FEBS Letters 580 (2006) 4784–4792

TRIM11 binds to and destabilizes a key component of the activator-mediated cofactor complex (ARC105) through the ubiquitin–proteasome system

Hideaki Ishikawa, Hiroyuki Tachikawa1, Yutaka Miura, Nobuhiro Takahashi* Department of Bioengineering, Applied Life Science, United Graduate School of Agriculture, Tokyo University of Agriculture and Technology, 3-5-8 Saiwai-cho, Fuchu-shi, Tokyo 183-8509, Japan

Received 16 May 2006; revised 3 July 2006; accepted 19 July 2006

Available online 4 August 2006

Edited by Ivan Sadowski

RET, or B-raf, respectively [1–3]. Some of the TRIM family Abstract TRIM11 is a member of the tripartite-motif-contain- ing family and is known to destabilize humanin, an inhib- members form homo- and/or heterooligomers and function itor of Alzheimer-like neuronal insults. In this study, we in multi-protein complexes [4]. demonstrate that TRIM11 interacts with activator-recruited TRIM11 (with 468 amino acid residues) is a typical RBCC cofactor 105-kDa component (ARC105) that mediates chroma- protein and forms homooligomers [5]. A green fluorescent pro- tin-directed activation and is a key regulatory tein (GFP)-TRIM11 fusion protein localizes to both the cyto- factor for transforming growth factor b (TGFb) signaling. Co- plasm and nucleus in mammalian cells (http://www.tigem.it/ expression of TRIM11 increased ARC105 degradation but a TRIM/). TRIM11 also interacts with humanin, a neuroprotec- proteasome inhibitor suppressed this. Co-expression of TRIM11 tive peptide that specifically suppresses Alzheimer’s disease-re- and ARC105 also increased ubiquitination of ARC105. In addi- lated neurotoxicity, and TRIM11 is proposed to help regulate tion, TRIM11 suppressed ARC105-mediated transcriptional intracellular levels of humanin through ubiquitin-mediated pro- activation induced with TGFb in a reporter assay. These results suggest that TRIM11, with the ubiquitin–proteasome pathway, tein degradation in neural tissues [6]. Humanin interacts with regulates ARC105 function in TGFb signaling. apoptotic factor Bax and induces antiapoptotic effects by inhib- Ó 2006 Federation of European Biochemical Societies. Pub- iting translocation of Bax to the mitochondria [7]; thus, lished by Elsevier B.V. All rights reserved. TRIM11 may regulate apoptosis by controlling humanin stability. TRIM11 mRNA is ubiquitously expressed in adult Keywords: TRIM/RBCC; Ubiquitin-proteasome system; mouse tissues, including the thymus, testis, spleen, and liver, Transcription mediator; TGFb and is highly expressed in embryonic kidney and nerve systems (http://www.tigem.it/TRIM/); however, the function of TRIM11 remains explored. In this study, we identified potential TRIM11-interacting 1. Introduction using a combination of pull-down assays and mass- spectrometry-based analysis and found that activator-recruited The tripartite motif-containing (TRIM) family is a subgroup cofactor 105-kDa component (ARC105) is a novel TRIM11- of the really interesting new gene (RING) finger-containing interacting protein. We also showed evidence that ARC105- protein family. TRIM proteins contain a RING finger, a B- mediated transcriptional activation induced by transforming box, and a coiled-coil domain, making them members of the growth factor b (TGFb) signaling is regulated by TRIM11 RING B-box coiled-coil (RBCC) protein family as well. The through an ubiquitin-mediated degradation pathway. TRIM proteins, promyelocytic leukemia protein (PML, TRIM19), ret finger protein (RFP, TRIM27), and transcrip- tion intermediary factor 1 (Tif1, TRIM24) have been well- 2. Materials and methods studied and shown to be oncogenic when fused by chromo- somal translocation to retinoic acid receptor a (RARa), 2.1. Materials Anti-FLAG-M2, anti-HA, anti-b-tubulin, anti-GFP and anti-mouse *Corresponding author. Fax: +81 42 367 5709. IgG Cy3-conjugated antibody was obtained from Sigma–Aldrich (To- E-mail address: [email protected] (N. Takahashi). kyo, Japan). Anti-GAPDH antibody was obtained from Ambion (Aus- tin, TX, USA). Anti-Myc rabbit polyclonal antibody was purchased 1 Present address: Graduate School of Agricultural and Life Sciences, from BioVision Research Products (Mountain View, CA, USA). University of Tokyo, Japan. Anti-mouse IgG and anti-rabbit IgG alkaline phosphatase-conjugated antibody was obtained from Cell Signaling Technology (Beverly, MA, Abbreviations: TRIM, tripartite motif-containing protein; RBCC, R- USA). MG132 was obtained from Calbiochem (La Jolla, CA, USA). ING B-box coiled-coil; RING, really interesting new gene; ARC105, recombinant TGF-b1 was obtained from Nakalai Tesque (Kyo- activator recruited cofactor 105-kDa component; GFP, green fluores- to, Japan). p3TP-Lux vector was kindly provided by Dr. Miyazono. cent protein; HEK293, human embryonic kidney 293 cells; Ub, ubiq- uitin; E2s, ubiquitin conjugating ; TGFb, transforming growth 2.2. Cloning and construction for FLAG-tagged, HA-tagged, and factor b; MALDI-TOF/MS, matrix assisted laser desorption/ioniza- Myc-tagged fusion proteins tion time of flight mass spectrometry; PMSF, phenylmethylsulfonyl RT-PCR was performed using the Super Script II kit (Invitrogen, fluoride; PMF, peptide mass fingerprint; CMV, cyto megalo virus Carlsbad, CA, USA) according to the manufacturer’s protocol using

0014-5793/$32.00 Ó 2006 Federation of European Biochemical Societies. Published by Elsevier B.V. All rights reserved. doi:10.1016/j.febslet.2006.07.066 H. Ishikawa et al. / FEBS Letters 580 (2006) 4784–4792 4785 total mRNA prepared from human embryonic kidney (HEK) with 10% heat-inactivated fetal calf serum in a humidified atmosphere 293EBNA cells (Invitrogen). cDNA for C-terminally FLAG-tagged of 95% air and 5% CO2 [8]. Transfection was performed by either the TRIM11 (TRIM11-FLAG) (GenBank Accession: AF327056) and calcium phosphate method or using LipofectAMINE (Invitrogen) [8]. FLAG-tagged ARC105 (ARC105-FLAG) (GenBank Accession: 0 BC013985) were obtained by PCR with the following primer sets; 5 - 2.5. Pull-down analysis GAAGAAAAGCTTCACCATGGGAGCCGCCCCCGACCTGTC- Cells were harvested at 48 h after transfection, washed twice with ice- 0 0 CACC-3 and 5 -CAAGAATTCTTACTTATCGTCGTCATCCTTG- cold PBS, and centrifuged at 1500 rpm for 5 min at 4 °C. Cell pellets 0 TAATCGGCACCAGAACCCTGGG GAGCCAGGGTGTCCCC-3 were resuspended in lysis buffer (50 mM Tris–HCl, pH 8.0, 150 mM 0 for TRIM11, and 5 -GAAGAAGGATCCCACCATGGACGTTTC- NaCl, containing 0.5% IGEPAL-CA630, protease inhibitors; 0.1 mM 0 0 CGGGCAAGAG-3 and 5 -CAAGAATTCTTACTTATCGTCGT- phenylmethylsulfonyl fluoride (PMSF), 2 lg/ml aprotinin, 2 lg/ml leu- CATCCTTGTAATCGGCACCAGAACCGGCGGCTGAGAGGC- peptin and 2 lg/ml pepstatin) and incubated on ice for 30 min. Cell deb- 0 AGGCCTG-3 for ARC105. The PCR product was cloned into the ris was removed by centrifugation at 15000 rpm for 30 min at 4 °C. HindIII/EcoRI sites of the expression vector pcDNA3.1(+) for Anti-FLAG-M2-conjugated agarose beads (Sigma–Aldrich, Tokyo, TRIM11 and into the BamHI/EcoRI sites for ARC105. Using the Japan) were added to the cleared lysate, and the mixture was rotated cloned FLAG-tagged TRIM11 as template, TRIM11 lacking the for 4–6 h at 4 °C. The anti-FLAG-M2 agarose beads were collected, 0 FLAG tag was amplified by PCR with a primer set, 5 - washed five times with lysis buffer and once with detergent-free lysis buf- GAAGAAAAGCTTCACCATGGGAGCCGCCCCCGACCTGTC- fer, and then bound proteins were eluted with 500 lg/ml FLAG peptide. CACC-30 and 50-CAAGAATTCCCTGGGGAGCCAGGGTGTCC- CC-30. The PCR product was then ligated into the HindIII and EcoRI sites of the pEGFP-N1 vector (Invitrogen) to generate GFP-tagged 2.6. SDS–PAGE and Western blotting TRIM11 (TRIM11-GFP). Using the cloned FLAG-tagged ARC105 SDS–PAGE gels were subjected to Western blotting after electro- as template, HA-tagged ARC105 was amplified by PCR with the pri- transfer to PVDF membranes according to methods described previ- mer set, 50-GAAGAAGGATCCCACCATGGACGTTTCCGGGCA- ously [8]. The membranes were blocked with 5% non-fat dried skim AGAG-30 and 50-CAAGAATTCTTAGGCGTAGTCCGGGACGT- milk in TBST (50 mM Tris–HCl, pH 7.4, 0.1% (w/v) Tween 20, CATATGGGTAGGCACCAGAACCGGCGGCTGAGAGGCAG- 150 mM NaCl) for 30 min, and incubated with the appropriate pri- GCCTG-30. The PCR product was then ligated into the BamHI and mary antibody for 1 h. The membranes were rinsed three times with EcoRI sites of the pcDNA3.1(+) vector to generate ARC105-HA. TBST for 10 min, incubated with secondary antibody conjugated with Myc-tagged TRIM11 (TRIM11-Myc) was obtained by PCR amplifica- alkaline phosphatase for 1 h and washed four times with TBST. Stain- tion of the cloned FLAG-tagged TRIM11 and a set of primers; 50- ing was performed in NBT/BCIP solution, prepared by a (1:50) dilu- GAAGAAAAGCTTCACCATGGGAGCCGCCCCCGACCTGTCC- tion of NBT/BCIP stock solution (Roche, Diagnostics, Manheim, ACC-30 and 50-CAAGAATTCTTACAAGTCCTCTTCAGAAAT- Germany) with alkaline phosphatase buffer (100 mM Tris–HCl, pH GAGCTTTTGCTCGGCACCAGAACCCTGGGGAGCCAGGGT- 9.5, 100 mM NaCl, and 50 mM MgCl2). GTCCCC-30. Amplified product was ligated into the HindIII and EcoRI sites of the pcDNA3.1(+) vector. All constructs were verified 2.7. Matrix assisted laser desorption/ionization time of flight mass by DNA sequence analysis. spectrometry (MALDI-TOF/MS) and peptide mass fingerprint (PMF) analysis 2.3. Construction of TRIM11 deletion mutants Each of protein staining bands on an SDS–PAGE gel was excised The domain structure of TRIM11 was analyzed with Pfam (http:// and subjected to in-gel protease digestion as described previously [9]. www.sanger.ac.uk/Software/Pfam/) and Smart (http://smart.emblhei- The peptides generated were recovered with ZipChip and analyzed delberg.de/smart/) Sequence Analyzing Tools. A coiled-coil-specific by the PMF method using a PE Biosystems MALDI-TOF mass spec- analyzing tool (http://www.ch.embnet.org/software/COILS_- trometer (Voyager DE-STR, PE Biosystems, Foster City, CA). Using form.html) was used to determine the location of the coiled-coil region. 50 ppm mass accuracy, the peptide masses were compared to the NCBI The expression plasmids for TRIM11-FLAG deletion mutants were nr database using the database fitting program MS-Fit (http://prospec- constructed using PCR with pcDNA3.1(+)-TRIM11-FLAG as tem- tor.ucsf.edu/ucsfhtml4.0/msfit.htm), and protein identification was car- plate. DNA fragments for the TDN deletion series were amplified by ried out using criteria described previously [8]. PCR with a combination of the TRIM11 reverse cloning primer de- scribed above and the forward primer; 50-GAAGAAAAGCTTCAC- 2.8. Immunocytochemistry CATGGTCCCGCAGGGCGTGTGCCC-30 for TDN-R, 50-GAAG- Human 293EBNA cells were cultured on collagen-coated culture AAAAGCTTCACCATGGGACTGCAGGACGCGGCCGAAGAC- glass slides (Biocoat, Becton Dickinson Labware) and transfected with 30 for TDN-RB, or 50-GAAGAAAAGCTTCACCATGGGACG- expression plasmid DNA using LipofectAMINE. The transfected and AGGGGACGTGACCTTGGAC-30 for TDN-RBCC. The amplified intact cells were fixed with 3.7% formaldehyde in PBS. After washing DNA fragments were ligated into the HindIII and EcoRI sites of with PBST (PBS containing 0.05% (w/v) Tween 20), the cells were per- pcDNA3.1(+). meabilized with 0.1% (w/v) Triton X-100 for 5 min, and treated with DNA fragments for the TDC deletion series were amplified by PCR 3% non-fat dried skim milk in PBS at room temperature for 30 min. with a combination of the forward primer 50-GAAGGTACC- The cells were then incubated with the appropriate primary antibody GGTGCCGATTACAAGGATGACGACGAT-30 and the reverse for 1 h at room temperature. After a wash with PBST, the cells were primers 50-GAAGGTACCGGTGAAGCGCTCCTGGCCCAG-30 for further incubated with fluorochrome-conjugated secondary antibody TDC-SP, 50-GAAGGTACCAAACCTCCGCAGTGTCTCTAC-30 for for 1 h at room temperature. After incubation, the slides were washed TDC-SASP, and 50-GAAGGTACCCAGCGGCCGCACGCGGTG- and mounted with Vectashield (Vector Laboratories, Burlingame, CA, CG-30 for TDC-CCSASP. The amplified fragments were digested with USA). Fluorescence images were visualized with an Axiovert 200 M KpnI and self-ligated. microscope (Carl Zeiss, Germany). Exogenously expressed FLAG- For the construction of Myc-tagged TDN-R or TDN-RB deletion tagged ARC105 was visualized with anti-FLAG-M2 (1:500) and mutants, the DNA fragment encoding the N-terminal region of anti-mouse IgG conjugate Cy3 antibody (1:1000). Native ARC105 TRIM11 was removed by cleavage at the HindIII and BamHI site of was visualized with anti-ARC105 (PAQ6C9, 1:50) and anti-mouse the pcDNA3.1(+) plasmid containing the Myc-tagged TRIM11, and IgG conjugate Cy3 antibody (1:1000). the plasmid was ligated to the DNA fragment encoding the N-terminal region of TDN-R or TDN-RB that had been cleaved from the FLAG- 2.9. Establishment of cell lines for stable expression of ARC105 tagged TDN-R or TDN-RB pcDNA3.1(+) plasmid at the HindIII and HEK293 cells were transfected with ScaI-linearized pcDNA3.1(+)- BamHI site. ARC105-HA plasmid to establish strains with stable expression of the HA-tagged ARC105 fusion protein. The transfected cells were selected 2.4. Cell culture and transfection for at least two weeks with 500 lg/ml G418 (Gibco, Grand Island, NY, Human embryonic kidney 293 (HEK293) cells and HEK293 cells USA). The resulting G418-resistant cells were seeded on 96-well plates that express the Epstein-Barr virus nuclear antigen (293EBNA) were and incubated for at least one week for secondary clone selection. Iso- maintained according to standard methods in Dulbecco’s modified Ea- lated colonies were propagated in 24-well plates and the protein expres- gle’s medium (Nissui Pharmaceuticals, Tokyo, Japan) supplemented sion level of ARC105-HA was monitored by Western blotting. 4786 H. Ishikawa et al. / FEBS Letters 580 (2006) 4784–4792

2.10. Destabilization assay for ARC105 Three expression vectors, pcDNA3.1(+)-ARC105-HA, pcDNA3.1(+)-TRIM11-FLAG, and pEGFP-N1-GFP, were mixed in a ratio of 8:1:1 and transfected into 293EBNA cells. After 24 h cultiva- tion, the transfected cells were incubated further with or without 10 lM MG132 for 12 h. Whole-cell extract (100 lg) was subjected to Western blot analysis with anti-FLAG, anti-HA, anti-tubulin, and anti-GFP antibodies as described above (SDS–PAGE and Western blotting). As a control or antagonistic experiment, pcDNA3.1(+)- empty vector or pcDNA3.1(+)-TDN-R-FLAG was used, respectively, in place of pcDNA3.1(+)-TRIM11-FLAG. For analysis of the dose- dependent effect of TRIM11 on ARC105 stabilization, a mixture of pcDNA3.1(+)-empty vector and pcDNA3.1(+)-TRIM11-FLAG in a ratio of 1:0, 3:1, 1:1 or 0:1 was transfected with pcDNA3.1(+)- ARC105-HA and pEGFP-N1-GFP into 293EBNA cells. The total amount of vector in each experiment was adjusted to 0.625 lg. For the effect of the TRIM11 domain mutant on the stabilization of ARC105, pcDNA3.1(+)-TDN-R-FLAG was used instead of pcDNA3.1(+)-TRIM11-FLAG.

2.11. In vivo ubiquitination assay A mixture of expression vectors, pCGN-HA-Ub (0.375 lg), pcDNA3.1(+)-ARC105-FLAG (0.75 lg) and, pcDNA3.1(+)-empty vector (0.1 lg) or pcDNA3.1(+)-TRIM11-Myc (0.1 lg), was transfec- ted into 293EBNA cells in 3.5-cm dishes, and expressed for 36 h. The cultured cells were incubated further with or without 10 lM MG132 for 12 h. Collected cells were lysed in RIPA Buffer (50 mM Tris–HCl, pH 8.0, 150 mM NaCl, containing 0.1% SDS, 1.0% deoxy- cholate, 0.5% IGEPAL-CA630, and protease inhibitors; 0.1 mM PMSF, 2 lg/ml aprotinin, 2 lg/ml leupeptin and 2 lg/ml pepstatin) and incubated on ice for 30 min. Cell lysates were cleared by centrifu- gation at 15000 rpm for 30 min at 4 °C and the supernatants were sub- jected to FLAG pull-down analysis.

2.12. Reporter assay for TGFb-ARC105 signaling pathway A chimeric intron region containing Renilla Luciferase fragment was excised from pRL-TK vector (Promega) with HindIII and BamHI and ligated into the HindIII and BamHI sites of pcDNA3.1(+) to construct CMV driven internal control vector, pcDNA3.1(+)-Chi- m.Int.-RL. 293EBNA cells were plated in 24 wells plate, and cultured until cell density became 80–90%. At these points, a plasmid mixture of p3TP-Lux vector as a TGF-b specific reporter plasmid [24], pcDNA3.1(+)-Chim.Int.-RL as an internal control plasmid and a mix- ture of pcDNA3.1(+)-ARC105-FLAG, pcDNA3.1(+)-TRIM11-Myc plasmid, and pcDNA3.1(+) empty vector in the ratio indicated in Fig. 6 was transfected using Lipofectamine 2000 reagent (Invitrogen) according to manufacturer’s protocol. The total amount of vector in each experiment was adjusted to 0.626 lg. Ten hours after transfec- tion; 40 ng/ml TGF-b1 in OPTI-MEM (Invitrogen) was added to each Fig. 1. Association of TRIM11 with ARC105. (A) SDS–PAGE of well to the final concentration of 5 ng/ml. After another 6 h incubation, TRIM11-FLAG-associated proteins. Pull-down analysis of TRIM11- cells were harvested and luciferase activity was measured using Prome- FLAG in transfected cells. Protein bands were detected by silver ga’s Dual Luciferase Assay Kit and luminometer (Lumicounter 700; staining. The protein bands identified by MS analysis after in-gel Microtech Nichi-on, Chiba, Japan). protease digestion are indicated. An asterisk indicates a protein band reproducibly observed in the pull-down assay. Molecular weight markers are shown at left. (B) Pull-down analysis of ARC105-FLAG 3. Results with TRIM11-GFP in co-transfected cells. IP indicates that immuno- precipitation of the indicated protein as affinity bait was done. Protein bands were detected by Western blotting with anti-FLAG (1:10000) or 3.1. TRIM11 interacts with ARC105 anti-GFP (1:1000) antibody. +, plasmid containing a DNA fragment To identify possible binding partners of TRIM11 in the cell, encoding ARC105-FLAG, TRIM11-GFP, or GFP was transfected; , we expressed FLAG-tagged TRIM11 (TRIM11-FLAG) in hu- untransfected. Lanes 1 and 4; pcDNA3.1 vector without any insertion man embryonic kidney cells expressing the Epstein-Barr virus was transfected (control). An aliquot of whole-cell extract (50 lg) was loaded onto the WCE lanes 1–3. Molecular weight markers are shown nuclear antigen (293EBNA cells) and immunoprecipitated cell at left. lysates with anti-FLAG antibody. At least two protein bands that were not TRIM11 corresponding to the molecular weights a doublet pattern in SDS–PAGE analysis [10]. The larger of 90 kDa and 95 kDa on SDS–PAGE were reproducibly 95 kDa band may correspond to one of the doublet bands re- found associated with TRIM11-FLAG in at least three inde- ported. To further confirm the physical association between pendent experiments (Fig. 1A). Peptide mass fingerprint TRIM11 and ARC105, we co-transfected ARC105-FLAG (PMF) analysis identified the smaller band as ARC105 (mass and TRIM11-GFP into 293EBNA cells, immunoprecipitated matched 14/25, sequence coverage 21%, mean error the cell lysates with anti-FLAG antibody, and confirmed that 3.59 ppm, data tolerance 14 ppm, accession no. gi14276857: the immunoprecipitate contained TRIM11-GFP (Fig. 1B); molecular weight 82325). ARC105 has been reported to show thus, a reciprocal interaction was confirmed. H. Ishikawa et al. / FEBS Letters 580 (2006) 4784–4792 4787

3.2. TRIM11 and ARC105 co-localize mainly in the nucleoplasm predominantly in the nucleoplasm (Fig. 2B). We next exam- of the cell ined the cellular localization of endogenous ARC105 with We also examined the cellular localization of TRIM11 and anti-ARC105 antibody and confirmed that endogenous ARC105 to assess their specific association in vivo. When we ARC105 also localizes predominantly in the nucleus transfected either TRIM11-GFP or ARC105-FLAG into (Fig. 2C). Thus, the cellular localization of exogenously ex- 293EBNA cells and performed immunocytochemical analysis, pressed FLAG-tagged ARC105 was indistinguishable from we observed that TRIM11-GFP localized diffusely in the cyto- that of native ARC105. When TRIM11-GFP and ARC105- plasm and the nucleoplasm as reported previously by Rey- FLAG were co-transfected, we noted that they co-localized mond et al. ([5], Fig. 2A), whereas ARC105-FLAG localized in the nucleoplasm of most of the co-transfected cells

Fig. 2. Cellular localization of TRIM11 and ARC105. (A) Localization of TRIM11-GFP in 293EBNA cells. DAPI, DAPI image showing the position of the nuclei; TRIM11-GFP, corresponding immunolocalization of exogenously expressed TRIM11; Merge, merge of TRIM11-GFP and DAPI. (B) Cellular localization of ARC105-FLAG. ARC105-FLAG, typical immunostaining pattern of exogenously expressed ARC105-FLAG; Merge, merge of ARC105-FLAG and DAPI staining; DIC, a transmitted image of transfected 293EBNA cells. (C) Immunostaining of endogenously expressed ARC105 in 293EBNA cells. ARC105: visualized image of native ARC105. Merge: merge of endogenous ARC105 and DAPI. (D) Double immunostaining of TRIM11-GFP and ARC105-FLAG in 293EBNA cells. Merge, merged image of TRIM11-GFP and ARC105-FLAG. (E,F) Nucleolar colocalization of TRIM11-GFP and ARC105-FLAG, scale bar indicates 10 lm. All immunocytochemical observation was done at 24 h after the transfection of the plasmid for expressing the protein indicated. 4788 H. Ishikawa et al. / FEBS Letters 580 (2006) 4784–4792

(Fig. 2D). This was somewhat contradictory to the result that TRIM11 was localized mainly in the cytoplasm without co- expression of ARC105 (Fig. 2A and D–F). We, therefore, examined a possibility that the expression of ARC105 induced re-location of TRIM11 from the cytoplasm to the nucleo- plasm. We constructed the cells that expressed stably TRIM11-GFP, confirmed that almost 100% of the cells were expressed TRIM11-GFP dominantly in the cytoplasm, and found that more than 70% of the ARC105-HA expressing cells showed the dominant nucleoplasmic localization of TRIM11- GFP (supplementary Fig. 1A and 1B). These results strongly suggest that the expression level of ARC105 regulates the nucleoplasmic localization of TRIM11. In addition, we ob- served that the two proteins co-localized in the nucleolus in less than 5% of the co-transfected cell population (Fig. 2E and F). These results suggest that the association between TRIM11 and ARC105 occur at least in the nucleoplasm and possibly in the nucleolus under some physiological conditions, and im- ply that ARC105 regulates the nucleoplasmic and/or nucleolar localization of TRIM11 either through the ability of ARC105 to translocate TRIM11 from the cytoplasm to the nucleus or to retain TRIM11 in the nucleoplasm.

3.3. The coiled-coil and C-terminal domains of TRIM11 are required for association with ARC105 We next examined which TRIM11 domains are responsible for interacting with ARC105. Based on the domain structure of TRIM11 (containing a RING finger, a B-Box, a coiled-coil domain, a SPRY-associated and a SPRY domain), we first constructed five FLAG-fused domain mutants, TDN-R (87- 468), TDN-RB (128-468), TDN-RBCC (286-468), TDC-SP (1- 338) and TDC-SASP (1-285) (Fig. 3A) and expressed each of these in HEK293 cells stably expressing HA-tagged ARC105 (ARC105-HA). Each of the immunoprecipitates prepared with anti-FLAG-M2 beads was analyzed by Western blotting with anti-FLAG and anti-HA antibodies (Fig. 3B). ARC105 was found to be associated with two mutants, TDN-R (87-468) and TDN-RB (128-468), but not with the other mutants. In addition, we detected endogenous ARC105 in the immunopre- cipitates obtained from the 293EBNA cells transfected with the Fig. 3. Identification of ARC105-associating domains in the TRIM11 molecule. (A) Schematic representation of the domains of the full- TDN-R (87-468) or TDN-RB (128-468) mutants by Western length and truncated TRIM11 proteins. Hatched boxes represent the blot analysis with anti-ARC105 antibody, thereby demonstrat- RING finger, B-Box, coiled-coil, SPRY-associated and SPRY ing that the two mutants also associate with endogenous domains, as indicated. The amino acid residue numbers of TRIM11 ARC105 (Fig. 3C). For safety in case that the inability of and the deletion mutants are shown to the right of the corresponding constructs. A FLAG tag was added to the C-terminus of each peptide the other truncated mutants, TDN-RBCC, TDC-SP, TDC- (not indicated in the diagram). (B) Pull-down analysis of TRIM11 SASP, to bind to ARC105 were due to the delocalization of deletion mutants. Each of the deletion mutants was transfected into those mutants in the cell, we examined the cellular localization HEK293 cells stably expressing ARC105-HA, and pulled down with of those mutants by immunocytochemical analysis and showed anti-FLAG antibody-conjugated beads. Western blotting was per- that those truncated mutants localized in the nucleus as well as formed with either anti-HA (1:10000) or anti-FLAG (1:10000) antibody after SDS–PAGE separation of proteins associated with the cytoplasm similarly to full-length TRIM11 (supplementary FLAG-TRIM11 deletion mutants. The asterisks indicate TRIM11 Fig. 2). These results indicate that the coiled-coil, SPRY-asso- deletion mutant proteins as determined by calculation of their ciated and SPRY domains are required for binding of molecular weight. (C) Each of the deletion mutants was transfected TRIM11 to ARC105, whereas the RING finger and B-Box do- into 293EBNA cells and pulled down with anti-FLAG antibody- conjugated beads. Western blotting was performed with either anti- mains are not. The coiled-coil, SPRY-associated and SPRY ARC105 (1:100) or anti-FLAG antibody after SDS–PAGE separation domains are also reported to interact with humanin [6]. of proteins associated with FLAG-TRIM11 deletion mutants. Aster- isks show TRIM11 deletion mutants as determined by calculation of their molecular weight. 3.4. TRIM11 destabilizes ARC105 probably through the ubiquitin–proteasome degradation pathway The RING finger domain is proposed to be a consensus do- considered to be ubiquitin ligase E3s [12–14]. In fact, some main for binding to ubiquitin conjugating enzymes [11]; TRIM family proteins interact with ubiquitin ligase E2 and accordingly, proteins containing a RING finger domain are have proved to be E3 ubiquitin ligases [15–18]. Given the re- H. Ishikawa et al. / FEBS Letters 580 (2006) 4784–4792 4789 port that TRIM11 binds to and destabilizes humanin as a probable E3 ubiquitin ligase [6], we investigated whether TRIM11 destabilizes ARC105 through the ubiquitin–protea- some degradation pathway. We first examined whether co- expression of TRIM11 destabilizes ARC105 and, if so, whether the proteasome inhibitor MG132 inhibits the TRIM11-induced ARC105 destabilization. Co-expression of TRIM11-FLAG decreased the amount of ARC105; further- more, MG132 treatment reversed the effect of TRIM11 on ARC105 levels (Fig. 4A). Current report showed that the application of MG132 induces specific gene through CHOP , which binds a consensus sequence, 50- GNGKATTGCNNYY-30 (ex. GAGGATTGCGATC) [19]. Although we did not find such consensus sequence in the expression vector with CMV promoter we used, we examined a possibility that MG132 induced ARC105 gene expression and caused the increase of ARC105 in our experiment. How- ever, we did not detect any significant increase of the expres- sion of the ARC105 mRNA (supplementary Fig. 3). We also examined the effect of expression of TRIM11-GFP on the ARC105-HA by immunocytochemical method, and found that the staining intensity of ARC105-HA was certainly reduced in the cells expressed TRIM11-GFP at higher level when com- pared with in the cells expressed TRIM11-GFP at lower level (supplementary Fig. 1B, indicated by arrows). Those results suggest that proteasomal degradation of ARC105 is acceler- ated by co-expression of TRIM11. Given that TRIM11 has a RING finger domain and is a possible E3 ubiquitin ligase, we also tested the possible involvement of the RING finger do- main in destabilization of ARC105 by co-expressing ARC105 and a TRIM11 domain mutant lacking the RING finger do- main (TDN-R). We found that expression of this mutant pre- vented degradation of ARC105, and that MG132 had little effect on the stability of ARC105 in the TDN-R-transfected cells (Fig. 4A, lanes 7 and 8). In addition, we transfected the ARC105-HA plasmid with a mixture of pcDNA3.1 vector Fig. 4. Destabilization of ARC105 by co-expression of TRIM11. (A) and full-length TRIM11 plasmid or a mixture of pcDNA3.1 Effect of TRIM11 expression on ARC105 stability examined by an immunoblot. Each of the antibodies used for immunoblot is indicated vector and the TDN-R deletion mutant plasmid to examine next to the abbreviation IB. ARC105-HA was co-transfected with dose dependent effect of TRIM11 or TDN-R on degradation pcDNA3.1(+)-empty vector (lanes 3 and 4), TRIM11-FLAG (lanes 5 of ARC105. We found that full-length TRIM11 dramatically and 6), or TDN-R-FLAG (TDN-R; lanes 7 and 8) with (+) or without decreased expression of ARC105 (Fig. 4B). Although treat- () MG132. Lanes 1 and 2 show whole-cell extracts of cells without l ment with MG132 could restore ARC105 expression at low any expression vector. MG132 (10 M) was added 24 h post transfection (+) and incubated for 12 h. An aliquot of 100 lgof levels of full-length TRIM11 plasmid, it could not completely whole-cell extract was loaded in each lane. Expression of GFP from restore ARC105 expression at higher doses (Fig. 4B). On the pEGFP-N1-GFP was used as a control for transfection. (B) Dose- other hand, the TDN-R mutant showed no effect or somewhat dependent effect of TRIM11 on ARC105 stability. Expression vector elevated ARC105-HA levels regardless of MG132 treatment containing TRIM11-FLAG was mixed with pcDNA3.1(+)-empty vector in the ratios indicated, and co-transfected with ARC105-HA (Fig. 4C). We also observed very similar results for the domain and GFP (as a transfection control). MG132 (10 lM) was added 24 h mutant TDN-RB that lacks the RING finger and B-Box post transfection (+) and incubated for 12 h. Western blotting was domains but is still able to interact with ARC105 (data not performed with anti-HA, anti-FLAG, and anti-GFP antibodies as shown). Furthermore, we examined the effect of the other indicated at the right. An aliquot of 50 lg of whole-cell extract was truncated mutants, TDN-RBCC, TDC-SP, TDC-SASP, which loaded in each lane. (C) Dose-dependent effect of TDN-R on ARC105 stability. Expression vector containing TDN-R was mixed with lack RING finger domain or had no binding activity to pcDNA3.1(+)-empty vector in the ratios indicated, and co-transfected ARC105 (Fig. 3), on the expression level of ARC105. Cer- with ARC105-HA and GFP. MG132 (10 lM) was added 24 h post tainly, those mutants did not have any significant effect on transfection (+) and incubated for 12 h. Western blotting was that (supplementary Fig. 4). These results indicate that the performed with anti-HA (1:10000), anti-FLAG (1:10000), anti-GFP (1:1000), and anti-tubulin (1:10000) antibodies as indicated at the RING finger domain and the ARC105-binding domains right. are required for destabilizing ARC105. In order to examine the effect of TRIM11 expression on ubiquitination of ARC105, we transfected HA-tagged ubiquitin (HA-Ub) in the immunoprecipitates by Western blotting with anti-HA addition to ARC105-FLAG and Myc-tagged TRIM11 antibody (Fig. 5). Poly-ubiquitination of ARC105-FLAG (TRIM11-Myc) into 293EBNA cells, performed a pull-down was observed in all ARC105-transfected cells, regardless of assay with FLAG antibody-conjugated beads and analyzed the expression of TRIM11-Myc (Fig. 5); however, the degree 4790 H. Ishikawa et al. / FEBS Letters 580 (2006) 4784–4792

Fig. 5. Effect of TRIM11 expression on ARC105 ubiquitination. +* indicates that pcDNA3.1(+)-empty vector was co-transfected to keep constant the total amount of vector added to the cells. Pull-down assays were performed with anti-FLAG antibody-conjugated beads followed by Western blotting with the antibodies indicated. WCE; aliquots of whole cell extract of 293EBNA cells were loaded on to SDS–PAGE gel. GAPDH was used as a loading control. Western blotting was performed with anti-HA (1:10000), anti-FLAG (1:10000), anti-Myc (1:100), and anti-GAPDH (1:10000) antibodies as indicated at the right.

of poly-ubiquitination was enhanced by the co-expression of TRIM11-Myc and incubation with MG132 (Fig. 5). In addi- tion, anti-FLAG antibody detected very weak bands corre- Fig. 6. Effect of TRIM11 expression on ARC105 function mediated by sponding to some poly-ubiquitinated forms of ARC105 in TGFb signaling. Expression vector encoding ARC105-FLAG was mixed with pcDNA3.1(+)-empty vector or with pcDNA 3.1(+)- the TRIM11-Myc-expressing cells (Fig. 5). These results sug- TRIM11-Myc plus pcDNA3.1(+)-empty vector, and adjusted the gest that TRIM11 destabilizes ARC105 through the ubiqui- total amount of the vectors to 0.5 lg. A mixture of reporter plasmid, tin–proteasome degradation pathway. 125 ng of p3TP-Lux plasmid (as TGFb reporter vector) and 1.25 ng of We noted that there was a band, whose size was a little bit pcDNA3.1 (+)-Chim.Int.-RL plasmid (as internal control vector) were co-transfected with those. The values of relative luciferase activity higher than that of ARC105-FLAG in ARC105-untransfected measured are normalized to those obtained from TGFb untreated samples (Fig. 5, lanes 1 and 2). This band was also observed in control. The vertical line within each histogram indicates the means the ARC105-transfected samples when the amounts of the and standard deviations of the triplicated experiments. , untransfec- loading samples for SDS–PAGE are reduced (supplementary ted or untreated; +, treated with TGFb. Western blotting was Fig. 5). We have identified the protein corresponding to this performed with anti-FLAG (1:10000), anti-Myc (1:100), and anti- GAPDH (1:10000) antibodies as indicated at the right. band as kinesin-like spindle protein (HKSP) by mass-based analysis after in-gel-protease digestion (data not shown). This undesired association of HKSP protein and several other pro- teins with FLAG-antibody had been observed when the anti- conventional reporter assay. Using p3TP-Lux reporter plas- FLAG antibody beads were used for immunoisolation [8,20]. mid, we confirmed that the transcriptional activity was in- The extent of the association varied depending on the lots of creased in proportion to the amount of the ARC105 the anti-FLAG antibodies purchased because of unknown rea- expression vector cotransfected under TGFb stimulation son; we had reported this problem previously [20]. Because the (Fig. 6, lanes 1–4, supplementary Fig. 6). On the other hand, size of HKSP is very similar to ARC105, the HKSP and the expression of TRIM11 reduced the effect of ARC105 on ARC105 bands overlapped each other in the ARC105-trans- the TGFb-induced transcriptional activity in a dose-dependent fected samples shown in Fig. 5. manner (Fig. 6, lanes 5 and 6). We also examined the expres- sion levels of ARC105 and TRIM11, and showed that they were consistent with the luciferase activities obtained by using 3.5. TRIM11 suppresses ARC105 mediated transcriptional reporter assay (Fig. 6, upper inset). These results suggest activation induced with TGFb strongly that TRIM11 regulates ARC105 function induced Given the fact that ARC105 has a role in linking TGFb/ by TGFb signaling through ubiquitin–proteasome pathway, Activin/Nodal/Smad2/3 signaling to ARC complex-dependent though we cannot exclude a possibility at present that transcription [24], we next examined the effect of TRIM11 on TRIM11 may have the targeted degradation activity on the ex- TGFb-induced ARC105-dependent transcription by using pressed luciferase itself. H. Ishikawa et al. / FEBS Letters 580 (2006) 4784–4792 4791

4. Discussion nucleoplasm through the interaction with the ARC105. A sim- ple scenario of this is that TRIM11 can shuttle between the In this study, we showed that TRIM11 interacts with cytoplasm and the nucleoplasm. In either case, our results sug- ARC105 through a region containing a coiled-coil, a SPRY- gest the presence of unknown regulatory mechanism by which associated domain and a SPRY domain and causes the desta- TRIM11 regulates a process that leads TGFb/Activin/Nodal/ bilization of ARC105 through the RING finger domain. Deg- Smad2/3 signaling to ARC complex-dependent transcription. radation of ARC105 was suppressed by treatment with the Certainly, further investigation of those possibilities will open proteasome inhibitor MG132. In addition, we demonstrated up new insights for the function of TRIM11 as well as the reg- that truncated mutants of TRIM11 lacking the RING finger ulatory roles of ARC105 in ARC complex dependent tran- domain stabilized ARC105, and that poly-ubiquitination of scription. ARC105 increased upon expression of TRIM11. Given the TRIM11 also interacts with humanin, a neuroprotective fact that the RING finger domain of TRIM11 is required for peptide that specifically suppresses Alzheimer’s disease-related ARC105 destabilization and that TRIM11 is likely an ubiqui- neurotoxicity, and TRIM11 is proposed to help regulate intra- tin ligase E3 [6], these results suggest that the function of cellular levels of humanin through ubiquitin-mediated protein ARC105 is regulated by TRIM11 through the ubiquitin–pro- degradation in neural tissues [6]. Our result coupled with this teasome degradation pathway. report establishes that TRIM11 is the E3 ubiquitin ligase, ARC105 was identified initially as a phorbol ester 12-O-tet- and that ARC105 is the second substrate for this iden- radecanoylphorbol-13-acetate (TPA)-inducible gene-1 product tified to date, though we do not know the functional relevance (TIG1) in the human leukemia K562 cell line [21]. It is also between those substrates in their physiological roles. In conclu- known as positive cofactor 2 glutamine/Q-rich-associated pro- sion, the present study defines a unique function of TRIM11 in tein (PC2) [10]. ARC105 is a component of the ARC complex the TGFb signaling pathway linking to ARC complex-depen- that mediates -directed transcription activation, and dent transcription. has roles in basal transcription [22,23]. A recent report showed that ARC105 stimulates Activin/Nodal/Smad2 signaling in Acknowledgements: We thank Drs. Keiichi Nakayama and Masaki Xenopus laevis embryos, inducing axis duplication and Matsumoto for providing pCGN-HA-Ub, Drs. Kouhei Miyazono and Tetsuro Watabe for TGFb-specific p3TP-Lux reporter vector mesoendoderm differentiation, and enhances TGFb and Acti- and Dr. Michael Meisterernst for anti-ARC105 (PAQ6C9) antibody. vin response in human cells; thus, ARC105 links TGFb/Acti- This work was partially supported by a grant from Pioneer Research vin/Nodal/Smad2/3 signaling to ARC complex-dependent on Genome the Frontier, Ministry of Education, Culture, Sports, Sci- transcription [24]. Because TRIM11 suppresses the ARC105- ence and Technology of Japan, and partially by a grant-in-aid for sci- ence research, Japan Society for the Promotion of Science. dependent transcriptional activity induced by TGFb in the conventional reporter assay we used (Fig. 6), it is strongly sug- gested that TRIM11 is involved in a process that links TGFb/ Appendix A. Supplementary data Activin/Nodal/Smad2/3 signaling to ARC complex-dependent transcription. Our present results also suggest that this process Supplementary data associated with this article can be is the ubiquitination of ARC105 specifically targeted by found, in the online version, at doi:10.1016/j.febslet. TRIM11, leading to the proteasome-dependent degradation. 2006.07.066. Given the result that the increase of ARC105 expression in- duced the translocation of TRIM11 from the cytoplasm to the nucleus, it is likely that the proteasome-dependent degra- dation of ARC105 occurs in the nucleoplasm. At present there References are two possibilities in which cellular compartment the interac- [1] de The`, H., Lavau, C., Marchio, A., Chomienne, C., Degos, L. tion of TRIM11 with ARC105 takes place first; one is that and Dejean, A. (1991) The PML-RAR alpha fusion mRNA takes place in the cytoplasm and another is in the nucleoplasm. generated by the t(15;17) translocation in acute promyelocytic In the former case, the interaction of the two proteins leads to leukemia encodes a functionally altered RAR. Cell 66, 675–684. the translocation from the cytoplasm to the nucleoplasm, but [2] Takahashi, M. and Cooper, G.M. (1986) ret transforming gene will not cause the ubiquitination or poly-ubiquitination of encodes a fusion protein homologous to tyrosine . Mol. Cell. Biol. 7, 1378–1385. ARC105 at this process. Since the binding domain of TRIM11 [3] Le Douarin, B., Zechel, C., Garnier, J.M., Lutz, Y., Tora, L., to ARC105 is different from that of ubiquitin ligase, the inter- Pierrat, B., Heery, D., Gronemeyer, H., Chambon, P. and action between the two proteins may be regulated indepen- Losson, R. (1995) The N-terminal part of TIF1, a putative dently of the ubiquitination of ARC105 with TRIM11. The mediator of the ligand-dependent activation function (AF-2) of nuclear receptors, is fused to B-raf in the oncogenic protein T18. ability of TRIM11 to form homo-oligomer may be related to EMBO J. 14, 2020–2033. this function. If this is the case, ARC105 may regulate ARC [4] Peng, H., Feldman, I. and Rauscher 3rd., F.J. (2002) Hetero- complex-dependent transcription as a complex with TRIM11. oligomerization among the TIF family of RBCC/TRIM domain- It is very intriguing to speculate that TRIM11 dissociates the containing nuclear cofactors: a potential mechanism for regulat- transcriptional machinery working on a gene after the termina- ing the switch between coactivation and coexpression. J. Mol. Biol. 320, 629–644. tion of the transcription through the ubiquitin–proteasome [5] Reymond, A., Meroni, G., Fantozzi, A., Merla, G., Cairo, S., dependent degradation of ARC105. On the other hand, if Luzi, L., Riganelli, D., Zanaria, E., Messali, S., Cainarca, S., the interaction of TRIM11 takes place in the nucleoplasm after Guffanti, A., Minucci, S., Pelicci, P.G. and Ballabio, A. (2001) translocation of ARC105, ARC105 may functions as a nucle- The identifies cell compartments. EMBO J. 20, 2140–2151. oplasmic anchor of TRIM11. The increase of ARC105 expres- [6] Niikura, T., Hashimoto, Y., Tajima, H., Ishizaka, M., Yamagishi, sion will cause the increase of the presence of ARC105 in the Y., Kawasumi, M., Nawa, M., Terashita, K., Aiso, S. and nucleoplasm, leading to the accumulation of TRIM11 in the Nishimoto, I. (2003) A tripartite motif protein TRIM11 binds and 4792 H. Ishikawa et al. / FEBS Letters 580 (2006) 4784–4792

destabilizes humanin, a neuroprotective peptide against Alzhei- tional ARD1 (ADP-ribosylation factor domain protein 1). Proc. mer’s disease-relevant insults. Eur. J. Neurosci. 17, 1150–1158. Natl. Acad. Sci. USA 102, 1945–1950. [7] Guo, B., Zhai, D., Cabezas, E., Welsh, K., Nouraini, S., [16] Xu, L., Yang, L., Moitra, P.K., Hashimoto, K., Rallabhandi, P., Satterthwait, A.C. and Reed, J.C. (2003) Humanin peptide Kaul, S., Meroni, G., Jensen, J.P., Weissman, A.M. and D’Arpa, suppresses apoptosis by interfering with Bax activation. Nature P. (2003) BTBD1 and BTBD2 colocalize to cytoplasmic bodies 423, 456–461. with the RBCC/tripartite motif protein, TRIM5d. Exp. Cell Res. [8] Yanagida, M., Shimamoto, A., Nishikawa, K., Furuichi, Y., 288, 84–93. Isobe, T. and Takahashi, N. (2001) Isolation and proteomic [17] Urano, T., Saito, T., Tsukui, T., Fujita, M., Hosoi, T., Murama- characterization of the major proteins of the nucleolin-binding tsu, M., Ouchi, Y. and Inoue, S. (2002) Efp targets 14-3-3r for ribonucleoprotein complexes. Proteomics 1, 1390–1404. proteolysis and promotes breast tumour growth. Nature 417, 871– [9] Yanagida, M., Miura, Y., Yagasaki, K., Taoka, M., Isobe, T. and 875. Takahashi, N. (2000) Matrix assisted laser desorption/ionization- [18] Dupont, S., Zacchigna, L., Cordenonsi, M., Soligo, S., Adorno, time of flight mass spectrometry analysis of proteins detected by M., Rugge, M. and Piccolo, S. (2005) Germ-layer specification anti-phosphotyrosine antibody on two-dimensional gels of fibro- and control of cell growth by ectodermin, a Smad4 ubiquitin blast cell lysates after tumor necrosis factor-alpha stimulation. ligase. Cell 121, 87–99. Electrophoresis 21, 1890–1898. [19] Yoshida, T., Shiraishi, T., Nakata, S., Horinaka, M., Wakada, [10] Berti, L., Mittler, G., Przemeck, G.K., Stelzer, G., Gu¨nzler, B., M., Mizutani, Y., Miki, T. and Sakai, T. (2005) Proteasome Amati, F., Conti, E., Dallapiccola, B., Hrabe´ de Angelis, M., inhibitor MG132 induces death receptor 5 through CCAAT/ Novelli, G. and Meisterernst, M. (2001) Isolation and chara- -binding protein homologous protein. Cancer Res. 65 cterization of a novel gene from the DiGeorge chromosomal (13), 5662–5667. region that encodes for a mediator subunit. Genomics 74, 320– [20] Yanagida, M., Hayano, T., Yamauchi, Y., Shinkawa, T., 332. Natsume, T., Isobe, T. and Takahashi, N. (2004) Human [11] Lorick, K.L., Jensen, J.P., Fang, S., Ong, A.M., Hatakeyama, S. fibrillarin forms a sub-complex with splicing factor 2-associated and Weissman, A.M. (1999) RING fingers mediate ubiquitin- p32, protein arginine methyltransferases, and tubulins alpha 3 and conjugating enzyme (E2)-dependent ubiquitination. Proc. Natl. beta 1 that is independent of its association with preribosomal Acad. Sci. USA 96, 11364–11369. ribonucleoprotein complexes. J. Biol. Chem. 279, 1607–1614. [12] Fang, S., Jensen, J.P., Ludwig, R.L., Vousden, K.H. and [21] Abraham, S. and Solomon, W.B. (2000) A novel glutamine-rich Weissman, A.M. (2000) Mdm2 is a RING finger-dependent putative transcriptional adaptor protein (TIG-1), preferentially ubiquitin protein ligase for itself and p53. J. Biol. Chem. 275, expressed in placental and bonemarrow tissues. Gene 255, 389– 8945–8951. 400. [13] Fukuchi, M., Imamura, T., Chiba, T., Ebisawa, T., Kawabata, [22] Na¨a¨r, A.M., Beaurang, P.A., Zhou, S., Abraham, S., Solomon, M., Tanaka, K. and Miyazono, K. (2001) Ligand-dependent W. and Tjian, R. (1999) Composite co-activator ARC mediates degradation of Smad3 by a ubiquitin ligase complex of ROC1 and chromatin-directed transcriptional activation. Nature 398, 828– associated proteins. Mol. Biol. Cell 12, 1431–1443. 832. [14] Ohta, T., Michel, J.J., Schottelius, A.J. and Xiong, Y. (1999) [23] Mittler, G., Kremmer, E., Timmers, H.T. and Meisterernst, M. ROC1, a homolog of APC11, represents a family of cullin (2001) Novel critical role of a human mediator complex for basal partners with an associated ubiquitin ligase activity. Mol. Cell 3, RNA polymerase II transcription. EMBO Rep. 2, 808–813. 535–541. [24] Kato, Y., Habas, R., Katsuyama, Y., Na¨a¨r, A.M. and He, X. [15] Vichi, A., Payne, D.M., Pacheco-Rodriguez, G., Moss, J. and (2002) A component of the ARC/Mediator complex required for Vaughan, M. (2005) E3 ubiquitin ligase activity of the trifunc- TGFb/Nodal signalling. Nature 418, 641–646.