PRMT1-Mediated Arginine Methylation of PIAS1 Regulates STAT1 Signaling

Total Page:16

File Type:pdf, Size:1020Kb

PRMT1-Mediated Arginine Methylation of PIAS1 Regulates STAT1 Signaling Downloaded from genesdev.cshlp.org on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press PRMT1-mediated arginine methylation of PIAS1 regulates STAT1 signaling Susanne Weber,1 Florian Maaß,1 Michael Schuemann,2 Eberhard Krause,2 Guntram Suske,1 and Uta-Maria Bauer1,3 1Institute of Molecular Biology and Tumor Research (IMT), Philipps-University of Marburg, 35032 Marburg, Germany; 2Leibniz Institute of Molecular Pharmacology, 13125 Berlin, Germany To elucidate the function of the transcriptional coregulator PRMT1 (protein arginine methyltranferase 1) in interferon (IFN) signaling, we investigated the expression of STAT1 (signal transducer and activator of transcription) target genes in PRMT1-depleted cells. We show here that PRMT1 represses a subset of IFNg- inducible STAT1 target genes in a methyltransferase-dependent manner. These genes are also regulated by the STAT1 inhibitor PIAS1 (protein inhibitor of activated STAT1). PIAS1 is arginine methylated by PRMT1 in vitro as well as in vivo upon IFN treatment. Mutational and mass spectrometric analysis of PIAS1 identifies Arg 303 as the single methylation site. Using both methylation-deficient and methylation-mimicking mutants, we find that arginine methylation of PIAS1 is essential for the repressive function of PRMT1 in IFN-dependent transcription and for the recruitment of PIAS1 to STAT1 target gene promoters in the late phase of the IFN response. Methylation-dependent promoter recruitment of PIAS1 results in the release of STAT1 and coincides with the decline of STAT1-activated transcription. Accordingly, knockdown of PRMT1 or PIAS1 enhances the anti- proliferative effect of IFNg. Our findings identify PRMT1 as a novel and crucial negative regulator of STAT1 activation that controls PIAS1-mediated repression by arginine methylation. [Keywords: Transcriptional repression; interferon signaling; STAT1; PIAS1; PRMT1; arginine methylation] Supplemental material is available at http://www.genesdev.org. Received May 26, 2008; revised version accepted October 27, 2008. Interferons (IFNs) constitute a family of secreted auto- mammalian STAT proteins STAT1 and STAT2 are medi- crine and paracrine proteins that regulate innate and ators of the IFN signaling pathway, whereas the remain- adaptive immunity in response to viral and microbial in- ing STAT family members respond to distinct cytokines. fections and modulate proliferation, survival, and apo- Type I IFNs, such as IFNa and IFNb, activate STAT1/ ptosis of normal cells as well as tumor cells (Borden et al. STAT2 heterodimers, and type II IFNg mainly signals via 2007). IFNs exert their effects by binding to cell surface STAT1 homodimers (Borden et al. 2007). receptors, which triggers receptor dimerization/oligomer- Proper duration and magnitude of IFN-induced gene ization, allowing transphosphorylation and activation of activation is crucial, as abnormal and imbalanced JAK– intracellular receptor-associated tyrosine kinases of the STAT activation is associated with immune disorders and JAK (Janus kinase) family. Activated JAKs phosphorylate cancer (Darnell 2002; Levy and Darnell 2002). Therefore, critical tyrosine residues on the receptor, which results in cells have evolved mechanisms to allow rapid and tran- the recruitment of a familyRETRACTED of nucleo–cytoplasmic shut- sient activation and to prevent excess responses to IFNs tling transcription factors, the STAT (signal transducer and other cytokines. Restriction and inactivation of the and activator of transcription) proteins (Levy and Darnell STAT pathway is accomplished by regulators such as 2002; Schindler et al. 2007). In the following, receptor- protein tyrosine phosphatases, which inactivate JAKs and bound STATs are activated by phosphorylation of a single STATs in the cytoplasm or in the nucleus (Levy and tyrosine residue within the C terminus leading to STAT Darnell 2002). In addition, SOCS (suppressors of cytokine dimerization via a reciprocal phosphotyrosine-SH2 (src signaling) proteins switch off the activity of JAKs by acting homology 2) interaction. The STAT dimers are retained in as pseudosubstrates or by recruiting the ubiquitination the nucleus, where they bind specific DNA sequences in machinery (Alexander and Hilton 2004). The PIAS (protein promoter regions of target genes and induce gene tran- inhibitor of activated STAT) protein family operates in scription (Meyer and Vinkemeier 2004). Among the seven the nucleus and the family member PIAS1 interacts with activated STAT1 and suppresses the DNA-binding ability of the transcription factor (Liu et al. 1998; Shuai and Liu 3Corresponding author. E-MAIL [email protected]; FAX 0049-6421-2865196. 2005). PIAS1 depletion causes increased expression of a Article is online at http://www.genesdev.org/cgi/doi/10.1101/gad.489409. subset of STAT1 target genes (Liu et al. 2004). In agreement 118 GENES & DEVELOPMENT 23:118–132 Ó 2009 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/09; www.genesdev.org Downloaded from genesdev.cshlp.org on September 25, 2021 - Published by Cold Spring Harbor Laboratory Press Arginine methylation of PIAS1 with this observation, PIAS1 knockout mice reveal en- PRMT1 in HeLa cells using transient transfection of two hanced protection against pathogenic infections (Liu et al. different siRNAs directed against the PRMT1 transcript 2004). Whether the Sumo E3 ligase activity of PIAS1 and (siPRMT1_1 and siPRMT1_2). The efficient depletion of sumoylation of STAT1 is required for its repressive activity endogenous PRMT1 by the two alternative siRNAs in on IFN-inducible gene expression is controversial in the comparison with the control siRNA (siScr) was deter- literature (Rogers et al. 2003; Ungureanu et al. 2003, 2005; mined on RNA (Fig. 1A) and protein level (Fig. 1B). The Song et al. 2006). Until now, little was known about the expression of STAT1 or PIAS1 was not altered by knock- molecular mechanism leading to activation of PIAS1, down of PRMT1 (Fig. 1A,B). To examine the effect of which then restrains STAT1 activity, and how PIAS1 PRMT1 on IFNg-inducible STAT1-dependent transcrip- interferes with the chromatin binding of STAT1. tion we performed RT-QPCR of known STAT1 target Arginine methylation is catalyzed by a family of en- genes (Ramana et al. 2002) in control (siScr) and PRMT1- zymes, called PRMTs (protein arginine methyltransferases), depleted HeLa cells either unstimulated (0 h) or stimu- consisting of 11 members (Boisvert et al. 2005; Pal and Sif lated with IFNg for 2, 4, and 6 h. As shown in Figure 1C, 2007). These enzymes share a central highly conserved the IFNg-inducible transcription of several STAT1 tar- catalytic domain and perform mono- and dimethylation gets, including CXCL9 (Cxc chemokine ligand 9), of the guanidino group of the arginine residues using S- CXCL10 (Cxc chemokine ligand 10), and GBP1 (guany- adenosylmethionine (SAM) as methyl donor (Bedford and late-binding protein 1), was enhanced threefold to 10-fold Richard 2005). PRMTs regulate various cellular processes upon knockdown of PRMT1 using either of the two such as RNA processing, nucleo–cytoplasmic transport, siRNAs against PRMT1. In contrast, the basal transcript DNA repair, transcription, and signal transduction. Sev- level of these genes was not affected by PRMT1 depletion. eral reports implicate arginine methylation in the regu- IFN induction of other STAT1 target genes, like IRF1 (IFN lation of IFN signaling: PRMT1 interacts with the response factor 1) and TAP1 (transporter associated with cytoplasmic domain of the IFNa receptor, and its de- anti-gene presentation 1), was not significantly influ- pletion alters IFNa-induced growth arrest (Abramovich enced by PRMT1 knockdown, suggesting that tran- et al. 1997; Altschuler et al. 1999). Tyrosine phosphory- scriptional activation of only a subset of STAT1 target lation, transcriptional activity, and protein stability of genes is repressed by PRMT1 (Fig. 1C). The PRMT1- STAT transcription factors were suggested to be influ- dependent STAT1 targets identified here appeared to enced by arginine methylation (Mowen et al. 2001; Zhu match the subset of IFN-induced genes selectively con- et al. 2002; Chen et al. 2004). However, arginine methyl- trolled by the inhibitor of STAT1 signaling, PIAS1 (Liu ation of STAT transcription factors itself was not confirmed et al. 2004). We asked whether siRNA-mediated depletion by other groups, and the nonspecific methyltransferase of the PIAS1 protein would affect the same subset of inhibitor MTA, which was commonly used to study the IFNg-dependent STAT1 target genes as PRMT1 depletion in vivo relevance of arginine methylation in this pathway, does. Indeed, we found that PIAS1 knockdown, which executes strong side effects and inhibits IFN-induced was verified by Western blot (Fig. 1D), enhanced the IFN- phosphorylation and activation of STATs (Meissner inducible transcription of the PRMT1-dependent STAT1 et al. 2004; Komyod et al. 2005). Consequently, how ex- targets CXCL9, CXCL10, and GBP1, whereas PRMT1- actly PRMTs and arginine methylation might regulate independent genes, like IRF1 and TAP1, were not im- IFN signaling, is so far unclear. paired by suppression of PIAS1 (Fig. 1E). We obtained Here, we investigated the influence of PRMT1 on IFN similar results for the IFN-responsive A375 melanoma signaling and found that PRMT1 represses a subset of (Supplemental Fig. S1) and HEK293 cells (data not shown). STAT1 target genes that overlaps with genes regulated by Taken together, these data identify
Recommended publications
  • Activation of Smad Transcriptional Activity by Protein Inhibitor of Activated STAT3 (PIAS3)
    Activation of Smad transcriptional activity by protein inhibitor of activated STAT3 (PIAS3) Jianyin Long*†‡, Guannan Wang*†‡, Isao Matsuura*†‡, Dongming He*†‡, and Fang Liu*†‡§ *Center for Advanced Biotechnology and Medicine, †Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, and ‡Cancer Institute of New Jersey, 679 Hoes Lane, Piscataway, NJ 08854 Communicated by Allan H. Conney, Rutgers, The State University of New Jersey, Piscataway, NJ, November 17, 2003 (received for review August 22, 2003) Smad proteins play pivotal roles in mediating the transforming of many transcription factors through distinct mechanisms. growth factor ␤ (TGF-␤) transcriptional responses. We show in this PIAS1 and PIAS3 bind and inhibit STAT1 and STAT3 DNA- report that PIAS3, a member of the protein inhibitor of activated binding activities, respectively (19, 20). PIASx␣ and PIASx␤ STAT (PIAS) family, activates TGF-␤͞Smad transcriptional re- were identified through interactions with the androgen receptor sponses. PIAS3 interacts with Smad proteins, most strongly with and the homeodomain protein Msx2, respectively (21, 22). Smad3. PIAS3 and Smad3 interact with each other at the endog- PIASx␣ and PIASx␤ inhibit IL12-mediated and STAT4- enous protein level in mammalian cells and also in vitro, and the dependent gene activation (23). PIAS1, PIAS3, PIASx␣, and association occurs through the C-terminal domain of Smad3. We PIASx␤ also regulate transcriptional activation by various ste- further show that PIAS3 can interact with the general coactivators roid receptors (21, 24–26). PIASy has been shown to antagonize p300͞CBP, the first evidence that a PIAS protein can associate with the activities of STAT1 (27), androgen receptor (28), p53 (29), p300͞CBP.
    [Show full text]
  • An Immunoevasive Strategy Through Clinically-Relevant Pan-Cancer Genomic and Transcriptomic Alterations of JAK-STAT Signaling Components
    bioRxiv preprint doi: https://doi.org/10.1101/576645; this version posted March 14, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. An immunoevasive strategy through clinically-relevant pan-cancer genomic and transcriptomic alterations of JAK-STAT signaling components Wai Hoong Chang1 and Alvina G. Lai1, 1Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, OX3 7FZ, United Kingdom Since its discovery almost three decades ago, the Janus ki- Although cytokines are responsible for inflammation in nase (JAK)-signal transducer and activator of transcription cancer, spontaneous eradication of tumors by endoge- (STAT) pathway has paved the road for understanding inflam- nous immune processes rarely occurs. Moreover, the matory and immunity processes related to a wide range of hu- dynamic interaction between tumor cells and host immu- man pathologies including cancer. Several studies have demon- nity shields tumors from immunological ablation, which strated the importance of JAK-STAT pathway components in overall limits the efficacy of immunotherapy in the clinic. regulating tumor initiation and metastatic progression, yet, the extent of how genetic alterations influence patient outcome is far from being understood. Focusing on 133 genes involved in Cytokines can be pro- or anti-inflammatory and are inter- JAK-STAT signaling, we found that copy number alterations dependent on each other’s function to maintain immune underpin transcriptional dysregulation that differs within and homeostasis(3).
    [Show full text]
  • 1541-7786.MCR-09-0417.Full.Pdf
    Published Online First on April 6, 2010 Cell Cycle, Cell Death, and Senescence Molecular Cancer Research The Intracellular Delivery of a Recombinant Peptide Derived from the Acidic Domain of PIAS3 Inhibits STAT3 Transactivation and Induces Tumor Cell Death Corina Borghouts, Hanna Tittmann, Natalia Delis, Marisa Kirchenbauer, Boris Brill, and Bernd Groner Abstract Signaling components, which confer an “addiction” phenotype on cancer cells, represent promising drug tar- gets. The transcription factor signal transducers and activators of transcription 3 (STAT3) is constitutively ac- tivated in many different types of tumor cells and its activity is indispensible in a large fraction. We found that the expression of the endogenous inhibitor of STAT3, protein inhibitor of activated STAT3 (PIAS3), positively correlates with STAT3 activation in normal cells. This suggests that PIAS3 controls the extent and the duration of STAT3 activity in normal cells and thus prevents its oncogenic function. In cancer cells, however, the ex- pression of PIAS3 is posttranscriptionally suppressed, possibly enhancing the oncogenic effects of activated STAT3. We delimited the interacting domains of STAT3 and PIAS3 and identified a short fragment of the COOH-terminal acidic region of PIAS3, which binds strongly to the coiled-coil domain of STAT3. This PIAS3 fragment was used to derive the recombinant STAT3-specific inhibitor rPP-C8. The addition of a protein trans- duction domain allowed the efficient internalization of rPP-C8 into cancer cells. This resulted in the suppression of STAT3 target gene expression, in the inhibition of migration and proliferation, and in the induction of μ apoptosis at low concentrations [half maximal effective concentration (EC50), <3 mol/L].
    [Show full text]
  • Modulation of STAT Signaling by STAT-Interacting Proteins
    Oncogene (2000) 19, 2638 ± 2644 ã 2000 Macmillan Publishers Ltd All rights reserved 0950 ± 9232/00 $15.00 www.nature.com/onc Modulation of STAT signaling by STAT-interacting proteins K Shuai*,1 1Departments of Medicine and Biological Chemistry, University of California, Los Angeles, California, CA 90095, USA STATs (signal transducer and activator of transcription) play important roles in numerous cellular processes Interaction with non-STAT transcription factors including immune responses, cell growth and dierentia- tion, cell survival and apoptosis, and oncogenesis. In Studies on the promoters of a number of IFN-a- contrast to many other cellular signaling cascades, the induced genes identi®ed a conserved DNA sequence STAT pathway is direct: STATs bind to receptors at the named ISRE (interferon-a stimulated response element) cell surface and translocate into the nucleus where they that mediates IFN-a response (Darnell, 1997; Darnell function as transcription factors to trigger gene activa- et al., 1994). Stat1 and Stat2, the ®rst known members tion. However, STATs do not act alone. A number of of the STAT family, were identi®ed in the transcription proteins are found to be associated with STATs. These complex ISGF-3 (interferon-stimulated gene factor 3) STAT-interacting proteins function to modulate STAT that binds to ISRE (Fu et al., 1990, 1992; Schindler et signaling at various steps and mediate the crosstalk of al., 1992). ISGF-3 consists of a Stat1:Stat2 heterodimer STATs with other cellular signaling pathways. This and a non-STAT protein named p48, a member of the article reviews the roles of STAT-interacting proteins in IRF (interferon regulated factor) family (Levy, 1997).
    [Show full text]
  • Loss of Protein Inhibitors of Activated STAT-3 Expression in Glioblastoma Multiformetumors: Implications for STAT-3 Activation and Gene Expression Emily C
    Human Cancer Biology Loss of Protein Inhibitors of Activated STAT-3 Expression in Glioblastoma MultiformeTumors: Implications for STAT-3 Activation and Gene Expression Emily C. Brantley,1L. Burton Nabors,2 G. Yancey Gillespie,3 Youn-Hee Choi,5 Cheryl Ann Palmer,2,4 Keith Harrison,4 Kevin Roarty,1and Etty N. Benveniste1 Abstract Purpose: STATs activate transcription in response to numerous cytokines, controlling prolifera- tion, gene expression, and apoptosis. Aberrant activation of STAT proteins, particularly STAT-3, is implicated in the pathogenesis of many cancers, including GBM, by promoting cell cycle progres- sion, stimulating angiogenesis, and impairing tumor immune surveillance. Little is known about the endogenous STAT inhibitors, the PIAS proteins, in human malignancies.The objective of this study was to examine the expression of STAT-3 and its negative regulator, PIAS3, in human tissue samples from control and GBM brains. Experimental Design: Control and GBM human tissues were analyzed by immunoblotting and immunohistochemistry to determine the activation status of STAT-3 and expression of the PIAS3 protein.The functional consequence of PIAS3 inhibition by small interfering RNA or PIAS3 over- expression in GBM cells was determined by examining cell proliferation, STAT-3 transcriptional activity, and STAT-3 target gene expression.This was accomplished using [3H]TdR incorporation, STAT-3 dominant-negative constructs, reverse transcription-PCR, and immunoblotting. Results and Conclusions: STAT-3 activation, as assessed by tyrosine and serine phosphoryla- tion, was elevated in GBM tissue compared with control tissue. Interestingly, we observed expression of PIAS3 in control tissue, whereas PIAS3 protein expression in GBM tissue was greatly reduced.
    [Show full text]
  • Downloaded from As a Tab-Delimited file, in Which Genes Are Represented in Rows and Phenotypes in Columns
    cells Article Network of Interactions between ZIKA Virus Non-Structural Proteins and Human Host Proteins 1, 1,2, 2 Volha A. Golubeva y , Thales C. Nepomuceno y , Giuliana de Gregoriis , Rafael D. Mesquita 3, Xueli Li 1, Sweta Dash 1,4, Patrícia P. Garcez 5 , Guilherme Suarez-Kurtz 2 , Victoria Izumi 6, John Koomen 7, Marcelo A. Carvalho 2,8,* and Alvaro N. A. Monteiro 1,* 1 Cancer Epidemiology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; [email protected] (V.A.G.); [email protected] (T.C.N.); xueli.li@moffitt.org (X.L.); sweta.dash@moffitt.org (S.D.) 2 Divisão de Pesquisa Clínica, Instituto Nacional de Câncer, Rio de Janeiro 20230-130, Brazil; [email protected] (G.d.G.); [email protected] (G.S.-K.) 3 Departamento de Bioquímica, Instituto de Química, Federal University of Rio de Janeiro, Rio de Janeiro 21941-909, Brazil; [email protected] 4 Cancer Biology PhD Program, University of South Florida, Tampa, FL 33612, USA 5 Institute of Biomedical Science, Federal University of Rio de Janeiro, Rio de Janeiro 20230-130, Brazil; [email protected] 6 Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; victoria.izumi@moffitt.org 7 Chemical Biology and Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA; john.koomen@moffitt.org 8 Instituto Federal do Rio de Janeiro-IFRJ, Rio de Janeiro 20270-021, Brazil * Correspondence: [email protected] (M.A.C.); alvaro.monteiro@moffitt.org (A.N.A.M.); Tel.: +55-21-2566-7774 (M.A.C.); +813-7456321 (A.N.A.M.) These authors contributed equally to this work.
    [Show full text]
  • Interferon-Regulatory Factors Determine Macrophage Phenotype Polarization
    Hindawi Publishing Corporation Mediators of Inflammation Volume 2013, Article ID 731023, 8 pages http://dx.doi.org/10.1155/2013/731023 Review Article Interferon-Regulatory Factors Determine Macrophage Phenotype Polarization Roman Günthner and Hans-Joachim Anders Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universitat¨ Munchen,¨ Ziemssenstraße 1, 80336 Munchen,¨ Germany Correspondence should be addressed to Hans-Joachim Anders; [email protected] Received 5 August 2013; Revised 28 October 2013; Accepted 28 October 2013 Academic Editor: Eduardo Lopez-Collazo´ Copyright © 2013 R. Gunthner¨ and H.-J. Anders. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. The mononuclear phagocyte system regulates tissue homeostasis as well as all phases of tissue injury and repair. To do sochanging tissue environments alter the phenotype of tissue macrophages to assure their support for sustaining and amplifying their respective surrounding environment. Interferon-regulatory factors are intracellular signaling elements that determine the maturation and gene transcription of leukocytes. Here we discuss how several among the 9 interferon-regulatory factors contribute to macrophage polarization. 1. Introduction polarize macrophages toward a classically activated M1-like phenotype [9, 10]. This process is associated with NF-B During development mononuclear phagocyte progenitors and STAT1 pathway activation [2]. Macrophages apoptosis populate most tissues where they differentiate into tran- or their phenotype switches towards alternatively activated, scriptionally and functionally diverse phenotypes [1–3]; for M2-like macrophages that produce IL-10 and TGF-, induce example, bone marrow, liver, and lung harbor macrophages resolution of inflammation, and enforce tissue regeneration with an enormous capacity to clear airborne particles, gut- [11–15].
    [Show full text]
  • The Association and Nuclear Translocation of the PIAS3-STAT3 Complex Is Ligand and Time Dependent
    Published OnlineFirst November 10, 2009; DOI: 10.1158/1541-7786.MCR-09-0313 The Association and Nuclear Translocation of the PIAS3-STAT3 Complex Is Ligand and Time Dependent Snehal Dabir, AmyKluge, and Afshin Dowlati Division of Hematology/Oncology, Department of Medicine, Case Western Reserve University, Cleveland, Ohio Abstract nuclear translocation and modulation of gene transcription of The epidermal growth factor (EGF) receptor activation of its target genes. This pathway is of importance because of its downstream signal transducers and activators of functions in hematopoiesis, immune response, and oncogenesis transcription 3 (STAT3) plays a crucial role in the (1). Of these seven STATs (STAT1, STAT2, STAT3, STAT4, pathogenesis of lung cancer. STAT3 transcriptional activity STAT5a, STAT5b, and STAT6), STAT3 is of particular impor- can be negativelyregulated byprotein inhibitor of activated tance due to its involvement in a wide spectrum of biological STAT3 (PIAS3). We investigated the time-dependent PIAS3 functions. It is activated (phosphorylated) by a variety of cyto- shuffling and binding to STAT3 in an EGF-dependent kines and growth factors, such as platelet-derived growth factor model in lung cancer byusing confocal microscopy, and epidermal growth factor (EGF), in a large number of hu- immunoprecipitation, luciferase reporter assay, and protein man malignancies (2). STAT proteins have three families of analysis of segregated cellular components. We also natural inhibitors: the protein inhibitors of activated STAT explored the role of phosphorylation at Tyr705 of STAT3 in (PIAS; ref. 3), the suppressors of cytokine signaling (4-6), the formation and intracellular shuffling of the PIAS3-STAT3 and the Src homology 2–containing phosphatase (7).
    [Show full text]
  • Distinct E€Ects of PIAS Proteins on Androgen-Mediated Gene Activation
    Oncogene (2001) 20, 3880 ± 3887 ã 2001 Nature Publishing Group All rights reserved 0950 ± 9232/01 $15.00 www.nature.com/onc Distinct eects of PIAS proteins on androgen-mediated gene activation in prostate cancer cells Mitchell Gross1, Bin Liu1, Jiann-an Tan3, Frank S French3, Michael Carey2 and Ke Shuai*,1,2 1Division of Hematology-Oncology, Department of Medicine, University of California, Los Angeles, California, CA 90095, USA; 2Department of Biological Chemistry, University of California, Los Angeles, California, CA 90095, USA; 3The Laboratories for Reproductive Biology, Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, North Carolina, NC 27599-7500, USA Androgen signaling in¯uences the development and The androgen receptor (AR) is a member of the growth of prostate carcinoma. The transcriptional nuclear receptor (NR) superfamily (Bentel and Tilley, activity of androgen receptor (AR) is regulated by 1996). NRs have conserved domain structures (Freed- positive or negative transcriptional cofactors. We report man, 1999). At the N-terminus is the ligand- here that PIAS1, PIAS3, and PIASy of the protein independent transcriptional activation domain (AF- inhibitor of activated STAT (PIAS) family, which are 1). The central domain contains two zinc ®nger expressed in human prostate, display distinct eects on structures that are involved in DNA binding (DBD). AR-mediated gene activation in prostate cancer cells. The C-terminal region is the ligand binding domain While PIAS1 and PIAS3 enhance the transcriptional (LBD). In addition, an essential ligand-dependent activity of AR, PIASy acts as a potent inhibitor of AR transactivation domain (AF-2) is localized in the C- in prostate cancer cells.
    [Show full text]
  • PIAS1 Potentiates the Anti-EBV Activity of SAMHD1 Through Sumoylation
    Saiada et al. Cell Biosci (2021) 11:127 https://doi.org/10.1186/s13578-021-00636-y Cell & Bioscience RESEARCH Open Access PIAS1 potentiates the anti-EBV activity of SAMHD1 through SUMOylation Farjana Saiada1, Kun Zhang1 and Renfeng Li1,2,3* Abstract Background: Sterile alpha motif and HD domain 1 (SAMHD1) is a deoxynucleotide triphosphohydrolase (dNTPase) that restricts the infection of a variety of RNA and DNA viruses, including herpesviruses. The anti-viral function of SAMHD1 is associated with its dNTPase activity, which is regulated by several post-translational modifcations, includ- ing phosphorylation, acetylation and ubiquitination. Our recent studies also demonstrated that the E3 SUMO ligase PIAS1 functions as an Epstein-Barr virus (EBV) restriction factor. However, whether SAMHD1 is regulated by PIAS1 to restrict EBV replication remains unknown. Results: In this study, we showed that PIAS1 interacts with SAMHD1 and promotes its SUMOylation. We identifed three lysine residues (K469, K595 and K622) located on the surface of SAMHD1 as the major SUMOylation sites. We demonstrated that phosphorylated SAMHD1 can be SUMOylated by PIAS1 and SUMOylated SAMHD1 can also be phosphorylated by viral protein kinases. We showed that SUMOylation-defcient SAMHD1 loses its anti-EBV activity. Furthermore, we demonstrated that SAMHD1 is associated with EBV genome in a PIAS1-dependent manner. Conclusion: Our study reveals that PIAS1 synergizes with SAMHD1 to inhibit EBV lytic replication through protein– protein interaction and SUMOylation. Keywords: SAMHD1, PIAS1, Restriction factor, Epstein-Barr virus, Cytomegalovirus, SUMOylation, Herpesvirus, Deoxynucleotide triphosphohydrolase, Phosphorylation Background and human simplex virus 1 (HSV-1) [2–7], vaccinia virus Host restriction factors serve as the frst line of defense [2], human T cell leukemia virus type 1 [8], hepatitis B against viral infection through blocking virus entry, virus [9] and human papillomavirus 16 [10].
    [Show full text]
  • S41467-018-04936-9.Pdf
    ARTICLE DOI: 10.1038/s41467-018-04936-9 OPEN Nuclear Smad6 promotes gliomagenesis by negatively regulating PIAS3-mediated STAT3 inhibition Jiantong Jiao1,2,3, Rui Zhang1,2, Zheng Li1,2,3, Ying Yin1,3, Xiangming Fang4, Xiaopeng Ding1,2, Ying Cai5, Shudong Yang5, Huijun Mu1,3, Da Zong2, Yuexin Chen1,3, Yansong Zhang6, Jian Zou1,3, Junfei Shao2,3 & Zhaohui Huang7 1234567890():,; To date, the molecular mechanism underlying constitutive signal transducer and activator of transcription 3 (STAT3) activation in gliomas is largely unclear. In this study, we report that Smad6 is overexpressed in nuclei of glioma cells, which correlates with poor patient survival and regulates STAT3 activity via negatively regulating the Protein Inhibitors of Activated STAT3 (PIAS3). Mechanically, Smad6 interacts directly with PIAS3, and this interaction is mediated through the Mad homology 2 (MH2) domain of Smad6 and the Ring domain of PIAS3. Smad6 recruits Smurf1 to facilitate PIAS3 ubiquitination and degradation, which also depends on the MH2 domain and the PY motif of Smad6. Consequently, Smad6 reduces PIAS3-mediated STAT3 inhibition and promotes glioma cell growth and stem-like cell initiation. Moreover, the Smad6 MH2 transducible protein restores PIAS3 expression and subsequently reduces gliomagenesis. Collectively, we conclude that nuclear-Smad6 enhances glioma development by inducing PIAS3 degradation and subsequent STAT3 activity upregulation. 1 Center of Clinical Research, Wuxi People’s Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, China. 2 Department of Neurosurgery, Wuxi People’s Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, China. 3 Wuxi Institute of Translational Medicine, Wuxi People’s Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, China.
    [Show full text]
  • Regulation of Interferon Regulatory Factor 1 by Protein
    REGULATION OF INTERFERON REGULATORY FACTOR 1 BY PROTEIN INHIBITOR OF ACTIVATED STAT3 by Danyang Xu A thesis submitted to the School of Graduate Studies in partial fulfillment of the requirements for the degree of Master of Science in Medicine (Cancer and Development) BioMedical Sciences Faculty of Medicine October 2020 Memorial University of Newfoundland St. John’s Newfoundland and Labrador Abstract Oncolytic viruses exploit tumor-specific cellular changes for selective replication, inducing cancer cell death. Our previous research showed that Ras/mitogen-activated protein kinase kinase (MEK) downregulation of interferon regulatory factor 1 (IRF1) is a major mechanism underlying viral oncolysis. Protein inhibitor of activated STAT 3 (PIAS3) is known as the E3 ligase of IRF1 sumoylation. The objective of this study was to identify if, and how, PIAS3 modulates cellular sensitivity to oncolytic viruses via regulation of IRF1. By conducting co-immunoprecipitation, I found that IRF1 has no direct interaction with PIAS3 found within HT1080 cells. However, CRISPR knockdown of PIAS3 increases IRF1 expression as well as transcription of IRF1-responsive anti-viral genes. Furthermore, PIAS3 knockdown HT1080 cells were equally sensitive to viral infection as their parent HT1080 cells. Together, these results demonstrate that PIAS3 does not directly interact with IRF1 but regulates expression and transcriptional activity of IRF1. Moreover, as CRISPR knockdown of PIAS3 did not change cellular sensitivity to viral infection, IRF1 modulation via PIAS3 does not play very critical roles in host innate antiviral responses. ii General Summary Oncolytic viruses selectively kill cancer cells but not normal cells. Our previous research has shown that the expression of antiviral protein, interferon regulatory factor 1 (IRF1), is suppressed in cancer cells, which increases their susceptibility to oncolytic virus infection.
    [Show full text]