2,060
Views
18
CrossRef citations to date
0
Altmetric
Review

The nuclear phosphoinositide response to stress

, , ORCID Icon, , , , ORCID Icon & show all

References

  • Hokin MR, Hokin LE. Enzyme secretion and the incorporation of P32 into phospholipides of pancreas slices. J Biol Chem. 1953;203:967–977.
  • Di Paolo G, De Camilli P. Phosphoinositides in cell regulation and membrane dynamics. Nature. 2006;443:651–657.
  • Rose HG, Frenster JH. Composition and metabolism of lipids within repressed and active chromatin of interphase lymphocytes. Biochim Biophys Acta. 1965;106:577–591.
  • Manzoli FA, Maraldi NM, Cocco L, et al. Chromatin phospholipids in normal and chronic lymphocytic leukemia lymphocytes. Cancer Res. 1977;37:843–849.
  • Smith CD, Wells WW. Phosphorylation of rat liver nuclear envelopes. II. Characterization of in vitro lipid phosphorylation. J Biol Chem. 1983;258:9368–9373.
  • Cocco L, Gilmour RS, Ognibene A, et al. Synthesis of polyphosphoinositides in nuclei of friend cells. Evidence for polyphosphoinositide metabolism inside the nucleus which changes with cell differentiation. Biochem J. 1987;248:765–770.
  • Boronenkov IV, Loijens JC, Umeda M, et al. Phosphoinositide signaling pathways in nuclei are associated with nuclear speckles containing pre-mRNA processing factors. Mol Biol Cell. 1998;9:3547–3560.
  • Barlow CA, Laishram RS, Anderson RA. Nuclear phosphoinositides: a signaling enigma wrapped in a compartmental conundrum. Trends Cell Biol. 2010;20:25–35.
  • Sztacho M, Sobol M, Balaban C, et al. Nuclear phosphoinositides and phase separation: important players in nuclear compartmentalization. Adv Biol Regul. 2019;71:111–117.
  • Castano E, Yildirim S, Faberova V, et al. Nuclear phosphoinositides-versatile regulators of genome functions. Cells. 2019;8(7):649.
  • Dickson EJ, Hille B. Understanding phosphoinositides: rare, dynamic, and essential membrane phospholipids. Biochem J. 2019;476:1–23.
  • Shisheva A. Regulating Glut4 vesicle dynamics by phosphoinositide kinases and phosphoinositide phosphatases. Front Biosci. 2003;8:s945–s946.
  • Rameh LE, Cantley LC. The role of phosphoinositide 3-kinase lipid products in cell function. J Biol Chem. 1999;274:8347–8350.
  • Liu Y, Bankaitis VA. Phosphoinositide phosphatases in cell biology and disease. Prog Lipid Res. 2010;49:201–217.
  • Sarkes D, Rameh LE. A novel HPLC-based approach makes possible the spatial characterization of cellular PtdIns5P and other phosphoinositides. Biochem J. 2010;428:375–384.
  • Bryant JM, Blind RD. Signaling through non-membrane nuclear phosphoinositide binding proteins in human health and disease. J Lipid Res. 2019;60:299–311.
  • Fu P, Ebenezer DL, Ha AW, et al. Nuclear lipid mediators: role of nuclear sphingolipids and sphingosine-1-phosphate signaling in epigenetic regulation of inflammation and gene expression. J Cell Biochem. 2018;119:6337–6353.
  • Poli A, Billi AM, Mongiorgi S, et al. Nuclear phosphatidylinositol signaling: focus on phosphatidylinositol phosphate kinases and phospholipases C. J Cell Physiol. 2016;231:1645–1655.
  • Hamann BL, Blind RD. Nuclear phosphoinositide regulation of chromatin. J Cell Physiol. 2018;233:107–123.
  • Jacobsen RG, Mazloumi Gavgani F, Edson AJ, et al. Polyphosphoinositides in the nucleus: roadmap of their effectors and mechanisms of interaction. Adv Biol Regul. 2019;72:7–21.
  • Shah ZH, Jones DR, Sommer L, et al. Nuclear phosphoinositides and their impact on nuclear functions. Febs J. 2013;280:6295–6310.
  • Gillooly DJ, Morrow IC, Lindsay M, et al. Localization of phosphatidylinositol 3-phosphate in yeast and mammalian cells. Embo J. 2000;19:4577–4588.
  • Kalasova I, Faberova V, Kalendova A, et al. Tools for visualization of phosphoinositides in the cell nucleus. Histochem Cell Biol. 2016;145:485–496.
  • Sindic A, Aleksandrova A, Fields AP, et al. Presence and activation of nuclear phosphoinositide 3-kinase C2beta during compensatory liver growth. J Biol Chem. 2001;276:17754–17761.
  • Choi S, Chen M, Cryns VL, et al. A nuclear phosphoinositide kinase complex regulates p53. Nat Cell Biol. 2019;21:462–475.
  • Clarke JH, Letcher AJ, D’Santos CS, et al. Inositol lipids are regulated during cell cycle progression in the nuclei of murine erythroleukaemia cells. Biochem J. 2001;357:905–910.
  • Zhang L, Malik S, Pang J, et al. Phospholipase Cepsilon hydrolyzes perinuclear phosphatidylinositol 4-phosphate to regulate cardiac hypertrophy. Cell. 2013;153:216–227.
  • Wang YH, Hariharan A, Bastianello G, et al. DNA damage causes rapid accumulation of phosphoinositides for ATR signaling. Nat Commun. 2017;8:2118.
  • Gozani O, Karuman P, Jones DR, et al. The PHD finger of the chromatin-associated protein ING2 functions as a nuclear phosphoinositide receptor. Cell. 2003;114:99–111.
  • Jones DR, Bultsma Y, Keune WJ, et al. Nuclear PtdIns5P as a transducer of stress signaling: an in vivo role for PIP4Kbeta. Mol Cell. 2006;23:685–695.
  • Bua DJ, Martin GM, Binda O, et al. Nuclear phosphatidylinositol-5-phosphate regulates ING2 stability at discrete chromatin targets in response to DNA damage. Sci Rep. 2013;3:2137.
  • Bunce MW, Boronenkov IV, Anderson RA. Coordinated activation of the nuclear ubiquitin ligase Cul3-SPOP by the generation of phosphatidylinositol 5-phosphate. J Biol Chem. 2008;283:8678–8686.
  • Poli A, Zaurito AE, Abdul-Hamid S, et al. Phosphatidylinositol 5 phosphate (PI5P): from behind the scenes to the front (nuclear) stage. Int J Mol Sci. 2019;20(9):2080.
  • Van der Kaay J, Beck M, Gray A, et al. Distinct phosphatidylinositol 3-kinase lipid products accumulate upon oxidative and osmotic stress and lead to different cellular responses. J Biol Chem. 1999;274:35963–35968.
  • Deleris P, Bacqueville D, Gayral S, et al. SHIP-2 and PTEN are expressed and active in vascular smooth muscle cell nuclei, but only SHIP-2 is associated with nuclear speckles. J Biol Chem. 2003;278:38884–38891.
  • Ehm P, Nalaskowski MM, Wundenberg T, et al. The tumor suppressor SHIP1 colocalizes in nucleolar cavities with p53 and components of PML nuclear bodies. Nucleus. 2015;6:154–164.
  • Yokogawa T, Nagata S, Nishio Y, et al. Evidence that 3 ‘-phosphorylated polyphosphoinositides are generated at the nuclear surface: use of immunostaining technique with monoclonal antibodies specific for PI 3,4-P-2. Febs Lett. 2000;473:222–226.
  • Osborne SL, Thomas CL, Gschmeissner S, et al. Nuclear PtdIns(4,5)P2 assembles in a mitotically regulated particle involved in pre-mRNA splicing. J Cell Sci. 2001;114:2501–2511.
  • Yoo SH, Huh YH, Huh SK, et al. Localization and projected role of phosphatidylinositol 4-kinases IIalpha and IIbeta in inositol 1,4,5-trisphosphate-sensitive nucleoplasmic Ca(2)(+) store vesicles. Nucleus. 2014;5:341–351.
  • Watt SA, Kular G, Fleming IN, et al. Subcellular localization of phosphatidylinositol 4,5-bisphosphate using the pleckstrin homology domain of phospholipase C delta1. Biochem J. 2002;363:657–666.
  • Sobol M, Krausova A, Yildirim S, et al. Nuclear phosphatidylinositol 4,5-bisphosphate islets contribute to efficient RNA polymerase II-dependent transcription. J Cell Sci. 2018;131:8.
  • Li W, Laishram RS, Ji Z, et al. Star-PAP control of BIK expression and apoptosis is regulated by nuclear PIPKIalpha and PKCdelta signaling. Mol Cell. 2012;45:25–37.
  • Mellman DL, Gonzales ML, Song C, et al. A PtdIns4,5P2-regulated nuclear poly(A) polymerase controls expression of select mRNAs. Nature. 2008;451:1013–1017.
  • Mishkind M, Vermeer JE, Darwish E, et al. Heat stress activates phospholipase D and triggers PIP accumulation at the plasma membrane and nucleus. Plant J. 2009;60:10–21.
  • Lindsay Y, McCoull D, Davidson L, et al. Localization of agonist-sensitive PtdIns(3,4,5)P3 reveals a nuclear pool that is insensitive to PTEN expression. J Cell Sci. 2006;119:5160–5168.
  • Karlsson T, Altankhuyag A, Dobrovolska O, et al. A polybasic motif in ErbB3-binding protein 1 (EBP1) has key functions in nucleolar localization and polyphosphoinositide interaction. Biochem J. 2016;473:2033–2047.
  • Tanaka K, Horiguchi K, Yoshida T, et al. Evidence that a phosphatidylinositol 3,4,5-trisphosphate-binding protein can function in nucleus. J Biol Chem. 1999;274:3919–3922.
  • Kobayashi M, Muroyama A, Ohizumi Y. Phosphatidylinositol 4,5-bisphosphate enhances calcium release from sarcoplasmic reticulum of skeletal muscle. Biochem Biophys Res Commun. 1989;163:1487–1491.
  • Prole DL, Taylor CW. Structure and function of IP3 receptors. Cold Spring Harb Perspect Biol. 2019;11(4).
  • Romanauska A, Kohler A. The inner nuclear membrane is a metabolically active territory that generates nuclear lipid droplets. Cell. 2018;174:700-+.
  • Payrastre B, Nievers M, Boonstra J, et al. A differential location of phosphoinositide kinases, diacylglycerol kinase, and phospholipase-C in the nuclear matrix. J Biol Chem. 1992;267:5078–5084.
  • Zima AV, Bare DJ, Mignery GA, et al. IP3-dependent nuclear Ca2+ signalling in the mammalian heart. J Physiol. 2007;584:601–611.
  • Humbert JP, Matter N, Artault JC, et al. Inositol 1,4,5-trisphosphate receptor is located to the inner nuclear membrane vindicating regulation of nuclear calcium signaling by inositol 1,4,5-trisphosphate. Discrete distribution of inositol phosphate receptors to inner and outer nuclear membranes (vol 271, pg 478, 1996). J Biol Chem. 1996;271:5287.
  • Echevarria W, Leite MF, Guerra MT, et al. Regulation of calcium signals in the nucleus by a nucleoplasmic reticulum. Nat Cell Biol. 2003;5:440–446.
  • Tabellini G, Bortul R, Santi S, et al. Diacylglycerol kinase-theta is localized in the speckle domains of the nucleus. Exp Cell Res. 2003;287:143–154.
  • Neri LM, Bortul R, Borgatti P, et al. Proliferating or differentiating stimuli act on different lipid-dependent signaling pathways in nuclei of human leukemia cells. Mol Biol Cell. 2002;13:947–964.
  • Zhang H, Shao Z, Alibin CP, et al. Liganded peroxisome proliferator-activated receptors (PPARs) preserve nuclear histone deacetylase 5 levels in endothelin-treated Sprague-Dawley rat cardiac myocytes. PLoS One. 2014;9:e115258.
  • Arantes LAM, Aguiar CJ, Amaya MJ, et al. Nuclear inositol 1,4,5-trisphosphate is a necessary and conserved signal for the induction of both pathological and physiological cardiomyocyte hypertrophy. J Mol Cell Cardiol. 2012;53:475–486.
  • Balla A, Balla T. Phosphatidylinositol 4-kinases: old enzymes with emerging functions. Trends Cell Biol. 2006;16:351–361.
  • Boura E, Nencka R. Phosphatidylinositol 4-kinases: function, structure, and inhibition. Exp Cell Res. 2015;337:136–145.
  • Kakuk A, Friedlander E, Vereb G Jr., et al. Nucleolar localization of phosphatidylinositol 4-kinase PI4K230 in various mammalian cells. Cytometry A. 2006;69:1174–1183.
  • de Graaf P, Klapisz EE, Schulz TK, et al. Nuclear localization of phosphatidylinositol 4-kinase beta. J Cell Sci. 2002;115:1769–1775.
  • Szivak I, Lamb N, Heilmeyer LM. Subcellular localization and structural function of endogenous phosphorylated phosphatidylinositol 4-kinase (PI4K92). J Biol Chem. 2006;281:16740–16749.
  • Heilmeyer LM Jr., Vereb G Jr., Vereb G, et al. Mammalian phosphatidylinositol 4-kinases. IUBMB Life. 2003;55:59–65.
  • Schill NJ, Anderson RA. Two novel phosphatidylinositol-4-phosphate 5-kinase type Igamma splice variants expressed in human cells display distinctive cellular targeting. Biochem J. 2009;422:473–482.
  • Chakrabarti R, Sanyal S, Ghosh A, et al. Phosphatidylinositol-4-phosphate 5-kinase 1alpha modulates ribosomal RNA gene Silencing through its interaction with histone H3 lysine 9 trimethylation and heterochromatin protein HP1-alpha. J Biol Chem. 2015;290:20893–20903.
  • Ciruela A, Hinchliffe KA, Divecha N, et al. Nuclear targeting of the beta isoform of type II phosphatidylinositol phosphate kinase (phosphatidylinositol 5-phosphate 4-kinase) by its alpha-helix 7. Biochem J. 2000;346(Pt 3):587–591.
  • Richardson JP, Wang MC, Clarke JH, et al. Genomic tagging of endogenous Type II beta phosphatidylinositol 5-phosphate 4-kinase in DT40 cells reveals a nuclear localisation. Cell Signal. 2007;19:1309–1314.
  • Kouchi Z, Fujiwara Y, Yamaguchi H, et al. Phosphatidylinositol 5-phosphate 4-kinase type II beta is required for vitamin D receptor-dependent E-cadherin expression in SW480 cells. Biochem Biophys Res Commun. 2011;408:523–529.
  • Stijf-Bultsma Y, Sommer L, Tauber M, et al. The basal transcription complex component TAF3 transduces changes in nuclear phosphoinositides into transcriptional output. Mol Cell. 2015;58:453–467.
  • Jean S, Kiger AA. Classes of phosphoinositide 3-kinases at a glance. J Cell Sci. 2014;127:923–928.
  • Kumar A, Redondo-Munoz J, Perez-Garcia V, et al. Nuclear but not cytosolic phosphoinositide 3-kinase beta has an essential function in cell survival. Mol Cell Biol. 2011;31:2122–2133.
  • Kumar A, Fernandez-Capetillo O, Carrera AC. Nuclear phosphoinositide 3-kinase beta controls double-strand break DNA repair. Proc Natl Acad Sci U S A. 2010;107:7491–7496.
  • Marques M, Kumar A, Poveda AM, et al. Specific function of phosphoinositide 3-kinase beta in the control of DNA replication. Proc Natl Acad Sci U S A. 2009;106:7525–7530.
  • Didichenko SA, Thelen M. Phosphatidylinositol 3-kinase c2alpha contains a nuclear localization sequence and associates with nuclear speckles. J Biol Chem. 2001;276:48135–48142.
  • Banfic H, Visnjic D, Mise N, et al. Epidermal growth factor stimulates translocation of the class II phosphoinositide 3-kinase PI3K-C2beta to the nucleus. Biochem J. 2009;422:53–60.
  • Resnick AC, Snowman AM, Kang BN, et al. Inositol polyphosphate multikinase is a nuclear P13-kinase with transcriptional regulatory activity. Proc Natl Acad Sci U S A. 2005;102:12783–12788.
  • Kim E, Beon J, Lee S, et al. IPMK: A versatile regulator of nuclear signaling events. Adv Biol Regul. 2016;61:25–32.
  • Blind RD, Suzawa M, Ingraham HA. Direct modification and activation of a nuclear receptor-PIP(2) complex by the inositol lipid kinase IPMK. Sci Signal. 2012;5:ra44.
  • Xu R, Sen N, Paul BD, et al. Inositol polyphosphate multikinase is a coactivator of p53-mediated transcription and cell death. Sci Signal. 2013;6:ra22.
  • Lee YR, Chen M, Pandolfi PP. The functions and regulation of the PTEN tumour suppressor: new modes and prospects. Nat Rev Mol Cell Biol. 2018;19:547–562.
  • Li P, Wang D, Li H, et al. Identification of nucleolus-localized PTEN and its function in regulating ribosome biogenesis. Mol Biol Rep. 2014;41:6383–6390.
  • Shen WH, Balajee AS, Wang J, et al. Essential role for nuclear PTEN in maintaining chromosomal integrity. Cell. 2007;128:157–170.
  • Choi BH, Chen Y, Dai W. Chromatin PTEN is involved in DNA damage response partly through regulating Rad52 sumoylation. Cell Cycle. 2013;12:3442–3447.
  • Chang CJ, Mulholland DJ, Valamehr B, et al. PTEN nuclear localization is regulated by oxidative stress and mediates p53-dependent tumor suppression. Mol Cell Biol. 2008;28:3281–3289.
  • Trotman LC, Pandolfi PP. PTEN and p53: who will get the upper hand?. Cancer Cell. 2003;3:97–99.
  • Freeman DJ, Li AG, Wei G, et al. PTEN tumor suppressor regulates p53 protein levels and activity through phosphatase-dependent and -independent mechanisms. Cancer Cell. 2003;3:117–130.
  • Mayo LD, Donner DB. The PTEN, Mdm2, p53 tumor suppressor-oncoprotein network. Trends Biochem Sci. 2002;27:462–467.
  • Mayo LD, Dixon JE, Durden DL, et al. PTEN protects p53 from Mdm2 and sensitizes cancer cells to chemotherapy. J Biol Chem. 2002;277:5484–5489.
  • Stambolic V, MacPherson D, Sas D, et al. Regulation of PTEN transcription by p53. Mol Cell. 2001;8:317–325.
  • Elong Edimo W, Derua R, Janssens V, et al. Evidence of SHIP2 Ser132 phosphorylation, its nuclear localization and stability. Biochem J. 2011;439:391–401.
  • Zou J, Marjanovic J, Kisseleva MV, et al. Type I phosphatidylinositol-4, 5-bisphosphate 4-phosphatase regulates stress-induced apoptosis. Proc Natl Acad Sci U S A. 2007;104:16834–16839.
  • Cocco L, Rubbini S, Manzoli L, et al. Inositides in the nucleus: presence and characterisation of the isozymes of phospholipase beta family in NIH 3T3 cells. Biochim Biophys Acta. 1999;1438:295–299.
  • Lee YH, Kim SY, Kim JR, et al. Overexpression of phospholipase Cbeta-1 protects NIH3T3 cells from oxidative stress-induced cell death. Life Sci. 2000;67:827–837.
  • Stallings JD, Tall EG, Pentyala S, et al. Nuclear translocation of phospholipase C-delta(1) is linked to the cell cycle and nuclear phosphatidylinositol 4,5-bisphosphate. J Biol Chem. 2005;280:22060–22069.
  • Kunrath-Lima M, de Miranda MC, Ferreira ADF, et al. Phospholipase C delta 4 (PLCdelta4) is a nuclear protein involved in cell proliferation and senescence in mesenchymal stromal stem cells. Cell Signal. 2018;49:59–67.
  • Chi B, Wang Q, Wu G, et al. Aly and THO are required for assembly of the human TREX complex and association of TREX components with the spliced mRNA. Nucleic Acids Res. 2013;41:1294–1306.
  • Okada M, Jang SW, Ye K. Akt phosphorylation and nuclear phosphoinositide association mediate mRNA export and cell proliferation activities by ALY. Proc Natl Acad Sci U S A. 2008;105:8649–8654.
  • Wickramasinghe VO, Savill JM, Chavali S, et al. Human inositol polyphosphate multikinase regulates transcript-selective nuclear mRNA export to preserve genome integrity. Mol Cell. 2013;51:737–750.
  • Toska E, Campbell HA, Shandilya J, et al. Repression of transcription by WT1-BASP1 requires the myristoylation of BASP1 and the PIP2-dependent recruitment of histone deacetylase. Cell Rep. 2012;2:462–469.
  • Wu C, Wang Y, Xia Y, et al. Wilms’ tumor 1 enhances Cisplatin-resistance of advanced NSCLC. Febs Lett. 2014;588:4566–4572.
  • Gadadhar S, Bora N, Tiwari V, et al. Sequestration of the abrin A chain to the nucleus by BASP1 increases the resistance of cells to abrin toxicity. Biochem J. 2014;458:375–385.
  • Kadoch C, Crabtree GR. Mammalian SWI/SNF chromatin remodeling complexes and cancer: mechanistic insights gained from human genomics. Sci Adv. 2015;1:e1500447.
  • Khan P, Drobic B, Perez-Cadahia B, et al. Mitogen- and stress-activated protein kinases 1 and 2 are required for maximal trefoil factor 1 induction. PLoS One. 2013;8:e63189.
  • Li F, Liang J, Tang D. Brahma-related gene 1 ameliorates the neuronal apoptosis and oxidative stress induced by oxygen-glucose deprivation/reoxygenation through activation of Nrf2/HO-1 signaling. Biomed Pharmacother. 2018;108:1216–1224.
  • Porter EG, Dhiman A, Chowdhury B, et al. PBRM1 regulates stress response in epithelial cells. iScience. 2019;15:196–210.
  • Zhao K, Wang W, Rando OJ, et al. Rapid and phosphoinositol-dependent binding of the SWI/SNF-like BAF complex to chromatin after T lymphocyte receptor signaling. Cell. 1998;95:625–636.
  • Rando OJ, Zhao K, Janmey P, et al. Phosphatidylinositol-dependent actin filament binding by the SWI/SNF-like BAF chromatin remodeling complex. Proc Natl Acad Sci U S A. 2002;99:2824–2829.
  • Shubina MY, Musinova YR, Sheval EV. Nucleolar methyltransferase fibrillarin: evolution of structure and functions. Biochemistry (Mosc). 2016;81:941–950.
  • Yildirim S, Castano E, Sobol M, et al. Involvement of phosphatidylinositol 4,5-bisphosphate in RNA polymerase I transcription. J Cell Sci. 2013;126:2730–2739.
  • Tiku V, Kew C, Mehrotra P, et al. Nucleolar fibrillarin is an evolutionarily conserved regulator of bacterial pathogen resistance. Nat Commun. 2018;9(1):3607.
  • Sobol M, Yildirim S, Philimonenko VV, et al. UBF complexes with phosphatidylinositol 4,5-bisphosphate in nucleolar organizer regions regardless of ongoing RNA polymerase I activity. Nucleus. 2013;4:478–486.
  • Kodiha M, Banski P, Stochaj U. Computer-based fluorescence quantification: a novel approach to study nucleolar biology. BMC Cell Biol. 2011;12:25.
  • Nagashima M, Shiseki M, Miura K, et al. DNA damage-inducible gene p33ING2 negatively regulates cell proliferation through acetylation of p53. Proc Natl Acad Sci U S A. 2001;98:9671–9676.
  • Lindstrom MS. NPM1/B23: A multifunctional chaperone in ribosome biogenesis and chromatin remodeling. Biochem Res Int. 2011;2011:195209.
  • Li J, Zhang X, Sejas DP, et al. Negative regulation of p53 by nucleophosmin antagonizes stress-induced apoptosis in human normal and malignant hematopoietic cells. Leuk Res. 2005;29:1415–1423.
  • Kurki S, Peltonen K, Latonen L, et al. Nucleolar protein NPM interacts with HDM2 and protects tumor suppressor protein p53 from HDM2-mediated degradation. Cancer Cell. 2004;5:465–475.
  • Borer RA, Lehner CF, Eppenberger HM, et al. Major nucleolar proteins shuttle between nucleus and cytoplasm. Cell. 1989;56:379–390.
  • Ahn JY, Liu X, Cheng D, et al. Nucleophosmin/B23, a nuclear PI(3,4,5)P(3) receptor, mediates the antiapoptotic actions of NGF by inhibiting CAD. Mol Cell. 2005;18:435–445.
  • Feric M, Vaidya N, Harmon TS, et al. Coexisting liquid phases underlie nucleolar subcompartments. Cell. 2016;165:1686–1697.
  • Mitrea DM, Cika JA, Stanley CB, et al. Self-interaction of NPM1 modulates multiple mechanisms of liquid-liquid phase separation. Nat Commun. 2018;9:842.
  • Ulicna L, Kalendova A, Kalasova I, et al. PIP2 epigenetically represses rRNA genes transcription interacting with PHF8. Biochim Biophys Acta Mol Cell Biol Lipids. 2018;1863:266–275.
  • Wang Q, Ma S, Song N, et al. Stabilization of histone demethylase PHF8 by USP7 promotes breast carcinogenesis. J Clin Invest. 2016;126:2205–2220.
  • Viiri KM, Janis J, Siggers T, et al. DNA-binding and -bending activities of SAP30L and SAP30 are mediated by a zinc-dependent module and monophosphoinositides. Mol Cell Biol. 2009;29:342–356.
  • Viiri KM, Korkeamaki H, Kukkonen MK, et al. SAP30L interacts with members of the Sin3A corepressor complex and targets Sin3A to the nucleolus. Nucleic Acids Res. 2006;34:3288–3298.
  • Krylova IN, Sablin EP, Moore J, et al. Structural analyses reveal phosphatidyl inositols as ligands for the NR5 orphan receptors SF-1 and LRH-1. Cell. 2005;120:343–355.
  • Blind RD, Sablin EP, Kuchenbecker KM, et al. The signaling phospholipid PIP3 creates a new interaction surface on the nuclear receptor SF-1. Proc Natl Acad Sci U S A. 2014;111:15054–15059.
  • Bland ML, Jamieson CA, Akana SF, et al. Haploinsufficiency of steroidogenic factor-1 in mice disrupts adrenal development leading to an impaired stress response. Proc Natl Acad Sci U S A. 2000;97:14488–14493.
  • Mortier E, Wuytens G, Leenaerts I, et al. Nuclear speckles and nucleoli targeting by PIP2-PDZ domain interactions. Embo J. 2005;24:2556–2565.
  • Liu XL, Gao QQ, Li PS, et al. UHRF1 targets DNMT1 for DNA methylation through cooperative binding of hemi-methylated DNA and methylated H3K9. Nat Commun. 2013;4:1563.
  • Bronner C, Fuhrmann G, Chedin FL, et al. UHRF1 links the histone code and DNA methylation to ensure faithful epigenetic memory inheritance. Genet Epigenet. 2010;2009:29–36.
  • Wang F, Yang YZ, Shi CZ, et al. UHRF1 promotes cell growth and metastasis through repression of p16(ink4a) in colorectal cancer. Ann Surg Oncol. 2012;19:2753–2762.
  • Gelato KA, Tauber M, Ong MS, et al. Accessibility of different histone H3-binding domains of UHRF1 is allosterically regulated by phosphatidylinositol 5-phosphate. Mol Cell. 2014;54:905–919.
  • Tian YY, Paramasivam M, Ghosal G, et al. UHRF1 contributes to DNA damage repair as a lesion recognition factor and nuclease scaffold. Cell Rep. 2015;10:1957–1966.
  • Abu-Alainin W, Gana T, Liloglou T, et al. UHRF1 regulation of the Keap1-Nrf2 pathway in pancreatic cancer contributes to oncogenesis. J Pathol. 2016;238:423–433.
  • Bronner C, Achour M, Arima Y, et al. The UHRF family: oncogenes that are drugable targets for cancer therapy in the near future? Pharmacol Ther. 2007;115:419–434.
  • Unoki M, Brunet J, Mousli M. Drug discovery targeting epigenetic codes: the great potential of UHRF1, which links DNA methylation and histone modifications, as a drug target in cancers and toxoplasmosis. Biochem Pharmacol. 2009;78:1279–1288.
  • Trippe R, Guschina E, Hossbach M, et al. Identification, cloning, and functional analysis of the human U6 snRNA-specific terminal uridylyl transferase. RNA. 2006;12:1494–1504.
  • Li W, Li W, Laishram RS, et al. Distinct regulation of alternative polyadenylation and gene expression by nuclear poly(A) polymerases. Nucleic Acids Res. 2017;45:8930–8942.
  • Laishram RS, Anderson RA. The poly A polymerase star-PAP controls 3ʹ-end cleavage by promoting CPSF interaction and specificity toward the pre-mRNA. Embo J. 2010;29:4132–4145.
  • Laishram RS, Barlow CA, Anderson RA. CKI isoforms α and ε regulate star-PAP target messages by controlling star-PAP poly(A) polymerase activity and phosphoinositide stimulation. Nucleic Acids Res. 2011;39:7961–7973.
  • Dai C, Gu W. p53 post-translational modification: deregulated in tumorigenesis. Trends Mol Med. 2010;16:528–536.
  • Bieging KT, Mello SS, Attardi LD. Unravelling mechanisms of p53-mediated tumour suppression. Nat Rev Cancer. 2014;14:359–370.
  • Kruiswijk F, Labuschagne CF, Vousden KH. p53 in survival, death and metabolic health: a lifeguard with a licence to kill. Nat Rev Mol Cell Biol. 2015;16:393–405.
  • Freed-Pastor WA, Prives C. Mutant p53: one name, many proteins. Genes Dev. 2012;26:1268–1286.
  • Muller PA, Vousden KH. Mutant p53 in cancer: new functions and therapeutic opportunities. Cancer Cell. 2014;25:304–317.
  • Weibrecht I, Leuchowius KJ, Clausson CM, et al. Proximity ligation assays: a recent addition to the proteomics toolbox. Expert Rev Proteomics. 2010;7:401–409.
  • Sehat B, Tofigh A, Lin Y, et al. SUMOylation mediates the nuclear translocation and signaling of the IGF-1 receptor. Sci Signal. 2010;3:ra10.
  • Chen M, Qiu T, Wu J, et al. Extracellular anti-angiogenic proteins augment an endosomal protein trafficking pathway to reach mitochondria and execute apoptosis in HUVECs. Cell Death Differ. 2018;25:1905–1920.
  • Chen M, Zhang Y, Yu VC, et al. Isthmin targets cell-surface GRP78 and triggers apoptosis via induction of mitochondrial dysfunction. Cell Death Differ. 2014;21:797–810.
  • Semenas J, Hedblom A, Miftakhova RR, et al. The role of PI3K/AKT-related PIP5K1alpha and the discovery of its selective inhibitor for treatment of advanced prostate cancer. Proc Natl Acad Sci U S A. 2014;111:E3689–E3698.
  • Arrigo AP, Gibert B. HspB1, HspB5 and HspB4 in human cancers: potent oncogenic role of some of their client proteins. Cancers (Basel). 2014;6:333–365.
  • Malin D, Petrovic V, Strekalova E, et al. alphaB-crystallin: portrait of a malignant chaperone as a cancer therapeutic target. Pharmacol Ther. 2016;160:1–10.
  • Fiume R, Faenza I, Sheth B, et al. Nuclear phosphoinositides: their regulation and roles in nuclear functions. Int J Mol Sci. 2019;20:2991.
  • Choi S, Hedman AC, Sayedyahossein S, et al. Agonist-stimulated phosphatidylinositol-3,4,5-trisphosphate generation by scaffolded phosphoinositide kinases. Nat Cell Biol. 2016;18:1324–1335.
  • Chen M, Choi S, Jung O, et al. The specificity of EGF-stimulated IQGAP1 scaffold towards the PI3K-Akt pathway is defined by the IQ3 motif. Sci Rep. 2019;9:9126.
  • Kakuk A, Friedlander E, Vereb G Jr., et al. Nuclear and nucleolar localization signals and their targeting function in phosphatidylinositol 4-kinase PI4K230. Exp Cell Res. 2008;314:2376–2388.
  • Visnjic D, Crljen V, Curic J, et al. The activation of nuclear phosphoinositide 3-kinase C2beta in all-trans-retinoic acid-differentiated HL-60 cells. Febs Lett. 2002;529:268–274.
  • Oh SM, Liu Z, Okada M, et al. Ebp1 sumoylation, regulated by TLS/FUS E3 ligase, is required for its anti-proliferative activity. Oncogene. 2010;29:1017–1030.
  • Geeraerts A, Hsiu-Fang F, Zimmermann P, et al. The characterization of the nuclear dynamics of syntenin-2, a PIP2 binding PDZ protein. Cytometry A. 2013;83:866–875.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.