Protein Synthesis and Degradation: ISG15 and ISG-15 linked proteins Can Associate with Members of the Selective Autophagic process, HDAC6 and SQSTM1/p62 Hiroshi Nakashima, Tran Nguyen, William F. Goins and Ennio Antonio Chiocca J. Biol. Chem. published online November 26, 2014 Find articles, minireviews, Reflections and Classics on similar topics on the JBC Affinity Sites. Alerts: • When this article is cited • When a correction for this article is posted Click here to choose from all of JBC's e-mail alerts This article cites 0 references, 0 of which can be accessed free at http://www.jbc.org/content/early/2014/11/26/jbc.M114.593871.full.html#ref-list-1 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 Access the most updated version of this article at doi: 10.1074/jbc.M114.593871 JBC Papers in Press. Published on November 26, 2014 as Manuscript M114.593871 The latest version is at http://www.jbc.org/cgi/doi/10.1074/jbc.M114.593871 ISG15 associates with HDAC6 and p62 ISG15 and ISG-15 linked proteins Can Associate with Members of the Selective Autophagic process, HDAC6 and SQSTM1/p62* Hiroshi Nakashima1, Tran Nguyen1 , William F. Goins2 and Ennio Antonio Chiocca1,3 1 Harvey Cushing Neuro-oncology Laboratories, Harvard Institutes of Medicine 9th floor, Department of Neurosurgery and Institute for the Neurosciences at the Brigham and Women's Hospital, 4 Blackfan Circle, Boston, MA 02115 2 Department of Microbiology & Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA *Running title: ISG15 associates with HDAC6 and p62. 3 To whom correspondence should be addressed: E. Antonio Chiocca, Department of Neurosurgery, Brigham and Women's Hospital, 75 Francis Street, Boston, MA USA, Tel.: (617) 732-6939; Fax: (617) 734-8342; E-mail: [email protected] the molecular events that occur in this process are not well known. Here, we show that the C-terminal LRLRGG of ISG15 interacts with the ubiquitin zinc finger (BUZ) domain of Histone Deacetylase 6 (HDAC6). Since HDAC6 is involved in the autophagic clearance of ubiquitinated (Ub) aggregates, during which SQSTM1/p62 plays a major role as a cargo adapter, we also were able to confirm that p62 binds to ISG15 protein and its conjugated proteins, upon forced expression. Both HDAC6 and p62 colocalize with ISG15 in an insoluble fraction of the cytosol and this colocalization was magnified by the proteasome inhibitor, MG132. In addition, ISG15 was degraded via the lysosome. Overexpression of ISG15, which leads to an increased conjugation level of the cellular proteome, enhanced autophagic degradation, independent of IFN signaling transduction. These results thus indicate that ISG15 conjugation marks proteins for interaction with HDAC6 and p62 upon forced stressful conditions, likely as a step towards autophagic clearance. The type I interferon (IFN) signaling pathway is the quintessential intracellular innate defense mechanism against invading pathogens. It sets up a robust antiviral state, rendering cells non-permissive for infection Capsule Background: ISG15 is a Ub-like protein that conjugates cellular and pathogenic proteins during the innate immune response. Results: ISG15 associates with the selective autophagic factors HDAC6 and p62, leading to degradation of ISG15-conjugates. Conclusion: The IFN response leads to ISG15 expression allowing for its association with HDAC and p62. This may mark proteins for autophagy. Significance: This finding provides evidence of an interferon-stimulated pathway linked to autophagy. ABSTRACT The ubiquitin-like interferon (IFN)stimulated gene 15 (ISG15), and its specific E1, E2 and E3 enzymes are transcriptionally induced by type I Interferons (IFNs). ISG15 conjugates newly synthesized proteins. ISG15 linkage to proteins appears to be an important downstream IFN-signaling event that discrimininates cellular and pathogenic proteins synthesized during IFN stimulation, from existing proteins. This eliminates potentially pathogenic proteins, as the cell attempts to return to normal homeostasis after IFN “stressed” conditions. However, 1 Copyright 2014 by The American Society for Biochemistry and Molecular Biology, Inc. Downloaded from http://www.jbc.org/ by guest on December 4, 2014 Keywords: Autophagy, HDAC6, ISG15, SQSTM1, p62, post-translational modification (PTM), ubiquitin, lysosome, virus, interferon ISG15 associates with HDAC6 and p62 autophagy, which plays a role in cellular homeostasis through sequestration of intracellular materials including pathogens within double membrane vesicles (autophagosomes) that fuse with lysosomes leading to degradation of delivered cellular contents (7). Although starvation-induced, non-selective autophagy recycles cytosolic contents and organelles during limited nutrient supply, nutrient-independent, basal or selective autophagy eliminates misfolded proteins and damaged organelles before the accumulation of excessive amounts of cytosolic aggregates or inclusion bodies at the microtubule organizing center (MTOC) (8-10). It has been reported that aggregate-prone proteins become marked by the lysine 63 (K63)-linked polyubiquitin chain that functionally distinguishes them from UPS-targeted lysine 48 (K48)-linked ones (11,12). In this context, the cargo adaptor protein SQSTM1/p62 plays an important role by preferentially binding K63-linked polyubiquitinated proteins via an ubiquitin associated (UBA) domain at its C-terminus. To facilitate Ub-protein aggregates and autophagic degradation, p62 forms self- and/or hetero-oligomerization at its N-terminal PB1 domain and recruits the LC3 membrane protein that is anchored in phagophore membranes for autophagosome maturation (13-15). During selective autophagy, the cytosolic deacetylase HDAC6 is involved in clearance of ubiquitin-prone aggregates (10,16). Its C-terminal BUZ (a binder of ubiquitin zinc finger) domain can bind free mono- and poly-Ub chains (17,18). Both the catalytic activity and the BUZ domain are required for HDAC6 to facilitate clearance of ubiquitin-prone aggregates (also referred to as "aggrephagy") and HDAC6 promotes autophagosome-lysosome fusion through the cortactin-dependent actin remodeling machinery in mouse embryonic fibroblasts (MEF) cells (19). In this report, we report that forced expression of HDAC6 leads to interaction with p62 and ISG15 that structurally resembles K63-diUb, and its ISGylated substrates, rendering them targets for lysosomal degradation. ISG15 localized to cytosolic inclusion bodies with HDAC6 and p62. 2 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 and replication. Upon IFNs’ binding to their receptors (type I interferon receptors, IFNARs), plasma-membrane resident STATs are activated through phosphorylation by Jak1/TYK2 kinases. This leads to subsequent translocation of STATs into nuclei where they bind to a cis-acting DNA element upstream of IFN stimulated genes (ISGs), leading to ISGs transcriptional activation. The ISG family of proteins activates IFN signaling, virus recognition factors or pattern-recognition receptors (PRRs), protein kinase R (PKR), ribonuclease L (RNaseL), Mx1 and ISG15 (1). ISG15 is a diubiquitin-like protein, able to conjugate to lysine residues of its substrate via isopeptide bonds, utilizing the same enzymatic cascade as that of Ubiquitin (2). Conjugation by ISG15 (referred to as "ISGylation") onto de novo synthesizing polypeptides on polysomes proceeds through a series of enzymatic reactions, mediated by the E1-activating UbE1L, E2-conjugating UbcH8 and HECTtype E3 ligase Herc5, which are also all IFNinducible. There is also an inverse reaction where the IFN-inducible UBP43/USP18 protease deconjugates ISG15 from conjugated substrates (3). ISG15 broadly conjugates IFNinduced ISG genes and progeny viral proteins (2). It has been shown that ISG15-knockout mice were broadly susceptible to RNA and DNA viruses (4). Several studies have found that free ISG15 and ISGylated host and viral proteins interfere in the processes of virion egress and release from the cell during the viral life cycle (4). The fate(s) of ISG15 and ISG15-conjugated proteins, particularly those that belong to pathogenic organelles and particles, is assumed to be elimination, but the pathway of this degradation is not well understood. The 26S proteasome, responsible for degrading ubiquinated proteins as part of the canonical ubiquitin-proteasome system (UPS), is not thought to play a role of degradation of ISG15 conjugates (5,6). While in general the UPS is involved primarily in the degradation of single molecules, the lysosome in the process of (macro-) autophagy is more involved in eliminating large multiple molecules, organelles and microbes/viruses. Type I interferons have also been reported to induce ISG15 associates with HDAC6 and p62 Pharmacological induction of aggrephagy became more prominent if ISG15 was overexpressed. Therefore, our results suggest that ISG15 augments p62-mediated aggresome formation and their autophagic degradation under conditions of cellular stress, such as forced expression of genes, implying an important role during intrinsic cellular defense. EXPERIMENTAL PROCEDURES Materials – Reagents: Human IFNβ 1a (11410) was obtained from PBL Assay Science. Proteasome inhibitor MG132 (40mM stock in DMSO) was from EMD Millipore. Lysosomal protease inhibitors pepstatin A (20mM in DMSO), E64d (10mg/ml in DMSO) were from Enzo Life Science. Tubacin was kindly obtained from Dr. Stuart Schreiber’s (Broad Institute, MIT/Harvard, Cambridge, MA) (20). Doxcycline hyclate was purchased from Sigma. Protease inhibitor mixtures, complete EDTA-free and phosphatase inhibitor mixtures, PhosSTOP were from Roche. Recombinant GST-tagged HDAC6 protein was from EMD Millipore. GST-tagged p62 was from Enzo Life Science. Human ISG15 protein was from Boston Biochem. Anti-FLAG M1 Agarose Affinity gel and monoclonal anti-FLAG M1 antibody were from Sigma-Aldrich. Immobilized GST agarose and GST protein were from Thermo. cDNA and lentiviral vectors: HDAC6 Flag (13823), pcDNA-hISG15 (12447), pFlagCMV2-UbcH8 (12442), pCAGGS-HAhUBE1L (12438) and HA-p62 (28027) were purchased from Addgene. Human HERC5 cDNA (GenBank #BC140716) was obtained from MGC cDNA library. Tet3G system and lentivirus vectors (pLenti CMV rtTA3G Blast, pLenti CMVTRE3G Neo DEST and pLenti CMVTRE3G Puro DEST) were purchased from Addgene. pGIPZ vector was purchased from Open Biosystems, Inc. Antibodies: Antibodies against ISG15 (sc-50366) and HDAC6 (sc-28386) were from Santa Cruz. Antibodies against FLAG M2 (F3165), αTubulin (T6074), Acetylated αTubulin (T6793) and SQSTM1/p62 (WH0008878M1) were from Sigma. Antibodies against Ubiquitin (3936), LC3B (2775) and Lamin A/C (2032) were from Cell signaling. 3 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 Antibodies against HA (11-867) and GST (ab34589) were from Roche and Abcam, respectively, and GFP (598) from MBL. Cell Culture and Transfection – 293A (Life technologies) and U251 cells (ATCC) were cultured in DMEM supplemented with 10% FBS, 100µg/ml penicillin/streptomycin and 10mM HEPES at 37 ºC in a humidified incubator of 5% CO2. For transfections, 293A cells were seeded at 2 x 106 cells on 60-mm dishes and transfected with vectors using Lipofectamine 2000 (Life technologies) following the supplier’s protocol. DNA constructs - For mutant constructs: HDAC6 point mutation at histidine 611 to alanine (H611A), and deletions from residues 841-1195 (ΔCter), from 2-837 (ΔCD1/2), from 841-1053 (ΔSE14) and from 1133-1195 (ΔBUZ) were generated from a HDAC6 Flag plasmid DNA by PCR (QuickChnge siteDirected Mutagenesis kit, Agilent Technologies). For lentivirus packaging, some HDAC6 constructs were also cloned in pGIPZ at blunt-ended NotI and XbaI sites by inserting the SpeI-XbaI fragments that contains a CMV IE promoter and a HDAC6 CDS. ISG15 truncated mutants in the C-terminal end peptides LRLRGG and aspartic acid (D) insert (LRLRGGD) were also generated from a pcDNA-hISG15 plasmid by PCR. The sequences of all mutant constructs were verified by DNA sequencing analyses. For the ISG15-CFP construct, three repeated FLAG sequences were added in ISG15 by PCR using the primers (5’caccctcgagctccatatggactacaaagaccatgacggtgatt a-3’ and 5’ttaggatcccgggcccgcccacccctcaggcgcaga-3’) and pcDNA-hISG15 as template. These were cloned in pENTR/D-TOPO (Life Technologies). The FLAG-ISG15 fragment was inserted in pmTurquoise2-Golgi (Addgene, #36205) at XhoI/BamHI sites. FLAG-tagged mTurquoise2 (referred to CFP in this study) was previously engineered (unpublished data). Gene transfer was performed either by lipofectoamine 2000 (for 293), lipofectoamine 3000 (for U251) or by lentiviral infection. For transduction of genes via lentiviral infection, vectors were packaged in lentiviral vector systems using 293T (ATCC) or 293FT cells ISG15 associates with HDAC6 and p62 followed by sonication and centrifugation to collect the supernatant as the insoluble fraction. Protein interaction assays – For the in vitro GST pull-down assay of p62, each 2µg of recombinant GST-p62 or GST protein was incubated with immobilized glutathione agarose in the binding buffer (20mM Tris-Cl [pH7.4], 50mM NaCl, 0.1% NP40, 25µg/ml BSA, 1mM ATP, 20µM MG-132 and protease inhibitors cocktail), followed by a three times wash with the binding buffer. The recombinant mature form of human ISG15 protein (2µg) was incubated with GST protein bound agarose over-night at 4°C before analysis by the immunoblot. The GST-HDAC6 pull down assay was performed using the cell lysate of 293T cells that were transfected with mutant ISG15 constructs. For coimmunoprecipitation assay, cells were lysed in a buffer containing 50mM Tris-Cl [pH7.4], 150mM NaCl, 1% Trition-X 100, 2mM EGTA, 40µM MG-132, PhosSTOP and protease inhibitors cocktail. The lysates were incubated with anti-FLAG M2 affinity gel over-night at 4°C. Immunocomplexes were collected by centrifugation and analyzed by immunoblots. Immunofluorescent microscopy analysis – Cells were cultured on German glass round coverslip (#1 thickness) and then were washed with 37°C PBS once before fixation with 4% paraformaldehyde (PFA) in PBS. For prepermeabilized cell preparation, washed cells were incubated in extraction buffer (100mM PIPES [pH6.9], 0.5mM MgCl2, 0.1mM EDTA and 0.5% Trition X-100) for ~20 sec before 4% PFA fixation. The samples were quenched with 50mM NH4Cl in PBS and permeabilized with 50 µg/ml digitonin (EMD) for nonprepermeabilized condition. The samples were washed with 0.1% gelatin in PBS and incubated with diluted antibodies (1/200, αTubulin and FLAG M; 1/50, p62 and ISG15) followed by Alexa Fluor antibodies (1/200, Life technologies). Nuclei were counterstained with DAPI during the wash step and then coverslips were mounted with VectaShield. Fluorescent images were acquired with a Zeiss LSM510 META or LSM710 confocal microscopy system and processed using Image J (1.48 or earlier version, NIH). Figure panels were assembled using Adobe Illustrator CS5. 4 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 (Life technologies) by co-transfection with packaging vectors, psPAX2 and pMD2.G (a gift of Dr. Didier Trono, Switzerland), followed by lentivirus infection. U251 expressing FLAG-HDAC6 WT and derivative mutants were stably selected by G418 treatment after infection. Dox-inducible ISG15 conjugation system - Construction of the Dox-inducible ISG15 conjugation system was as follows: 2A peptide linkage constructs between Ube1L and ISG15, and between Herc5 and UbcH8 were generated in pENTR/D-TOPO vectors with two-step PCR using the following primer sets, as described previously (21): UBE1L; 5’caccaagcttgccagcatggatgccctggacgcttcg-3’, 5’gtctcctgcttgctttaacagagagaagttcgtggctccggatccc agctcatagtgcagaggtgggaagg-3’, ISG15; 5’gccacgaacttctctctgttaaagcaagcaggagacgtggaaga aaaccccggtcctatggaattccatatgggctgggacc-3’ and 5’- caatgtatcttatcatgtctggatccccg-3’, Herc5; 5’caccgtcgacgccagcatggagcggaggtcgcggag-3’ and 5’gtctcctgcttgctttaacagagagaagttcgtggctccggatcc gccaaatcctctgttgttgttgatgg-3’, UbcH8; 5’gccacgaacttctctctgttaaagcaagcaggagacgtggaaga aaaccccggtcctgcgagcatgcgagtggtgaag-3’ and 5’- gctcgagttaggagggccggtccactcc-3’. Tet-on constructs of these 2A peptide-linked dicistronic vectors were generated in pLenti CMVTRE3G Neo DEST and pLenti CMVTRE3G Puro DEST using the gateway cloning system (Life technologies). Lentivirusmediated transformed cell lines along with rtTA3G were initially selected by G418, puromycin and blastosidin S. Clonal cells were analyzed by immunoblots using antibody against ISG15 and we used clone.7-8 of U251 (referred to U251.ISG7-8) and clone.8 of 293A (referred to 293.ISG8) in this study. Protein soluble and insoluble fractions – Cells were lysed in soluble lysis buffer (50mM Tris-Cl [pH7.4], 150mM NaCl, 2.5mM EDTA, 0.5% Trition X-100, 40µM MG-132, PhosSTOP and protease inhibitors cocktail) and incubated on ice for 5 min. After centrifugation at 13,000rpm, the supernatant was collected in the tube as the soluble fraction, whereas the pellet was dissolved in insoluble lysis buffer (1% SDS in soluble lysis buffer), ISG15 associates with HDAC6 and p62 RESULTS HDAC6 associates with IFN-induced ISG15 conjugated proteins –We first attempted to analyze whether an interaction between ISG15-conjugated substrates and HDAC6 occurred upon IFN treatment. We attempted to utilize two commercially available antibodies raised against endogenous HDAC6 to detect co-immunoprecipitates of HDAC6 with ISG15-conjugates in U251 exposed to type 1 IFN. However, we were unable to visualize co-immunoprecipitation of endogenous HDAC6 with ISGylated substrates (data not shown). Although there could be multiple reasons for this, we reasoned that perhaps we needed to over-express HDAC6 and/or the HDAC6 antibody was not sufficiently avid to allow coimmunoprecipitations. We thus performed a second series of experiments whereby the lysates of human U251 glioma cells, forced to express FLAGtagged HDAC6 by lentivirus transduction, were immunoprecipitated with an antibody against FLAG-peptides and the coimmunoprecipitates of FLAG- HDAC6 were analyzed by Western blots using antibodies against ISG15 or Ubiquitin. Figure 1a shows that IFN treatment led to ISG15 conjugation. FLAG-HDAC6 co-immunoprecipated with ISGylated substrates and this was even more prominent when cell were treated by MG132, a protease inhibitor that induces formation of intracellular aggresomes. In comparison and in contrast, there was a specific effect of MG132 on FLAG-HDAC6 association with ubiquitinated substrates, independent of IFN treatment (17,18). These data thus suggested that there was a previously undescribed, yet visible, association of forcedly expressed FLAG-HDAC6 with IFN-induced ISGylated substrates even in the absence of MG132 (i.e. when the proteasome and the unfolded protein system - UPS- is functional). Next, we visualized the subcellular localization of FLAG-HDAC6 and ISG15 in IFNβ treated cells utilizing immunofluorescent staining with antibodies against FLAG and ISG15 (Fig. 1b). ISG15 displayed small 5 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 punctate stains in the cytoplasm with a concentration in the perinuclear region where FLAG-HDAC6 vacuoles also appeared to partly localize, although co-localization was minimal, if any. To examine whether this lack of localization reflected insoluble aggregates, live cells were permeabilized with a pulse of TritonX-100 to remove soluble proteins from cytosols prior to fixation (Fig. 1 c-d). In permeabilized cells in the absence of proteasomal inhibition, ISG15 punctate stains were visualized in the same perinuclear region as that of small HDAC6 subfractions (Fig. 1c). However, in the presence of MG132 proteasome inhibition there was visually intensified HDAC6 immunofluorescence with a perinuclear aggregation pattern that now also colocalized with ISG15 speckles (Fig. 1d). Taken together, these results suggested that the interaction between HDAC6 and ISG15 was localized to detergent-resistant aggregates in the perinuclear region and that proteasome inhibition led to a prominent co-localization of ISG15 and HDAC6 in the same aggregates. HDAC6 binds ISG15 at the C-terminal LRLRGG motif via a BUZ domain – To find out which motif of HDAC6 interacts with ISG15, we engineered U251 cells that stably overexpress its FLAG-tagged full-length wildtype HDAC6 (WT), or its C-terminal deletion mutant (ΔCter; Δ841-1195) or its catalytic subsite mutant (H611A) (Fig. 2a). Subsequently, we performed co-IP assays with an antibody against FLAG peptides, using cell lysates that had been treated with IFNβ. Figure 2b shows that ISG15 was bound to FLAGHDAC6 WT and FLAG-HDAC6H611A, but not to FLAG-HDAC6ΔCter. To further map the ISG15 binding site in residues 841-1195 of the HDAC6 C-terminal region, we transiently transfected FLAG-tagged full-length HDAC6 and truncation mutants constructs of HDAC6 that lackedthe BUZ domain (BUZ), or the SE14 domain (SE14) or both catalytic domains (CD1/2) (see Figure 2a) together with the HA-tagged mature form of ISG15 in 293 cells. In addition to FLAG-HDAC6 WT, figure 2c shows that ISG15 coimmunoprecipitated with the CD1/2 and SE14, but not with the BUZ truncation ISG15 associates with HDAC6 and p62 ISG15 also bound to p62. In fact, a pull-down assay using recombinant GST-tagged p62 did lead to direct binding of GST-p62 to ISG15 (Fig. 3a). This indicated that ISG15, like K63linked diUb, also directly bound to p62. In order to test if p62 was involved in the formation and clearance of aggregates of ISGylated proteins, we engineered U251 cells that expressed a doxycycline (Dox)-inducible transcriptionally regulated ISG15, as well as UbE1L, UbcH8 and Herc5, the enzymatic components of the ISG15 conjugation cascade (Fig. 3b). These cells thus would express the ISG15 conjugation system upon Dox treatment, independent of IFN. Figure 3c shows that one of the isolated clones (U251.ISG7-8) showed evidence of significant induction of ISGylation under Dox treatment. We then sought to determine if ISGylation affected proteins associated with p62 in soluble vs. insoluble cellular subfractions. Uninduced or Doxinduced cells were treated with pepstatin A and E64d (PepA/E64d, inhibitors of lysosomal proteases) and then soluble and insoluble fractions of cell lysates were analyzed by Western blots. Figure 3d shows that Doxtreatment induced ISGylation of the proteome in both soluble and insoluble fractions and that this was even more evident when cells were treated with PepA/E64d. Interestingly, Dox treatment resulted in perhaps a mild increase in p62 levels in the soluble fraction, while LC3-II levels were increased a bit more convincingly. However, there was a clear increase by Dox of p62 and LC3-II in the insoluble fraction which was even more visible in PepA/E64d treated cells. Similar findings were also obtained in IFNβ-treated U251 cells (see Fig. 6). These results suggested that induction of ISG15 and ISGylation leads to upregulation of p62 and LC3-II association with IGS15-conjugated proteins ending up in the lysosomes, under condition of both Dox-induced ISGylation and IFN-stimulation (from Fig. 6). The association with p62 and LC3-II strongly suggests an autophagic mechanism for degradation of ISG15. To analyze the in vivo interaction between p62 and ISG15-conjugated proteins, we stably transduced 293 cells to express the ISG15 conjugation system in response to Dox 6 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 mutant of HDAC6. Therefore, these findings indicated that residues 1133-1195, corresponding to a BUZ domain, were responsible for ISG15 binding. Based on this, we then sought to find the motif in ISG15 responsible for binding to HDAC6. It has been reported that the BUZ domain of HDAC6 interacts with a terminal conjugation motif of ubiquitin (ub), composed of LRLRGG. This motif appears in Ubaggregates that contain free mono- or polyubiquitin chains (22). The mature form of ISG15 also represents a carboxyl-terminus LRLRGG motif and thus we asked if it also bound to HDAC6 BUZ. We utilized a fulllength mature ISG15 (ISG15-WT) and four mutants of ISG15 (ISG15-LRLRG; ISG15LRLR; ISG15-LRLRGGD; and ISG15-LRGG) with deletions or additions at the C-terminus residues (Fig. 2d). In vitro protein affinity assays revealed that the full-length, wild-type LRLRGG motif was the most efficient at interacting with recombinant GST-tagged HDAC6, while all the other mutant forms of ISG15 failed to bind to GST-HDAC6 (Fig. 2e). There was some relatively weak interaction between the LRLRGG-D mutant with HDAC6. Overall, these findings thus confirmed that the ISG15 LRLRGG peptide motif is indispensable for binding to the BUZ domain of HDAC6. ISG15 associates with p62, likely to facilitate autophagosome/lysosome clearance – The Ub-binding property of HDAC6 is associated with formation and clearance of organized aggregates (also called aggresomes), as part of the autophagy-lysosome degradation pathway (12,17,23). During this process, the SQSTM1/p62 cargo protein forms aggregates of ubiquitinated proteins and recruits LC3, anchored in the phagophore membrane (24). p62 preferentially binds with higher affinity to K63-linked diUb, when compared to binding to mono-ubiquitin and K48-linked diUb (25). Our in silico analysis using the flexible structure alignment (available on the PDB site: http://www.rcsb.org/pdb/), predicted that the structure of ISG15 (PDB ID: 1z2m)4 resembles that of K63-linked diUb (PDB IDs; 2w9n) rather than that of K48-linked diUb (PDB IDs; 3m3j) (26-29). We thus sought to determine if ISG15 associates with HDAC6 and p62 contributes to the formation of ISG15containing aggregates in the MTOC. An engineered ISG15 protein is a target of lysosomal degradation – We also attempted to detect ISG15-conjugate degradation using an engineered ISG15 conjugate. For this, we engineered a cyan- fluorescence protein (CFP) fused ISG15 expression vector, in which the C-terminal LRLRGG is linked to the Nterminal of mTurquoise2 (CFP) protein (Fig. 5a). As shown in Figure 5b, we could not detect the fusion protein upon DNA transfection in the absence of interferon, but instead observed separate immunopositive bands for ISG15 and mTurquoise2 proteins, suggesting fairly prompt cleavage of the expressed fusion protein, after transfection into cells. This observation appears to agree with that describing precursor processing of ISG15 to expose LRLRGG-terminal peptides by proteases (30). We then took advantage of this observed cleavage of ISG15-CFP protein to evaluate its lysosomal degradation (Fig. 5c). Similar to results of Fig.3, cleaved ISG15 protein was observed in the detergent insoluble fraction and PepA/E64d treatment caused accumulation of the ISG15 protein. These findings thus suggested that ISG15 protein can be a target of lysosomal degradation. The deacetylase activity of HDAC6 contributes to ISG15 degradation – We had shown In Fig. 2 that the BUZ domain of HDAC6 was essential for binding to ISG15 protein and it has also been shown that HDAC6’s deacetylase activity is involved in the degradation of the aggresome, containing ubiquitinated proteins via autophagy-lysosome (10). In a similar fashion, we thus sought to determine if HDAC6 activity was involved in ISG15 degradation. Fig. 6 shows that type 1 IFN, as expected, visibly increased ISG15 levels and also mildly increased ISG15conjugates in both soluble and insoluble fractions. When PepA/E64D lysosomal protease inhibitors were added, there was a visible increase in ISG15 levels in the insoluble fraction. This suggests that ISG15 can be degraded by lysosomal proteases. We then utilized Tubacin (20), a relatively selective HDAC6 deacetylase inhibitor, and observed a visible decrease of 7 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 (293.ISG8 cells), as shown in Fig. 3e. These 293.ISG8 cells were then transiently transfected with human p62 containing a Cterminal FLAG epitope tag (FLAG-p62) or empty vector (empty). Figure 3f shows that FLAG-p62 coimmunoprecipitated with ISG15conjugated substrates. Interestingly, MG132 led to a visible decrease in FLAG-p62 levels, in the input lysates that were extracted from the detergent-containing lysis buffer (Fig. 3f). To further try to understand this, we decided to analyze the detergent-insoluble fraction for p62 levels as a function of MG132 doses, with and without Dox treatment. Figure 3g shows that, in the absence of Dox, p62 was present primarily in the soluble fraction without an effect from proteasome inhibition. With Dox (thus increasing ISGylation activity) and increased doses of MG132, there was a visible shift of p62 from the soluble to insoluble fractiosn. Since MG132 treatment blocks autophagic degradation pathway of aggregates, there was also a MG132 dose-dependent increase in LC3, ISGylated proteins and HDAC6 in the insoluble fraction, although a lot of these were still persistent in the soluble fraction. These findings thus suggested that ISGylation led to a shift of p62, HDAC6 and LC3 into the insoluble aggregate fraction of cells, which was detectable once the proteasome was inhibited. Therefore, to determine if p62 interacted and directed ISG15 to accumulate in inclusion bodies in response to IFN, we prepared prepermeabilized IFN-treated U251 cells and immunostained them using antibodies against p62, ISG15 and α-tubulin (Fig. 4). In the absence of MG132, cells contained numerous small punctuated areas of staining for p62 bodies in the cytosol, most of which also colocalized with ISG15 (Fig. 4a). The p62positive puncta became visibly larger in MG132 treated cells, and this was also true for puncta in which ISG15 and p62 colocalized (Fig. 4b). ISG15 puncta were also observed on detergent-resistant stable microtubules, regardless of MG132 (Fig. 4c-d), which accumulated ISG15-positive large aggregates at the microtubule-organizing center (MTOC) (Fig. 4d). These results suggested that p62 ISG15 associates with HDAC6 and p62 ISG15-conjugation marks target substrates for clearance via the autophagy-lysosomal pathway, as proposed in the model shown in Fig. 7. We should note that the reported data was mostly obtained under experimental conditions of forced over-expression and have not been able to detect this under normal, homeostatic physiological conditions. This implies that the observed results may occur primarily when the cell is stressed (forced over-expression of cDNAs, interferon stimulation, inhibition of the proteasome and lysosomal proteases). The structural conformation of ISG15 resembles that of K63-linked diUb rather than that of K48-linked diUb, known to associate with the proteasomal pathway. K63-linkage appears to function in homeostatic cellular processes, including signal transduction (31). In addition, when molecular chaperons and the proteasome-degradation systems are unable to unfold and thread misfolded/aggregated proteins through the narrow interior proteolytic chamber of the 26S proteasome, these proteins get tagged with K63, designating them for the autophagy-lysosome pathway (11,13). Molecularly, this happens through p62 that preferentially binds to polyubiquitinated proteins with K63-linked chains instead of those with K48-linked chains. This ends up forming oligomeric aggregates of ubiquinated proteins, organelles and intracellular pathogens that lead to autophagic rather than proteasomal degradation (15,25,32,33). This is also in agreement with evidence indicating that K63linked polyubiquitin substrates are enriched in intracellular inclusion bodies, which are also sequestered into p62-LC3 marked autophagosomes (34). Results in the present study show that p62 also binds to non-covalent ISG15 by pull-down assay and to ISG15conjugated proteins by coIP assay. In addition, using a ISG15 fusion protein with CFP via linkage of LRLRGG, we clearly showed that ISG15 itself is a target of lysosomal degradation (Fig. 5c). Therefore, this suggests that p62 also directly targets ISG15 and ISGylated peptides to form aggregates and autophagosomes, in a manner that is similar to p62 interaction with K63-linked poly- DISCUSSION The ubiquitin-like post-translational modifier ISG15 is up-regulated during type 1 IFN stimulation of cells and it becomes conjugated to newly synthesized proteins of host cell and invading pathogens. Although this conjugation can be reversed by a specific protease UBP43/USP18 (3), it is not known whether other cellular pathways may also be operative. In this report, for the first time, we show that: 1- ISG15 interacts via its LRLRGG C-terminus motif with a BUZ domain of cytosolic Ub-binding HDAC6, implicating HDAC6 for a relevant role in ISG15’s function. Since this interaction is the same as that known to occur for (poly-) ubiquitin chains binding to HDAC6, this also suggests that the ISG15 pathway may represent the IFN-stimulated counterpart to the normal homeostatic ubiquitin clearance pathway; 2- ISG15 also interacts with the cargo-adapter protein, SQSTM1/p62. Knowing that both p62 and HDAC6 facilitate aggregate formation (aggresomes) and clearance of ubiquitinated cargo through the LC3-II marked autophagosome and lysosome, this also suggests that a similar process may be occurring with ISGylation of proteins as Ublike cargo at innate immune response. Therefore, our data strongly suggests that 8 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 ISG15 levels in both soluble and insoluble fractions without much of a change in ISG15 conjugate levels. However, when PepA/E64D lysosomal protease inhibitors were also added there was no change in ISG15 levels. This suggested that HDAC6 was needed for the degradation of ISG15 by lysosomal proteases. Tubacin also led to an increase of p62 and LC3 levels in the insoluble fraction. Therefore inhibition of HDAC6 activity led to the accumulation of p62 and LC3-II in the insoluble fraction, suggesting inhibition of lysosome-autophagosome fusion. It also reduced total ISG15 levels in cells in response to type 1 IFN, but this did not lead to an increase in ISG15 when lysosomal proteases were inhibited. Those results suggested that HDAC6's deacetylase activity is important for degradation of ISG15 via the lysosome. ISG15 associates with HDAC6 and p62 evidence that HDAC6 directly binds to Ubconjugated proteins in vitro (12). We also identify for the first time in this report that the BUZ domain of HDAC6 interacts with the Cterminal LRLRGG peptide of free ISG15. Coupled with the coIP evidence of FLAGHDAC6 interaction with ISG15 conjugates, we speculate that HDAC6 may recognize and associate with unanchored free ISG15 in the p62 cargo, which also sequesters the substrates of ISG15 conjugation. The multifaceted adapter p62 plays critical roles in the regulation of a broad range of signaling pathways that respond to inflammatory and oxidative stresses through its binding to K63-linked Ub-proteins and related binding partners such as tumor necrosis factor receptor-associated factor (TRAF) 6 and Kelch-like ECH-associated protein 1 (Keap1) (31,41). ISG15 is also involved in innate immune signaling pathway. In fact, IFNinduced ISGs proteins including IRF3 are known substrates of ISG15 conjugation and their function is modulated by ISGylation (42). This report also shows for the first time that p62 is involved in ISG15-mediated signaling pathways, in a manner similar to published reports related to p62 involvement in K63linked Ub-mediated signaling. However, it is not known how p62 distinguishes ISG15conjugated proteins that remain active to perform a function from those that are targets for a degradation tag. One possible explanation is that, for ISGylated proteins to remain active, p62-scaffolding with ISG15 conjugates forms without free ISG15 and/or free K63-Ub chains. On the other hand, for proteins destined for degradation, sequestration into disorganized aggresomes occurs (and this could be induced by blocking the UPS with MG132), in combination with HDAC6 and LC3 recruitment (12). In fact, we did find accumulation of free ISG15 in the detergentinsoluble fraction of cells with p62 and lipid bound LC3-II as a function of MG132 dose. Further, the catalytic inhibition of HDAC6 with Tubacin also suppressed lysosomal ISG15 degradation. Therefore, it is likely that type 1 IFN signaling induces ISG15, which in turn strengthens the basal activity of selective autophagy, leading to efficient removal of 9 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 ubiquitinated proteins being targeted to autophagosomes. ISG15 and its conjugates have been shown to possess activity against several viruses using cultured cells or animal models (4). The antiviral mechanisms of ISG15 include inhibition of viral release and budding, as well as direct ISG15 conjugation of cellular proteins including antiviral factors (e.g. IRF3, RIG-I, PKR) and other ISG proteins. Also, ISG15 can be directly conjugated with capsid proteins of papillomavirus (HPV) and Sindbis virus (SIN) (2,35). However, it is unknown how ISG15 modification of viral proteins acts mechanistically. Our findings show that ISG15 not only colocalizes with p62 and HDAC6 in the same intracellular punctuate areas (puncta), but also associates with them. p62 and other sequestrome-1-like receptors (SLRs) function as cargo-adaptor proteins that target substrates to autophagosomes via the LC3-interacting region (LIR) motif. In xenophagy, ubiquitination of invading bacteria is required for antimicrobial autophagy through p62-cargo recognition and degradation (36). In a similar manner to antiviral autophagy (virophagy), capsids of SIN and Chikungunya virus (CHIKV) are targets of autophagosomes through binding of p62 and CHIKV capsids are also modified by ubiquitin (37,38). Therefore, future studies should elucidate if ISG15 enhances degradation or sequestration of viral proteins via p62 and HDAC6 mediated selective autophagy. HDAC6 functions to deacetylate cytoplasmic proteins (e.g. α-tubulin) and catalytic inhibition leads to hyper-acetylation of α-tubulin, which stimulates canonical (bulk) autophagy by kinesin-1 recruitment to microtubules (MT) for autophagosome transport toward the MTOC (39,40). In noncanonical (selective) autophagy, HDAC6 regulates the fusion of autophagosome and lysosome through both catalytical deacetylase and ubiquitin binding activities (10). We also observed that this deacetylase activity is associated with ISG15 degradation via lysosome-autophagosome (Fig. 6). HDAC6 preferentially interacts with K63-linked polyubiquitinated protein aggregates in vivo (16). However, there is no biochemical ISG15 associates with HDAC6 and p62 ubiquitin and ISG15-tagged unwelcome proteins and pathogens through p62 and HDAC6. In summary, our studies provide novel evidence for ISG15 association with HDAC6 and p62, implying that ISGylation can lead to autophagic degradation, in pathways that are similar to those utilized by the Ub system. ISG15 mediated protein conjugation may thus be the IFN based system that, together with or in lieu of Ub-based selective autophagy, allows the cell to use autophagy as an intrinsic antipathogen defense. Downloaded from http://www.jbc.org/ by guest on December 4, 2014 10 ISG15 associates with HDAC6 and p62 REFERENCES 1. 2. 3. 4. 5. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 11 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 6. Sadler, A. J., and Williams, B. R. (2008) Interferon-inducible antiviral effectors. Nature reviews. Immunology 8, 559-568 Durfee, L. A., Lyon, N., Seo, K., and Huibregtse, J. M. (2010) The ISG15 conjugation system broadly targets newly synthesized proteins: implications for the antiviral function of ISG15. Molecular cell 38, 722-732 Malakhov, M. P., Malakhova, O. A., Kim, K. I., Ritchie, K. J., and Zhang, D. E. (2002) UBP43 (USP18) specifically removes ISG15 from conjugated proteins. The Journal of biological chemistry 277, 9976-9981 Morales, D. J., and Lenschow, D. J. (2013) The antiviral activities of ISG15. Journal of molecular biology 425, 4995-5008 Romijn, R. A., Westein, E., Bouma, B., Schiphorst, M. E., Sixma, J. J., Lenting, P. J., and Huizinga, E. G. (2003) Mapping the collagen-binding site in the von Willebrand factor-A3 domain. The Journal of biological chemistry 278, 15035-15039 Liu, M., Li, X. L., and Hassel, B. A. (2003) Proteasomes modulate conjugation to the ubiquitinlike protein, ISG15. The Journal of biological chemistry 278, 1594-1602 Schmeisser, H., Bekisz, J., and Zoon, K. C. (2014) New function of type I IFN: induction of autophagy. Journal of interferon & cytokine research : the official journal of the International Society for Interferon and Cytokine Research 34, 71-78 Johansen, T., and Lamark, T. (2011) Selective autophagy mediated by autophagic adapter proteins. Autophagy 7, 279-296 Shaid, S., Brandts, C. H., Serve, H., and Dikic, I. (2013) Ubiquitination and selective autophagy. Cell death and differentiation 20, 21-30 Lee, J. Y., Koga, H., Kawaguchi, Y., Tang, W., Wong, E., Gao, Y. S., Pandey, U. B., Kaushik, S., Tresse, E., Lu, J., Taylor, J. P., Cuervo, A. M., and Yao, T. P. (2010) HDAC6 controls autophagosome maturation essential for ubiquitin-selective quality-control autophagy. The EMBO journal 29, 969-980 Tan, J. M., Wong, E. S., Kirkpatrick, D. S., Pletnikova, O., Ko, H. S., Tay, S. P., Ho, M. W., Troncoso, J., Gygi, S. P., Lee, M. K., Dawson, V. L., Dawson, T. M., and Lim, K. L. (2008) Lysine 63-linked ubiquitination promotes the formation and autophagic clearance of protein inclusions associated with neurodegenerative diseases. Human molecular genetics 17, 431-439 Hao, R., Nanduri, P., Rao, Y., Panichelli, R. S., Ito, A., Yoshida, M., and Yao, T. P. (2013) Proteasomes activate aggresome disassembly and clearance by producing unanchored ubiquitin chains. Molecular cell 51, 819-828 Pankiv, S., Clausen, T. H., Lamark, T., Brech, A., Bruun, J. A., Outzen, H., Overvatn, A., Bjorkoy, G., and Johansen, T. (2007) p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. The Journal of biological chemistry 282, 24131-24145 Babu, J. R., Geetha, T., and Wooten, M. W. (2005) Sequestosome 1/p62 shuttles polyubiquitinated tau for proteasomal degradation. Journal of neurochemistry 94, 192-203 Long, J., Gallagher, T. R., Cavey, J. R., Sheppard, P. W., Ralston, S. H., Layfield, R., and Searle, M. S. (2008) Ubiquitin recognition by the ubiquitin-associated domain of p62 involves a novel conformational switch. The Journal of biological chemistry 283, 5427-5440 Olzmann, J. A., Li, L., Chudaev, M. V., Chen, J., Perez, F. A., Palmiter, R. D., and Chin, L. S. (2007) Parkin-mediated K63-linked polyubiquitination targets misfolded DJ-1 to aggresomes via binding to HDAC6. The Journal of cell biology 178, 1025-1038 Kawaguchi, Y., Kovacs, J. J., McLaurin, A., Vance, J. M., Ito, A., and Yao, T. P. (2003) The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 115, 727-738 ISG15 associates with HDAC6 and p62 18. 19. 20. 21. 22. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 12 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 23. Pai, M. T., Tzeng, S. R., Kovacs, J. J., Keaton, M. A., Li, S. S., Yao, T. P., and Zhou, P. (2007) Solution structure of the Ubp-M BUZ domain, a highly specific protein module that recognizes the C-terminal tail of free ubiquitin. Journal of molecular biology 370, 290-302 Zuin, A., Carmona, M., Morales-Ivorra, I., Gabrielli, N., Vivancos, A. P., Ayte, J., and Hidalgo, E. (2010) Lifespan extension by calorie restriction relies on the Sty1 MAP kinase stress pathway. The EMBO journal 29, 981-991 Haggarty, S. J., Koeller, K. M., Wong, J. C., Grozinger, C. M., and Schreiber, S. L. (2003) Domain-selective small-molecule inhibitor of histone deacetylase 6 (HDAC6)-mediated tubulin deacetylation. Proceedings of the National Academy of Sciences of the United States of America 100, 4389-4394 Szymczak, A. L., Workman, C. J., Wang, Y., Vignali, K. M., Dilioglou, S., Vanin, E. F., and Vignali, D. A. (2004) Correction of multi-gene deficiency in vivo using a single 'self-cleaving' 2A peptide-based retroviral vector. Nature biotechnology 22, 589-594 Reyes-Turcu, F. E., Horton, J. R., Mullally, J. E., Heroux, A., Cheng, X., and Wilkinson, K. D. (2006) The ubiquitin binding domain ZnF UBP recognizes the C-terminal diglycine motif of unanchored ubiquitin. Cell 124, 1197-1208 Ouyang, H., Ali, Y. O., Ravichandran, M., Dong, A., Qiu, W., MacKenzie, F., Dhe-Paganon, S., Arrowsmith, C. H., and Zhai, R. G. (2012) Protein aggregates are recruited to aggresome by histone deacetylase 6 via unanchored ubiquitin C termini. The Journal of biological chemistry 287, 2317-2327 Kirkin, V., McEwan, D. G., Novak, I., and Dikic, I. (2009) A role for ubiquitin in selective autophagy. Molecular cell 34, 259-269 Seibenhener, M. L., Babu, J. R., Geetha, T., Wong, H. C., Krishna, N. R., and Wooten, M. W. (2004) Sequestosome 1/p62 is a polyubiquitin chain binding protein involved in ubiquitin proteasome degradation. Molecular and cellular biology 24, 8055-8068 Ye, Y., and Godzik, A. (2003) Flexible structure alignment by chaining aligned fragment pairs allowing twists. Bioinformatics 19 Suppl 2, ii246-255 Narasimhan, J., Wang, M., Fu, Z., Klein, J. M., Haas, A. L., and Kim, J. J. (2005) Crystal structure of the interferon-induced ubiquitin-like protein ISG15. The Journal of biological chemistry 280, 27356-27365 Komander, D., Reyes-Turcu, F., Licchesi, J. D., Odenwaelder, P., Wilkinson, K. D., and Barford, D. (2009) Molecular discrimination of structurally equivalent Lys 63-linked and linear polyubiquitin chains. EMBO reports 10, 466-473 Trempe, J. F., Brown, N. R., Noble, M. E., and Endicott, J. A. (2010) A new crystal form of Lys48-linked diubiquitin. Acta crystallographica. Section F, Structural biology and crystallization communications 66, 994-998 Potter, J. L., Narasimhan, J., Mende-Mueller, L., and Haas, A. L. (1999) Precursor processing of pro-ISG15/UCRP, an interferon-beta-induced ubiquitin-like protein. The Journal of biological chemistry 274, 25061-25068 Komatsu, M., Kurokawa, H., Waguri, S., Taguchi, K., Kobayashi, A., Ichimura, Y., Sou, Y. S., Ueno, I., Sakamoto, A., Tong, K. I., Kim, M., Nishito, Y., Iemura, S., Natsume, T., Ueno, T., Kominami, E., Motohashi, H., Tanaka, K., and Yamamoto, M. (2010) The selective autophagy substrate p62 activates the stress responsive transcription factor Nrf2 through inactivation of Keap1. Nature cell biology 12, 213-223 Itakura, E., and Mizushima, N. (2011) p62 Targeting to the autophagosome formation site requires self-oligomerization but not LC3 binding. The Journal of cell biology 192, 17-27 Searle, M. S., Garner, T. P., Strachan, J., Long, J., Adlington, J., Cavey, J. R., Shaw, B., and Layfield, R. (2012) Structural insights into specificity and diversity in mechanisms of ubiquitin recognition by ubiquitin-binding domains. Biochemical Society transactions 40, 404-408 ISG15 associates with HDAC6 and p62 34. 35. 36. 37. 38. 39. 41. 42. 13 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 40. Wooten, M. W., Geetha, T., Babu, J. R., Seibenhener, M. L., Peng, J., Cox, N., Diaz-Meco, M. T., and Moscat, J. (2008) Essential role of sequestosome 1/p62 in regulating accumulation of Lys63ubiquitinated proteins. The Journal of biological chemistry 283, 6783-6789 Lenschow, D. J., Giannakopoulos, N. V., Gunn, L. J., Johnston, C., O'Guin, A. K., Schmidt, R. E., Levine, B., and Virgin, H. W. t. (2005) Identification of interferon-stimulated gene 15 as an antiviral molecule during Sindbis virus infection in vivo. Journal of virology 79, 13974-13983 Boyle, K. B., and Randow, F. (2013) The role of 'eat-me' signals and autophagy cargo receptors in innate immunity. Current opinion in microbiology 16, 339-348 Orvedahl, A., MacPherson, S., Sumpter, R., Jr., Talloczy, Z., Zou, Z., and Levine, B. (2010) Autophagy protects against Sindbis virus infection of the central nervous system. Cell host & microbe 7, 115-127 Judith, D., Mostowy, S., Bourai, M., Gangneux, N., Lelek, M., Lucas-Hourani, M., Cayet, N., Jacob, Y., Prevost, M. C., Pierre, P., Tangy, F., Zimmer, C., Vidalain, P. O., Couderc, T., and Lecuit, M. (2013) Species-specific impact of the autophagy machinery on Chikungunya virus infection. EMBO reports 14, 534-544 Geeraert, C., Ratier, A., Pfisterer, S. G., Perdiz, D., Cantaloube, I., Rouault, A., Pattingre, S., Proikas-Cezanne, T., Codogno, P., and Pous, C. (2010) Starvation-induced hyperacetylation of tubulin is required for the stimulation of autophagy by nutrient deprivation. The Journal of biological chemistry 285, 24184-24194 Reed, N. A., Cai, D., Blasius, T. L., Jih, G. T., Meyhofer, E., Gaertig, J., and Verhey, K. J. (2006) Microtubule acetylation promotes kinesin-1 binding and transport. Current biology : CB 16, 2166-2172 Sanz, L., Diaz-Meco, M. T., Nakano, H., and Moscat, J. (2000) The atypical PKC-interacting protein p62 channels NF-kappaB activation by the IL-1-TRAF6 pathway. The EMBO journal 19, 1576-1586 Shi, H. X., Yang, K., Liu, X., Liu, X. Y., Wei, B., Shan, Y. F., Zhu, L. H., and Wang, C. (2010) Positive regulation of interferon regulatory factor 3 activation by Herc5 via ISG15 modification. Molecular and cellular biology 30, 2424-2436 ISG15 associates with HDAC6 and p62 FOOTNOTES FIGURE LEGENDS FIGURE 1. HDAC6 interacts with ISG15-conjugated cellular proteins. (a) U251 expressing FLAGHDAC6 WT (full-length) were treated with IFN (1,000 unit/ml; 16h), with and without MG132 (40 µM, 4h). Anti-FLAG -immunoprecipitated (IP) proteins and input cell lysates were immunoblotted with antiFLAG, anti-ISG15 and anti-Ubiquitin (Ub) antibodies. (b-d) U251 cells expressing FLAG-HDAC6 WT were treated with IFNβ (1,000 units/ml, 16h) in the absence (for b, c) or the presence (for d) of MG132 (40 µM, 5h). Cytoskeletal insoluble fractions were visible in pre-permeabilized cell with Triton-X 100 (c,d) Fluorescence signals for Alexa-488 (green) for FLAG, Alexa-555 (red) for ISG15 and DAPI (blue) were visualized using confocal microscopy. High magnification views highlighted by broken line boxes are shown in the bottom panels of the image (b). Scale bar: 20µm FIGURE 2. Mapping the interacting domains of HDAC6 and ISG15. (a) Schematic of FLAG-tagged HDAC6 and its deletion mutants. The two catalytic domains (CD1, CD2), a Ser-Glu-containing tetradecapeptide repeat domain (SE14), and a binding of Ubiquitin zinc finger domain (BUZ) are shown as closed boxes. Asterisk (*) represents the H611A point mutation in WT (b) U251 cells that express wild-type FLAG-HDAC6 (WT), mutant without an active site histidine (H611A), an SE14-BUZ truncated mutant (ΔCter), or empty vector (Vec), were treated with IFNβ (1,000 unit/ml; 16h). Lyates were subjected to the co-immunoprecipitation with antibody against FLAG and analysis using antibodies against FLAG and ISG15. (c) 293 cells were transiently co-transfected with expression vectors for a FLAG-HDAC6 WT, a C-terminal truncation (ΔCter) mutant, a N-terminal truncation (ΔCD1/2) mutant, a BUZ domain truncation (ΔBUZ) mutant, a SE14 domain truncation (ΔSE14) mutant or an empty vector (Vec), along with an HA-tagged ISG15 expression vector. Lysates and immunoprecipitates were analyzed with antibodies against FLAG and HA. (d) Schematic of HA-tagged ISG15 with wild type or mutants. The two Ub-like domains are shown as closed boxes. HA peptides were tagged at the N-terminal and deleted amino acid terminal of the LRLRGG peptide sequences at the C-terminal end are described as (*). (e) 293A cells were transiently transfected with the plasmids carrying HA-tagged ISG15 vectors with mutants. In a pull-down assay, cell lysates were incubated with purified recombinant GST-tagged 14 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 *This work was supported by funds from NIH grant P01 CA069246 to EAC and from a sundry fund by the Brigham and Women’s Hospital to EAC. 1 Harvey Cushing Neuro-oncology Laboratories, Harvard Institutes of Medicine 9th floor, Department of Neurosurgery and Institute for the Neurosciences at the Brigham and Women's Hospital, 4 Blackfan Circle, Boston, MA 02115 2 To whom correspondence should be addressed: E. Antonio Chiocca, Department of Neurosurgery, Brigham and Women's Hospital, 75 Francis Street, Boston, MA USA, Tel.: (617) 732-6939; Fax: (617) 734-8342; E-mail: [email protected] 3 The abbreviations used are: ISG15, interferon-stimulated gene 15; IFN, interferon; HDAC, histone deacetylase; BUZ, a binder of ubiquitin zinc finger; Ub, ubiquitin; IFNAR, interferon α/β receptor; PRR, pattern-recognition receptor; PKR, protein kinase R; RNaseL, ribonuclease L; ISGylation, ISG15 conjugation; UPS, ubiquitin proteasome system; MTOC, microtubule organizing center; K63, lysine 63; K48, lysine 48; UBA, ubiquitin associated; MEF, mouse embryonic fibroblast; CD, catalytic domain; SE14, serine-glutamate containing tetradecapeptide; PepA/E64d, pepstatin A and E64d; SLR, sequestrome-1 like receptor; LIR, LC3 interacting region; HPV, human papillomavirus; SIN, Sindbis virus; CHIKV, Chikungunya virus; MT, microtubule; TRAF6, tumor necrosis factor receptor-associated factor 6; Kerp1, Kelch-like ECH-associated protein 1 4 Research Collaboratory for Structural Bioinformatics Protein Databank = PDB #1z2m, 2w9n, 3m3j ISG15 associates with HDAC6 and p62 HDAC6 or control GST, bound to glutathione-Sepharose beads, subjecting to the analysis with antibodies against HA and GST. FIGURE 4. ISG15 and p62 colocalization in insoluble fraction. (a-d) U251 cells were treated with IFNβ (1000 units/ml, 16h) before treatment with MG132 (40 µM, 5h) (b,d) or without it (a,c). Prepermealized cells were fixed with 4% PFA and subjected to fluorescence microscopy analysis using alexa-488 (green) against p62 and αTubulin, alexa-555 (red) against ISG15 and DAPI (blue). High magnification rescans of the broken line box ROIs are shown in the panels on the right of each sample image. Scale bar: 50µm FIGURE 5. Lysosomal degradation of cleaved ISG15. (a) Schematic of ISG15-CFP and CFP expressing cDNA vector and its product protein (a.a.). the LRLRGG c-terminal end of FLAG-tagged human ISG15 is linked to the N-terminal of mTurquoise2 (referred to as CFP). Control CFP is tagged with FLAG at the C-terminal end. (b) Cell lysates were prepared in RIPA buffer three days after transient transfection of the vectors as indicated, and were immunoblotted using antibodies against GFP, ISG15, HDAC6 and p62. (c) Immunoblots of detergent soluble (Soluble) and –insoluble (Insoluble) fractions of U251 with 24h transient transfection of the indicated vectors in the presence or absence of pepstatin A and E64d (PepA/E64d; each 20 µg/ml, 6h), were probed for the indicated proteins. Volumes of insoluble fractions are 5-fold (x5) higher concentration than those of the soluble fractions. Numbers shown on the left images indicate protein sizes (kDa). FIGURE 6. The HDAC6 deacetylase inhibitor suppresses ISG15 degradation. U251 cells were treated with IFNβ (1000 units/ml, 12h) and Tubacin (5µM, 13h) before treatment with PepA/E64d (20 µg/ml, 6h). Cell lysates prepared as soluble and insoluble fraction was immunoblotted using antibodies against indicated proteins. An immunoblot against ISG15 shows both a short-exposure (short) and a longexposure (long) time to compare the levels in non-conjugated ISG15. FIGURE 7. Schematic model of ISG15-mediated degradation. a. IFNs are known to stimulate via the STAT-JAKs pathway a number of interferon-stimulated genes including ISG15, UbE1L, UbcH8, Herc5 that lead to ISGylation process and UBP43 that deconjugates ISGylated proteins. b. The K63-linked diUb-like protein, ISG15, conjugates to lysine (K) residues of newly synthesized proteins at the LRLRGG terminal end (2). Some ISG15-conjugates are also deconjugated to remove ISG15. c. Our findings indicate that HDAC6 and SQSTM1/p62 can independently bind ISG15 to recognize ISG15 and ISG15conjugates directly or indirectly. ISG15-aggregates or aggresomes would be formed through 15 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 FIGURE 3. ISG15 up-regulation of aggregate formation of p62 (a) Purified recombinant ISG15 proteins were pulled down with glutathione-Sepharose beads bound purified recombinant GST-tag fusion p62 or control GST and analyzed as indicated. (b) Schematic inducible co-expression vectors of ISG15 and the core enzyme genes, ISG15 E1 UbE1L, E2 UbcH8 and E3 Herc5 using self-cleavage peptide sequences (P2A) under the control of the tet-on promoter (Ptet-on). The details of the vector constructs are described in Materials and Methods. (c) U251.ISG7-8 exhibited overexpression of ISG15 and ISG15 conjugation to cellular proteins in the presence of doxycycline (Dox; 100 ng/ml, 24h) prior to IB with anti-ISG15 antibody. (d) Immunoblots of detergent soluble (Soluble) and –insoluble (Insoluble) fractions of U251.ISG7-8 cells with and without Dox treatment (100 ng/ml, 24h) in the presence or absence of pepstatin A and E64d (PepA/E64d; each 20 µg/ml, 4h), were probed for the indicated proteins. (e) 293.ISG8 exhibited overexpression of ISG15 and conjugation in the presence of Dox (200 ng/ml, 24h). (f) 293-ISG8 cells were transiently transfected with FLAG-tagged p62 or control empty vector and were treated with MG132 (40 µM, 6h). Anti-FLAG immunoprecipitated (IP) proteins and input cell lysates were immunoblotted (IB) with anti-FLAG and anti-ISG15 antibodies. g, Immunoblots of 0.5% Triton X100 detergent soluble (Soluble) and –insoluble (Insoluble) lysate fraction of 293.ISG8 cells treated with Dox (200 ng/ml, 24h) in the presence or absence of MG132 (6h) at indicated doses prior to collecting the cells, were probed for the indicated proteins. ISG15 associates with HDAC6 and p62 oligomerization and recruitment of autophagosomes-bound LC3 by p62. The [roteasome inhibitor, MG132, induces and enhances this aggregate formation and accumulation in the insoluble fraction d. The deacetylase activity of HDAC6 can mediate lysosomal fusion of ISG15-containing aggregates for clearance and this is inhibited by the specific HDAC6 inhibitor, Tubacin. Pepstatin A and E64d (pepA/E64d) inhibit lysosomal digestion of peptide contents. . Downloaded from http://www.jbc.org/ by guest on December 4, 2014 16 ISG15 associates with HDAC6 and p62 Figure 1 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 17 ISG15 associates with HDAC6 and p62 Figure2 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 18 ISG15 associates with HDAC6 and p62 Figure 3 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 19 ISG15 associates with HDAC6 and p62 Figure 4 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 20 ISG15 associates with HDAC6 and p62 Figure 5 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 21 ISG15 associates with HDAC6 and p62 Figure 6 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 22 ISG15 associates with HDAC6 and p62 Figure 7 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 23 ISG15 associates with HDAC6 and p62 Downloaded from http://www.jbc.org/ by guest on December 4, 2014 24
© Copyright 2024