ASS1 metabolically contributes to the nuclear and cytosolic p53-mediated DNA damage response.


Journal

Nature metabolism
ISSN: 2522-5812
Titre abrégé: Nat Metab
Pays: Germany
ID NLM: 101736592

Informations de publication

Date de publication:
10 Jun 2024
Historique:
received: 23 07 2023
accepted: 30 04 2024
medline: 11 6 2024
pubmed: 11 6 2024
entrez: 10 6 2024
Statut: aheadofprint

Résumé

Downregulation of the urea cycle enzyme argininosuccinate synthase (ASS1) in multiple tumors is associated with a poor prognosis partly because of the metabolic diversion of cytosolic aspartate for pyrimidine synthesis, supporting proliferation and mutagenesis owing to nucleotide imbalance. Here, we find that prolonged loss of ASS1 promotes DNA damage in colon cancer cells and fibroblasts from subjects with citrullinemia type I. Following acute induction of DNA damage with doxorubicin, ASS1 expression is elevated in the cytosol and the nucleus with at least a partial dependency on p53; ASS1 metabolically restrains cell cycle progression in the cytosol by restricting nucleotide synthesis. In the nucleus, ASS1 and ASL generate fumarate for the succination of SMARCC1, destabilizing the chromatin-remodeling complex SMARCC1-SNF5 to decrease gene transcription, specifically in a subset of the p53-regulated cell cycle genes. Thus, following DNA damage, ASS1 is part of the p53 network that pauses cell cycle progression, enabling genome maintenance and survival. Loss of ASS1 contributes to DNA damage and promotes cell cycle progression, likely contributing to cancer mutagenesis and, hence, adaptability potential.

Identifiants

pubmed: 38858597
doi: 10.1038/s42255-024-01060-5
pii: 10.1038/s42255-024-01060-5
doi:

Types de publication

Journal Article

Langues

eng

Sous-ensembles de citation

IM

Subventions

Organisme : EC | EU Framework Programme for Research and Innovation H2020 | H2020 Priority Excellent Science | H2020 European Research Council (H2020 Excellent Science - European Research Council)
ID : 818943
Organisme : Israel Cancer Research Fund (Israel Cancer Research Fund, Inc.)
ID : 837124

Informations de copyright

© 2024. The Author(s).

Références

Erez, A. et al. Requirement of argininosuccinate lyase for systemic nitric oxide production. Nat. Med. 17, 1619–1626 (2011).
pubmed: 22081021 pmcid: 3348956 doi: 10.1038/nm.2544
Ah Mew, N et al. Urea cycle disorders overview in GeneReviews (eds. Adam M. P. et al.) (University of Washington, 2017).
Delage, B. et al. Arginine deprivation and argininosuccinate synthetase expression in the treatment of cancer. Int. J. Cancer 126, 2762–2772 (2010).
pubmed: 20104527 doi: 10.1002/ijc.25202
Rabinovich, S. et al. Diversion of aspartate in ASS1-deficient tumours fosters de novo pyrimidine synthesis. Nature 527, 379–383 (2015).
pubmed: 26560030 pmcid: 4655447 doi: 10.1038/nature15529
Keshet, R. et al. Targeting purine synthesis in ASS1-expressing tumors enhances the response to immune checkpoint inhibitors. Nat. Cancer 1, 894–908 (2020).
pubmed: 35121952 doi: 10.1038/s43018-020-0106-7
Lee, J. S. et al. Urea cycle dysregulation generates clinically relevant genomic and biochemical signatures. Cell 174, 1559–1570.e22 (2018).
pubmed: 30100185 pmcid: 6225773 doi: 10.1016/j.cell.2018.07.019
Miyamoto, T. et al. Argininosuccinate synthase 1 is an intrinsic Akt repressor transactivated by p53. Sci. Adv. 3, e1603204 (2017).
pubmed: 28560349 pmcid: 5438217 doi: 10.1126/sciadv.1603204
Yang, F., Teves, S. S., Kemp, C. J. & Henikoff, S. Doxorubicin, DNA torsion, and chromatin dynamics. Biochim. Biophys. Acta 1845, 84–89 (2014).
pubmed: 24361676
Zhan, T., Rindtorff, N., Betge, J., Ebert, M. P. & Boutros, M. CRISPR/Cas9 for cancer research and therapy. Semin. Cancer Biol. 55, 106–119 (2019).
pubmed: 29673923 doi: 10.1016/j.semcancer.2018.04.001
Swayden, M. et al. Activation of autophagy following [HuArgI (Co)-PEG5000]-induced arginine deprivation mediates cell death in colon cancer cells. Hum. cell 34, 152–164 (2021).
pubmed: 32979152 doi: 10.1007/s13577-020-00437-4
Suijkerbuijk, S. J. E. & van Rheenen, J. From good to bad: intravital imaging of the hijack of physiological processes by cancer cells. Dev. Biol. 428, 328–337 (2017).
pubmed: 28473106 doi: 10.1016/j.ydbio.2017.04.015
Sliwinska, M. A. et al. Induction of senescence with doxorubicin leads to increased genomic instability of HCT116 cells. Mech. Ageing Dev. 130, 24–32 (2009).
pubmed: 18538372 doi: 10.1016/j.mad.2008.04.011
Mah, L. J., El-Osta, A. & Karagiannis, T. C. γh2AX: a sensitive molecular marker of DNA damage and repair. Leukemia 24, 679–686 (2010).
pubmed: 20130602 doi: 10.1038/leu.2010.6
Lu, Y., Liu, Y. & Yang, C. Evaluating in vitro DNA damage using comet assay. J. Vis. Exp. 128, e56450 (2017).
dos Santos, Á. & Toseland, C. P. Regulation of nuclear mechanics and the impact on DNA damage. Int. J. Mol. Sci. 22, 3178 (2021).
pubmed: 33804722 pmcid: 8003950 doi: 10.3390/ijms22063178
Lin, R. et al. CLOCK acetylates ASS1 to drive circadian rhythm of ureagenesis. Mol. Cell 68, 198–209.e6 (2017).
pubmed: 28985504 doi: 10.1016/j.molcel.2017.09.008
Bunz, F. et al. Requirement for p53 and p21 to sustain G2 arrest after DNA damage. Science 282, 1497–1501 (1998).
pubmed: 9822382 doi: 10.1126/science.282.5393.1497
Pramil, E., Dillard, C. & Escargueil, A. E. Colorectal cancer and immunity: from the wet lab to individuals. Cancers (Basel) 13, 1713 (2021).
pubmed: 33916641 doi: 10.3390/cancers13071713
Sole, L. et al. p53 wild-type colorectal cancer cells that express a fetal gene signature are associated with metastasis and poor prognosis. Nat. Commun. 13, 2866 (2022).
pubmed: 35606354 pmcid: 9126967 doi: 10.1038/s41467-022-30382-9
Flores, K. & Seger, R. Stimulated nuclear import by β-like importins. F1000Prime Rep. 5, 1–7 (2013).
doi: 10.12703/P5-41
Golomb, L. et al. Importin 7 and exportin 1 link c-Myc and p53 to regulation of ribosomal biogenesis. Mol. Cell 45, 222–232 (2012).
pubmed: 22284678 pmcid: 3270374 doi: 10.1016/j.molcel.2011.11.022
Xu, J. et al. Pancreatic cancer progression is regulated by IPO7/p53/LncRNA MALAT1/MiR-129-5p positive feedback loop. Front. Cell Dev. Biol. 9, 630262 (2021).
pubmed: 34660566 pmcid: 8517143 doi: 10.3389/fcell.2021.630262
Soria, L. R. et al. Beclin-1-mediated activation of autophagy improves proximal and distal urea cycle disorders. EMBO Mol. Med. 13, e13158 (2021).
pubmed: 33369168 doi: 10.15252/emmm.202013158
Leshets, M., Silas, Y. B. H., Lehming, N. & Pines, O. Fumarase: from the TCA cycle to DNA damage response and tumor suppression. Front. Mol. Biosci. 5, 68 (2018).
pubmed: 30090811 pmcid: 6068284 doi: 10.3389/fmolb.2018.00068
Centore, R. C., Sandoval, G. J., Soares, L. M. M., Kadoch, C. & Chan, H. M. Mammalian SWI/SNF chromatin remodeling complexes: emerging mechanisms and therapeutic strategies. Trends Genet.: TIG 36, 936–950 (2020).
pubmed: 32873422 doi: 10.1016/j.tig.2020.07.011
Xiao, Z. M. et al. Front. Cell Dev. Biol. https://doi.org/10.3389/fcell.2021.678967 (2021).
Yan, L., Xie, S., Du, Y. & Qian, C. Structural insights into BAF47 and BAF155 complex formation. J. Mol. Biol. 429, 1650–1660 (2017).
pubmed: 28438634 doi: 10.1016/j.jmb.2017.04.008
Kulkarni, R. A. et al. A chemoproteomic portrait of the oncometabolite fumarate. Nat. Chem. Biol. 15, 391–400 (2019).
pubmed: 30718813 pmcid: 6430658 doi: 10.1038/s41589-018-0217-y
Merkley, E. D., Metz, T. O., Smith, R. D., Baynes, J. W. & Frizzell, N. The succinated proteome. Mass Spectrom. Rev. 33, 98–109 (2014).
pubmed: 24115015 doi: 10.1002/mas.21382
Grandi, F. C., Modi, H., Kampman, L. & Corces, M. R. Chromatin accessibility profiling by ATAC–seq. Nat. Protoc. 17, 1518–1552 (2022).
pubmed: 35478247 pmcid: 9189070 doi: 10.1038/s41596-022-00692-9
Peng, Q. et al. ZNF385A and ZNF346 serve as prognostic biomarkers associated with an inflamed immunosuppressive tumor microenvironment in hepatocellular carcinoma. 24, 3155 (2023).
Wingert, S. et al. DNA-damage response gene GADD45A induces differentiation in hematopoietic stem cells without inhibiting cell cycle or survival. Stem Cells 34, 699–710 (2016).
pubmed: 26731607 doi: 10.1002/stem.2282
DelBove, J. et al. Identification of a core member of the SWI/SNF complex, BAF155/SMARCC1, as a human tumor suppressor gene. Epigenetics 6, 1444–1453 (2011).
pubmed: 22139574 pmcid: 3256333 doi: 10.4161/epi.6.12.18492
Noronha, A. et al. AXL and error-prone DNA replication confer drug resistance and offer strategies to treat EGFR-mutant lung cancer. Cancer Discov. 12, 2666–2683 (2022).
pubmed: 35895872 pmcid: 9627128 doi: 10.1158/2159-8290.CD-22-0111
Allen, M. A. et al. Global analysis of p53-regulated transcription identifies its direct targets and unexpected regulatory mechanisms. eLife 3, e02200 (2014).
pubmed: 24867637 pmcid: 4033189 doi: 10.7554/eLife.02200
Lee, D. et al. SWI/SNF complex interacts with tumor suppressor p53 and is necessary for the activation of p53-mediated transcription. J. Biol. Chem. 277, 22330–22337 (2002).
pubmed: 11950834 doi: 10.1074/jbc.M111987200
Ribeiro-Silva, C., Vermeulen, W. & Lans, H. SWI/SNF: complex complexes in genome stability and cancer. DNA Repair 77, 87–95 (2019).
pubmed: 30897376 doi: 10.1016/j.dnarep.2019.03.007
Kafkia, E. et al. Operation of a TCA cycle subnetwork in the mammalian nucleus. Sci. Adv. 8, eabq5206 (2022).
pubmed: 36044572 pmcid: 9432838 doi: 10.1126/sciadv.abq5206
Jiang, Y. et al. Author correction: local generation of fumarate promotes DNA repair through inhibition of histone H3 demethylation. Nat. Cell Biol. 20, 1226 (2018).
pubmed: 29632342 doi: 10.1038/s41556-018-0074-7
Sulkowski, P. L. et al. Krebs-cycle-deficient hereditary cancer syndromes are defined by defects in homologous-recombination DNA repair. Nat. Genet. 50, 1086–1092 (2018).
pubmed: 30013182 pmcid: 6072579 doi: 10.1038/s41588-018-0170-4
Golino, P. et al. Simultaneous administration of thromboxane A2- and serotonin S2-receptor antagonists markedly enhances thrombolysis and prevents or delays reocclusion after tissue-type plasminogen activator in a canine model of coronary thrombosis. Circulation 79, 911–919 (1989).
pubmed: 2494005 doi: 10.1161/01.CIR.79.4.911
Vande Voorde, J. et al. Improving the metabolic fidelity of cancer models with a physiological cell culture medium. Sci. Adv. 5, eaau7314 (2019).
pubmed: 30613774 pmcid: 6314821 doi: 10.1126/sciadv.aau7314
Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR–Cas9. Nat. Biotechnol. 34, 184–191 (2016).
pubmed: 26780180 pmcid: 4744125 doi: 10.1038/nbt.3437
Hsu, P. D. et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat. Biotechnol. 31, 827–832 (2013).
pubmed: 23873081 pmcid: 3969858 doi: 10.1038/nbt.2647
Concordet, J.-P. & Haeussler, M. CRISPOR: intuitive guide selection for CRISPR/Cas9 genome editing experiments and screens. Nucleic Acids Res. 46, W242–W245 (2018).
pubmed: 29762716 pmcid: 6030908 doi: 10.1093/nar/gky354
Gasperini, M. et al. A genome-wide framework for mapping gene regulation via cellular genetic screens. Cell 176, 377–390.e19 (2019).
pubmed: 30612741 pmcid: 6690346 doi: 10.1016/j.cell.2018.11.029
Sun, R. C. et al. Nuclear glycogenolysis modulates histone acetylation in human non-small cell lung cancers. Cell Metab. 30, 903–916.e7 (2019).
pubmed: 31523006 pmcid: 6834909 doi: 10.1016/j.cmet.2019.08.014
Soria, L. R. et al. O-GlcNAcylation enhances CPS1 catalytic efficiency for ammonia and promotes ureagenesis. Nat. Commun. 13, 5212 (2022).
pubmed: 36064721 pmcid: 9445089 doi: 10.1038/s41467-022-32904-x
Millard, P. et al. IsoCor: isotope correction for high-resolution MS labeling experiments. Bioinformatics https://doi.org/10.1093/bioinformatics/btz209 (2019).
Han, J. et al. Metabolomic analysis of key central carbon metabolism carboxylic acids as their 3-nitrophenylhydrazones by UPLC/ESI-MS. Electrophor. https://doi.org/10.1002/elps.201200601 (2013).
Elinger, D., Gabashvili, A. & Levin, Y. Suspension trapping (S-Trap) is compatible with typical protein extraction buffers and detergents for bottom-up proteomics. J. Proteome Res. 18, 1441–1445 (2019).
pubmed: 30761899 doi: 10.1021/acs.jproteome.8b00891
Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).
pubmed: 19029910 doi: 10.1038/nbt.1511
Fischer, M. Census and evaluation of p53 target genes. Oncogene 36, 3943–3956 (2017).
pubmed: 28288132 pmcid: 5511239 doi: 10.1038/onc.2016.502
Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).
pubmed: 24097267 pmcid: 3959825 doi: 10.1038/nmeth.2688
Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).
pubmed: 22388286 pmcid: 3322381 doi: 10.1038/nmeth.1923
Zhang, Y. et al. Model-based analysis of ChIP–seq (MACS). Genome Biol. 9, R137 (2008).
pubmed: 18798982 pmcid: 2592715 doi: 10.1186/gb-2008-9-9-r137
Morales, J. et al. A joint NCBI and EMBL-EBI transcript set for clinical genomics and research. Nature 604, 310–315 (2022).
pubmed: 35388217 pmcid: 9007741 doi: 10.1038/s41586-022-04558-8
Rom, A. et al. Regulation of CHD2 expression by the Chaserr long noncoding RNA gene is essential for viability. Nat. Commun. 10, 5092 (2019).
pubmed: 31704914 pmcid: 6841665 doi: 10.1038/s41467-019-13075-8
Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).
pubmed: 20513432 pmcid: 2898526 doi: 10.1016/j.molcel.2010.05.004
Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).
pubmed: 25516281 pmcid: 4302049 doi: 10.1186/s13059-014-0550-8
Kent, W. J. et al. The human genome browser at UCSC. Genome Res. 12, 996–1006 (2002).
pubmed: 12045153 pmcid: 186604 doi: 10.1101/gr.229102
Hammal, F., de Langen, P., Bergon, A., Lopez, F. & Ballester, B. ReMap 2022: a database of human, mouse, Drosophila and Arabidopsis regulatory regions from an integrative analysis of DNA-binding sequencing experiments. Nucleic Acids Res. 50, D316–D325 (2022).
pubmed: 34751401 doi: 10.1093/nar/gkab996
Cartharius, K. et al. MatInspector and beyond: promoter analysis based on transcription factor binding sites. Bioinformatics 21, 2933–2942 (2005).
pubmed: 15860560 doi: 10.1093/bioinformatics/bti473
Lim, L. Q. J. et al. ASS1 KO vs EV ctrl cells after DNA damage using Dox—RNA sequencing. Dryad https://datadryad.org/stash/share/_SN3vlavn00iwe_oKns4f16TItXUlsbbzxpt0cQlrhM (2024).
Lim, L. Q. J. et al. Proteomic analysis—ASS1 binders. Dryad https://datadryad.org/stash/share/HMwv5E4ZfHAieT3X5K5l6mLq-LogVb-HnTQF3VinSWo (2024).

Auteurs

Lisha Qiu Jin Lim (LQJ)

Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.

Lital Adler (L)

Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.

Emma Hajaj (E)

Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
Department of Medicine D, Beilinson Hospital, Petah Tikva, Israel.

Leandro R Soria (LR)

Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.

Rotem Ben-Tov Perry (RB)

Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel.

Naama Darzi (N)

Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.

Ruchama Brody (R)

Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.

Noa Furth (N)

Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.

Michal Lichtenstein (M)

Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.

Elizabeta Bab-Dinitz (E)

Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.

Ziv Porat (Z)

Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel.

Tevie Melman (T)

Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel.

Alexander Brandis (A)

Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel.

Yael Aylon (Y)

Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.

Shifra Ben-Dor (S)

Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel.

Irit Orr (I)

Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel.

Amir Pri-Or (A)

The De Botton Protein Profiling Institute of the Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel.

Rony Seger (R)

Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.

Yoav Shaul (Y)

Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel-Canada, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.

Eytan Ruppin (E)

Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, USA.

Moshe Oren (M)

Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.

Minervo Perez (M)

Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, USA.

Jordan Meier (J)

Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institute of Health, Bethesda, MD, USA.

Nicola Brunetti-Pierri (N)

Telethon Institute of Genetics and Medicine, Pozzuoli, Italy.
Department of Translational Medicine, Medical Genetics, University of Naples Federico II, Naples, Italy.
Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, University of Naples Federico II, Naples, Italy.

Efrat Shema (E)

Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.

Igor Ulitsky (I)

Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
Department of Molecular Neuroscience, Weizmann Institute of Science, Rehovot, Israel.

Ayelet Erez (A)

Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel. ayelet.erez@weizmann.ac.il.

Sergey Malitsky (S)

Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel.

Maxim Itkin (M)

Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel.

Classifications MeSH