KAT2A/KAT2B-targeted acetylome reveals a role for PLK4 acetylation in preventing centrosome amplification

Marjorie Fournier, Meritxell Orpinell, Cédric Grauffel, Elisabeth Scheer, Jean-Marie Garnier, Tao Ye, Virginie Chavant, Mathilde Joint, Fumiko Esashi, Annick Dejaegere, Pierre Gönczy, László Tora, Marjorie Fournier, Meritxell Orpinell, Cédric Grauffel, Elisabeth Scheer, Jean-Marie Garnier, Tao Ye, Virginie Chavant, Mathilde Joint, Fumiko Esashi, Annick Dejaegere, Pierre Gönczy, László Tora

Abstract

Lysine acetylation is a widespread post-translational modification regulating various biological processes. To characterize cellular functions of the human lysine acetyltransferases KAT2A (GCN5) and KAT2B (PCAF), we determined their acetylome by shotgun proteomics. One of the newly identified KAT2A/2B substrate is polo-like kinase 4 (PLK4), a key regulator of centrosome duplication. We demonstrate that KAT2A/2B acetylate the PLK4 kinase domain on residues K45 and K46. Molecular dynamics modelling suggests that K45/K46 acetylation impairs kinase activity by shifting the kinase to an inactive conformation. Accordingly, PLK4 activity is reduced upon in vitro acetylation of its kinase domain. Moreover, the overexpression of the PLK4 K45R/K46R mutant in cells does not lead to centrosome overamplification, as observed with wild-type PLK4. We also find that impairing KAT2A/2B-acetyltransferase activity results in diminished phosphorylation of PLK4 and in excess centrosome numbers in cells. Overall, our study identifies the global human KAT2A/2B acetylome and uncovers that KAT2A/2B acetylation of PLK4 prevents centrosome amplification.

Figures

Figure 1. Identification of the KAT2A- and…
Figure 1. Identification of the KAT2A- and KAT2B-dependent acetylome.
(a) KAT2A/2B knockdown efficiency. Tetracycline (Tet) inducible stable HeLa cell lines were used in which shRNAs either not targeting any endogenous transcript (Tet-shCtrl) or targeting KAT2A (Tet-shKAT2A) were expressed under doxycycline (Dox) induction. In the Tet-shKAT2A cell line, a siRNA against KAT2B (K2B) was also transfected. The knockdown efficiency of KAT2A, KAT2B and the acetylation of histone H3K9 was tested by western blot analyses. (b) In all, 398 potential KAT2A/2B acetylated protein targets were identified as being present in control samples (3/5), but absent in all KAT2A/2B KD-depleted cell lysates. (c) Analysis of the frequency of amino acids surrounding the acetylated lysines targeted by KAT2A and KAT2B, as compared with that of amino acids surrounding non-acetylated lysines. In all, 1,569 distinct ‘Ks' from the list of 398 proteins were used in this analysis, with 10 aa upstream (−10 on the x axis) and 10 aa downstream (+10 on the x axis). The K itself (at position 0) is not shown in these logos. A random selection of 5,243 sequences (K in the middle, 21 aa length, 0.05% of the total K) from the total human proteins was used for the comparison. The overall height of the stack indicates the sequence conservation at that position, while the height of symbols within the stack indicates the relative frequency of each amino acid at that position. Note that the y axes on the upper and lower graphs are different. (d) Gene ontology (GO) term enrichment analysis for biological processes using Manteia of the 398 identified proteins. Pathways at a false discovery rate below 1% are represented. The x axis represents the reverse P value obtained after GO term enrichment analysis in Manteia. Txn,transcription. (e) Examples of proteins acetylated by KAT2A/2B belonging to the same biological pathways targeted by KAT2A/2B (see GO term analysis in d).
Figure 2. KAT2A and KAT2B acetylate the…
Figure 2. KAT2A and KAT2B acetylate the kinase domain of PLK4.
(a) Alignment of the first 55 amino acids of human PLK4 with the corresponding sequences from several other species. Danre, Danio rerio; Drome, Drosophila melanogaster; Human, H. sapiens; Mouse, Mus musculus. The well-conserved acetylated K45 and K46 residues are highlighted with red dots. (b) In vitro AT assay on recombinant PLK4. Molecular weight markers are indicated on the left in kDa. (c) PLK4 kinase domain (KinDo) was acetylated by KAT2A, KAT2Amut or KAT2B, and the acetylation sites determined after LC–MS/MS analysis. Acetylation abundance factor (see Methods) values are depicted using the indicated colour code. (d) HEK293 cells were transfected with expression vectors coding for only the Flag tag, Flag-tagged KAT2A or Flag-KAT2B. Western blots were probed with the indicated antibodies. (e) PLK4ac (green) and Centrin-2 localization (red) were probed by immunofluorescence. Scale bars represent 5 μm for the high-magnification view, 1 μm for the inset.
Figure 3. KAT2A/2B and PLK4 co-localize at…
Figure 3. KAT2A/2B and PLK4 co-localize at centrosomes.
(a) The co-localization of KAT2A (red) or KAT2B (green) with several centriolar proteins, HsSAS-6, Centrin-2, CP110 and PLK4ac was tested by immunofluorescence in U2OS cells. Scale bars represent 5 μm, and insets are high-magnification views of the regions indicated in the low-magnification images. (b) PLK4 and KAT2A localization probed by immunofluorescence at different cell cycle stages following synchronization. U2OS cells were tested by immunofluorescence with antibodies against the kinase domain of PLK4 or KAT2A. (c,d) The histograms report the average frequency of cells displaying detectable PLK4 and KAT2A (c), or PLK4ac and KAT2A (d) Signal at centrosomes in different cell cycle stages. Data were collected from three experiments. n>100 cells per cell cycle stage. Error bars: s.e.m. (e) Cell cycle-staged HeLa cell extracts were prepared. The cell cycle distribution was verified by western blot. (f) Using these cell cycle-staged protein extracts IP experiments were carried out with either anti-KAT2A antibodies (KAT2A IP) or control antibodies (mock IP). The presence of KAT2A and PLK4 in the IPs was tested by western blot analysis (WB). M, molecular weight marker in kDa.
Figure 4. KAT2A and the ATAC complex…
Figure 4. KAT2A and the ATAC complex regulate centrosome duplication.
(a) U2OS cells were transfected with vectors either to overexpress (OE) or to knockdown the indicated proteins using shRNA-mediated depletion. Centrosomes were visualized by immunofluorescence. Cen-2, Centrin-2; γ-Tub, γ-tubulin. Note that all cells were analysed, without knowledge of their transfection status thus, the values reported here are probably underestimates. (b) The histograms report the average frequency of interphase cells displaying aberrant Centrin-2 (>4) and γ-tubulin (>2) numbers. Data from three experiments, n=50 cells per condition. Error bars: s.e.m.
Figure 5. Molecular modelling of PLK4 kinase…
Figure 5. Molecular modelling of PLK4 kinase domain suggests that acetylation shifts the dynamics of the acetylated PLK4 to the inactive conformation.
(a) Active (PDB ID 3COK) and (b) inactive (PDB ID 4JXF) conformations of the kinase domain (KinDo) of PLK4 are shown in the middle of the panels. The region where the structure was reconstructed is shown in grey. In both conformations, zoomed-in views have been generated to highlight the changes caused by K45/K46 acetylation. Zooms on the left show the non-modified WT KinDo structures, zooms on the right the KinDO structures with K45/K46 acetylations (ac). The zooms are rotated with the indicated angles when compared with the structures in the middle. On the right, the atoms of the backbone are included. Dotted lines are plotted from nitrogen atoms to oxygen atoms. The left side zooms give an overview of the most stable H-bonds of the WT KinDo, while the right side zoom only shows the H-bonds that differ in the acetylated forms. The thicker lines in a show the local H-bonds that are lost on acetylation: K45-E80 (red) and K46-D44 (yellow), whereas weakened H-bonds are shown in green. If an H-bond is not reported from left to right, its stability does not vary. In b, a distant H-bond newly formed in the acetylated structure (D154-L157) is highlighted in blue. The ATP and residues of the DFG motif are shown in sticks, whereas the αC helix and other structural motifs are in ribbons. (cf) Ramachandran representation of the backbone dihedral angles of D154 (of the DFG motif), in the WT (c,e) and acetylated (d,f) forms of the active and inactive conformations. (d) In the active acetylated form, D154 explores conformations characteristic of the inactive structure, whereas this is not observed for the WT (c).
Figure 6. Acetylation of PLK4 kinase domain…
Figure 6. Acetylation of PLK4 kinase domain by KAT2A/2B inhibits its kinase activity.
(a,b) Kinase activity of the PLK4 kinase domain (KinDo) in the presence of 32Pγ-ATP after being acetylated by Flag-KAT2A (a) or Flag-KAT2B (b). KinDo acetylation was tested by western blot analyses (WB). Kinase activity was measured by the autophosphorylation reaction (32P) of the KinDo. M, molecular weight marker in kDa. (c,d) Kinase activity normalized to protein amount was measured in three independent experiments. Mean values from the three replicate experiments and the corresponding s.d.'s are shown. (e) Kinase activity of the KinDo was tested on His-CPB of PLK4. KinDo was first acetylated by Flag-KAT2A, Flag-KAT2Amut, Flag-KAT2B or Flag-KAT2Bmut and then 32Pγ-ATP was added to the reactions. The level of acetylated KinDo was tested by WB. Kinase activity was measured by phosphorylation (32P) of His-CPB by the KinDo. M, molecular weight marker in kDa. (f) Kinase activity of the recombinant purified WT or mutated (K45R/K46R) KinDos. Protein levels and autophosphorylation activities were analysed as in a,b. (e) The kinase activities of WT and mutated K45R/K46R KinDos were normalized and measured from three independent experiments as in c,d. In g, kinase activity of the K45/K46R KinDo was calculated relative to that of the WT KinDo, which was normalized to 100% in all replicates. In a,b,e,f, the reaction mixes were analysed by Coomassie blue (CB) staining. In a,b,e,f, a dotted line has been inserted to indicate where unnecessary lanes were cut out from the gels.
Figure 7. KAT2A/2B-mediated acetylation of PLK4 prevents…
Figure 7. KAT2A/2B-mediated acetylation of PLK4 prevents abnormal centrosome amplification in human cells.
(a) Distribution of PLK4 phosphorylated at Ser305 (pSer305) upon KAT2A/2B knockdown. U2OS cells were transfected with control siRNAs (control), or siRNAs targeting PLK4, KAT2A or KAT2B to knock down (KD) these factors. Cells were stained with anti-PLK4-pS305 antibodies (green) and γ-tubulin (red). Scale bar represents 5 μm, and insets are high-magnification views of the regions indicated in the low-magnification images. (b) Quantification of total centrosomal pS305 signal in cells of the indicated conditions (reflected in the images shown in a). n=10 cells for each condition. Student's two-tailed paired T-test (comparing with WT): KAT2A depletion <0.001; KAT2B depletion 0.038. Note that we found no statistically significant increase on KAT2A or KAT2B depletion if each focus of PLK4-pS305 was considered instead of the total centrosomal signal per cell. (c) U2OS cells were transfected with control siRNAs (upper row), or siRNAs targeting the 3′-untranslated region (UTR) of PLK4 (PLK4 3′-UTR siRNA, lower row). Twenty-four hours later, cells were transfected again with control-empty vectors, a vector encoding the full-length PLK4-GFP, or a vector encoding the full-length PLK4-K45R/K46R-GFP double point mutant. Cells were stained for GFP (green) and Centrin-2 (Cen-2). (d) The histogram reports the average frequency of mitotic cells displaying >4 centrioles in the siRNA control condition. (e) The histogram reports the average frequency of mitotic cells displaying <3 centrioles in the siPLK4 3'-UTR condition. Data from three experiments, n=50 cells per condition. Error bars: s.e.m.

References

    1. Choudhary C. et al.. Lysine acetylation targets protein complexes and co-regulates major cellular functions. Science 325, 834–840 (2009).
    1. Kim S. C. et al.. Substrate and functional diversity of lysine acetylation revealed by a proteomics survey. Mol. Cell 23, 607–618 (2006).
    1. Zhang J. et al.. Lysine acetylation is a highly abundant and evolutionarily conserved modification in Escherichia coli. Mol. Cell. Proteomics 8, 215–225 (2009).
    1. Jeffers V. & Sullivan W. J. Jr Lysine acetylation is widespread on proteins of diverse function and localization in the protozoan parasite Toxoplasma gondii. Eukaryot. Cell 11, 735–742 (2012).
    1. Henriksen P. et al.. Proteome-wide analysis of lysine acetylation suggests its broad regulatory scope in Saccharomyces cerevisiae. Mol. Cell. Proteomics 11, 1510–1522 (2012).
    1. Weinert B. T. et al.. Proteome-wide mapping of the Drosophila acetylome demonstrates a high degree of conservation of lysine acetylation. Sci. Signal. 4, ra48 (2011).
    1. You L., Nie J., Sun W. J., Zheng Z. Q. & Yang X. J. Lysine acetylation: enzymes, bromodomains and links to different diseases. Essays Biochem. 52, 1–12 (2012).
    1. Kouzarides T. Chromatin modifications and their function. Cell 128, 693–705 (2007).
    1. Yang X. J. & Seto E. Lysine acetylation: codified crosstalk with other posttranslational modifications. Mol. Cell 31, 449–461 (2008).
    1. Hornbeck P. V. et al.. PhosphoSitePlus: a comprehensive resource for investigating the structure and function of experimentally determined post-translational modifications in man and mouse. Nucleic Acids Res. 40, D261–D270 (2012).
    1. Sadoul K., Wang J., Diagouraga B. & Khochbin S. The tale of protein lysine acetylation in the cytoplasm. J. Biomed. Biotechnol. 2011, 970382 (2011).
    1. Kelly T. J., Lerin C., Haas W., Gygi S. P. & Puigserver P. GCN5-mediated transcriptional control of the metabolic coactivator PGC-1beta through lysine acetylation. J. Biol. Chem. 284, 19945–19952 (2009).
    1. Paolinelli R., Mendoza-Maldonado R., Cereseto A. & Giacca M. Acetylation by GCN5 regulates CDC6 phosphorylation in the S phase of the cell cycle. Nat. Struct. Mol. Biol. 16, 412–420 (2009).
    1. Orpinell M. et al.. The ATAC acetyl transferase complex controls mitotic progression by targeting non-histone substrates. EMBO J. 29, 2381–2394 (2010).
    1. Espinosa M. C., Rehman M. A., Chisamore-Robert P., Jeffery D. & Yankulov K. GCN5 is a positive regulator of origins of DNA replication in Saccharomyces cerevisiae. PloS ONE 5, e8964 (2010).
    1. Xie A. Y., Bermudez V. P. & Folk W. R. Stimulation of DNA replication from the polyomavirus origin by PCAF and GCN5 acetyltransferases: acetylation of large T antigen. Mol. Cell. Biol. 22, 7907–7918 (2002).
    1. Pai C. C. et al.. A histone H3K36 chromatin switch coordinates DNA double-strand break repair pathway choice. Nat. Commun. 5, 4091 (2014).
    1. Lin W. et al.. Developmental potential of Gcn5(−/−) embryonic stem cells in vivo and in vitro. Dev. Dyn. 236, 1547–1557 (2007).
    1. Riss A. et al.. Subunits of ADA-two-A-containing (ATAC) or Spt-Ada-Gcn5-acetyltrasferase (SAGA) coactivator complexes enhance the acetyltransferase activity of GCN5. J. Biol. Chem. 290, 28997–29009 (2015).
    1. Guelman S. et al.. Host cell factor and an uncharacterized SANT domain protein are stable components of ATAC, a novel dAda2A/dGcn5-containing histone acetyltransferase complex in Drosophila. Mol. Cell. Biol. 26, 871–882 (2006).
    1. Wang Y. L., Faiola F., Xu M., Pan S. & Martinez E. Human ATAC Is a GCN5/PCAF-containing acetylase complex with a novel NC2-like histone fold module that interacts with the TATA-binding protein. J. Biol. Chem. 283, 33808–33815 (2008).
    1. Spedale G., Timmers H. T. & Pijnappel W. W. ATAC-king the complexity of SAGA during evolution. Genes Dev. 26, 527–541 (2012).
    1. Godinho S. A. & Pellman D. Causes and consequences of centrosome abnormalities in cancer. Phil. Trans. R. Soc. B 369, 20130467 (2014).
    1. Nigg E. A., Cajanek L. & Arquint C. The centrosome duplication cycle in health and disease. FEBS Lett. 588, 2366–2372 (2014).
    1. Gönczy P. Centrosomes and cancer: revisiting a long-standing relationship. Nat. Rev. Cancer 15, 639–652 (2015).
    1. Habedanck R., Stierhof Y. D., Wilkinson C. J. & Nigg E. A. The Polo kinase Plk4 functions in centriole duplication. Nat. Cell Biol. 7, 1140–1146 (2005).
    1. Bettencourt-Dias M. et al.. SAK/PLK4 is required for centriole duplication and flagella development. Curr. Biol. 15, 2199–2207 (2005).
    1. Sillibourne J. E. & Bornens M. Polo-like kinase 4: the odd one out of the family. Cell Div. 5, 25 (2010).
    1. Holland A. J., Lan W., Niessen S., Hoover H. & Cleveland D. W. Polo-like kinase 4 kinase activity limits centrosome overduplication by autoregulating its own stability. J. Cell Biol. 188, 191–198 (2010).
    1. Klebba J. E., Buster D. W., McLamarrah T. A., Rusan N. M. & Rogers G. C. Autoinhibition and relief mechanism for Polo-like kinase 4. Proc. Natl Acad. Sci. USA 112, E657–E666 (2015).
    1. Jin Q. et al.. Distinct roles of GCN5/PCAF-mediated H3K9ac and CBP/p300-mediated H3K18/27ac in nuclear receptor transactivation. EMBO J. 30, 249–262 (2011).
    1. Suo S. B. et al.. Position-specific analysis and prediction for protein lysine acetylation based on multiple features. PloS ONE 7, e49108 (2012).
    1. Lundby A. et al.. Proteomic analysis of lysine acetylation sites in rat tissues reveals organ specificity and subcellular patterns. Cell Rep. 2, 419–431 (2012).
    1. Tassy O. & Pourquie O. Manteia, a predictive data mining system for vertebrate genes and its applications to human genetic diseases. Nucleic Acids Res. 42, D882–D891 (2014).
    1. Nagy Z. & Tora L. Distinct GCN5/PCAF-containing complexes function as co-activators and are involved in transcription factor and global histone acetylation. Oncogene 26, 5341–5357 (2007).
    1. Bu P., Evrard Y. A., Lozano G. & Dent S. Y. Loss of Gcn5 acetyltransferase activity leads to neural tube closure defects and exencephaly in mouse embryos. Mol. Cell. Biol. 27, 3405–3416 (2007).
    1. Strnad P. et al.. Regulated HsSAS-6 levels ensure formation of a single procentriole per centriole during the centrosome duplication cycle. Dev. Cell 13, 203–213 (2007).
    1. Sanders M. A. & Salisbury J. L. Centrin plays an essential role in microtubule severing during flagellar excision in Chlamydomonas reinhardtii. J. Cell Biol. 124, 795–805 (1994).
    1. Chen Z., Indjeian V. B., McManus M., Wang L. & Dynlacht B. D. CP110, a cell cycle-dependent CDK substrate, regulates centrosome duplication in human cells. Dev. Cell 3, 339–350 (2002).
    1. Sillibourne J. E. et al.. Autophosphorylation of polo-like kinase 4 and its role in centriole duplication. Mol. Biol. Cell 21, 547–561 (2010).
    1. Sonnen K. F., Schermelleh L., Leonhardt H. & Nigg E. A. 3D-structured illumination microscopy provides novel insight into architecture of human centrosomes. Biol. Open 1, 965–976 (2012).
    1. Zitouni S. et al.. CDK1 prevents unscheduled PLK4-STIL complex assembly in centriole biogenesis. Curr. Biol. 26, 1127–1137 (2016).
    1. Lake R. J. & Jelinek W. R. Cell cycle- and terminal differentiation-associated regulation of the mouse mRNA encoding a conserved mitotic protein kinase. Mol. Cell. Biol. 13, 7793–7801 (1993).
    1. Dulic V., Lees E. & Reed S. I. Association of human cyclin E with a periodic G1-S phase protein kinase. Science 257, 1958–1961 (1992).
    1. Tokarski J. S. et al.. The structure of Dasatinib (BMS-354825) bound to activated ABL kinase domain elucidates its inhibitory activity against imatinib-resistant ABL mutants. Cancer Res. 66, 5790–5797 (2006).
    1. Jura N. et al.. Catalytic control in the EGF receptor and its connection to general kinase regulatory mechanisms. Mol. Cell 42, 9–22 (2011).
    1. Aad G. et al.. Search for diphoton events with large missing transverse energy in 7 TeV proton-proton collisions with the ATLAS detector. Phys. Rev. Lett. 106, 121803 (2011).
    1. Fujimoto H. et al.. A possible overestimation of the effect of acetylation on lysine residues in KQ mutant analysis. J. Comput. Chem. 33, 239–246 (2012).
    1. Mateo F. et al.. The transcriptional co-activator PCAF regulates cdk2 activity. Nucleic Acids Res. 37, 7072–7084 (2009).
    1. Lee J. et al.. Acetylation of cyclin-dependent kinase 5 is mediated by GCN5. Biochem. Biophys. Res. Commun. 447, 121–127 (2014).
    1. Sabo A., Lusic M., Cereseto A. & Giacca M. Acetylation of conserved lysines in the catalytic core of cyclin-dependent kinase 9 inhibits kinase activity and regulates transcription. Mol. Cell. Biol. 28, 2201–2212 (2008).
    1. Hatch E. M., Kulukian A., Holland A. J., Cleveland D. W. & Stearns T. Cep152 interacts with Plk4 and is required for centriole duplication. J. Cell Biol. 191, 721–729 (2010).
    1. Moyer T. C., Clutario K. M., Lambrus B. G., Daggubati V. & Holland A. J. Binding of STIL to Plk4 activates kinase activity to promote centriole assembly. J. Cell Biol. 209, 863–878 (2015).
    1. Ling H., Peng L., Seto E. & Fukasawa K. Suppression of centrosome duplication and amplification by deacetylases. Cell Cycle 11, 3779–3791 (2012).
    1. Yamauchi Y. et al.. Histone deacetylase 8 is required for centrosome cohesion and influenza A virus entry. PLoS Pathog. 7, e1002316 (2011).
    1. Rosario C. O. et al.. Plk4 is required for cytokinesis and maintenance of chromosomal stability. Proc. Natl Acad. Sci. USA 107, 6888–6893 (2010).
    1. Yamashita Y. et al.. Sak serine-threonine kinase acts as an effector of Tec tyrosine kinase. J. Biol. Chem. 276, 39012–39020 (2001).
    1. Brand M. et al.. UV-damaged DNA-binding protein in the TFTC complex links DNA damage recognition to nucleosome acetylation. EMBO J. 20, 3187–3196 (2001).
    1. Esashi F. et al.. CDK-dependent phosphorylation of BRCA2 as a regulatory mechanism for recombinational repair. Nature 434, 598–604 (2005).
    1. Schorl C. & Sedivy J. M. Analysis of cell cycle phases and progression in cultured mammalian cells. Methods 41, 143–150 (2007).
    1. Keller D. et al.. Mechanisms of HsSAS-6 assembly promoting centriole formation in human cells. J. Cell Biol. 204, 697–712 (2014).
    1. Wong Y. L. et al.. Cell biology. Reversible centriole depletion with an inhibitor of Polo-like kinase 4. Science 348, 1155–1160 (2015).
    1. Sampson P. B. et al.. The discovery of Polo-like kinase 4 inhibitors: design and optimization of spiro[cyclopropane-1,3′[3H]indol]-2′(1′H).ones as orally bioavailable antitumor agents. J. Med. Chem. 58, 130–146 (2015).
    1. Eswar N. et al.. Tools for comparative protein structure modeling and analysis. Nucleic Acids Res. 31, 3375–3380 (2003).
    1. Gustafson W. C. et al.. Drugging MYCN through an allosteric transition in Aurora kinase A. Cancer Cell 26, 414–427 (2014).
    1. Shen M. Y. & Sali A. Statistical potential for assessment and prediction of protein structures. Protein Sci. 15, 2507–2524 (2006).
    1. Babu C. S., Dudev T. & Lim C. Differential role of the protein matrix on the binding of a catalytic aspartate to Mg2+ vs Ca2+: application to ribonuclease H. J. Am. Chem. Soc. 135, 6541–6548 (2013).
    1. Brunger A. & Karplus M. Polar hydrogen positions in proteins: empirical energy placement and neutron diffraction comparison. Proteins 4, 148–156 (1988).
    1. Brooks B. R. et al.. CHARMM: the biomolecular simulation program. J. Comput. Chem. 30, 1545–1614 (2009).
    1. Phillips J. C. et al.. Scalable molecular dynamics with NAMD. J. Comput. Chem. 26, 1781–1802 (2005).
    1. Huang J. & MacKerell A. D. Jr CHARMM36 all-atom additive protein force field: validation based on comparison to NMR data. J. Comput. Chem. 34, 2135–2145 (2013).
    1. Grauffel C., Stote R. H. & Dejaegere A. Force field parameters for the simulation of modified histone tails. J. Comput. Chem. 31, 2434–2451 (2010).
    1. Florens L. et al.. Analyzing chromatin remodeling complexes using shotgun proteomics and normalized spectral abundance factors. Methods 40, 303–311 (2006).
    1. Eng J. K., McCormack A. L. & Yates J. R. An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database. J. Am. Soc. Mass Spectrom. 5, 976–989 (1994).
    1. Vizcaino J. A. et al.. 2016 update of the PRIDE database and its related tools. Nucleic Acids Res. 44, D447–D456 (2016).

Source: PubMed

3
Suscribir