Transcription factor Sp3 as target for SUMOylation in vivo

A group of sequence-specific DNA-binding proteins related to the transcription factor Sp1 (specificity protein 1) has been implicated in the regulation of many different genes, since binding sites for these transcription factors (GC/GT boxes) are a recurrent motif in regulatory sequences of these ge...

Ausführliche Beschreibung

Gespeichert in:
Bibliographische Detailangaben
1. Verfasser: Rischitor, Grigore
Beteiligte: Renkawitz-Pohl, Renate (Prof. Dr.) (BetreuerIn (Doktorarbeit))
Format: Dissertation
Sprache:Englisch
Veröffentlicht: Philipps-Universität Marburg 2005
Schlagworte:
Online Zugang:PDF-Volltext
Tags: Tag hinzufügen
Keine Tags, Fügen Sie den ersten Tag hinzu!
Beschreibung
Zusammenfassung:A group of sequence-specific DNA-binding proteins related to the transcription factor Sp1 (specificity protein 1) has been implicated in the regulation of many different genes, since binding sites for these transcription factors (GC/GT boxes) are a recurrent motif in regulatory sequences of these genes. In contrast to the transcriptional activators Sp1 and Sp4, the ubiquitously expressed Sp3 protein can both activate and repress transcription. The complex activity of Sp3 depends on two glutamine-rich activation domains, similar to those found in Sp1 and Sp4, and, adjacent to these, on an inhibitory domain unique to Sp3. The critical lysine residue in the Sp3 inhibitory domain lies within a consensus motif (IK551EE) that targets proteins for SUMO modification. SUMO (small ubiquitin-related modifier) is covalently attached to lysine residues in target proteins via an isopeptide linkage in a multi-step process that is analogous to ubiquitination. The present work analyses various aspects of SUMO conjugation to Sp3 in vivo. Studying modification of Sp3 by SUMO is complicated by the existence of a number of Sp3 isoforms. Immunoblot analyses revealed four distinct Sp3 proteins, two slow migrating of more than 100 kDa and two fast migrating species. Seven to eight Sp3 bands appeared, when cells were lysed in denaturing conditions. The additional protein species represent SUMO modified Sp3 isoforms. Currently, it is not known whether the relative distribution of the different Sp3 isoforms is regulated. However, a significant shift towards the long isoforms of Sp3, however, is observed in Sp1-/- ES cells demonstrating that Sp3 isoform expression principally can change in vivo. In addition, this observation suggests that the long isoforms of Sp3 may take over Sp1 functions under Sp1 knockout conditions. When Sp3 is overexpressed along with SUMO1 and SUMO2 in cells in culture, attachment of both SUMO paralogues to Sp3 occurred with almost equal efficiency. Beside lysine 551 within the inhibitory domain, there are two other potential SUMOylation sites in Sp3 (VKQE at position 9 and IKDE at position 120). This study revealed that SUMOylation takes place exclusively at K551, present in all four isoforms. Visualization of endogenous Sp3 by immumofluorescence showed a sponge-like, diffuse appearance, located predominantly in the nucleus. Evolutionally closely related Sp family members Sp1 and Sp2 are also located in the nucleus and the subcellular localization patterns are similar to Sp3. Ectopic expression of SUMO1 fused to GFP (green fluorescent protein) led to the accumulation of this fusion protein within subnuclear “dots” or PODs (promyelocytic leukemia oncogenic domains), whereas endogenous Sp3 remained diffusely distributed throughout nuclei. In addition, the wild-type Sp3 isoforms and the SUMOylation-deficient mutants of Sp3were located in the nucleus exhibiting also a sponge-like, diffuse appearance. Analyzing the Sp3 expression in different cell lines and mouse organs revealed that the relative level of Sp3 modification by SUMO is not cell line or organ dependent. In addition, no variation in Sp3 expression pattern after serum starvation, serum induction and heat shock was observed. Ultraviolet radiation or Tumor Necrosis Factor alpha and Cycloheximide treatment of mammalian cells did not alter the SUMOylation level of Sp3 protein in our experimental conditions. A significant reduction in Sp3 SUMO modification was observed upon treatment with MG-132, a cell-permeable inhibitor of the proteasome. Possibly this proteasome inhibitor prevents proteasome degradation of SUMO specific isopeptidase, which subsequently remove the Sp3-SUMO moiety. PIAS1 (protein inhibitor of activated STAT) was previously cloned in a two-hybrid screen by using the inhibitory domain of Sp3. Moreover, it was shown that PIAS1 strongly enhances SUMO-modification of Sp3 in vitro and thus acts as an SUMO E3 ligase towards Sp3. PIAS1-associated proteins might confer substrate specificity towards Sp3 and other transcription factors and/or regulate PIAS1 activity in vivo. For the purification and identification of PIAS1-associated proteins, a number of C-terminal tagged expression plasmids were constructed for constitutive and inducible expression. The dual-tag affinity purification system established in this thesis work contains a small 15 amino acid artificial tag (BiotinTAG) that becomes biotinylated by the BirA ligase upon co-transfection of an appropriate expression construct. To enhance specificity, a second tag was included in the expression vectors (Calmodulin Binding Peptide or alternatively FLAG or Triple-FLAG). In addition, dual tags expression plasmids for Sp3 were constructed. The establishment of stable cell lines expressing these fusion proteins in an inducible manner was initiated. Such cell lines might be ideal for further analyzes of PIAS1 activities and to purify PIAS1 (Sp3) associated factors.
DOI:10.17192/z2005.0095