25.01.2013 Views

Role of the ubiquitin-like modifier FAT10 in protein degradation and ...

Role of the ubiquitin-like modifier FAT10 in protein degradation and ...

Role of the ubiquitin-like modifier FAT10 in protein degradation and ...

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

Chapter 3<br />

precipitates with HDAC6 (Fig. 21E). Moreover, a mutant <strong>of</strong> <strong>FAT10</strong> <strong>in</strong> which all<br />

lys<strong>in</strong>es were mutated to arg<strong>in</strong><strong>in</strong>e (Hipp et al., 2005) – thus prevent<strong>in</strong>g it from<br />

be<strong>in</strong>g ubiquitylated or acetylated – was still able to <strong>in</strong>teract with HDAC6 as well<br />

as, if not better than, wild-type <strong>FAT10</strong> (Fig. 21D). Still, <strong>the</strong> ability <strong>of</strong> catalyti-<br />

cally dead or <strong>in</strong>hibited HDAC6 to <strong>in</strong>teract with <strong>FAT10</strong> might solely be mediated<br />

through <strong>the</strong> C-term<strong>in</strong>al BUZ doma<strong>in</strong>. To exclude this possibility, we performed<br />

co-immunoprecipitation experiments with an isolated, catalytically dead CAT1<br />

doma<strong>in</strong>. As can be seen <strong>in</strong> figure 27A, <strong>the</strong> mutated CAT1 doma<strong>in</strong> was still able<br />

to pull down <strong>FAT10</strong>. In addition, <strong>the</strong> isolated CAT1 doma<strong>in</strong> was still capable <strong>of</strong><br />

<strong>in</strong>teract<strong>in</strong>g with <strong>the</strong> lys<strong>in</strong>e-less mutant <strong>of</strong> <strong>FAT10</strong>. Figure 27B demonstrates that<br />

recomb<strong>in</strong>ant GST-<strong>FAT10</strong> was able to pull down similar amounts <strong>of</strong> <strong>the</strong> wild-type<br />

<strong>and</strong> mutated <strong>in</strong> vitro transcribed <strong>and</strong> translated [ 35 S]-methion<strong>in</strong>e labeled CAT1<br />

doma<strong>in</strong>s. Toge<strong>the</strong>r, <strong>the</strong>se f<strong>in</strong>d<strong>in</strong>gs suggest that, while a small portion <strong>of</strong> <strong>the</strong> <strong>in</strong>tra-<br />

cellular <strong>FAT10</strong> is acetylated, acetylation is not required for its b<strong>in</strong>d<strong>in</strong>g to HDAC6<br />

nor is <strong>FAT10</strong> a substrate <strong>of</strong> HDAC6 deacetylase activity.<br />

<strong>FAT10</strong> <strong>and</strong> <strong>FAT10</strong>-GFP localize to <strong>the</strong> aggresome after proteasome <strong>in</strong>hibition<br />

As <strong>FAT10</strong> <strong>in</strong>teracts with HDAC6 after proteasome <strong>in</strong>hibition, <strong>and</strong> as HDAC6 is<br />

<strong>in</strong>volved <strong>in</strong> <strong>the</strong> transport <strong>of</strong> polyubiquitylated prote<strong>in</strong>s to <strong>the</strong> aggresome upon pro-<br />

teasome impairment (Kawaguchi et al., 2003), we <strong>in</strong>vestigated whe<strong>the</strong>r HDAC6<br />

may <strong>like</strong>wise transport <strong>FAT10</strong> to <strong>the</strong> aggresome. To test this hypo<strong>the</strong>sis, we<br />

<strong>in</strong>vestigated whe<strong>the</strong>r treatment <strong>of</strong> transiently HA-<strong>FAT10</strong>-tranfected HEK293T<br />

cells with proteasome <strong>in</strong>hibitor had an <strong>in</strong>fluence on <strong>the</strong> subcellular localization<br />

<strong>of</strong> <strong>FAT10</strong> as analyzed by confocal immun<strong>of</strong>luorescence microscopy. In untreated<br />

cells, <strong>FAT10</strong> was evenly distributed throughout <strong>the</strong> cytoplasm <strong>and</strong> showed vary-<br />

<strong>in</strong>g degrees <strong>of</strong> localization to <strong>the</strong> nucleus (Fig. 22A, a <strong>and</strong> i). HDAC6, on <strong>the</strong><br />

o<strong>the</strong>r h<strong>and</strong>, was completely excluded from <strong>the</strong> nucleus <strong>and</strong> showed only cytoplas-<br />

matic localization (Fig. 22A, b). As reported by Kawaguchi et al., proteasome<br />

<strong>in</strong>hibition <strong>in</strong>duced relocalization <strong>of</strong> HDAC6 to a s<strong>in</strong>gle prom<strong>in</strong>ent juxtanuclear<br />

structure – <strong>and</strong> <strong>in</strong>deed also caused <strong>FAT10</strong> to localize to <strong>the</strong> very same structure<br />

(Fig. 22A, e-h). To determ<strong>in</strong>e <strong>the</strong> identity <strong>of</strong> <strong>the</strong> observed <strong>in</strong>clusion bodies, we<br />

analyzed <strong>the</strong>m for <strong>the</strong> presence <strong>of</strong> known aggresome markers o<strong>the</strong>r than HDAC6.<br />

We never observed formation <strong>of</strong> more than one <strong>of</strong> <strong>the</strong>se <strong>in</strong>clusion bodies per cell,<br />

<strong>and</strong> comb<strong>in</strong>ed with <strong>the</strong> observation that <strong>the</strong>y colocalizd with γ-tubul<strong>in</strong> (Fig. 22A,<br />

m-p), a component <strong>of</strong> centromeres, this leads us to <strong>in</strong>fer that <strong>the</strong>y form at <strong>the</strong><br />

microtubule organiz<strong>in</strong>g center. In addition, we observed a characteristic distor-<br />

tion <strong>and</strong> enlargement <strong>of</strong> <strong>the</strong> centromeres upon proteasome <strong>in</strong>hibition (compare<br />

Fig. 22A, j <strong>and</strong> n), which is ano<strong>the</strong>r hallmark <strong>of</strong> aggresome formation (Johnston<br />

88

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!