A. Aguzzi, A. , and M. , Phase separation: linking cellular compartmentalization to disease, Trends Cell Biol, vol.26, pp.547-558, 2016.

M. Altmeyer, K. J. Neelsen, F. Teloni, I. Pozdnyakova, S. Pellegrino et al., Liquid demixing of intrinsically disordered proteins is seeded by poly(ADP-ribose), Nat. Commun, vol.6, p.8088, 2015.

J. C. Amé, É. Hé-berlé, B. Camuzeaux, F. Dantzer, and V. Schreiber, Purification of recombinant human PARG and activity assays, Methods Mol, 2017.

, Biol, vol.1608, pp.395-413

S. F. Banani, H. O. Lee, A. A. Hyman, and M. K. Rosen, Biomolecular condensates: organizers of cellular biochemistry, Nat. Rev. Mol. Cell Biol, vol.18, pp.285-298, 2017.

L. P. Bergeron-sandoval, N. Safaee, and S. W. Michnick, Mechanisms and consequences of macromolecular phase separation, Cell, vol.165, pp.1067-1079, 2016.

F. J. Bock, T. T. Todorova, C. , and P. , RNA regulation by poly(ADPribose) polymerases, Mol. Cell, vol.58, pp.959-969, 2015.

E. Bogaert, S. Boeynaems, M. Kato, L. Guo, T. R. Caulfield et al., Molecular Cell Reports, vol.27, pp.1809-1821, 2018.

O. Bounedjah, B. Desforges, T. D. Wu, C. Pioche-durieu, S. Marco et al., , 2014.

, Free mRNA in excess upon polysome dissociation is a scaffold for protein multimerization to form stress granules, Nucleic Acids Res, vol.42, pp.8678-8691

S. Britton, E. Dernoncourt, C. Delteil, C. Froment, O. Schiltz et al., DNA damage triggers SAF-A and RNA biogenesis factors exclusion from chromatin coupled to R-loops removal, Nucleic Acids Res, vol.42, pp.9047-9062, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01191434

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.

Q. Deng, C. J. Holler, G. Taylor, K. F. Hudson, W. Watkins et al., FUS is phosphorylated by DNA-PK and accumulates in the cytoplasm after DNA damage, J. Neurosci, vol.34, pp.7802-7813, 2014.

A. Doller, J. Pfeilschifter, and W. Eberhardt, Signalling pathways regulating nucleo-cytoplasmic shuttling of the mRNA-binding protein HuR, Cell. Signal, vol.20, pp.2165-2173, 2008.

D. Dormann, R. Rodde, D. Edbauer, E. Bentmann, I. Fischer et al., ALS-associated fused in sarcoma (FUS) mutations disrupt Transportin-mediated nuclear import, EMBO J, vol.29, pp.2841-2857, 2010.

D. Dormann, T. Madl, C. F. Valori, E. Bentmann, S. Tahirovic et al.,

, Arginine methylation next to the PY-NLS modulates Transportin binding and nuclear import of FUS, EMBO J, vol.31, pp.4258-4275

M. Feric, N. Vaidya, T. S. Harmon, D. M. Mitrea, L. Zhu et al., Coexisting liquid phases underlie nucleolar subcompartments, Cell, vol.165, pp.1686-1697, 2016.

J. P. Gagné, E. Pic, M. Isabelle, J. Krietsch, C. Ethier et al., Quantitative proteomics profiling of the poly(ADP-ribose)-related response to genotoxic stress, Nucleic Acids Res, vol.40, pp.7788-7805, 2012.

L. Hamon, D. Pastré, P. Dupaigne, C. Le-breton, E. Le-cam et al., High-resolution AFM imaging of single-stranded DNA-binding (SSB) protein-DNA complexes, Nucleic Acids Res, vol.35, p.58, 2007.
URL : https://hal.archives-ouvertes.fr/hal-02295472

S. Hennig, G. Kong, T. Mannen, A. Sadowska, S. Kobelke et al., Prion-like domains in RNA binding proteins are essential for building subnuclear paraspeckles, 2015.

, J. Cell Biol, vol.210, pp.529-539

N. C. Hoch, H. Hanzlikova, S. L. Rulten, M. Té-treault, E. Komulainen et al., XRCC1 mutation is associated with PARP1 hyperactivation and cerebellar ataxia, Nature, vol.541, pp.87-91, 2017.

M. Hofweber, S. Hutten, B. Bourgeois, E. Spreitzer, A. Niedner-boblenz et al., Phase separation of FUS is suppressed by its nuclear import receptor and arginine methylation, Cell, vol.173, pp.706-719, 2018.

G. Illuzzi, E. Fouquerel, J. C. Amé, A. Noll, K. Rehmet et al., PARG is dispensable for recovery from transient replicative stress but required to prevent detrimental accumulation of poly(ADP-ribose) upon prolonged replicative stress, Nucleic Acids Res, vol.42, pp.7776-7792, 2014.

L. Izhar, B. Adamson, A. Ciccia, J. Lewis, L. Pontano-vaites et al., A systematic analysis of factors localized to damaged chromatin reveals PARP-dependent recruitment of transcription factors, Cell Rep, vol.11, pp.1486-1500, 2015.

B. Jakob, J. Splinter, M. Durante, and G. Taucher-scholz, Live cell microscopy analysis of radiation-induced DNA double-strand break motion, Proc. Natl. Acad. Sci. USA, vol.106, pp.3172-3177, 2009.

S. Jungmichel, F. Rosenthal, M. Altmeyer, J. Lukas, M. O. Hottiger et al., Proteome-wide identification of poly(ADP-ribosyl)ation targets in different genotoxic stress responses, Mol. Cell, vol.52, pp.272-285, 2013.

H. M. Kaneb, P. A. Dion, and G. A. Rouleau, The FUS about arginine methylation in ALS and FTLD, EMBO J, vol.31, pp.4249-4251, 2012.

Y. Ke, Y. Han, X. Guo, J. Wen, K. Wang et al., PARP1 promotes gene expression at the post-transcriptiona level by modulating the RNA-binding protein HuR, Nat. Commun, vol.8, p.14632, 2017.

U. K. Laemmli, Cleavage of structural proteins during the assembly of the head of bacteriophage T4, Nature, vol.227, pp.680-685, 1970.

A. K. Leung, Poly(ADP-ribose): an organizer of cellular architecture, J. Cell Biol, vol.205, pp.613-619, 2014.

A. K. Leung, S. Vyas, J. E. Rood, A. Bhutkar, P. A. Sharp et al., Poly(ADP-ribose) regulates stress responses and microRNA activity in the cytoplasm, Mol. Cell, vol.42, pp.489-499, 2011.

P. Li, S. Banjade, H. C. Cheng, S. Kim, B. Chen et al., Phase transitions in the assembly of multivalent signalling proteins, Nature, vol.483, pp.336-340, 2012.

Y. R. Li, O. D. King, J. Shorter, and A. D. Gitler, Stress granules as crucibles of ALS pathogenesis, J. Cell Biol, vol.201, pp.361-372, 2013.

J. Liu, P. A. Vidi, S. A. Leliè-vre, and J. M. Irudayaraj, Nanoscale histone localization in live cells reveals reduced chromatin mobility in response to DNA damage, J. Cell Sci, vol.128, pp.599-604, 2015.

Y. Lorenzo, A. Azqueta, L. Luna, F. Bonilla, G. Domínguez et al., The carotenoid beta-cryptoxanthin stimulates the repair of DNA oxidation damage in addition to acting as an antioxidant in human cells. Carcinogenesis, vol.30, pp.308-314, 2009.

A. S. Mastrocola, S. H. Kim, A. T. Trinh, L. A. Rodenkirch, and R. S. Tibbetts, The RNA-binding protein fused in sarcoma (FUS) functions downstream of poly(ADP-ribose) polymerase (PARP) in response to DNA damage, J. Biol. Chem, vol.288, pp.24731-24741, 2013.

Z. Monahan, V. H. Ryan, A. M. Janke, K. A. Burke, S. N. Rhoads et al., Phosphorylation of the FUS low-complexity domain disrupts phase separation, aggregation, and toxicity, EMBO J, vol.36, pp.2951-2967, 2017.

N. Mukherjee, D. L. Corcoran, J. D. Nusbaum, D. W. Reid, S. Georgiev et al., , 2011.

, Integrative regulatory mapping indicates that the RNA-binding protein HuR couples pre-mRNA processing and mRNA stability, Mol. Cell, vol.43, pp.327-339

D. T. Murray, M. Kato, Y. Lin, K. R. Thurber, I. Hung et al., Structure of FUS protein fibrils and its relevance to selfassembly and phase separation of low-complexity domains, Cell, vol.171, pp.615-627, 2017.

M. Naumann, A. Pal, A. Goswami, X. Lojewski, J. Japtok et al., Impaired DNA damage response signaling by FUS-NLS mutations leads to neurodegeneration and FUS aggregate formation, Nat. Commun, vol.9, p.335, 2018.

A. Patel, H. O. Lee, L. Jawerth, S. Maharana, M. Jahnel et al., A liquid-to-solid phase transition of the ALS protein FUS accelerated by disease mutation, Cell, vol.162, pp.1066-1077, 2015.

S. Qamar, G. Wang, S. J. Randle, F. S. Ruggeri, J. A. Varela et al., FUS phase separation is modulated by a molecular chaperone and methylation of arginine cation-p interactions, Cell, vol.173, pp.720-734, 2018.

S. N. Rhoads, Z. T. Monahan, D. S. Yee, A. Y. Leung, C. G. Newcombe et al., The prion-like domain of FUS is multiphosphorylated following DNA damage without altering nuclear localization, Mol. Biol. Cell, vol.29, pp.1786-1797, 2018.

S. L. Rulten, A. Rotheray, R. L. Green, G. J. Grundy, D. A. Moore et al., PARP-1 dependent recruitment of the amyotrophic lateral sclerosis-associated protein FUS/TLS to sites of oxidative DNA damage, Nucleic Acids Res, vol.42, pp.307-314, 2014.

R. R. Sama, C. L. Ward, and D. A. Bosco, Functions of FUS/TLS from DNA repair to stress response: implications for ALS, ASN Neuro, vol.6, p.1759091414544472, 2014.

B. R. Schrank, T. Aparicio, Y. Li, W. Chang, B. T. Chait et al., Nuclear ARP2/3 drives DNA break clustering for homology-directed repair, Nature, vol.559, pp.61-66, 2018.

V. Schreiber, F. Dantzer, J. C. Ame, and G. De-murcia, Poly(ADPribose): novel functions for an old molecule, Nat. Rev. Mol. Cell Biol, vol.7, pp.517-528, 2006.

G. Singh, G. Pratt, G. W. Yeo, and M. J. Moore, The clothes make the mRNA: past and present trends in mRNP fashion, Annu. Rev. Biochem, vol.84, pp.325-354, 2015.

P. Strzyz, Organelle dynamics: controlling phase separation of P granules, Nat. Rev. Mol. Cell Biol, vol.18, p.4, 2016.

M. V. Sukhanova, O. I. Lavrik, and S. N. Khodyreva, , 2004.

, Mol. Biol. (Mosk.), vol.38, pp.834-847

M. V. Sukhanova, S. Abrakhi, V. Joshi, D. Pastre, M. M. Kutuzov et al., Single molecule detection of PARP1 and PARP2 interaction with DNA strand breaks and their poly(ADPribosyl)ation using high-resolution AFM imaging, Readers of poly(ADP-ribose): designed to be fit for purpose, vol.44, pp.993-1006, 2016.

W. Y. Wang, L. Pan, S. C. Su, E. J. Quinn, M. Sasaki et al., Interaction of FUS and HDAC1 regulates DNA damage response and repair in neurons, Nat. Neurosci, vol.16, pp.1383-1391, 2013.

X. Wang, J. C. Schwartz, and T. R. Cech, Nucleic acid-binding specificity of human FUS protein, Nucleic Acids Res, vol.43, pp.7535-7543, 2015.

H. Wang, W. Guo, J. Mitra, P. M. Hegde, T. Vandoorne et al., , 2018.

, Mutant FUS causes DNA ligation defects to inhibit oxidative damage repair in amyotrophic lateral sclerosis, Nat. Commun, vol.9, p.3683

J. Wang, J. Choi, A. S. Holehouse, H. O. Lee, X. Zhang et al., A molecular grammar governing the driving forces for phase separation of prion-like RNA binding proteins, Cell, vol.174, pp.688-699, 2018.

H. Zhang, S. Elbaum-garfinkle, E. M. Langdon, N. Taylor, P. Occhipinti et al., RNA controls PolyQ protein phase transitions, Mol. Cell, vol.60, pp.220-230, 2015.

Y. Zhen, Y. Zhang, Y. , and Y. , A cell-line-specific atlas of PARP-mediated protein Asp/Glu-ADP-ribosylation in breast cancer, Cell Rep, vol.21, pp.2326-2337, 2017.

L. Zhu and C. P. Brangwynne, Nuclear bodies: the emerging biophysics of nucleoplasmic phases, Curr. Opin. Cell Biol, vol.34, pp.23-30, 2015.

, 100-300 nm at low frequency (1-2 kHz). PFT mode decreases the lateral and shear forces, Images were, 2048.

, and 20 mM MgCl 2 was incubated for 1 h at 37 C. The enzyme was denatured at 65 C for 10 min and precipitated proteins were removed by centrifugation. An in vitro poly(ADP-ribosyl)ation assay was performed in the reaction mixtures (20 mL) contained 20 mM Tris-HCl, pH 7.5, 25 mM NaCl, 1 mM DTT, 5 mM MgCl 2 , 400 mM Urea, 0.05 A260/mL of DNase I-activated calf thymus DNA, 50 nM PARP-1, 0.3 mM NAD + , 0.4 mCi [ 32 P]-NAD + and 2 mg FUS, FUSDLCD, G3BP1, HuR, TDP-43, or BSA as indicated at the figure legends. The reactions were initiated by the addition of NAD + . The reaction mixtures were incubated at 37 for 30 min and stopped by adding SDS-sample buffer and heating for 5 min at 90 C. The reaction mixtures were analyzed by 10% SDS-PAGE with subsequent phosphorimaging and/or colloidal Coomassie staining. For the analysis of the degradation of PARP-1-bound PAR by PARG, 30 nM PARP-1 was incubated with 3 nM damaged pBR in the buffer containing 12.5 mM HEPES-KOH, pH 8.0, 25 mM NaCl, 1 mM DTT, 5 mM MgCl 2 , 260 mM Urea, 400 mM, 0.3 mM NAD + , 0.4 mCi [ 32 P]-NAD + at 37 C for 30 min followed by addition of EDTA to the final concentration of 15 mM. After that, the samples were supplemented with 2.4 mM FUS, FUSDLCD, G3BP1, HuR or TDP-43 and incubated at 37 C for 5 min, and then PARG was added to final concentration 4 nM followed by incubation for 1-5 min at 37 C. The reaction mixture, NAD + labeled on the adenylate phosphate was synthesized in a reaction mixture (100mL) containing 2 mM b-Nicotinamide mononucleotide, 1 mM ATP and 0.25 mCi of

, After that the reactions were incubated with RNA-binding proteins (1 mM) for 5 min at 37 C. For sedimentation analysis, the reactions were centrifuged at 10,000 3 g for 5 min at 25 C. Supernatants were collected, and pellets were resuspended in 20 mL of the buffer with 0.25 volume of the loading solution containing 5% SDS, 5% 2-mercaptoethanol, 0.3 M Tris-HCl (pH 6.8), 50% glycerol, and 0.015% bromophenol blue. Supernatants and pellets were divided into two parts. One part (20 mL) was analyzed by separation in 10% SDS-PAGE and visualized with Coomassie staining. The other part (20 mL) was resolved in a 0.8% agarose gel in 0.5 3 TAE buffer at room temperature at 5 V/cm and stained with GelRed. Immunofluorescence analysis Cells were washed with PBS and fixed with 4% paraformaldehyde (PFA) in PBS for 45 min at 37 C. After washing with PBS, coverslips were kept with blocking buffer (50 mM Tris pH 7.5, 100 mM NaCl, BSA 2%, 0.15% Triton X-100) for 40 min at 37 C in order to permeabilize the cells and reduce nonspecific recognition by antibodies. Blocking buffer was removed and cells were washed and then incubated for 1 h at room temperature with primary antibody described in supplementary methods. The cells were washed 5 times with PBS and incubated for 1 h with fluorochrome (Alexa Fluorâ488 and À594)-coupled secondary antibodies in 50 mM Tris pH 7.5. After final washes with PBS, the cells were stained with 300 nM DAPI to visualize the nuclei and mounted for fluorescence microscopy analysis. Western blot analysis of PARG expression Cells were lysed in 50 mM Tris-HCl, pH 7.5, 150 mM NaCl, 0.1% Triton X-100, 1 mm EDTA, and protease inhibitor mixture. Lysates were centrifuged at 14,000 3 g for 15 min at 4 C, and supernatants were collected. Proteins (120 mg) of cell extracts were separated on 12% SDS-PAGE gels and transferred onto a PVDF membrane. The membranes were blocked in 5% (w/v) non fat dried milk, C, and the products were separated by denaturing electrophoresis in 20% polyacrylamide gel followed by visualization with phosphorimaging. Analysis of binding of FUS to RNA by EMSA To assay stability of RNA-protein complexes in the presence of PAR, the reactions were performed in a mixture 25 mL containing 12.5 mM HEPES, pH 8.0, 25 mM KCl, 260 mM urea, 0.8 nM mRNA, 30-120 nM FUS and 0.4-4 mM PAR, as indicated in the figure legends