R. D. Kornberg, Chromatin Structure: A Repeating Unit of Histones and DNA, Science, vol.184, issue.4139, pp.868-871, 1974.
DOI : 10.1126/science.184.4139.868

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

T. Kouzarides, Chromatin Modifications and Their Function, Cell, vol.128, issue.4, pp.693-705, 2007.
DOI : 10.1016/j.cell.2007.02.005

A. Zweidler, Core histone variants of the mouse : Primary structure and differential expression, in Histones genes, structure, organization and regulation, pp.339-371, 1984.

P. B. Talbert and S. Henikoff, Histone variants ??? ancient wrap artists of the epigenome, Nature Reviews Molecular Cell Biology, vol.8, issue.4, pp.264-275, 2010.
DOI : 10.1038/nrm2861

D. Koning, L. Corpet, A. Haber, J. E. Almouzni, and G. , Histone chaperones: an escort network regulating histone traffic, Nature Structural & Molecular Biology, vol.15, issue.11, pp.997-1007, 2007.
DOI : 10.1016/S0092-8674(00)80321-9

G. A. Orsi, P. Couble, and B. Loppin, Epigenetic and replacement roles of histone variant H3.3 in reproduction and development, The International Journal of Developmental Biology, vol.53, issue.2-3, pp.231-243, 2009.
DOI : 10.1387/ijdb.082653go

URL : https://hal.archives-ouvertes.fr/hal-00425198

S. J. Elsaesser, A. D. Goldberg, and C. Allis, New functions for an old variant: no substitute for histone H3.3, Current Opinion in Genetics & Development, vol.20, issue.2, 2010.
DOI : 10.1016/j.gde.2010.01.003

L. A. Banaszynski, C. D. Allis, and P. W. Lewis, Histone Variants in Metazoan Development, Developmental Cell, vol.19, issue.5, pp.662-674, 2010.
DOI : 10.1016/j.devcel.2010.10.014

R. C. Allshire and G. H. Karpen, Epigenetic regulation of centromeric chromatin: old dogs, new tricks?, Nature Reviews Genetics, vol.293, issue.12, pp.923-937, 2008.
DOI : 10.1038/nrg2466

O. Witt, W. Albig, and D. Doenecke, Testis-Specific Expression of a Novel Human H3 Histone Gene, Experimental Cell Research, vol.229, issue.2, pp.301-306, 1996.
DOI : 10.1006/excr.1996.0375

S. M. Wiedemann, S. N. Mildner, and C. Bonisch, Identification and characterization of two novel primate-specific histone H3 variants, H3.X and H3.Y, The Journal of Cell Biology, vol.259, issue.5, pp.777-791, 2010.
DOI : 10.1083/jcb.201002043.dv

W. F. Marzluff, P. Gongidi, K. R. Woods, J. Jin, and L. J. Maltais, The Human and Mouse Replication-Dependent Histone Genes, Genomics, vol.80, issue.5, pp.487-498, 2002.
DOI : 10.1006/geno.2002.6850

M. A. Osley, The Regulation of Histone Synthesis in the Cell Cycle, Annual Review of Biochemistry, vol.60, issue.1, pp.827-861, 1991.
DOI : 10.1146/annurev.bi.60.070191.004143

S. E. Polo, D. Roche, and G. Almouzni, New Histone Incorporation Marks Sites of UV Repair in Human Cells, Cell, vol.127, issue.3, pp.481-493, 2006.
DOI : 10.1016/j.cell.2006.08.049

D. Frank, D. Doenecke, and W. Albig, Differential expression of human replacement and cell cycle dependent H3 histone genes, Gene, vol.312, pp.135-143, 2003.
DOI : 10.1016/S0378-1119(03)00609-7

D. B. Krimer, G. Cheng, and A. Skoultchi, Induction of H3.3 replacement histone mRNAs during the precommitment period of murine erythroleukemia cell differentiation, Nucleic Acids Research, vol.21, issue.12, pp.2873-2879, 1993.
DOI : 10.1093/nar/21.12.2873

D. Castiglia, A. Cestelli, M. Scaturro, T. Nastasi, D. Liegro et al., H1?? and H3.3B mRNA levels in developing rat brain, Neurochemical Research, vol.112, issue.12, pp.1531-1537, 1994.
DOI : 10.1007/BF00969002

R. S. Wu, S. Tsai, and W. M. Bonner, Patterns of histone variant synthesis can distinguish go from G1 cells, Cell, vol.31, issue.2, pp.367-374, 1982.
DOI : 10.1016/0092-8674(82)90130-1

H. S. Malik and S. Henikoff, Phylogenomics of the nucleosome, Nature Structural Biology, vol.10, issue.11, pp.882-891, 2003.
DOI : 10.1038/nsb996

S. B. Hake, B. A. Garcia, and M. Kauer, Serine 31 phosphorylation of histone variant H3.3 is specific to regions bordering centromeres in metaphase chromosomes, Proceedings of the National Academy of Sciences, vol.102, issue.18, pp.6344-6349, 2005.
DOI : 10.1073/pnas.0502413102

K. Ahmad and S. Henikoff, The Histone Variant H3.3 Marks Active Chromatin by Replication-Independent Nucleosome Assembly, Molecular Cell, vol.9, issue.6, pp.1191-1200, 2002.
DOI : 10.1016/S1097-2765(02)00542-7

URL : https://hal.archives-ouvertes.fr/hal-00782360

A. D. Goldberg, L. A. Banaszynski, and K. M. Noh, Distinct Factors Control Histone Variant H3.3 Localization at Specific Genomic Regions, Cell, vol.140, issue.5, pp.678-691, 2010.
DOI : 10.1016/j.cell.2010.01.003

S. B. Hake and C. D. Allis, Histone H3 variants and their potential role in indexing mammalian genomes: The "H3 barcode hypothesis", Proceedings of the National Academy of Sciences, vol.103, issue.17, pp.6428-6435, 2006.
DOI : 10.1073/pnas.0600803103

C. Jin and G. Felsenfeld, Nucleosome stability mediated by histone variants H3.3 and H2A.Z, Genes & Development, vol.21, issue.12, pp.1519-1529, 2007.
DOI : 10.1101/gad.1547707

M. Xu, C. Long, and X. Chen, Partitioning of Histone H3-H4 Tetramers During DNA Replication-Dependent Chromatin Assembly, Science, vol.328, issue.5974, pp.94-98, 2010.
DOI : 10.1126/science.1178994

D. Ray-gallet and G. Almouzni, Mixing or Not Mixing, Science, vol.328, issue.5974, pp.56-57, 2010.
DOI : 10.1126/science.1188653

J. H. Waterborg, Sequence analysis of acetylation and methylation in two histone H3 variants of alfalfa, J Biol Chem, vol.265, pp.17157-17161, 1990.

E. Mckittrick, P. R. Gafken, K. Ahmad, and S. Henikoff, From The Cover: Histone H3.3 is enriched in covalent modifications associated with active chromatin, Proceedings of the National Academy of Sciences, vol.101, issue.6, pp.1525-1530, 2004.
DOI : 10.1073/pnas.0308092100

C. M. Chow, A. Georgiou, and H. Szutorisz, Variant histone H3.3 marks promoters of transcriptionally active genes during mammalian cell division, EMBO reports, vol.19, issue.4, pp.354-360, 2005.
DOI : 10.1073/pnas.0308506101

A. Loyola, T. Bonaldi, D. Roche, A. Imhof, and G. Almouzni, PTMs on H3 Variants before Chromatin Assembly Potentiate Their Final Epigenetic State, Molecular Cell, vol.24, issue.2, pp.309-316, 2006.
DOI : 10.1016/j.molcel.2006.08.019

S. B. Hake, B. A. Garcia, and E. M. Duncan, Expression Patterns and Post-translational Modifications Associated with Mammalian Histone H3 Variants, Journal of Biological Chemistry, vol.281, issue.1, pp.559-568, 2006.
DOI : 10.1074/jbc.M509266200

A. Loyola and G. Almouzni, Marking histone H3 variants: How, when and why?, Trends in Biochemical Sciences, vol.32, issue.9, pp.425-433, 2007.
DOI : 10.1016/j.tibs.2007.08.004

U. Braunschweig, G. J. Hogan, L. Pagie, and B. Van-steensel, Histone H1 binding is inhibited by histone variant H3.3, The EMBO Journal, vol.9, issue.23, pp.3635-3645, 2009.
DOI : 10.1126/science.1112178

B. E. Schwartz and K. Ahmad, Transcriptional activation triggers deposition and removal of the histone variant H3.3, Genes & Development, vol.19, issue.7, pp.804-814, 2005.
DOI : 10.1101/gad.1259805

E. Delbarre, B. M. Jacobsen, and A. H. Reiner, Chromatin Environment of Histone Variant H3.3 Revealed by Quantitative Imaging and Genome-scale Chromatin and DNA Immunoprecipitation, Molecular Biology of the Cell, vol.21, issue.11, pp.1872-1884, 2010.
DOI : 10.1091/mbc.E09-09-0839

C. Wirbelauer, O. Bell, and D. Schubeler, Variant histone H3.3 is deposited at sites of nucleosomal displacement throughout transcribed genes while active histone modifications show a promoter-proximal bias, Genes & Development, vol.19, issue.15, pp.1761-1766, 2005.
DOI : 10.1101/gad.347705

L. Daury, C. Chailleux, J. Bonvallet, and D. Trouche, Histone H3.3 deposition at E2F-regulated genes is linked to transcription, EMBO reports, vol.35, issue.1, pp.66-71, 2006.
DOI : 10.1016/S0092-8674(03)01064-X

URL : https://hal.archives-ouvertes.fr/hal-00021145

Y. Mito, J. G. Henikoff, and S. Henikoff, Genome-scale profiling of histone H3.3 replacement patterns, Nature Genetics, vol.35, issue.10, pp.1090-1097, 2005.
DOI : 10.1038/12640

T. Tamura, M. Smith, and T. Kanno, Inducible Deposition of the Histone Variant H3.3 in Interferon-stimulated Genes, Journal of Biological Chemistry, vol.284, issue.18, pp.12217-12225, 2009.
DOI : 10.1074/jbc.M805651200

C. Jin, C. Zang, and G. Wei, H3.3/H2A.Z double variant???containing nucleosomes mark 'nucleosome-free regions' of active promoters and other regulatory regions, Nature Genetics, vol.41, issue.8, pp.941-945, 2009.
DOI : 10.1038/76469

URL : https://hal.archives-ouvertes.fr/hal-01292426

T. Nakayama, K. Nishioka, Y. X. Dong, T. Shimojima, and S. Hirose, Drosophila GAGA factor directs histone H3.3 replacement that prevents the heterochromatin spreading, Genes & Development, vol.21, issue.5, pp.552-561, 2007.
DOI : 10.1101/gad.1503407

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC1820897

B. E. Schwartz and K. Ahmad, Chromatin Assembly with H3 Histones: Full Throttle Down Multiple Pathways, Curr Top Dev Biol, vol.74, issue.2, pp.31-55, 2006.
DOI : 10.1016/S0070-2153(06)74002-9

S. Henikoff, Nucleosome destabilization in the epigenetic regulation of gene expression, Nature Reviews Genetics, vol.7, issue.1, pp.15-26, 2008.
DOI : 10.1038/nrg2206

R. K. Ng and J. B. Gurdon, Epigenetic memory of an active gene state depends on histone H3.3 incorporation into chromatin in the absence of transcription, Nature Cell Biology, vol.101, issue.1, pp.102-109, 2008.
DOI : 10.1038/ncb1674

A. Sakai, B. E. Schwartz, S. Goldstein, and K. Ahmad, Transcriptional and Developmental Functions of the H3.3 Histone Variant in Drosophila, Current Biology, vol.19, issue.21, pp.1816-1820, 2009.
DOI : 10.1016/j.cub.2009.09.021

P. Drané, K. Ouararhni, A. Depaux, M. Shuaib, and A. Hamiche, The deathassociated protein DAXX is a novel histone chaperone involved in the replication-independent deposition of H3, Genes Dev, vol.3, issue.24, pp.1253-1265, 2010.

A. Santenard, C. Ziegler-birling, and M. Koch, Heterochromatin formation in the mouse embryo requires critical residues of the histone variant H3.3, Nature Cell Biology, vol.13, issue.9, pp.853-862, 2010.
DOI : 10.1038/ncb2089

L. H. Wong, H. Ren, and E. Williams, Histone H3.3 incorporation provides a unique and functionally essential telomeric chromatin in embryonic stem cells Chromosome-wide nucleosome replacement and H3.3 incorporation during mammalian meiotic sex chromosome inactivation, Genome Res Nat Genet, vol.19, issue.39, pp.404-414251, 2007.

R. Balhorn, The protamine family of sperm nuclear proteins, Genome Biology, vol.8, issue.9, pp.227-53, 2007.
DOI : 10.1186/gb-2007-8-9-227

B. Loppin, E. Bonnefoy, and C. Anselme, The histone H3.3 chaperone HIRA is essential for chromatin assembly in the male pronucleus, Nature, vol.15, issue.7063, pp.1386-13901008, 2005.
DOI : 10.1126/science.1092727

URL : https://hal.archives-ouvertes.fr/hal-00391456

M. E. Torres-padilla, A. J. Bannister, P. J. Hurd, T. Kouzarides, and M. Zernicka-goetz, Dynamic distribution of the replacement histone variant H3.3 in the mouse oocyte and preimplantation embryos, The International Journal of Developmental Biology, vol.50, issue.Next, pp.455-461, 2006.
DOI : 10.1387/ijdb.052073mt

A. V. Probst, I. Okamoto, and M. Casanova, A Strand-Specific Burst in Transcription of Pericentric Satellites Is Required for Chromocenter Formation and Early Mouse Development, Developmental Cell, vol.19, issue.4, pp.625-638, 2010.
DOI : 10.1016/j.devcel.2010.09.002

URL : https://hal.archives-ouvertes.fr/hal-00685732

H. Tagami, D. Ray-gallet, G. Almouzni, and Y. Nakatani, Histone H3.1 and H3.3 Complexes Mediate Nucleosome Assembly Pathways Dependent or Independent of DNA Synthesis, Cell, vol.116, issue.1, pp.51-61, 2004.
DOI : 10.1016/S0092-8674(03)01064-X

B. Stillman, Chromatin assembly during SV40 DNA replication in vitro, Cell, vol.45, issue.4, pp.555-565, 1986.
DOI : 10.1016/0092-8674(86)90287-4

P. H. Gaillard, E. M. Martini, and P. D. Kaufman, Chromatin Assembly Coupled to DNA Repair: A New Role for Chromatin Assembly Factor I, Cell, vol.86, issue.6, pp.887-896, 1996.
DOI : 10.1016/S0092-8674(00)80164-6

D. Ray-gallet, J. P. Quivy, and C. Scamps, HIRA Is Critical for a Nucleosome Assembly Pathway Independent of DNA Synthesis, Molecular Cell, vol.9, issue.5, pp.1091-1100, 2002.
DOI : 10.1016/S1097-2765(02)00526-9

B. Loppin, M. Docquier, F. Bonneton, and P. Couble, The Maternal Effect Mutation s??same Affects the Formation of the Male Pronucleus in Drosophila melanogaster, Developmental Biology, vol.222, issue.2, pp.392-404, 2000.
DOI : 10.1006/dbio.2000.9718

P. Hajkova, K. Ancelin, and T. Waldmann, Chromatin dynamics during epigenetic reprogramming in the mouse germ line, Nature, vol.171, issue.7189, pp.877-881, 2008.
DOI : 10.1038/nature06714

G. Banumathy, N. Somaiah, and R. Zhang, Human UBN1 Is an Ortholog of Yeast Hpc2p and Has an Essential Role in the HIRA/ASF1a Chromatin-Remodeling Pathway in Senescent Cells, Molecular and Cellular Biology, vol.29, issue.3, pp.758-770, 2009.
DOI : 10.1128/MCB.01047-08

S. Balaji, L. M. Iyer, and L. Aravind, HPC2 and ubinuclein define a novel family of histone chaperones conserved throughout eukaryotes, Molecular BioSystems, vol.31, issue.3, pp.269-275, 2009.
DOI : 10.1039/b816424j

S. J. Elsaesser and C. D. Allis, HIRA and Daxx Constitute Two Independent Histone H3.3-Containing Predisposition Complexes, Cold Spring Harb Symp Quant Biol, 2010.
DOI : 10.1101/sqb.2010.75.008

P. Salomoni and A. F. Khelifi, Daxx: death or survival protein?, Trends in Cell Biology, vol.16, issue.2, pp.97-104, 2006.
DOI : 10.1016/j.tcb.2005.12.002

R. J. Gibbons, D. J. Picketts, L. Villard, and D. R. Higgs, Mutations in a putative global transcriptional regulator cause X-linked mental retardation with ??-thalassemia (ATR-X syndrome), Cell, vol.80, issue.6, pp.837-845, 1995.
DOI : 10.1016/0092-8674(95)90287-2

T. L. Mcdowell, R. J. Gibbons, and H. Sutherland, Localization of a putative transcriptional regulator (ATRX) at pericentromeric heterochromatin and the short arms of acrocentric chromosomes, Proceedings of the National Academy of Sciences, vol.96, issue.24, pp.13983-13988, 1999.
DOI : 10.1073/pnas.96.24.13983

Y. Xue, R. Gibbons, and Z. Yan, The ATRX syndrome protein forms a chromatin-remodeling complex with Daxx and localizes in promyelocytic leukemia nuclear bodies, Proceedings of the National Academy of Sciences, vol.100, issue.19, pp.10635-10640, 2003.
DOI : 10.1073/pnas.1937626100

P. W. Lewis, S. J. Elsaesser, K. M. Noh, S. C. Stadler, and C. D. Allis, Daxx is an H3.3-specific histone chaperone and cooperates with ATRX in replication-independent chromatin assembly at telomeres, Proceedings of the National Academy of Sciences, vol.107, issue.32, pp.14075-14080, 2010.
DOI : 10.1073/pnas.1008850107

M. J. Law, K. M. Lower, and H. P. Voon, ATR-X Syndrome Protein Targets Tandem Repeats and Influences Allele-Specific Expression in a Size-Dependent Manner, Cell, vol.143, issue.3, pp.367-378, 2010.
DOI : 10.1016/j.cell.2010.09.023

R. S. Rogers, A. Inselman, M. A. Handel, and M. J. Matunis, SUMO modified proteins localize to the XY body of pachytene spermatocytes, Chromosoma, vol.2, issue.Pt 14, pp.233-243, 2004.
DOI : 10.1007/s00412-004-0311-7

A. Y. Konev, M. Tribus, and S. Y. Park, CHD1 Motor Protein Is Required for Deposition of Histone Variant H3.3 into Chromatin in Vivo, Science, vol.317, issue.5841, pp.1087-1090, 2007.
DOI : 10.1126/science.1145339

S. Sawatsubashi, T. Murata, and J. Lim, A histone chaperone, DEK, transcriptionally coactivates a nuclear receptor, Genes & Development, vol.24, issue.2, pp.159-170, 2010.
DOI : 10.1101/gad.1857410

A. D. Hollenbach, C. J. Mcpherson, E. J. Mientjes, R. Iyengar, and G. Grosveld, Daxx and histone deacetylase II associate with chromatin through an interaction with core histones and the chromatin-associated protein Dek, J Cell Sci, vol.115, pp.3319-3330, 2002.

E. Bonnefoy, G. A. Orsi, P. Couble, and B. Loppin, The essential role of Drosophila HIRA for de novo assembly of paternal chromatin at fertilization, PLoS Genet, vol.3, pp.1991-2006, 2007.
URL : https://hal.archives-ouvertes.fr/hal-00176483

J. Postberg, S. Forcob, W. J. Chang, and H. J. Lipps, The evolutionary history of histone H3 suggests a deep eukaryotic root of chromatin modifying mechanisms, BMC Evolutionary Biology, vol.10, issue.1, pp.259-78, 2010.
DOI : 10.1186/1471-2148-10-259

E. S. Choi, J. A. Shin, H. S. Kim, and Y. K. Jang, Dynamic regulation of replication independent deposition of histone H3 in fission yeast, Nucleic Acids Research, vol.33, issue.22, pp.7102-7110, 2005.
DOI : 10.1093/nar/gki1011

A. Jamai, R. M. Imoberdorf, and M. Strubin, Continuous Histone H2B and Transcription-Dependent Histone H3 Exchange in Yeast Cells outside of Replication, Molecular Cell, vol.25, issue.3, pp.345-355, 2007.
DOI : 10.1016/j.molcel.2007.01.019

A. Rufiange, P. E. Jacques, W. Bhat, F. Robert, and A. Nourani, Genome-Wide Replication-Independent Histone H3 Exchange Occurs Predominantly at Promoters and Implicates H3 K56 Acetylation and Asf1, Molecular Cell, vol.27, issue.3, pp.393-405, 2007.
DOI : 10.1016/j.molcel.2007.07.011

M. F. Dion, T. Kaplan, and M. Kim, Dynamics of Replication-Independent Histone Turnover in Budding Yeast, Science, vol.315, issue.5817, pp.1405-1408, 2007.
DOI : 10.1126/science.1134053

P. Prochasson, L. Florens, S. K. Swanson, M. P. Washburn, and J. L. Workman, The HIR corepressor complex binds to nucleosomes generating a distinct protein/DNA complex resistant to remodeling by SWI/SNF, Genes & Development, vol.19, issue.21, pp.2534-2539, 2005.
DOI : 10.1101/gad.1341105

E. M. Green, A. J. Antczak, and A. O. Bailey, Replication-Independent Histone Deposition by the HIR Complex and Asf1, Current Biology, vol.15, issue.22, pp.2044-2049, 2005.
DOI : 10.1016/j.cub.2005.10.053

H. E. Anderson, A. Kagansky, and J. Wardle, Silencing Mediated by the Schizosaccharomyces pombe HIRA Complex Is Dependent upon the Hpc2-Like Protein, Hip4, SPT16 or POB3 (yFACT) in Saccharomyces cerevisiae cause dependence on the Hir/Hpc pathway: polymerase passage may degrade chromatin structure, pp.1557-1571, 2002.
DOI : 10.1371/journal.pone.0013488.t002

B. Cui, Y. Liu, and M. A. Gorovsky, Deposition and Function of Histone H3 Variants in Tetrahymena thermophila, Molecular and Cellular Biology, vol.26, issue.20, pp.7719-7730, 2006.
DOI : 10.1128/MCB.01139-06

M. Hodl and K. Basler, Transcription in the Absence of Histone H3.3, Current Biology, vol.19, issue.14, pp.1221-1226, 2009.
DOI : 10.1016/j.cub.2009.05.048

U. Gunesdogan, H. Jackle, and A. Herzig, A genetic system to assess in vivo the functions of histones and histone modifications in higher eukaryotes, EMBO reports, vol.115, issue.10, pp.772-776, 2010.
DOI : 10.1242/jcs.01523

J. Wysocka, T. Swigut, and T. A. Milne, WDR5 Associates with Histone H3 Methylated at K4 and Is Essential for H3 K4 Methylation and Vertebrate Development, Cell, vol.121, issue.6, pp.859-872, 2005.
DOI : 10.1016/j.cell.2005.03.036

J. B. Gurdon, The developmental capacity of nuclei taken from intestinal epithelium cells of feeding tadpoles, J Embryol Exp Morphol, vol.10, pp.622-640, 1962.

C. Couldrey, M. Carlton, P. Nolan, X. Colledge, and M. Evans, A retroviral gene trap insertion into the histone 3.3A gene causes partial neonatal lethality, stunded growth, neuromuscular deficits and male sub-fertility in transgenic mice, pp.2489-2495, 1999.

C. Roberts, H. F. Sutherland, and H. Farmer, Targeted Mutagenesis of the Hira Gene Results in Gastrulation Defects and Patterning Abnormalities of Mesoendodermal Derivatives Prior to Early Embryonic Lethality, Molecular and Cellular Biology, vol.22, issue.7, pp.2318-2328, 2002.
DOI : 10.1128/MCB.22.7.2318-2328.2002

J. S. Michaelson, D. Bader, F. Kuo, C. Kozak, and P. Leder, Loss of Daxx, a promiscuously interacting protein, results in extensive apoptosis in early mouse development, Genes & Development, vol.13, issue.15, pp.1918-1923, 1999.
DOI : 10.1101/gad.13.15.1918

D. Garrick, J. A. Sharpe, and R. Arkell, Loss of Atrx Affects Trophoblast Development and the Pattern of X-Inactivation in Extraembryonic Tissues, PLoS Genetics, vol.14, issue.4, pp.58-95, 2006.
DOI : 10.1371/journal.pgen.0020058.st001

R. Zhang, W. Chen, and P. Adams, Molecular Dissection of Formation of Senescence-Associated Heterochromatin Foci, Molecular and Cellular Biology, vol.27, issue.6, pp.2343-2358, 2007.
DOI : 10.1128/MCB.02019-06