acad. Sci. U. S. A, vol.94, pp.8010-8015, 1997.

P. B. Rahl, C. Y. Lin, A. C. Seila, R. A. Flynn, S. Mccuine et al., C-Myc regulates transcriptional pause release, Cell, vol.141, pp.432-445, 2010.

D. B. Ramsden, P. W. Ho, J. W. Ho, H. F. Liu, D. H. So et al., Human neuronal uncoupling proteins 4 and 5 (UCP4 and UCP5): Structural properties, regulation, and physiological role in protection against oxidative stress and mitochondrial dysfunction, Brain Behav, vol.2, pp.468-478, 2012.

S. Rao, M. Huntley, N. Durand, E. Stamenova, I. Bochkov et al., A three-dimensional map of the genome at kilobase resolution reveals principles of chromatin looping, Cell, vol.159, pp.1665-80, 2014.

V. K. Rao, A. Pal, and R. Taneja, A drive in SUVs: From development to disease, Epigenetics, vol.12, pp.177-186, 2017.

J. T. Reardon, T. Bessho, H. C. Kung, P. H. Bolton, and A. Sancar, In vitro repair of oxidative DNA damage by human nucleotide excision repair system: possible explanation for neurodegeneration in xeroderma pigmentosum patients, Proc. Natl. Acad. Sci. U. S. A, vol.94, pp.9463-9468, 1997.

E. Revenkova, M. Eijpe, C. Heyting, C. A. Hodges, P. A. Hunt et al., Cohesin SMC1? is required for meiotic chromosome dynamics, sister chromatid cohesion and DNA recombination, Nat. Cell Biol, vol.6, pp.555-562, 2004.

E. Revenkova, K. Hermann, C. Adelfalk, and R. Jessberger, Ovocyte cohesin expression restricted to predictyate stages provides full fertility and prevents aneuploidy, Curr Biol, vol.20, pp.1529-1562, 2010.

H. Risheg, J. M. Graham, R. D. Clark, R. C. Rogers, J. M. Opitz et al., A recurrent mutation in MED12 leading to R961W causes Opitz-Kaveggia syndrome, Nat. Genet, vol.39, pp.451-453, 2007.

K. Ritchie, C. Seah, J. Moulin, C. Isaac, F. Dick et al., Loss of ATRX leads to chromosome cohesion and congression defects, J. Cell Biol, vol.180, pp.315-324, 2008.

P. J. Robinson, M. J. Trnka, R. Pellarin, C. H. Greenberg, D. A. Bushnell et al., Molecular architecture of the yeast Mediator complex. Elife, vol.4, pp.1-29, 2015.

P. P. Rocha, W. Bleiss, and H. Schrewe, Mosaic expression of Med12 in female mice leads to exencephaly, spina bifida, and craniorachischisis, Birth Defects Res. Part A -Clin. Mol. Teratol, vol.88, pp.626-632, 2010.

J. Ropa, N. Saha, Z. Chen, J. Serio, W. Chen et al., PAF1 complex interactions with SETDB1 mediate promoter H3K9 methylation and transcriptional repression of Hoxa9 and Meis1 in acute myeloid leukemia, Oncotarget, vol.9, pp.22123-22136, 2018.

S. G. Rudd, N. C. Valerie, and T. Helleday, Pathways controlling dNTP pools to maintain genome stability, DNA Repair (Amst), vol.44, pp.193-204, 2016.

M. T. Russo, G. De-luca, P. Degan, E. Parlanti, E. Dogliotti et al., Accumulation of the oxidative base lesion 8-hydroxyguanine in DNA of tumor-prone mice defective in both the Myh and Ogg1 DNA glycosylases, Cancer Res, vol.64, pp.4411-4414, 2004.

U. Sahin, O. Ferhi, M. Jeanne, S. Benhenda, C. Berthier et al., Oxidative stress-induced assembly of PML nuclear bodies controls sumoylation of partner proteins, J. Cell Biol, vol.204, pp.931-945, 2014.
URL : https://hal.archives-ouvertes.fr/hal-00992287

S. Sainsbury, C. Bernecky, and P. Cramer, Structural basis of transcription initiation by RNA polymerase II, Nat. Rev. Mol. Cell Biol, vol.16, pp.129-143, 2015.

K. Sakamoto, Y. Tominaga, K. Yamauchi, Y. Nakatsu, K. Sakumi et al., MUTYH-null mice are susceptible to spontaneous and oxidative stress-induced intestinal tumorigenesis, Cancer Res, vol.67, pp.6599-6604, 2007.

K. Sakumi, Y. Tominaga, M. Furuichi, P. Xu, T. Tsuzuki et al., Ogg1 Knockout-associated Lung Tumorigenesis and Its Suppression by Mth 1 Gene Disruption, Cancer Res, vol.63, pp.902-905, 2003.

A. Sanyal, B. Lajoie, G. Jain, and J. Dekker, The long-range interaction landscape of gene promoters, Nature, vol.489, pp.109-113, 2012.

K. Satou, K. Kawai, H. Kasai, H. Harashima, and H. Kamiya, Mutagenic effects of 8-hydroxydGTP in live mammalian cells. Free Radic, Biol. Med, vol.42, pp.1552-1560, 2007.

U. Sattler, P. Frit, B. Salles, and P. Calsou, Long-patch DNA repair synthesis during base excision repair in mammalian cells, EMBO Rep, vol.4, pp.363-367, 2003.

O. D. Schärer, Nucleotide excision repair in eukaryotes, Cold Spring Harb Perspect Biol, vol.5, p.12609, 2013.

D. Schmidt, P. C. Schwalie, C. S. Ross-innes, A. Hurtado, G. D. Brown et al., A CTCF-independent role for cohesin in tissue-specific transcription, Genome Res, vol.20, pp.578-588, 2010.

J. Schmitz, E. Watrin, P. Lénárt, K. Mechtler, and J. M. Peters, Sororin Is Required for Stable Binding of Cohesin to Chromatin and for Sister Chromatid Cohesion in Interphase, Curr. Biol, vol.17, pp.630-636, 2007.

B. Schüle, A. Oviedo, K. Johnston, S. Pai, and U. Francke, Inactivating mutations in ESCO2 cause SC phocomelia and Roberts syndrome: no phenotype-genotype correlation, Am. J. Hum. Genet, vol.77, pp.1117-1145, 2005.

D. C. Schultz, K. Ayyanathan, D. Negorev, G. G. Maul, F. J. Rauscher et al., SETDB1 : a novel KAP-1 -associated histone H3 , lysine 9-specific methyltransferase that contributes to HP1-mediated silencing of euchromatic genes by KRAB zinc-finger proteins, Genes Dev, vol.16, pp.919-932, 2002.

D. C. Schultz, J. R. Friedman, and F. J. Rauscher, Targeting histone deacetylase complexes via KRAB-zinc finger proteins: The PHD and bromodomains of KAP-1 form a cooperative unit that recruits a novel isoform of the Mi-2? subunit of NuRD, Genes Dev, vol.15, pp.428-443, 2001.

R. C. Seet, C. Y. Lee, E. C. Lim, J. J. Tan, A. M. Quek et al., Oxidative damage in Parkinson disease: Measurement using accurate biomarkers. Free Radic, Biol. Med, vol.48, pp.560-566, 2010.

V. Seitan, B. Hao, K. Tachibana-konwalski, T. Lavagnolli, H. Mira-bontenbal et al., A role for cohesin in T-cell-receptor rearrangement and thymocyte differentiation, Nature, vol.476, pp.467-71, 2011.

S. Shibutani, M. Takeshita, and A. Grollman, Insertion of specific bases during DNA synthesis past the oxidation-damaged base 8-oxodG, Nature, vol.349, pp.431-435, 1991.

K. Shinmura, T. Kohno, H. Kasai, K. Koda, H. Sugimura et al., Infrequent mutations of the hOGG1 gene, that is involved in the excision of 8-hydroxyguanine in damaged DNA, in human gastric cancer, Japanese J. Cancer Res, vol.89, pp.825-828, 1998.

H. Sies, C. Berndt, and D. Jones, Oxidative stress, Annu. Rev. Biochem, vol.86, pp.715-763, 2017.

C. Sjögren and K. Nasmyth, Sister chromatid cohesion is required for postreplicative doublestrand break repair in Saccharomyces cerevisiae, Curr. Biol, vol.11, pp.271-279, 2001.

A. Sliwinska, P. Sitarek, M. Toma, P. Czarny, E. Synowiec et al., Decreased expression level of BER genes in Alzheimer's disease patients is not derivative of their DNA methylation status, Prog. Neuro-Psychopharmacology Biol. Psychiatry, vol.79, pp.311-316, 2017.

M. J. Smerdon, DNA repair and the role of chromatin structure, Curr. Opin. Cell Biol, vol.3, p.90069, 1991.

M. J. Smerdon and M. W. Lieberman, Nucleosome rearrangement in human chromatin during UV-induced DNA-repair synthesis, Proc. Natl. Acad. Sci. U. S. A, vol.75, pp.4238-4279, 1978.

D. Solomon, J. Kim, J. Bondaruk, S. Shariat, Z. Wang et al., Frequent truncating mutations of STAG2 in bladder cancer, Nat Genet, vol.45, 2013.

E. Sonoda, T. Matsusaka, C. Morrison, P. Vagnarelli, O. Hoshi et al., Scc1/Rad21/Mcd1 Is Required for Sister Chromatid Cohesion and Kinetochore Function in Vertebrate Cells, Dev. Cell, vol.1, pp.759-770, 2001.

G. Soria, S. E. Polo, and G. Almouzni, Prime, Repair, Restore: The Active Role of Chromatin in the DNA Damage Response, Mol. Cell, vol.46, pp.722-734, 2012.

M. Sossou, C. Flohr-beckhaus, I. Schulz, F. Daboussi, B. Epe et al., APE1 overexpression in XRCC1-deficient cells complements the defective repair of oxidative single strand breaks but increases genomic instability, Nucleic Acids Res, vol.33, pp.298-306, 2005.

J. Soutourina, Transcription regulation by the Mediator complex, Nat. Rev. Mol. Cell Biol, vol.19, pp.262-274, 2018.
URL : https://hal.archives-ouvertes.fr/cea-01886009

J. M. Spaeth, N. H. Kim, and T. G. Boyer, Mediator and human disease, Semin Cell Dev Biol, vol.22, pp.776-787, 2011.

D. L. Spector, Nuclear domains. J. Cell Sci, vol.114, pp.2891-2894, 2001.

D. L. Spector, The Dynamics of Chromosome Organization and Gene Regulation, Annu. Rev. Biochem, vol.72, pp.573-608, 2003.

G. Spivak, Nucleotide excision repair in humans, DNA Repair (Amst), vol.36, pp.13-18, 2015.

S. P. Sripathy, J. Stevens, and D. C. Schultz, The KAP1 Corepressor Functions To Coordinate the Assembly of De Novo HP1-Demarcated Microenvironments of Heterochromatin Required for KRAB Zinc Finger Protein-Mediated Transcriptional Repression, Mol. Cell. Biol, vol.26, pp.8623-8638, 2006.

D. Stan?k and A. Fox, Nuclear bodies: news insights into structure and function, Curr. Opin. Cell Biol, vol.46, pp.94-101, 2017.

B. D. Strahl and C. D. Allis, The language of covalent histone modifications, Nature, vol.403, pp.41-45, 2000.

L. Ström, H. B. Lindroos, K. Shirahige, and C. Sjögren, Postreplicative recruitment of cohesin to double-strand breaks is required for DNA repair, Mol. Cell, vol.16, pp.1003-1015, 2004.

A. Strunnikov, V. Larionov, and D. Koshland, SMCI: An Essential Yeast Gene Encoding a Putative Head-Rod-Tail Protein Is Required for Nuclear Division and Defines a New Ubiquitous Protein Family, J. Cell Biol, vol.123, pp.1635-1648, 1993.

D. M. Stults, M. W. Killen, H. H. Pierce, and A. J. Pierce, Genomic architecture and inheritance of human ribosomal RNA gene clusters, Genome Res, vol.18, pp.13-18, 2008.

C. Sung, Y. Hsu, C. Chen, Y. Lin, and C. Wu, Oxidative Stress and Nucleic Acid Oxidation in Patients with Chronic Kidney Disease, Oxid. Med. Cell. Longev, vol.2013, pp.1-15, 2013.

J. Q. Svejstrup, Y. Li, J. Fellows, A. Gnatt, S. Bjorklund et al., Evidence for a mediator cycle at the initiation of transcription, Proc. Natl. Acad. Sci. U. S. A, vol.94, pp.6075-6083, 1997.

D. J. Taatjes, The human Mediator complex: a versatile, genome-wide regulator of transcription, Trends Biochem. Sci, vol.35, pp.315-322, 2010.

D. J. Taatjes, A. M. Na, E. Nogales, and R. Tjian, Structure, Function, and Conformations of the CRSP Coactivator, Science, vol.295, pp.1058-1062, 2002.

Y. Tahara, D. Auld, D. Ji, A. Beharry, A. Kietrys et al., Potent and selective inhibitors of 8-oxoguanine DNA glycosylase, J Am Chem Soc, vol.140, pp.2105-2114, 2018.

H. Takahashi, T. Parmely, S. Sato, C. Tomoromi-sato, C. Banks et al., Human Mediator subunit MED26 functions as a docking site for transcription elongation factors, Cell, vol.146, pp.92-104, 2011.

T. Tanaka, J. Fuchs, J. Loidl, and K. Nasmyth, Cohesin ensures bipolar attachment of microtubules to sister centromeres and resists their precocious separation, Nat. Cell Biol, vol.2, pp.492-499, 2000.

C. M. Thompson, A. J. Koleske, D. M. Chao, and R. A. Young, A multisubunit complex associated with the RNA polymerase II CTD and TATA-binding protein in yeast, Cell, vol.73, pp.1361-1375, 1993.

R. E. Thurman, E. Rynes, R. Humbert, J. Vierstra, M. T. Maurano et al.,

V. Reynolds, A. Roach, M. E. Safi, A. Sanchez, A. Sanyal et al., The accessible chromatin landscape of the human genome, Nature, vol.489, pp.75-82, 2012.

P. Tropberger, S. Pott, C. Keller, K. Kamieniarz-gdula, M. Caron et al., Regulation of transcription through acetylation of H3K122 on the lateral surface of the histone octamer, Cell, vol.152, pp.859-872, 2013.

K. Tsai, S. Shigeo, C. Tomoromi-sato, R. Conaway, J. Conaway et al., A conserved Mediator-CDK8 kinase module association regulates Mediator-RNA polymerase II interaction, Nat Struc Mol Biol, vol.20, pp.611-619, 2013.

K. Tsai, C. Tomoromi-sato, S. Sato, C. Rc, C. Jw et al., Subunit architecture and functional modular rearrangements of the transcriptional Mediator complex, Cell, vol.157, pp.1430-1444, 2014.

J. Tuo, C. Chen, X. Zeng, M. Christiansen, and V. A. Bohr, Functional crosstalk between hOgg1 and the helicase domain of Cockayne syndrome group B protein, DNA Repair (Amst), vol.1, pp.913-927, 2002.

F. Uhlmann, SMC complexes: From DNA to chromosomes, Nat. Rev. Mol. Cell Biol, vol.17, pp.399-412, 2016.

F. Uhlmann and K. Nasmyth, Cohesion between sister chromatids must be established during DNA replication, Curr. Biol, vol.8, pp.1095-1102, 1998.

E. Ünal, A. Arbel-eden, U. Sattler, R. Shroff, M. Lichten et al., DNA damage response pathway uses histone modification to assemble a double-strand break-specific cohesin domain, Mol. Cell, vol.16, pp.991-1002, 2004.

B. Uttara, A. Singh, P. Zamboni, and R. T. Mahajan, Oxidative stress and neurodegenerative diseases: a review of upstream and downstream antioxidant therapeutic options, Curr. Neuropharmacol, vol.7, pp.65-74, 2009.

D. Valle-garcía, Z. A. Qadeer, D. S. Mchugh, F. G. Ghiraldini, A. H. Chowdhury et al., ATRX binds to atypical chromatin domains at the 3? exons of zinc finger genes to preserve H3K9me3 enrichment, Epigenetics, vol.11, pp.398-414, 2016.

A. Vannini and P. Cramer, Conservation between the RNA Polymerase I, II, and III Transcription Initiation Machineries, Mol. Cell, vol.45, pp.439-446, 2012.

J. M. Vaquerizas, S. K. Kummerfeld, S. A. Teichmann, and N. M. Luscombe, A census of human transcription factors: Function, expression and evolution, Nat. Rev. Genet, vol.10, pp.252-263, 2009.

H. Vega, A. H. Trainer, M. Gordillo, M. Crosier, H. Kayserili et al., Phenotypic variability in 49 cases of ESCO2 mutations, including novel missense and codon deletion in the acetyltransferase domain, correlates with ESCO2 expression and establishes the clinical criteria for Roberts syndrome, J. Med. Genet, vol.47, pp.30-37, 2010.

A. Vidal, I. Hickson, S. Boiteux, and J. Radiclla, Mechanism of stimulation of the DNA glycosylase activity of hOGG1 by the major human AP endonuclease: bypass of the AP lyase activity step, Nucleic Acids Res, vol.29, pp.1285-1292, 2001.

A. E. Vidal, S. Boiteux, I. D. Hickson, and J. P. Radicella, XRCC1 coordinates the initial and late stages of DNA abasic site repair through protein-protein interactions, EMBO J, vol.20, pp.6530-6539, 2001.

R. Vijayvargia, M. S. May, and J. D. Fondell, A coregulatory role for the mediator complex in prostate cancer cell proliferation and gene expression, Cancer Res, vol.67, pp.4034-4041, 2007.

M. G. Vrouwe, E. Elghalbzouri-maghrani, M. Meijers, P. Schouten, B. C. Godthelp et al., Increased DNA damage sensitivity of Cornelia de Lange syndrome cells: Evidence for impaired recombinational repair, Hum. Mol. Genet, vol.16, pp.1478-1487, 2007.

S. S. Wallace, D. L. Murphy, and J. B. Sweasy, Base excision repair and cancer, Cancer Lett, vol.327, pp.73-89, 2012.

X. Q. Wang and J. Dostie, Chromosome folding and its regulation in health and disease, Curr. Opin. Genet. Dev, vol.43, pp.23-30, 2017.

E. Watrin, F. J. Kaiser, and K. S. Wendt, Gene regulation and chromatin organization: Relevance of cohesin mutations to human disease, Curr. Opin. Genet. Dev, vol.37, pp.59-66, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01263104

J. M. Weiss, E. L. Goode, W. C. Ladiges, and C. M. Ulrich, Polymorphic variation in hOGG1 and risk of cancer: A review of the functional and epidemiologic literature, Mol. Carcinog, vol.42, pp.127-141, 2005.

K. S. Wendt, K. Yoshida, T. Itoh, M. Bando, B. Koch et al., Cohesin mediates transcriptional insulation by CCCTC-binding factor, Nature, vol.451, pp.796-801, 2008.

A. G. West, M. Gaszner, and G. Felsenfeld, Insulators : many functions , many mechanisms Insulators : many functions, many mechanisms, Genes Dev, vol.16, pp.271-288, 2002.

O. Will, E. Gocke, I. Eckert, I. Schulz, M. Pflaum et al., Oxidative DNA damage and mutations induced by a polar photosensitizer, Ro19-8022, Mutat. Res. -DNA Repair, vol.435, pp.89-101, 1999.

J. L. Witztum and D. Steinberg, Role of oxidized low density lipoprotein in atherogenesis, J. Clin. Invest, vol.88, pp.1785-1792, 1991.

H. K. Wong, M. Muftuoglu, G. Beck, S. Z. Imam, V. A. Bohr et al., Cockayne syndrome B protein stimulates apurinic endonuclease 1 activity and protects against agents that introduce base excision repair intermediates, Nucleic Acids Res, vol.35, pp.4103-4113, 2007.

S. Xanthoudakis, G. Miao, F. Wang, Y. C. Pan, and T. Curran, Redox activation of Fos-Jun DNA binding activity is mediated by a DNA repair enzyme, EMBO J, vol.11, pp.3323-3358, 1992.

Y. Xie, H. Yang, C. Cunanan, K. Okamoto, D. Shibata et al., Deficiencies in Mouse Myh and Ogg1 Result in Tumor Predisposition and G to T Mutations in Codon 12 of the K-Ras Oncogene in Lung Tumors, Cancer Res, vol.64, pp.3096-3102, 2004.

F. Xu, K. Zhang, and M. Grunstein, Acetylation in histone H3 globular domain regulates gene expression in yeast, Cell, vol.121, pp.375-385, 2005.

M. Yan, H. Xu, N. Waddell, K. Shield-artin, I. Haviv et al., Enhanced RAD21 cohesin expression confers poor prognosis in BRCA2 and BRCAX, but not BRCA1 familial breast cancers, Breast Cancer Res, vol.14, p.69, 2012.

C. Yang, E. Bolotin, T. Jiang, F. Sladek, and E. Martinez, Prevalence of the initiator over the TATA box in human and yeast genes and identification of DNA motifs enriched in human TATA-less core promoters, Gene, vol.389, pp.52-65, 2007.

P. T. Yazdi, Y. Wang, S. Zhao, N. Patel, E. Y. Lee et al., SMC1 is a downstream effector in the ATM/NBS1 branch of the human S-phase checkpoint, Genes Dev, vol.16, pp.571-582, 2002.

P. Yeung, N. Denissova, C. Nasello, Z. Hakhverdyan, J. Chen et al., Promyelocytic leukemia nuclear bodies support a late step in DNA double-strand break repair by homologous recombination, J. Biol. Chem, vol.113, pp.1787-99, 2012.

M. Yoshihara, L. Jiang, S. Akatsuka, M. Suyama, and S. Toyokuni, Genome-wide profiling of 8-oxoguanine reveals its association with spatial positioning in nucleus, DNA Res, vol.21, pp.603-612, 2014.

B. Zhang, S. Jain, H. Song, M. Fu, R. O. Heuckeroth et al., Mice lacking sister chromatid cohesion protein PDS5B exhibit developmental abnormalities reminiscent of Cornelia de Lange syndrome, Development, vol.134, pp.3191-3201, 2007.

H. Zhou, J. M. Spaeth, N. H. Kim, X. Xu, M. J. Friez et al., MED12 mutations link intellectual disability syndromes with dysregulated GLI3-dependent Sonic Hedgehog signaling, Proc. Natl. Acad. Sci. U. S. A, vol.109, pp.19763-19768, 2012.

J. Zhou, M. Liu, A. M. Fleming, C. J. Burrows, and S. S. Wallace, Neil3 and NEIL1 DNA glycosylases remove oxidative damages from quadruplex DNA and exhibit preferences for lesions in the telomeric sequence context, J. Biol. Chem, vol.288, pp.27263-27272, 2013.

X. Zhou, W. Wang, C. Du, F. Yan, S. Yang et al., OGG1 regulates the level of symmetric dimethylation of histone H4 arginine-3 by interacting with PRMT5, Mol. Cell. Probes, vol.38, pp.19-24, 2018.

Y. Ziv, D. Bielopolski, Y. Galanty, C. Lukas, Y. Taya et al., Chromatin relaxation in response to DNA double-strand breaks is modulated by a novel ATM-and KAP-1 dependent pathway, Nat. Cell Biol, vol.8, pp.870-876, 2006.

M. Zolan, C. Smith, C. Nm, and H. Pc, Rearrangement of mammalian chromatin structure following excision repair, Nature, vol.299, pp.462-466, 1982.

G. Zou and A. Maitra, Small-molecule inhibitor of the AP endonuclease 1/REF-1 E3330 inhibits pancreatic cancer cell growth and migration, Mol. Cancer Ther, vol.7, pp.2012-2021, 2008.

J. Zuin, J. R. Dixon, M. I. Van-der-reijden, Z. Ye, P. Kolovos et al.,

. Wendt, Cohesin and CTCF differentially affect chromatin architecture and gene expression in human cells, Proc. Natl. Acad. Sci. U. S. A, vol.111, pp.996-1001, 2014.