Full Text (PDF)

Total Page:16

File Type:pdf, Size:1020Kb

Full Text (PDF) PIASy-Deficient Mice Display Modest Defects in IFN and Wnt Signaling Wera Roth, Claudio Sustmann, Matthias Kieslinger, Andrea Gilmozzi, Denis Irmer, Elisabeth Kremmer, Chris Turck and This information is current as Rudolf Grosschedl of October 2, 2021. J Immunol 2004; 173:6189-6199; ; doi: 10.4049/jimmunol.173.10.6189 http://www.jimmunol.org/content/173/10/6189 Downloaded from References This article cites 74 articles, 44 of which you can access for free at: http://www.jimmunol.org/content/173/10/6189.full#ref-list-1 http://www.jimmunol.org/ Why The JI? Submit online. • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average by guest on October 2, 2021 Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2004 by The American Association of Immunologists All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology PIASy-Deficient Mice Display Modest Defects in IFN and Wnt Signaling1 Wera Roth,* Claudio Sustmann,* Matthias Kieslinger,* Andrea Gilmozzi,* Denis Irmer,* Elisabeth Kremmer,† Chris Turck,‡ and Rudolf Grosschedl2* Protein inhibitors of activated STATs (PIAS) represent a small family of nuclear proteins that modulate the activity of many transcription factors and act as E3 ligases for covalent modification of proteins with the small ubiquitin-like modifier (SUMO). In particular, PIASy has been shown to inhibit the activation of gene expression by the IFN-responsive transcription factor STAT1 and the Wnt-responsive transcription factor LEF1. To assess the function of PIASy in vivo, we generated and analyzed mice carrying a targeted mutation of the Piasy gene. We find that homozygous mutant mice have no obvious morphological defects and have a normal distribution of lymphocyte populations. Molecular analysis of signaling in response to IFN-␥ and Wnt agonists revealed a modest reduction in the activation of endogenous and transfected target genes. Two-dimensional analysis of total Downloaded from proteins and SUMO-modified proteins in transformed pre-B cells showed no significant differences between wild-type mice and homozygous mutant mice. Taken together, our data indicate that PIASy has a modest effect on cytokine and Wnt signaling, suggesting a redundancy with other members of the family of PIAS proteins. The Journal of Immunology, 2004, 173: 6189–6199. rotein inhibitors of activated STATs (PIAS)3 have been with other proteins. PIAS1 and PIAS3 inhibit DNA binding of identified as interaction partners for multiple nuclear pro- STAT1 and STAT3, respectively (1, 2). In contrast, PIASy represses P teins, including the STATs and lymphoid enhancer bind- STAT1, LEF1, Smad4 and the AR without interfering with DNA http://www.jimmunol.org/ ing factor (LEF)1/T cell-specific factor (TCF) proteins that medi- binding by these proteins (3, 24, 26, 27). The Piasx gene encodes two ate nuclear responses to cytokines and Wnt signals, respectively splice variants, x␣ (ARIP3) and x␤ (Miz1), which interact with the (1–5). The cytokines IFN-␣ and IFN-␥ are the main cytokines for AR and with the homeodomain protein Msx2, respectively (28, 29). innate immune responses against viral infections. IFN-␣ is pro- In addition, PIASx␤ interacts with STAT4, following IL-12 stimula- duced by many cell types, whereas IFN-␥ is produced predomi- tion of T cells (5). In most cases, the association of PIAS proteins with nately by hemopoietic cells (reviewed in Refs. 6 and 7). The sig- transcription factors results in repression (reviewed in Ref. 30); how- naling pathways for both IFNs are similar and involve the Jak ever, PIAS proteins can also augment gene expression by synergizing tyrosine kinase-mediated activation of STAT proteins that stimu- with other transcriptional coactivators (4, 18). by guest on October 2, 2021 late transcription of target genes alone or in concert with IFN reg- PIAS proteins share a similar domain structure, including an ulatory factors (reviewed in Refs. 8 and 9). The response of cells N-terminal domain, termed SAP domain, which mediates, in part, to Wnt signals involves the stabilization and nuclear translocation the interaction with partner proteins (3, 31) and a central RING ␤ of -catenin, which associates with LEF/TCF proteins and acti- domain that resembles the catalytic domain of ubiquitin ligases vates Wnt-responsive genes (reviewed in Ref. 10). (32, 33). Recent experiments aimed at the mechanisms by which In addition to STATs and LEF1/TCF proteins, many other tran- PIAS proteins modulate the activity of their interaction partners, scription factors interact with PIAS proteins. These include nuclear have shown that PIAS proteins can augment the covalent modifi- hormone receptors, such as the androgen receptor (AR), p53, cation of proteins with the small ubiquitin-related modifier Smad4, Sp3, HMGI-C, Gfi-1, IRF-1, TFII-I and yeast septins (11– (SUMO) (3, 14–16, 22, 34–36). Two families of SUMO proteins, 25). In mouse and man, four Pias genes (Pias1, Pias3, Piasx, and SUMO-1 and SUMO-2/SUMO-3 resemble ubiquitin proteins and Piasy) have been identified, which encode proteins that share a are also attached to the ⑀-amino groups of lysines in target proteins similar domain structure but differ in their specificity of interaction (37). The sumoylation pathway resembles that of ubiquitination, with the activation of SUMO by an E1 protein and its transfer to an E2 enzyme (37–39). Multiple lines of evidence suggest that *Gene Center and Institute of Biochemistry, University of Munich, †GSF National Research Center for Environment and Health, and ‡Max-Planck-Institute of Psychi- PIAS proteins act as E3 ligases for SUMO. First, PIAS proteins atry, Munich, Germany can cooperate with E1 and E2 enzymes in reconstituted systems to Received for publication May 21, 2004. Accepted for publication August 20, 2004. augment SUMO modification of target proteins (3, 14, 22, 35). The costs of publication of this article were defrayed in part by the payment of page Second, PIAS proteins contain a cysteine-rich RING domain, charges. This article must therefore be hereby marked advertisement in accordance which is a hallmark for ubiquitin ligases and is functionally im- with 18 U.S.C. Section 1734 solely to indicate this fact. portant for the sumoylation activity of PIAS proteins (3, 22, 33– 1 This work was supported by a Grant of the German Research Foundation TR-5. 35). Finally, PIAS proteins display some specificity of protein sub- 2 Address correspondence and reprint requests to Dr. Rudolf Grosschedl at the current strates, although the specificity is not as pronounced as for address: Max-Planck-Institute of Immunobiology, Stuebeweg 51, 79108 Freiburg, Germany. E-mail address: [email protected] ubiquitin ligases (5, 22, 40). 3 Abbreviations used in this paper: PIAS, protein inhibitor of activated STAT; LEF, The function of PIAS proteins as SUMO E3 ligases appears lymphoid enhancer binding factor; TCF, T cell-specific factor; SUMO, small ubiq- to involve multiple mechanisms. The repression of transcription uitin-like modifier; ES, embryonic stem; IRF, IFN regulatory factor; AR, androgen factors by PIAS proteins often correlates with the subnuclear se- receptor; ␤-Gal, ␤-galactosidase; X-Gal, 5-bromo-4-chloro-3-indolyl ␤-D-galacto- side; MuLV, murine leukemia virus; MEF, mouse embryonic fibroblast. questration into speckles, such as promyelocytic leukemia nuclear Copyright © 2004 by The American Association of Immunologists, Inc. 0022-1767/04/$02.00 6190 FUNCTION OF THE SUMO-E3 LIGASE PIASy IN VIVO bodies (3, 20, 21, 23, 34, 41). This observation could account for and hybridized overnight at 42°C. Blots were washed to final stringency of the repression of transcription factor activity without changes in 0.2% SDS, 0.1% SDS at 65°C, and exposed to film. ␤ DNA binding. In addition, PIASx has been found to interact with Immunoblot analysis histone deacetylase-3, which is involved in repression of TFII-I family proteins (25). The activation function of PIAS proteins Total protein extracts were separated by 10% SDS-PAGE and transferred to nitrocellulose. Immunoblots were conducted by the ECL method ac- could also be accounted for by the sumoylation-dependent aug- cording to the manufacturer’s instructions (Amersham Pharmacia Biotech, mentation of protein-protein interactions. In nuclear hormone re- Piscataway, NJ), with the Abs rat anti-PIASy (mouse) mAb. ceptors, the SUMO modification sites coincide with the protein motifs that mediate synergy in the activation of reporters contain- Flow cytometry ing multimerized binding sites for the hormone receptors (42–44). Thymus, spleen, and bone marrow were isolated and dispersed in RPMI Genetic experiments in Drosophila indicated that the PIAS or- 1640, containing antibiotics and glutamine with 10% heat-inactivated FCS. ϫ 6 ␮ thologue dPIAS, also known as Zimp, interacts functionally with Cells (1 10 ) were resuspended in 100 l of FACS buffer (PBS, 1% FCS) and incubated with Ab for 20 min. Cells were washed three times the STAT orthologue stat92E to regulate blood cell and eye de- with FACS buffer, and the process was then repeated with secondary Abs velopment (45). An additional role of PIAS in the regulation of as necessary. After staining, cells were resuspended in 500 ␮l of FACS chromosome structure and function was inferred from the identi- buffer containing propidium iodine. Cells were then analyzed using a FACS- fication and characterization of dPIAS as a suppressor of position- Calibur (BD Biosciences, San Jose, CA).
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]
  • 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]
  • 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]
  • Gene Section Review
    Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS Gene Section Review PIAS3 (protein inhibitor of activated STAT, 3) Gilles Spoden, Werner Zwerschke Institute for Medical Microbiology and Hygiene, University Medical Center of the Johannes Gutenberg University Mainz, Hochhaus am Augustus-platz, 55131 Mainz, Germany (GS); Cell Metabolism and Differentiation Research Group, Institute for Biomedical Aging Research, Rennweg 10, 6020 Innsbruck, Austria (ZW); Tumorvirology Research Group, Tyrolean Cancer Research Institute at Medical University Innsbruck, Innrain 66, 6020 Innsbruck, Austria (ZW) Published in Atlas Database: April 2010 Online updated version : http://AtlasGeneticsOncology.org/Genes/PIAS3ID41709ch1q21.html DOI: 10.4267/2042/44937 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2011 Atlas of Genetics and Cytogenetics in Oncology and Haematology E3 ligase, catalyzing the covalent, post-translational Identity modification of specific target proteins with SUMO. Other names: FLJ14651, KChAP, ZMIZ5 Different splice variants of PIAS3 have been identified HGNC (Hugo): PIAS3 but the full-length sequence of some of these variants Location: 1q21.1 has not been described. Description DNA/RNA The human PIAS3 gene is 10559 bp long and consists Note of 14 exons and 13 introns. The gene codes for a protein of the PIAS family Transcription (protein inhibitor of activated STAT (signal transducer Transcript length 2902 bp (CDS 1887 bp ; residues 628 and activator of transcription)). PIAS3 regulates the aa). activity of several transcription factors by direct protein-protein interaction. Further, PIAS3 is a SUMO Pseudogene (small ubiquitin-like modifier)- No pseudogene reported. Figure 1.
    [Show full text]
  • NATURE Immunology
    RESOURCE Identification of transcriptional regulators in the mouse immune system Vladimir Jojic1,7, Tal Shay2,7, Katelyn Sylvia3, Or Zuk2, Xin Sun4, Joonsoo Kang3, Aviv Regev2,5,8, Daphne Koller1,8 & the Immunological Genome Project Consortium6 The differentiation of hematopoietic stem cells into cells of the immune system has been studied extensively in mammals, but the transcriptional circuitry that controls it is still only partially understood. Here, the Immunological Genome Project gene-expression profiles across mouse immune lineages allowed us to systematically analyze these circuits. To analyze this data set we developed Ontogenet, an algorithm for reconstructing lineage-specific regulation from gene-expression profiles across lineages. Using Ontogenet, we found differentiation stage–specific regulators of mouse hematopoiesis and identified many known hematopoietic regulators and 175 previously unknown candidate regulators, as well as their target genes and the cell types in which they act. Among the previously unknown regulators, we emphasize the role of ETV5 in the differentiation of gd T cells. As the transcriptional programs of human and mouse cells are highly conserved, it is likely that many lessons learned from the mouse model apply to humans. The Immunological Genome Project (ImmGen) is a consortium of ­programs of human and mouse cells are highly conserved4, many immunologists and computational biologists who aim, through the lessons learned from the mouse model will probably be applicable use of shared and rigorously controlled data-generation pipelines, to humans. Two key approaches for the identification of regulatory to exhaustively chart gene-expression profiles and their underlying networks5 are physical models based on the association of a transcrip- regulatory networks in the mouse immune system1.
    [Show full text]
  • 2653.Full.Pdf
    Characterization of a PIAS4 Homologue from Zebrafish: Insights into Its Conserved Negative Regulatory Mechanism in the TRIF, MAVS, and IFN Signaling Pathways during This information is current as Vertebrate Evolution of September 25, 2021. Ran Xiong, Li Nie, Li-xin Xiang and Jian-zhong Shao J Immunol 2012; 188:2653-2668; Prepublished online 17 February 2012; doi: 10.4049/jimmunol.1100959 Downloaded from http://www.jimmunol.org/content/188/6/2653 Supplementary http://www.jimmunol.org/content/suppl/2012/02/17/jimmunol.110095 http://www.jimmunol.org/ Material 9.DC1 References This article cites 72 articles, 33 of which you can access for free at: http://www.jimmunol.org/content/188/6/2653.full#ref-list-1 Why The JI? Submit online. by guest on September 25, 2021 • Rapid Reviews! 30 days* from submission to initial decision • No Triage! Every submission reviewed by practicing scientists • Fast Publication! 4 weeks from acceptance to publication *average Subscription Information about subscribing to The Journal of Immunology is online at: http://jimmunol.org/subscription Permissions Submit copyright permission requests at: http://www.aai.org/About/Publications/JI/copyright.html Email Alerts Receive free email-alerts when new articles cite this article. Sign up at: http://jimmunol.org/alerts The Journal of Immunology is published twice each month by The American Association of Immunologists, Inc., 1451 Rockville Pike, Suite 650, Rockville, MD 20852 Copyright © 2012 by The American Association of Immunologists, Inc. All rights reserved. Print ISSN: 0022-1767 Online ISSN: 1550-6606. The Journal of Immunology Characterization of a PIAS4 Homologue from Zebrafish: Insights into Its Conserved Negative Regulatory Mechanism in the TRIF, MAVS, and IFN Signaling Pathways during Vertebrate Evolution Ran Xiong, Li Nie, Li-xin Xiang, and Jian-zhong Shao Members of the protein inhibitor of activated STAT (PIAS) family are key regulators of various human and mammalian signaling pathways, but data on their occurrence and functions in ancient vertebrates are limited.
    [Show full text]