Clr4suv39h1 ubiquitination and non-coding rna mediate transcriptional silencing of heterochromatin via swi6 phase separation
Clr4suv39h1 ubiquitination and non-coding rna mediate transcriptional silencing of heterochromatin via swi6 phase separation"
- Select a language for the TTS:
- UK English Female
- UK English Male
- US English Female
- US English Male
- Australian Female
- Australian Male
- Language selected: (auto detect) - EN
Play all audios:
ABSTRACT Transcriptional silencing by RNAi paradoxically relies on transcription, but how the transition from transcription to silencing is achieved has remained unclear. The Cryptic Loci
Regulator complex (CLRC) in _Schizosaccharomyces pombe_ is a cullin-ring E3 ligase required for silencing that is recruited by RNAi. We found that the E2 ubiquitin conjugating enzyme Ubc4
interacts with CLRC and mono-ubiquitinates the histone H3K9 methyltransferase Clr4SUV39H1, promoting the transition from co-transcriptional gene silencing (H3K9me2) to transcriptional gene
silencing (H3K9me3). Ubiquitination of Clr4 occurs in an intrinsically disordered region (Clr4IDR), which undergoes liquid droplet formation in vitro, along with Swi6HP1 the effector of
transcriptional gene silencing. Our data suggests that phase separation is exquisitely sensitive to non-coding RNA (ncRNA) which promotes self-association of Clr4, chromatin association, and
di-, but not tri- methylation instead. Ubc4-CLRC also targets the transcriptional co-activator Bdf2BRD4, down-regulating centromeric transcription and small RNA (sRNA) production. The
deubiquitinase Ubp3 counteracts both activities. SIMILAR CONTENT BEING VIEWED BY OTHERS RIXOSOMAL RNA DEGRADATION CONTRIBUTES TO SILENCING OF POLYCOMB TARGET GENES Article Open access 30
March 2022 A PROTEIN ASSEMBLY MEDIATES _XIST_ LOCALIZATION AND GENE SILENCING Article 09 September 2020 THE YEAST ISW1B ATP-DEPENDENT CHROMATIN REMODELER IS CRITICAL FOR NUCLEOSOME SPACING
AND DINUCLEOSOME RESOLUTION Article Open access 18 February 2021 INTRODUCTION Heterochromatin has crucial roles in genome stability by silencing repetitive DNA and by suppressing
recombination1,2,3,4,5,6,7. In the fission yeast _Schizosaccharomyces pombe_, the H3K9 methyltransferase Clr4, the homolog of mammalian SUV39H1 and SUVH39H2, methylates histone H3 lysine 9
(H3K9) to maintain centromeric heterochromatin during the cell cycle8,9,10. Centromeric non-coding RNA (ncRNA) is transcribed by RNA Pol II during S phase, and is processed to make
centromeric sRNA by the concerted action of the RNAi transcriptional silencing complex (RITS), the RNA-directed RNA Polymerase Complex (RDRC) and Dicer (Dcr1)7,11,12,13,14,15,16.
Di-methylation of H3K9 by Clr4, guided in part by sRNA, mediates co-transcriptional gene silencing (CTGS) of heterochromatin, which remains permissive to transcription during S phase. The
subsequent tri-methylation of H3K9 by Clr4 and strong binding of Heterochromatin Protein 1 (HP1) homologs Swi6 and Chp2, suppresses RNA Pol II-dependent ncRNA transcription via
transcriptional gene silencing (TGS), which is essential for the epigenetic inheritance of heterochromatin1,17,18,19. However, the mechanism that promotes the transition from RNAi-dependent
CTGS to TGS is unclear. Ubiquitination can regulate enzyme activity by adding a single ubiquitin to its substrate (mono-ubiquitination), or target protein degradation via the 26S proteasome
by the addition of a ubiquitin chain (poly-ubiquitination). Ubiquitination requires ubiquitin, an E1 ubiquitin activating enzyme, an E2 ubiquitin conjugating enzyme and an E3 ubiquitin
ligase20,21,22,23. E2 enzymes interact with E1 and E3 enzymes and have essential roles in determination of ubiquitin linkage specificity and ubiquitin chain length24. The ubiquitination
complex Cul4-Rik1Raf1Raf2 (CLRC) of _S. pombe_ is composed of cullin Cul4, the RING-box protein Rbx1, the DNA damage binding protein 1 (DDB1) homolog Recombination in K 1 (Rik1) and two
substrate receptors, Raf1 and Raf2, as well as the eponymous Clr4, which is only weakly associated in vitro25,26,27,28,29,30. Cul4 is a highly conserved E3 ligase and interacts with
Rik1/Raf1/Raf2 for substrate recognition. All subunits of CLRC are essential for heterochromatin formation but its E2 enzyme and ubiquitination targets are not well characterized25,26,27,28.
Liquid-liquid phase separation (LLPS) is the mechanism by which biomolecular condensates like cellular granules, including membraneless organelles such as the nucleolus, P granules, and
stress granules form without a lipid bilayer. Multivalent macromolecular interactions, especially between proteins harboring intrinsically disordered regions (IDRs) and nucleic acids, drive
formation of phase-separated liquid droplets. These biomolecular condensates have higher protein density and reduced internal diffusion, which contributes to a myriad of cellular functions,
including heterochromatin formation31,32,33,34,35,36,37,38. Nucleosomes are post-translationally modified and many chromatin-related factors, such as heterochromatin protein 1 (HP1) and
bromo-domain containing proteins (e.g. BRD4), which recognize these histone modifications, have an intrinsic propensity for LLPS and form phase-separated liquid droplets in vitro and in
cells39,40,41,42,43. Here we set out to identify how the E3 ubiquitin ligase complex CLRC mediates the transition from RNAi-dependent CTGS to TGS. We found that Ubc4-CLRC mono-ubiquitinates
Clr4 in the IDR, reducing its chromatin- and ncRNA-binding activities. Importantly, we found that Clr4 undergoes phase-separated liquid droplet formation along with Swi6 through its IDR.
However, liquid droplet formation of Clr4 is blocked by the presence of ncRNA, which instead promotes self-association of Clr4, chromatin association of Clr4 and di-methylation of H3K9 but
not tri-methylation of H3K9. We also found that the Ubc4-CLRC complex poly-ubiquitinates Bdf2 and inhibits the recruitment of Epe1 and Bdf2 to centromeric heterochromatin, reducing ncRNA
transcription and sRNA production via RNAi. Both H3K9 tri-methylation by mono-ubiquitinated Clr4 and reduced sRNA production by targeted Epe1-Bdf2 are crucial for the transition between CTGS
to TGS and spreading of heterochromatin, which can be reversed by overexpression of deubiquitinase Ubp3. These observations implicate phase separation as a critical step in the transition
from RNAi to transcriptional gene silencing during heterochromatin formation. RESULTS UBC4 AND CUL4 REGULATE HETEROCHROMATIN SILENCING We previously showed that a specific mutation in the E2
ubiquitin conjugating enzyme Ubc4 (_ubc4-G48D_) reduces heterochromatic silencing at centromeres, at the mating-type locus and at telomeres (Fig. 1a)44. Overexpression of Ubc4 also causes
chromosome mis-segregation45. We found that the _ubc4-G48D_ (_ubc4-1_) mutation results in Ubc4 protein instability leading to an overall lower Ubc4 protein level (Fig. 1b; Supplementary
Fig. 1c). Replacing _ubc4_ with its mouse homolog _UBE2D2_ fully complemented _ubc4-1_, while _mUBE2D2-G48D_ (_mUBE2D2-1_) had similar silencing defects to _ubc4-1_, showing that the
function of Ubc4 is likely conserved throughout evolution (Fig. 1a; Supplementary Fig. 1a, b). We also isolated a spontaneous second-site suppressor S137T, which re-stabilizes Ubc4-1 protein
levels, thus restoring silencing (Fig. 1b–d). The E3 ubiquitin ligase Cul4 is an essential subunit of the CLRC, and _cul4-GFP_ (_cul4-1_) is a hypomorphic mutant with reduced
heterochromatic silencing27. _ubc4-1_ and the _cul4-1_ mutants have very similar phenotypes, including loss of silencing of _ade6__+_ and _ura4__+_ reporter genes in centromeric
heterochromatin, as well as increased binding of Rpb1, the main subunit of RNA polymerase II, to heterochromatin (Figs. 1a, 2a) and increased production of siRNA (Figs. 1e, 2f, 5a).
Interestingly, while H3K9me3 was reduced from both _ubc4-1_ and _cul4-1_ mutants at all heterochromatic loci tested, H3K9me2 was increased at the centromeric _dg_ and _dh_ repeats (Figs. 1f,
2a; Supplementary Fig. 2a). Increased H3K9me2 was also observed at the centromeric _ade6__+_ reporter gene, which accumulates small RNA, but not at the _ura4__+_ reporter gene, which does
not (Fig. 1f; Supplementary Fig. 10a). The Swi6HP1 protein, which coats and maintains heterochromatin, displayed intermediate binding levels at centromeres in _ubc4-1_ and _cul4-1_ mutants
presumably because it recognizes both H3K9me2 and H3K9me3 (Supplementary Fig. 2b)18,46. In contrast, H3K9me2/3 and Swi6 were completely lost from both _ubc4-1_ and _cul4-1_ mutants at
telomeres or mating-type loci where RNAi-dependent sRNA production is limited (Supplementary Fig. 2c, d). To determine whether RNAi is important for maintaining H3K9me2 in _ubc4-1_ mutant
cells, we introduced the _ubc4-1_ mutation in _dcr1Δ_ mutant cells and found that the _dcr1Δ ubc4-1_ double-mutant lost both H3K9me2 and H3K9me3 and was hyper-sensitive to the microtubule
destabilizing agent thiabendazole (TBZ), showing that centromeric heterochromatin was completely disrupted similarly to the _clr4_ deletion mutant (Fig. 2a; Supplementary Fig. 2e). The more
severe alleles _ubc4-3xHA_ and _cul4Δ_ abolished H3K9me2 and H3K9me3 altogether indicating that _ubc4-1_ and _cul4-1_ are hypomorphic and they might have other targets important for
heterochromatin silencing (Supplementary Fig. 2f, g). In addition to their similar phenotypes, we found that Ubc4 and Cul4 physically interact and form a stable complex by
co-immunoprecipitation (Co-IP) (Supplementary Fig. 3a). Ubc4 also forms a complex with Raf1 and Raf2, two substrate receptors of CLRC (Supplementary Fig. 3b) that mediate recruitment by
siRNA-to-chromatin 1 (Stc1) and the RNA-induced transcriptional gene silencing (RITS) complex. The interaction between Ubc4-Cul4 and Ubc4-Raf2 was independent of nuclease treatment
(Supplementary Fig. 3c, d), which indicates that Ubc4 and Cul4 directly form the E2/E3 complex by which CLRC targets substrates important for H3K9 methylation, in a RNAi-dependent manner.
MONO-UBIQUITINATION OF CLR4 BY UBC4-CLRC IS REQUIRED FOR H3K9ME2 TO H3K9ME3 TRANSITION Next, we set out to identify the substrates of this novel E2/E3 complex, Ubc4-CLRC, for the regulation
of H3K9 methylation. Interestingly, Clr4 co-purifies with the CLRC in vivo, but does not behave like a structural component of CLRC in vitro25,26,27,28. This led us to hypothesize that Clr4
is itself a target of the Ubc4-CLRC. Indeed, another Clr4 human homolog, SETDB1, is mono-ubiquitinated at its SET-Insertion domain and this modification is important for its H3K9
methyltransferase activity47,48. To test whether Clr4 is a substrate of Ubc4-CLRC, we purified 3xFlag-Clr4 protein from _S. pombe_ cells and detected a single higher molecular weight form
that reacted with ubiquitin antibodies in a Ubc4-dependent fashion (Fig. 2b; Supplementary Fig. 4a, b). A pulldown of ubiquitinated proteins using Ubiquitination Affinity Beads (UBA)
revealed a similar higher molecular weight Clr4 which is also dependent on Ubc4 (Supplementary Fig. 4c). In addition to Ubc4, the presence of this higher molecular weight Clr4 depends on
Cul4, Rik1, Raf1 and Raf2, the other components of CLRC, but not on the proteasome inhibitor MG-132 (Supplementary Fig. 4d, e). These results strongly suggest that Ubc4, together with CLRC,
mono-ubiquitinates Clr4 in vivo. We were unable to recover sufficient peptides in vivo for the identification of Clr4 ubiquitination sites by Mass Spectrometry because of the very low
abundance of Clr4 in _S. pombe_ cells49, so instead we ubiquitinated 6xHis-Clr4 recombinant protein in vitro. Clr4 was mono-ubiquitinated efficiently by UBE2D3, the human homolog of Ubc4,
independently of an E3 ligase complex (Fig. 3d; Supplementary Fig. 5a–c), similarly to SETDB148,50. This approach uncovered 9 possible ubiquitination sites, which mostly reside in the
intrinsically disordered region (IDR) between the chromodomain (CD) and the PreSET domain (Fig. 2c and Supplementary Fig. 5a–d) which is involved in nucleosome binding51. To identify which
of these represent functional sites in vivo, we partitioned the 36 lysines of Clr4 into 6 subgroups, which we then systematically mutated into arginines (R). We also included a mutant of all
36 lysines (_AllKtoR_). The all-lysine mutant (_AllKtoR_) and the second block mutant (_2-8KtoR_), corresponding to the middle of the IDR, completely abolished Clr4 ubiquitination in vivo
(Fig. 2c, d; Supplementary Fig. 5d, e), consistent with the results of in vitro ubiquitination and Mass Spectrometry. By this means, we could narrow down the possible sites to 4 lysines
(_clr4-2-4KtoR_; K109, K110, K113, and K114) (Fig. 2c, d; Supplementary Fig. 5f–i). These lysines are organized in tandem and are therefore very likely to compensate for each other. Using
ChIP-seq, we found that _clr4-AllKtoR_ lost both H3K9me2 and H3K9me3, presumably by reduced enzymatic activity52. _clr4-2-8KtoR_ and _clr4-2-4KtoR_ mutants had the same increase of H3K9me2
and decrease of H3K9me3 seen in _ubc4-1_ and _cul4-1_ hypomorphic mutants but loss of silencing of centromeric _ade6__+_ and _ura4__+_ reporter genes in _clr4-2-4KtoR_ mutant was weaker than
_clr4-AllKtoR_ and _cul4-1_ mutants (Fig. 2e; Supplementary Fig. 6a–c). In _ubc4-1_ and _cul4-1_ mutants, we observed more than 2-fold increase of centromeric sRNA, as previously observed
for _cul4-1_, but _clr4-2-8KtoR_ and _clr4-2-4KtoR_ mutants did not change centromeric sRNA production (Fig. 2f)53. These results indicate that Clr4 mono-ubiquitination by Ubc4-CLRC complex
mediates the transition from H3K9me2 to H3K9me3 (Fig. 2g). CLR4 MONO-UBIQUITINATION INDUCES ITS DISSOCIATION FROM CHROMATIN AND FROM CENTROMERIC NCRNA Remarkably, the lysines in Clr4-2-8
co-incided precisely with two adjacent patches in the IDR that were previously shown to bind to the nucleosome core, independently of H3K9me3 (KKVFS and KRQSRK)51. Consistently, Clr4 binding
to chromatin was increased in _ubc4-1_ and _cul4-1_ mutants, especially at _dh_ centromeric repeats and at the _ade6__+_ reporter gene, where sRNAs are over-produced (Fig. 3a; Supplementary
Figs. 7a, b, 10a). This increased binding of Clr4 to chromatin is not dependent on its chromodomain because there was no change in combination with a chromodomain mutant (_clr4-W31G
ubc4-_1, Fig. 3a; Supplementary Fig. 7a)18,53. Note that the _clr4-2-4KtoR_ mutant has much weaker binding than _ubc4-1_ or _cul4-1_ mutants presumably because _clr4-2-4KtoR_ mutation does
not affect ncRNA transcription and sRNA production (Figs. 2f, 3a; Supplementary Fig. 6c). Mutating all lysines (_AllKtoR_) resulted in an even stronger affinity of Clr4 to chromatin (Fig.
3a; Supplementary Fig. 7a). In contrast, Flag-Clr4 binding to centromeric heterochromatin decreased when H3K9me2/3 was completely abolished by _rik1Δ_, _raf1Δ_ and _raf2Δ_ (Fig. 3b)26. Clr4
also binds centromeric ncRNA54 and this activity was strongly increased in _ubc4-1_ and _cul4-1_ mutants and slightly but significantly increased in the _clr4-2-4KtoR_ mutant (Fig. 3c;
Supplementary Fig. 6d). These results indicate that both chromatin and ncRNA binding activities of Clr4 are down regulated by Clr4 mono-ubiquitination. To further explore the effect of Clr4
mono-ubiquitination on chromatin binding, we carried out nucleosome binding assays using in vitro ubiquitinated 6xHis-Clr4 recombinant protein and 3xFlag-Clr4 purified from _S. pombe_ cells,
with nucleosomes with either unmodified histone H3 (me0) or H3K9me3 (me3) tethered to streptavidin beads. In contrast to unmodified Clr4 protein, which had strong binding affinity to the
nucleosome, ubiquitinated Clr4 did not bind to nucleosomes (Fig. 3d; Supplementary Fig. 7c). These results suggest that Clr4 mono-ubiquitination promotes dissociation of Clr4 from chromatin
in agreement with the in vivo Clr4 ChIP-seq data. Next, we tested whether Ubc4-Cul4 ubiquitination activities are important in vivo in the context of a minimal silencing system, relying on
tethering Clr4 to a specific chromatin locus. To do this, we generated a reporter system in which _clr4-ΔCD_ lacking the chromodomain is fused to Gal4 DNA binding domain (GBD) and tethered
to 5 Gal4-binding sites (5xUAS) upstream of _ade6__+_, inserted at the endogenous _ura4_ locus, and with additional intact WT _clr4__+_ available55. Artificial tethering of GBD-_Clr4-ΔCD_
resulted in silencing of the _ade6__+_ reporter gene as indicated by red colony formation on low-adenine medium and accumulation of H3K9me2/3 by ChIP-qPCR (Fig. 3e), in accordance with
previous studies55,56,57,58,59,60. In contrast, _ade6__+_ silencing was greatly decreased by introduction of _ubc4-1_ and _cul4-1_ mutations, demonstrating that the ubiquitination activity
of Ubc4-CLRC is required for spreading of silencing into the _ade6__+_ reporter (Fig. 3e). PHASE-SEPARATED LIQUID DROPLET FORMATION OF CLR4 IS INHIBITED BY NON-CODING RNA IN VITRO Clr4 and
its mouse homolog, mSUV39H1, have an intrinsically disordered region (IDR), also known as a low-complexity domain (LCD), between its chromodomain (CD) and PreSET domain (Supplementary Fig.
8a, b)51, and IDR/LCD are typically responsible for phase separation31,33,35,37,38,61. We found that recombinant GFP-Clr4Full and GFP-Clr4IDR (51-220 amino acids of Clr4) proteins readily
form liquid droplets in vitro in a salt-dependent manner even without addition of crowding agents (Fig. 4a; Supplementary Fig. 8c)62. The formation of liquid droplets by GFP-Clr4IDR was
reduced by adding 1,6-hexanediol, which dissolves phase-separated liquid droplets by disrupting the hydrophobic interactions of protein-protein or protein-RNA (Supplementary Fig. 8d)37,63.
Furthermore, photobleaching of inside part of liquid droplets of GFP-Clr4IDR resulted in slow but steady recovery of fluorescence indicative of dynamic exchange of GFP-Clr4IDR protein inside
of liquid droplets (Fig. 4b and Supplementary Fig. 8e)31,39,43. These data indicate an involvement of phase separation for liquid droplet formation of Clr4. As the oligomerization of HP1 is
important to form phase-separated liquid droplets as well as during chromatin compaction, and as SUV39H1 is also able to tether together to form dimers40,42,64,65,66, we reasoned that Clr4
may self-associate to form phase-separated liquid droplets. To do this, we mixed recombinant GFP-Clr4 and mCherry-Clr4 and found that these proteins readily associate together to form
phase-separated liquid droplets, supporting the idea of Clr4 self-association (Fig. 4c). CTGS during G1/S phase of the cell cycle is accompanied by transcription of ncRNAs and accumulation
of sRNAs and the interaction of RNA-binding proteins with RNA are important for LLPS17,19,31,34. To investigate the possible role of centromeric ncRNA in Clr4 phase-separated liquid droplet
formation, we added 120 nt, synthetic centromeric ncRNA to GFP-Clr4/mCherry-Clr4. Surprisingly, ncRNA inhibited phase-separated liquid droplet formation of Clr4 and Clr4IDR in a dosage
dependent manner (Fig. 4c and Supplementary Fig. 8c), reminiscent of other IDR-containing proteins that interact with RNA67,68,69. We next examined Clr4 self-association via
immunoprecipitation using recombinant Flag-Clr4 and Myc-Clr4 proteins in the presence or absence of ncRNA. The weak interaction of Flag-Clr4 with Myc-Clr4 in the absence of ncRNA is
consistent with multivalent weak interactions that form phase-separated liquid droplets (Fig. 4d). In contrast, by addition of ncRNA, Clr4 self-association was greatly increased with
increasing concentration of ncRNA, indicating that Clr4 self-association induced by ncRNA actually inhibits its phase-separated liquid droplet formation (Fig. 4c, d). Furthermore,
mono-ubiquitinated Clr4 did not bind to the centromeric ncRNA compared to unmodified Clr4 protein from ncRNA binding assays using 3xFlag-Clr4 purified from _S. pombe_ cells (Supplementary
Fig. 8f). Thus, the RNA binding activity of Clr4 is key for its self-association and phase-separated liquid droplet formation, and is inhibited by mono-ubiquitination. Previously, ncRNA has
been shown to inhibit Clr4 enzyme activity51, but as nucleosomes were used as substrate, this was thought to be due to competition with nucleosome binding. To characterize the effect of
ncRNA-mediated self-association on Clr4 H3K9 methylation activity, we fused the N terminal tail of _S. pombe_ H3 (A1-K36) with GST (H3N-GST) and used it as a substrate of Clr4 in the absence
or presence of ncRNA70. Interestingly, Clr4 enzymatic activity for H3K9me3 decreased greatly as the concentration of ncRNA increases, while Clr4 enzymatic activity for H3K9me2 did not
change with increasing ncRNA and self-association of Clr4 (Fig. 4d, e). These results indicate that ncRNA binding and Clr4 self-association has an inhibitory effect on Clr4 enzymatic
activity for H3K9me3, independent of nucleosome binding. SUV39H1 forms a complex with HP1, and SUV39H1 and HP1 together colocalize to distinct heterochromatic subnuclear domains
(chromocenters) in human cells43,66,71,72. HP1α readily forms phase-separated liquid droplets without addition of DNA or nucleosomes in vitro, but HP1β only forms liquid droplets with
addition of the N-terminal chromo domain (CD) of SUV39H1 and nucleosomes modified on histone H3K9me343. Like other HP1 proteins, _S. pombe_ Swi6 has been shown to undergo liquid-liquid phase
separation (LLPS) which contributes to the highly dynamic features of heterochromatin40,42,73. Swi6 forms phase-separated liquid droplets only in combination with nucleosomes73. As in human
cells, we found that recombinant mCherry-Swi6 could not form phase-separated liquid droplets in vitro even in combination with full-length GFP-Clr4. However, GFP-Clr4IDR and mCherry-Swi6
could form phase-separated liquid droplets when mixed together (Fig. 4f). These droplets dissolved upon the addition of a much smaller amount of ncRNA compared to that required to dissolve
droplets formed by mixing GFP-Clr4 with mCherry-Clr4 (Fig. 4c, f). Thus, these results suggest the possibility that phase-separated liquid droplet formation of Swi6 can be promoted by
Clr4IDR independently of modified nucleosomes, but only during transcriptional silencing when RNA is absent. GFP-Swi6 localizes to heterochromatin in vivo in a H3K9me-dependent manner and we
used this to assess heterochromatin condensation in _ubc4_ mutant backgrounds (Fig. 4g). Compared to WT and _dcr1Δ_ strains which have up to 4 condensed foci representing the centromeric
chromocenter, telomere clusters and the mating type locus, _ubc4-1_ mutants retained only one visible chromocenter, presumably due to the complete loss of H3K9me2/3 and Swi6 at telomeres and
mating-type loci detected by ChIP (Supplementary Fig. 2c, d). The remaining GFP-Swi6 foci were abolished in _dcr1Δ ubc4-_1, _ubc4-3xHA_, _cul4Δ_ and _clr4Δ_ mutants in accordance with
complete loss of H3K9me2/3 from all heterochromatin in these mutants (Fig. 4g). When a strain which has both GFP-Clr4 and mCherry-Swi6 was used, GFP-Clr4 and mCherry-Swi6 co-localized to a
condensed focus in vivo in WT cells. Only one chromocenter was visible because the N-terminal GFP fusion to Clr4 causes a silencing defect (Supplementary Fig. 9a, b). GFP-Clr4 and
mCherry-Swi6 co-localization was significantly reduced in _ubc4-1_ and _dcr1Δ_ and completely abolished in _dcr1Δ ubc4-1_ double mutants (Supplementary Fig. 9a). These results indicate that
Clr4 and Swi6 form condensates in vitro and in vivo, which is promoted in the absence of RNA during the G2 phase of the cell cycle. After mitosis, phosphorylation of H3S10 evicts Swi6 from
heterochromatin, resulting in transient transcription in G1 and S phase19. RNA promotes self-association of Clr4 and rapid dissolution of heterochromatic condensates, consistent with
previously observed de-clustering of GFP-Swi6 chromocenters during the cell cycle74. Subsequent mono-ubiquitination by CLRC releases Clr4 from RNA and promotes phase-separated liquid droplet
formation together with H3K9me3 and Swi6 to drive the transition to TGS in G2. UBIQUITINATION AND DE-UBIQUITINATION OF EPE1 AND BDF2BRD4 REGULATES CENTROMERIC SRNA AND CLR4 FOR
HETEROCHROMATIN SILENCING It has recently been shown that Ubc4 and Cul4 are strongly enriched in Swi6-associated chromatin, along with other CLRC subunits, as well as in chromatin associated
with the BET family double bromodomain protein Bdf2 (a homolog of mammalian BRD4)49. Bdf2 is recruited by the putative H3K9 demethylase, Epe1, which localizes to centromeric heterochromatin
in a Swi6-dependent manner and contributes to sRNA production75,76,77, which also occurs mainly in S phase19. Bdf2 binds to RNA Pol II promoters at the centromeric heterochromatin
boundary78 and interacts with the TFIID transcription factor complex79,80 while Epe1 is confined to these boundaries by Cul4-Ddb1Cdt2 E3 ligase-dependent poly-ubiquitination75. We found that
the increased centromeric sRNA production in _ubc4-1_ and _cul4-1_ mutants was accompanied by spreading of Epe1 and Bdf2 to pericentromeric repeats, as well as Ago1 and Chp1, two components
of the RITS siRNA complex (Figs. 2f, 5a, b; Supplementary Fig. 10a–c). Moreover, _epe1Δ_ and _bdf2Δ_ deletion mutants suppressed the increased sRNA production in _ubc4-1_ and _cul4-1_
mutants back to wild-type levels (Fig. 5a), by reducing recruitment of RNA Pol II (Supplementary Fig. 10d). Consistent with the regulation of Epe1 by Cul475, we found that Ubc4 and CLRC
induced poly-ubiquitination and degradation of Bdf2 (Fig. 5c and Supplementary Fig. 10e, f). Bdf2BRD4 recruits TFIID and RNA Pol II to centromeric heterochromatin, an important step in
precursor ncRNA transcription for sRNA production79,80. To mimic the effect of _ubc4-1_ and _cul4-1_ on sRNA production, we overexpressed Epe1 (_epe1-OE_, Supplementary Fig. 10g). As
expected, _epe1-OE_ resulted in increased sRNA production by increased recruitment of Epe1 and Bdf2, but unlike _ubc4-1_ and _cul4-1_, it also resulted in decrease of both H3K9me2 and
H3K9me3 at centromeric heterochromatin (Fig. 5a, b; Supplementary Fig. 10h–j). Thus, Ubc4-CLRC regulates sRNA production separately through Epe1 and Bdf2, which in combination with Clr4,
regulates the proper transition from CTGS to TGS (Supplementary Fig. 10k). Ubiquitination is a reversible, highly dynamic and transient modification, and its removal is catalyzed by
deubiquitinating enzymes21,81,82. A deletion mutant of the deubiquitinating enzyme Ubp3 rescues the heterochromatic silencing defects of _ago1Δ_, and _ubp3Δ_ shows positive epistatic genetic
interactions with CLRC component mutants83,84. While deletion of Ubp3 did not increase Clr4 mono-ubiquitination most likely because of redundancy between de-ubiquitinating enzymes
(Supplementary Fig. 11a)85, the overexpression of Ubp3 (_ubp3-OE_) resulted in a silencing defect at the centromeric _ura4__+_ reporter gene and reduced mono-ubiquitination of Clr4,
supporting the idea that Ubp3 counteracts Clr4 mono-ubiquitination by Ubc4-CLRC (Fig. 5d and Supplementary Fig. 11b, c). The overexpression of Ubp3 also resulted in an increase of H3K9me2
and decrease of H3K9me3, as well as increased sRNA production, with increased binding of Epe1 and Bdf2 to centromeric heterochromatin, resembling the phenotypes of _ubc4-1_ or _cul4-1_
mutants at centromeric heterochromatin (Fig. 5e, left; Supplementary Fig. 11d). Similarly, at telomeres, _ubp3-OE_ resulted in increased H3K9me2 but decreased H3K9me3 as well as greatly
increased recruitment of Epe1 and Bdf2, resulting in ectopic sRNA production (Fig. 5e, right; Supplementary Fig. 11e). The 5’-nucleotide of these sRNAs from centromere and subtelomere
repeats had a strong U/A bias and the typical sRNA size was 23±1 nt, as is expected for bona fide RNAi products (Fig. 5f, g)12,86. These results indicate that Ubp3 counteracts the activities
of Ubc4-CLRC, both in terms of H3K9me2/3 transitions and sRNA production, with overexpression of Ubp3, making the chromatin structure of telomeres resemble that of the centromere.
DISCUSSION Our findings indicate that, while CTGS and H3K9 di-methylation depend strictly on RNAi, mono-ubiquitination of Clr4 promotes the transition from CTGS to TGS by first releasing
Clr4 from heterochromatin. The key lysine residues in the IDR form a β-sheet on binding of the nucleosome core51, which is likely disrupted by ubiquitination. Deubiquitination by Ubp3 then
allows re-engagement and spreading of H3K9 tri-methylation to adjacent chromatin, as demonstrated by ectopic heterochromatin formation at the _ade6__+_ reporter gene mediated by tethered
Clr4 (Fig. 3e). Spreading plays a similar role at the silent mating type locus and at telomeres where heterochromatic silencing is completely lost in _ubc4-1_ and _cul4-1_ mutants (Fig. 1c;
Supplementary Fig. 2c, d). The ability to induce the spreading of silencing by this mechanism may be critical at these genomic loci as these chromosomal locations have only limited capacity
for ncRNA transcription and RNAi. Ubc4-CLRC likely has additional functions, such as H3K14 ubiquitination, which was recently shown to enhance H3K9 methylation in vitro70. But H3K9me2 and
H3K9me3 are both lost at centromeres in H3K14R mutants, which suggests that H3K14 ubiquitination is essential for histone H3K9 methylation but not for transition of H3K9me2 to H3K9me3
(Supplementary Fig. 12a–c). Centromeric sRNA production was not affected by mutants in the chromo- (_clr4_-_W31G_), and SET domains (-_F449Y_ and -_I418P_) which are also defective in the
H3K9me2-to-H3K9me3 transition (Supplementary Fig. 12d, e)18. Interestingly, we found that _clr4_-_W31G_, -_F449Y_ and -_I418P_ mutants also have reduced Clr4 mono-ubiquitination indicating a
strong correlation between Clr4 mono-ubiquitination and transition of H3K9me2 to me3 (Supplementary Fig. 12f, g). Recently, phosphorylation of the Clr4 SET domain by Cdk1 was also shown to
be required for H3K9me3 during meiosis, and for proper gametogenesis, although the role in silencing, if any, was not examined87. The hierarchical relationship between phosphorylation,
auto-methylation and mono-ubiquitination of Clr4 remains to be explored52. Like Swi6, Clr4 forms biomolecular condensates in vitro and this property is mediated by the IDR.
Post-translational modifications of IDRs, and their association with RNA, can affect protein function significantly; including the induction of disorder-to-order transitions, alternative
complex formation and differential subcellular localization88,89,90,91,92,93,94. We found that ubiquitination of the IDR of Clr4 prevents RNA binding and protein self-association. This
promotes phase-separated liquid droplet formation, and is critical for the CTGS to TGS transition in vivo. These phase-separated liquid droplets of Clr4 and Clr4IDR/Swi6 are rapidly
dissolved in the presence of ncRNA and this RNA-dependent interaction promotes Clr4 self-association instead (Fig. 4c, d, f; Supplementary Fig. 8c). Intriguingly, binding of centromeric
ncRNA to the Swi6 hinge region, which is also disordered and lysine-rich, has previously been shown to prevent Swi6 binding to chromatin95, consistent with our results. In the absence of
mono-ubiquitination, Clr4 recruitment to genomic loci where ncRNA and sRNA are overproduced greatly increased in agreement with previous studies that mammalian SUV39H1 chromodomain binds to
pericentromeric RNA for the recruitment of SUV39H1 to heterochromatin, H3K9me3 deposition and heterochromatin assembly (Fig. 3a–c)96,97,98. We found that ncRNA binding inhibits Clr4
enzymatic activity for H3K9me3 likely through binding of ncRNA to the chromodomain and IDR of Clr4 and subsequent self-association (Fig. 4d, e and Supplementary Fig. 8c)54. In summary (Fig.
6), we have found that ncRNA transcription and sRNA production during G1/S promote CTGS and H3K9 di-methylation through recruitment of CLRC. The production of sRNA depends on the
transcriptional co-activator Epe1-Bdf2, which licenses recruitment of RNA Pol II to transcriptionally permissive heterochromatin13,14,15,18,46,53,59,99,100. Subsequently, removal of
Epe1-Bdf2 by Ubc4-CLRC-mediated poly-ubiquitination silences transcription, while Clr4 mono-ubiquitination is required for its dissociation from chromatin, and from ncRNA, triggering Clr4
dependent H3K9me3 and subsequent binding of HP1 proteins, Swi6 and Chp2 important for RNAi-independent TGS18,53,101. In the absence of RNAi, variable amounts of H3K9me2 are maintained at the
centromere, indicating that Clr4 must be recruited, at least in part independently of RNAi102. Based on our results, one explanation is that ncRNA, in the presence of existing H3K9
methylation, initially recruits Clr4 and promotes self-association and chromatin binding. In S phase, RNAi recruits the Ubc4-Cul4-Rik1Raf1Raf2 E3 ligase59, which mono-ubiquitinates Clr4
preventing RNA binding, and promoting H3K9me3, potentially by phase-separated liquid droplet formation and three-dimensional “spreading” along with Swi6 (Fig. 6). Romoval of ubiquitin
moieties by Ubp3 allows rapid recycling of Epe1-Bdf2 and of Clr4, ready for the next cell cycle. Given the high conservation of Ubc4, Clr4, Brd4 and Cul4, similar transitions between
RNAi-dependent gene silencing and heterochromatin assembly may be regulated by ubiquitination and RNA in other organisms, including mammals, where TRIM28, HP1 and SUVH39 form condensates
with H3K9me3 nucleosome arrays, similar to those reported here43. METHODS YEAST STRAINS AND PLASMIDS MANIPULATION Yeast strains, plasmids, oligonucleotide sequences and antibodies are listed
in Supplemental Tables S1–S4. All yeast strains were constructed by the PCR-based targeting method103 and lithium-acetate transformation method104. For N-terminus tagging of 3xFlag to
_clr4_, _rik1_ and _raf1_, endogenous promoters and terminators of each gene were used. For C-terminus tagging of 3xFlag, 3xFlag sequence and _adh1_ terminator with Kan resistant (KanR) gene
was inserted at the end of open reading frames (ORF). For some 3xFlag-Clr4 lysine mutants, _adh1_ terminator with Kan resistant (KanR) gene was inserted at the end of _clr4_ open reading
frames (ORF). For overexpression of _ubp3_, _ubp8_, and _epe1_, 500 bp of _adh1_ promoter was inserted between their promoter and ORF. To construct pET-14b-Clr4, the coding DNA sequence of
full-length Clr4 (amino acids 1-490) was amplified from _S. pombe_ genomic DNA, digested with _NdeI_ and cloned into _NdeI_-digested pET-14b. For pHis-parallel-GFP-Clr4Full and
pHis-parallel-GFP-Clr4IDR construction, the coding DNA sequence of full-length Clr4Full (amino acids 1-490) and Clr4IDR (amino acids 51-220) were amplified from _S. pombe_ genomic DNA,
digested with _BamHI_/_XhoI_ and cloned into _BamHI_/_XhoI_-digested pHis-parallel-GFP. For pHis-parallel-mCherry-Clr4, mCherry-Clr4 DNA sequence was synthesized (IDT), digested with
_NdeI_/_XhoI_ and cloned into _NdeI_/_XhoI_-digested pHis-parallel-GFP. For pHis-Clr4IDR-GFP, DNA sequence was synthesized (IDT) and cloned into pET-14b vector using In-Fusion HD cloning kit
(Takara). To construct pET-14b-Flag-Clr4 and pET-14b-Myc-Clr4, the coding DNA sequence of full-length Clr4 (amino acids 1-490) was amplified from _S. pombe_ genomic DNA using oligomers
containing 6xHis-1xFlag or 6xHis-1xMyc sequences respectively and cloned into pET-14b vector using In-Fusion HD cloning kit (Takara). SILENCING SPOT ASSAY Cells grown freshly on YES plate
were picked and resuspended in water to make a concentration of ~5 × 104 cells per μl. 8 μl of 10-fold serial dilutions were spotted on appropriate plates like YES, low adenine (Low Ade),
EMM-Ura (-Ura), and YES+TBZ (TBZ), and incubated 3 days at 30 °C for photography. _ura4__+_ reporter gene silencing was also tested on 5-fluoro-orotic acid (FOA) plates, which kills cells
expressing Ura4. IODINE STAINING Freshly grown yeast cells were spotted on SPA plates to induce sporulation, incubated at room temperature for 3 days, and then stained with iodine vapor for
2–3 min. Staining of starch which is abundant in mature spore produces a dark brown color. EMS MUTAGENESIS AND WHOLE-GENOME SEQUENCING FOR _UBC4-G48D SUPPRESSOR_ EMS mutagenesis was carried
out as previously described in ref. 105. Briefly, freshly grown 1 x 109 yeast cells were washed with distilled water, resuspended in 1.5 ml of 0.1 M sodium phosphate buffer (pH 7.0) and
treated with 50 μl EMS (Sigma M0880) for 1 h at 30 °C. 0.2 ml of treated cells was moved to 8 ml of sterile 5% sodium thiosulfate to inactivate EMS. Cells were washed, resuspended in sterile
water and plated on YES plate. Single colonies from plates were patched on SPA plates to induce sporulation, incubated at room temperature for 3 days, and then stained with iodine vapor for
2–3 min. For whole-genome sequencing for _ubc4-G48D_ suppressors, genomic DNA was purified from freshly grown yeast cells using Genomic-tip 20/G column (QIAGEN 10223). DNA library was
constructed using the TruSeq DNA PCR-Free Library Prep Kit (Illumina 20015962). Barcoded DNA library was sequenced using Illumina MiSeq platform, which generates paired-end 151 nt reads and
analyzed as previously described in ref. 106. Briefly, reads were adapter-trimmed and quality-filtered using Sickle (paired-end mode), then mapped to the _S. pombe_ genome using Bowtie2107.
Duplicate reads were removed using Samtools (≤ 0.14% per library). SNPs were called using FreeBayes to find SNPs present in suppressor strain but absent in the parental strain108. This
uncovered a G→C transversion at position chr2:716,192, a second-site mutation in the _ubc4_ gene. The mutation was verified by Sanger sequencing and confirmed to be causative by constructing
the same fresh mutation in WT and _ubc4-1_ strains. LIVE CELL IMAGING Images of freshly growing cells were collected using Zeiss Axio Imager M2 with DIC and EGFP channels and processed
using Zeiss Zen software. QUANTITATIVE REVERSE TRANSCRIPTION PCR (RT-QPCR) Total RNAs were purified from freshly grown yeast cells using _Quick_-RNA Fungal/Bacterial Miniprep Kit
(Zymoresearch R2014). Genomic DNAs were removed using DNase I (Roche 04716728001) and RNAs were cleaned using RNA Clean & Concentrator (Zymoresearch R1013). Reverse transcription was
carried out using SuperScript IV First-Strand Synthesis System (ThermoFisher 18091050) with random hexamer or oligo dT as primers. For quantitative PCR (qPCR), iQ SYBR Green Supermix
(Bio-rad 1708882) was used with the primers listed in Supplemental Table S3 and samples were run on CFX96 Real-Time PCR Detection System (Bio-rad). CHROMATIN IMMUNOPRECIPITATION (CHIP)
Chromatin Immunoprecipitation (ChIP) experiments were performed as previously described in ref. 79. Briefly, freshly grown 40 ml cells in YES were fixed with 1% formaldehyde from 37% stock
(Sigma F8775) for 20 min at room temperature for H3K9me2, H3K9me3, RNA Pol II, Ago1 and Chp1 ChIP. For Swi6 and 3xFlag-Clr4 ChIP, 40 ml cells were pre-incubated at 18 °C for 2 h and fixed
with 1.45% Formaldehyde from 16% stock (ThermoFisher 28908) for 30 min at 18 °C. Cells were quenched with 360 mM glycine and 2.4 mM Tris for 5 min. Whole-cell extracts (WCE) were prepared
using FastPrep-24 and sonicated using Covaris. 1~2 μg of antibodies like anti-H3K9me2 (Abcam ab1220), anti-H3K9me3 (Absolute Antibody Ab00700-1.26), anti-RNA polymerase II (BioLegend
904001), Swi6 (Abcam ab188276), Ago1 (Abcam Ab18190), Chp1 (Abcam ab18191) and anti-Flag (Sigma F1804) were pre-incubated with 50 μl Protein A and Protein G sepharose (GE 17513801 and
17061801) mix for 3~4 h and then incubated with sheared chromatin overnight at 4 °C. After washing, bead-antibody-chromatin mixture were treated with Proteinase K (ThermoFisher 100005393)
for 1 h at 42 °C and incubated 5~6 h at 65 °C to reverse cross-links. DNA was cleaned up using ChIP DNA Clean & Concentrator (Zymo Research D5205) and subsequently used for qPCR or
ChIP-Seq. CHIP-SEQ ChIP-Seq libraries were constructed using NEBNext Ultra II DNA Library Prep Kit for Illumina (NEB E7645) and barcoded multiplex libraries were sequenced using Illumina
MiSeq platform, which generates paired-end 101 bp reads. Raw reads were trimmed using Trimmomatic, mapped to _S. pombe_ genome using Bowtie2 and visualized in IGV genome browser. Total
ChIP-seq read counts for defined region were analyzed using MultiBigwigSummary or MultiBamSummary. SRNA-SEQ For sRNA-seq, total RNAs were purified from freshly grown yeast cells using
_Quick_-RNA Fungal/Bacterial Miniprep Kit (Zymoresearch R2014) and sRNA were enriched using RNA Clean & Concentrator (Zymoresearch R1013). sRNA libraries were constructed using NEBNext
Small RNA Library Prep Set for Illumina (NEB E7330) and barcoded multiplex libraries were sequenced using Illumina MiSeq platform, which generates single-end 36 nt reads. Reads were
adapter-trimmed using Cutadapt se, keeping a minimum read size of 15 nt, and mapped to the _S. pombe_ genome using Bowtie109 allowing up to 1 mismatch. The strand-specific genomic coverage
was calculated using Bedtools110. CO-IMMUNOPRECIPITATION (CO-IP) AND WESTERN BLOTTING (WB) Whole-cell extracts (WCE) were prepared from freshly grown yeast cells in lysis buffer (40mM
Tris-HCl, pH 7.5, 1 mM EDTA, 10% Glycerol, 0.1% NP-40, 300 mM NaCl) with Protease inhibitor cocktail (Roche 11836170001). For Co-IP of Cul4-GFP with Ubc4, 2 μg of GFP antibody (Roche
11814460001) was pre-incubated with 50 μl Protein A and Protein G Sepharose (GE Healthcare 17513801 and 17061801) mix for 3~4 h and then incubated with WCE overnight at 4 °C. After washing,
purified proteins were boiled and loaded on 4–20% Mini-PROTEAN TGX Gel (Bio-rad 456-1093) and Western blotting was performed with anti-GFP (Abcam 290) and anti-mUBE2D3 (Proteintech
11677-1-AP) antibodies to detect Cul4-GFP and Ubc4 using ECL (Thermo Fisher 32209) assay according to the manufacturer’s instruction. For Co-IP of Flag-Rik1, Flag-Dos1 or Dos2-Flag with
Ubc4, 50 μl of Anti-Flag M2 Affinity Gel (Sigma A2220) was incubated with WCE overnight at 4 °C. For detection of Flag tagged protein and Ubc4, anti-Flag (Sigma F7425) and anti-mUBE2D3
(Proteintech 11677-1-AP) antibodies were used. PURIFICATION OF UBIQUITINATED FLAG-CLR4 AND BDF2-FLAG PROTEINS For purification of ubiquitinated Flag-Clr4, WCE prepared from freshly grown
cells in lysis buffer (50 mM Hepes, pH 7.6, 150 mM NaCl, 300 mM KAc, 5 mM MgAc, 10% Glycerol, 20 mM β-GP, 1 mM EDTA, 1 mM EGTA, 0.1% NP-40, 1 mM DTT, 250 U/ml Dnase I (Roche 04716728001),
25~50 μg/ml Rnase A (Sigma R4642), 1 mM PMSF, 0.3 mM PR-619 (Sigma 662141)) with Protease inhibitor cocktail (Roche 11836170001) and De-ubiquitination/sumoylation Inhibitor (Cytoskeleton
NEM09BB) was incubated with Anti-Flag M2 Affinity Gel or Ubiquitination Affinity Beads (UAB, Cytoskeleton UBA01- Beads) overnight at 4 °C. After washing, purified proteins were loaded on
4–20% Mini-PROTEAN TGX Gel (Bio-rad 456-1093). For detection of ubiquitinated Clr4 protein, anti-Flag (Sigma F7425) or anti-ubiquitin (Cell Signalling 3936T) antibodies were used. Cells were
also treated with proteasome inhibitor 100 μM MG-132 (APExBIO A2585) for 4 h before harvesting to detect poly-ubiquitinated Bdf2-Flag or to see the effect of blocking proteasome pathway by
treating MG-132 on mono-ubiquitinated Flag-Clr4. CLRC COMPLEX PURIFICATION FROM _S. POMBE_ Protein purifications were performed as described previously15. Briefly, Rik1-TAP were purified
from 6 to 12 g of cells. Cells were lysed in 1 volume of lysis buffer (50 mM HEPES-KOH, pH 7.6, 300 mM KAc, 10% glycerol, 1 mM EGTA, 1 mM EDTA, 0.1% NP-40, 1 mM DTT, 5 mM MgAc, 1 mM NaF, 20
mM β-GP, 1 mM PMSF, 1 mM Benzamidine and 1 μg/ml of Leupeptin, Aprotinin, Bestatin and Pepstatin). 1 volume lysis buffer was added and the lysate was centrifuged at 16,500 rpm for 25 min at
4 °C in a SA600 rotor. The first affinity purification was performed using Ig-G Sepharose beads at 4 °C for 2 h. The beads were washed with lysis buffer and TEV-C buffer (10 mM Tris-HCl, pH
8.0, 150 mM KAc, 0.1% NP-40, 0.5 mM EDTA, 1 mM DTT) and bound protein was eluted using a GST-TEV protease. The eluate was diluted in 2 volumes of CAM-B buffer (10 mM Tris-HCL, pH 8.0, 150 mM
NaCl, 1 mM MgAc, 1 mM Imidazole, 2 mM CaCl2, 10 mM βME) and the second affinity purification was performed by adding Calmodulin-Sepharose at 4 °C for 1 h. The beads lysate was washed with
CAM-B buffer containing 0.1 % NP-40 and bound protein was eluted with CAM-E buffer (10 mM Tris-HCL, pH 8.0, 150 mM NaCl, 1 mM MgAc, 1 mM Imidazole, 10 mM EGTA, 10 mM βME). Purified CLRC
complex was added to in vitro ubiquitination assay of 6xHis-Clr4. RECOMBINANT PROTEIN PURIFICATION FROM E. COLI, IN VITRO UBIQUITINATION AND NUCLEOSOME BINDING ASSAY BL21-CodonPlus (DE3)-RIL
strains (Agilent Technologies 230245) that express recombinant Clr4 proteins were grown with 0.4 mM IPTG for 20 h at 18 °C. Induced proteins were purified on HisPur Ni-NTA Superflow Agarose
(ThermoFisher 25214) and eluted with elution buffer containing 150 mM Imidazole. The recombinant proteins purified from E. coli were further dialyzed against 1x TBS using Slide-A-Lyzer
Dialysis Cassettes (ThermoFisher 66003). For in vitro ubiquitination, recombinant 6xHis-Clr4 protein or 3xFlag-Clr4 purified form _S. pombe_ cells were mixed with human Ubiquitin-activating
Enzyme (UBE1, R&D systems E-305), human UbcH5c/UBE2D3 (R&D systems E2-627), human Ubiquitin (R&D systems U-100H) or human HA-Ubiquitin (R&D systems U-110), Mg-ATP (R&D
systems B-20) and 10x E3 Ligase Reaction Buffer (R&D systems B-71) with or without CLRC complex purified from _S. pombe_ cells. For nucleosome or ncRNA binding assay, biotin labeled
Recombinant Mononucleosomes (H3.1, Active Motif 31467) and Recombinant Mononucleosomes H3K9me3 (H3.2, Active Motif 31555) or biotin labeled ncRNA (synthesized from IDT) were pre-incubated
with Streptavidin beads (GE Healthcare 17-5113-01) and incubated in vitro ubiquitinated 6xHis-Clr4 or Flag-Clr4 proteins purified from _S. pombe_ cells. Samples were washed, boiled and
loaded on 4–20% Mini-PROTEAN TGX Gel (Bio-rad 456-1093) and WB was performed with anti-Flag (Abcam ab1791), anti-H3K9me3 (Abcam ab8898) and anti-H3 (Abcam ab1791), and using ECL (Thermo
Fisher 32209) assay according to the manufacturer’s instruction. MASS SPECTROMETRY The gel band sample was reduced and alkylated with _N_-ethylmaleimide (NEM), digested with trypsin for 1.5
h at 37 °C and washed with ammonium bicarbonate/acetonitrile to remove stain, SDS and other reagents. Peptides were extracted from the gel pieces, dried down and re-dissolved in 2.5%
acetonitrile, 0.1% formic acid. Each digest was run by nanoLC-MS/MS using a 2 h gradient on a 0.075 mm x 250 mm C18 column feeding into a Q-Exactive HF mass spectrometer. All MS/MS samples
were analyzed using Mascot (Matrix Science, London, UK; version 2.6.1). Mascot was set up to search the cRAP_20150130.fasta; custom4_20190215.fasta; SwissProt_2019_02 database (selected for
_Schizosaccharomyces pombe_, unknown version, 5269 entries) assuming the digestion enzyme trypsin. Mascot was searched with a fragment ion mass tolerance of 0.060 Da and a parent ion
tolerance of 10.0 PPM. Deamidated of asparagine and glutamine, oxidation of methionine, phospho of serine, threonine and tyrosine, GG of lysine, N-ethylmaleimide of cysteine were specified
in Mascot as variable modifications. Scaffold (version Scaffold_4.8.9, Proteome Software Inc., Portland, OR) was used to validate MS/MS based peptide and protein identifications. Peptide
identifications were accepted if they could be established at greater than 80.0% probability by the Peptide Prophet algorithm111 with Scaffold delta-mass correction. Protein identifications
were accepted if they could be established at greater than 99.0% probability and contained at least 2 identified peptides. Protein probabilities were assigned by the Protein Prophet
algorithm112. Proteins that contained similar peptides and could not be differentiated based on MS/MS analysis alone were grouped to satisfy the principles of parsimony. Proteins sharing
significant peptide evidence were grouped into clusters. LIQUID-LIQUID PHASE SEPARATION (LLPS) LLPS of GFP-Clr4, mCherry-Clr4, GFP-Clr4IDR and Clr4IDR-GFP proteins were induced by diluting
proteins to 100–500 nM range in 10 mM HEPES buffer (pH 7.6). To observe the effect of ncRNA on Clr4 LLPS, 120nt synthesized centromeric ncRNA (IDT) was added to the Clr4 proteins. To observe
the effect of 1,6-Hexanediol (Sigma H11807) on GFP-Clr4IDR LLPS, GFP-Clr4IDR protein was diluted to 10 mM HEPES buffers (pH 7.6) containing 5% Ethanol or 5% Hexanediol. LLPS was imaged on
Zeiss Axio Imager M2 with differential interference contrast (DIC) and EGFP channels and processed using Zeiss Zen software. FLUORESCENCE RECOVERY AFTER PHOTOBLEACHING (FRAP) EXPERIMENTS
FRAP experiments were performed as previously described in ref. 113. Briefly, 500 nM of GFP-Clr4IDR protein were diluted in 3.6% PEG8000. PEG8000 was used to stabilize phase-separated liquid
droplets during the experiments. FRAP experiments were carried out using Zeiss LSM710 Confocal Laser Scanning Microscope with 63x objective. All Bleaching experiments were performed with
488 nm argon laser at 100% intensity. Images before and after photobleaching were taken at 5 s interval and analyzed using standard methods. IN VITRO PULL DOWN ASSAY 4 μg of Flag antibody
(Sigma F1804) was pre-incubated with 50 μl Protein A and Protein G Sepharose (GE Healthcare 17513801 and 17061801) mix in 1 mL lysis buffer (40 mM Tris-HCl, pH 7.5, 1 mM EDTA, 10% Glycerol,
0.1% NP-40, 250 mM NaCl) for 1 h and then incunbated with recombinant Flag-Clr4 and Myc-Clr4 proteins (500 nM each) and different amount of ncRNA (0, 50, 100 and 200 nM) in 500 μL lysis
buffer at 4 °C for 3 h. After washing, purified proteins were boiled and loaded on 4–20% Mini-PROTEAN TGX Gel (Bio-rad 456-1093) and Western blotting was performed with anti-Myc (Abcam
ab9106), and anti-Flag (Sigma, F7425) using ECL (Thermo Fisher 32209) assay according to the manufacturer’s instruction. IN VITRO HISTONE METHYLATION (HMT) ASSAY Recombinant Flag-Clr4
protein (5 μM) were mixed with recombinant H3N-GST (10 μM), 200 μM _S_-(5′-Adenosyl)-_L_-methionine chloride dihydrochloride (SAM, Sigma 7007) and different amoount of ncRNA (0, 0.5, 1 and 2
μM) in 50 μL HMT buffer (50mM Tris, pH 8.0, 20 mM KCl, 10 mM MgCl2, 5% Glycerol, 1 mM DTT and 1 mM PMSF) and incubated at 30 °C for 1.25 h. Samples were boiled and loaded on 4–20%
Mini-PROTEAN TGX Gel (Bio-rad 456-1093) and Western blotting was performed with anti-H3K9me2 (Abcam ab1220), and anti-H3K9me3 (Absolute Antibody Ab00700-1.26) using ECL (Thermo Fisher 32209)
assay according to the manufacturer’s instruction. STATISTICS AND REPRODUCIBILITY The experiments for Figs. 1b, e, 2b, d, 3d, 4a, c–f, 5c–d and Supplementary Figs. 1c, 3, 4, 5e–I, 7c, 8d,
f, 10e, f, 11a, 12f, g were performed at least twice and similar results were obtained. The representative results were shown in the figures. SOURCE DATA FILE For an example of presentation
of full scan blots, see the Source Data file of https://www.nature.com/articles/s41467-020-16984-1#Sec35 and for more information, please refer to
https://www.nature.com/nature-research/editorial-policies/image-integrity. REPORTING SUMMARY Further information on research design is available in the Nature Portfolio Reporting Summary
linked to this article. DATA AVAILABILITY Genome-wide datasets are deposited in the Gene Expression Omnibus (GEO) under the accession number GSE156069. Source Data is provided. REFERENCES *
Castel, S. E. & Martienssen, R. A. RNA interference in the nucleus: roles for small RNAs in transcription, epigenetics and beyond. _Nat. Rev. Genet_ 14, 100–112 (2013). Article CAS
PubMed Google Scholar * Janssen, A., Colmenares, S. U. & Karpen, G. H. Heterochromatin: Guardian of the Genome. _Annu Rev. Cell Dev. Biol._ 34, 265–288 (2018). Article CAS PubMed
Google Scholar * Allshire, R. C. & Madhani, H. D. Ten principles of heterochromatin formation and function. _Nat. Rev. Mol. Cell Biol._ 19, 229–244 (2018). Article CAS PubMed Google
Scholar * Martienssen, R. & Moazed, D. RNAi and heterochromatin assembly. _Cold Spring Harb. Perspect. Biol._ 7, a019323 (2015). Article PubMed Google Scholar * Gutbrod, M. J. et al.
Dicer promotes genome stability via the bromodomain transcriptional co-activator BRD4. _Nat. Commun._ 13, 1001 (2022). Article ADS CAS PubMed Google Scholar * Hall, I. M., Noma, K.
& Grewal, S. I. RNA interference machinery regulates chromosome dynamics during mitosis and meiosis in fission yeast. _Proc. Natl Acad. Sci. USA_ 100, 193–198 (2003). Article ADS CAS
PubMed Google Scholar * Provost, P. et al. Dicer is required for chromosome segregation and gene silencing in fission yeast cells. _Proc. Natl Acad. Sci. USA_ 99, 16648–16653 (2002).
Article ADS CAS PubMed Google Scholar * Bannister, A. J. et al. Selective recognition of methylated lysine 9 on histone H3 by the HP1 chromo domain. _Nature_ 410, 120–124 (2001).
Article ADS CAS PubMed Google Scholar * Nakayama, J., Rice, J. C., Strahl, B. D., Allis, C. D. & Grewal, S. I. Role of histone H3 lysine 9 methylation in epigenetic control of
heterochromatin assembly. _Science_ 292, 110–113 (2001). Article ADS CAS PubMed Google Scholar * Rea, S. et al. Regulation of chromatin structure by site-specific histone H3
methyltransferases. _Nature_ 406, 593–599 (2000). Article ADS CAS PubMed Google Scholar * Colmenares, S. U., Buker, S. M., Buhler, M., Dlakić, M. & Moazed, D. Coupling of
double-stranded RNA synthesis and siRNA generation in fission yeast RNAi. _Mol. Cell_ 27, 449–461 (2007). Article CAS PubMed Google Scholar * Djupedal, I. et al. Analysis of small RNA in
fission yeast; centromeric siRNAs are potentially generated through a structured RNA. _Embo J._ 28, 3832–3844 (2009). Article CAS PubMed Google Scholar * Motamedi, M. R. et al. Two RNAi
complexes, RITS and RDRC, physically interact and localize to noncoding centromeric RNAs. _Cell_ 119, 789–802 (2004). Article CAS PubMed Google Scholar * Volpe, T. A. et al. Regulation
of heterochromatic silencing and histone H3 lysine-9 methylation by RNAi. _Science_ 297, 1833–1837 (2002). Article ADS CAS PubMed Google Scholar * Verdel, A. et al. RNAi-mediated
targeting of heterochromatin by the RITS complex. _Science_ 303, 672–676 (2004). Article ADS CAS PubMed Google Scholar * Gutbrod, M. J. & Martienssen, R. A. Conserved chromosomal
functions of RNA interference. _Nat. Rev. Genet_ 21, 311–331 (2020). Article CAS PubMed Google Scholar * Chen, E. S. et al. Cell cycle control of centromeric repeat transcription and
heterochromatin assembly. _Nature_ 451, 734–737 (2008). Article ADS CAS PubMed Google Scholar * Jih, G. et al. Unique roles for histone H3K9me states in RNAi and heritable silencing of
transcription. _Nature_ 547, 463–467 (2017). Article CAS PubMed Google Scholar * Kloc, A., Zaratiegui, M., Nora, E. & Martienssen, R. RNA interference guides histone modification
during the S phase of chromosomal replication. _Curr. Biol._ 18, 490–495 (2008). Article CAS PubMed Google Scholar * Cappadocia, L. & Lima, C. D. Ubiquitin-like Protein Conjugation:
Structures, Chemistry, and Mechanism. _Chem. Rev._ 118, 889–918 (2018). Article CAS PubMed Google Scholar * Hochstrasser, M. Origin and function of ubiquitin-like proteins. _Nature_ 458,
422–429 (2009). Article ADS CAS PubMed Google Scholar * Rape, M. Ubiquitylation at the crossroads of development and disease. _Nat. Rev. Mol. Cell Biol._ 19, 59–70 (2018). Article CAS
PubMed Google Scholar * Swatek, K. N. & Komander, D. Ubiquitin modifications. _Cell Res_. 26, 399–422 (2016). Article CAS PubMed Google Scholar * Ye, Y. & Rape, M. Building
ubiquitin chains: E2 enzymes at work. _Nat. Rev. Mol. Cell Biol._ 10, 755–764 (2009). Article CAS PubMed Google Scholar * Hong, E. J., Villén, J., Gerace, E. L., Gygi, S. P. &
Moazed, D. A cullin E3 ubiquitin ligase complex associates with Rik1 and the Clr4 histone H3-K9 methyltransferase and is required for RNAi-mediated heterochromatin formation. _RNA Biol._ 2,
106–111 (2005). Article CAS PubMed Google Scholar * Horn, P. J., Bastie, J. N. & Peterson, C. L. A Rik1-associated, cullin-dependent E3 ubiquitin ligase is essential for
heterochromatin formation. _Genes Dev._ 19, 1705–1714 (2005). Article CAS PubMed Google Scholar * Jia, S., Kobayashi, R. & Grewal, S. I. Ubiquitin ligase component Cul4 associates
with Clr4 histone methyltransferase to assemble heterochromatin. _Nat. Cell Biol._ 7, 1007–1013 (2005). Article CAS PubMed Google Scholar * Kuscu, C. et al. CRL4-like Clr4 complex in
Schizosaccharomyces pombe depends on an exposed surface of Dos1 for heterochromatin silencing. _Proc. Natl Acad. Sci. USA_ 111, 1795–1800 (2014). Article ADS CAS PubMed Google Scholar *
Thon, G. et al. The Clr7 and Clr8 directionality factors and the Pcu4 cullin mediate heterochromatin formation in the fission yeast Schizosaccharomyces pombe. _Genetics_ 171, 1583–1595
(2005). Article CAS PubMed Google Scholar * Li, F. et al. Two novel proteins, dos1 and dos2, interact with rik1 to regulate heterochromatic RNA interference and histone modification.
_Curr. Biol._ 15, 1448–1457 (2005). Article CAS PubMed Google Scholar * Banani, S. F., Lee, H. O., Hyman, A. A. & Rosen, M. K. Biomolecular condensates: organizers of cellular
biochemistry. _Nat. Rev. Mol. Cell Biol._ 18, 285–298 (2017). Article CAS PubMed Google Scholar * Brangwynne, C. P. et al. Germline P granules are liquid droplets that localize by
controlled dissolution/condensation. _Science_ 324, 1729–1732 (2009). Article ADS CAS PubMed Google Scholar * Kato, M. et al. Cell-free formation of RNA granules: low complexity
sequence domains form dynamic fibers within hydrogels. _Cell_ 149, 753–767 (2012). Article CAS PubMed Google Scholar * Rhine, K., Vidaurre, V. & Myong, S. RNA Droplets. _Annu Rev.
Biophys._ 49, 247–265 (2020). Article CAS PubMed Google Scholar * Rippe, K. Liquid-Liquid Phase Separation in Chromatin. _Cold Spring Harb Perspect Biol_ 14,
https://doi.org/10.1101/cshperspect.a040683 (2022). * Weber, S. C. & Brangwynne, C. P. Getting RNA and protein in phase. _Cell_ 149, 1188–1191 (2012). Article CAS PubMed Google
Scholar * Molliex, A. et al. Phase separation by low complexity domains promotes stress granule assembly and drives pathological fibrillization. _Cell_ 163, 123–133 (2015). Article CAS
PubMed Google Scholar * Mittag, T. & Parker, R. Multiple Modes of Protein-Protein Interactions Promote RNP Granule Assembly. _J. Mol. Biol._ 430, 4636–4649 (2018). Article CAS PubMed
Google Scholar * Gibson, B. A. et al. Organization of Chromatin by Intrinsic and Regulated Phase Separation. _Cell_ 179, 470–484.e421 (2019). Article CAS PubMed Google Scholar *
Larson, A. G. et al. Liquid droplet formation by HP1α suggests a role for phase separation in heterochromatin. _Nature_ 547, 236–240 (2017). Article ADS CAS PubMed Google Scholar *
Sabari, B. R. et al. Coactivator condensation at super-enhancers links phase separation and gene control. _Science_ 361, https://doi.org/10.1126/science.aar3958 (2018). * Strom, A. R. et al.
Phase separation drives heterochromatin domain formation. _Nature_ 547, 241–245 (2017). Article ADS CAS PubMed Google Scholar * Wang, L. et al. Histone Modifications Regulate Chromatin
Compartmentalization by Contributing to a Phase Separation Mechanism. _Mol. Cell_ 76, 646–659.e646 (2019). Article CAS PubMed Google Scholar * Irvine, D. V. et al. Mapping epigenetic
mutations in fission yeast using whole-genome next-generation sequencing. _Genome Res_. 19, 1077–1083 (2009). Article CAS PubMed Google Scholar * Javerzat, J. P., Cranston, G. &
Allshire, R. C. Fission yeast genes which disrupt mitotic chromosome segregation when overexpressed. _Nucleic Acids Res_. 24, 4676–4683 (1996). Article CAS PubMed Google Scholar *
Schalch, T. et al. High-affinity binding of Chp1 chromodomain to K9 methylated histone H3 is required to establish centromeric heterochromatin. _Mol. Cell_ 34, 36–46 (2009). Article CAS
PubMed Google Scholar * Ishimoto, K. et al. Ubiquitination of Lysine 867 of the Human SETDB1 Protein Upregulates Its Histone H3 Lysine 9 (H3K9) Methyltransferase Activity. _PLoS One_ 11,
e0165766 (2016). Article PubMed Google Scholar * Sun, L. & Fang, J. E3-Independent Constitutive Monoubiquitination Complements Histone Methyltransferase Activity of SETDB1. _Mol.
Cell_ 62, 958–966 (2016). Article CAS PubMed Google Scholar * Iglesias, N. et al. Native Chromatin Proteomics Reveals a Role for Specific Nucleoporins in Heterochromatin Organization and
Maintenance. _Mol. Cell_ 77, 51–66.e58 (2020). Article CAS PubMed Google Scholar * Hoeller, D. et al. E3-independent monoubiquitination of ubiquitin-binding proteins. _Mol. Cell_ 26,
891–898 (2007). Article CAS PubMed Google Scholar * Akoury, E. et al. Disordered region of H3K9 methyltransferase Clr4 binds the nucleosome and contributes to its activity. _Nucleic
Acids Res_. 47, 6726–6736 (2019). Article CAS PubMed Google Scholar * Iglesias, N. et al. Automethylation-induced conformational switch in Clr4 (Suv39h) maintains epigenetic stability.
_Nature_ 560, 504–508 (2018). Article ADS CAS PubMed Google Scholar * Zhang, K., Mosch, K., Fischle, W. & Grewal, S. I. Roles of the Clr4 methyltransferase complex in nucleation,
spreading and maintenance of heterochromatin. _Nat. Struct. Mol. Biol._ 15, 381–388 (2008). Article CAS PubMed Google Scholar * Ishida, M. et al. Intrinsic nucleic acid-binding activity
of Chp1 chromodomain is required for heterochromatic gene silencing. _Mol. Cell_ 47, 228–241 (2012). Article CAS PubMed Google Scholar * Kagansky, A. et al. Synthetic heterochromatin
bypasses RNAi and centromeric repeats to establish functional centromeres. _Science_ 324, 1716–1719 (2009). Article ADS CAS PubMed Google Scholar * Holla, S. et al. Positioning
Heterochromatin at the Nuclear Periphery Suppresses Histone Turnover to Promote Epigenetic Inheritance. _Cell_ 180, 150–164.e115 (2020). Article CAS PubMed Google Scholar * Ragunathan,
K., Jih, G. & Moazed, D. Epigenetics. Epigenetic inheritance uncoupled from sequence-specific recruitment. _Science_ 348, 1258699 (2015). Article PubMed Google Scholar * Shan, C. M.
et al. A histone H3K9M mutation traps histone methyltransferase Clr4 to prevent heterochromatin spreading. _Elife_ 5, https://doi.org/10.7554/eLife.17903 (2016). * Bayne, E. H. et al. Stc1:
a critical link between RNAi and chromatin modification required for heterochromatin integrity. _Cell_ 140, 666–677 (2010). Article CAS PubMed Google Scholar * Ban, H., Sun, W., Chen, Y.
H., Chen, Y. & Li, F. Dri1 mediates heterochromatin assembly via RNAi and histone deacetylation. _Genetics_ 218, https://doi.org/10.1093/genetics/iyab032 (2021). * Shin, Y. &
Brangwynne, C. P. Liquid phase condensation in cell physiology and disease. _Science_ 357, https://doi.org/10.1126/science.aaf4382 (2017). * Alberti, S., Gladfelter, A. & Mittag, T.
Considerations and Challenges in Studying Liquid-Liquid Phase Separation and Biomolecular Condensates. _Cell_ 176, 419–434 (2019). Article CAS PubMed Google Scholar * Kroschwald, S. et
al. Promiscuous interactions and protein disaggregases determine the material state of stress-inducible RNP granules. _Elife_ 4, e06807 (2015). Article PubMed PubMed Central Google
Scholar * Canzio, D. et al. Chromodomain-mediated oligomerization of HP1 suggests a nucleosome-bridging mechanism for heterochromatin assembly. _Mol. Cell_ 41, 67–81 (2011). Article CAS
PubMed Google Scholar * Hinde, E., Cardarelli, F. & Gratton, E. Spatiotemporal regulation of Heterochromatin Protein 1-alpha oligomerization and dynamics in live cells. _Sci. Rep._ 5,
12001 (2015). Article ADS CAS PubMed Google Scholar * Müller-Ott, K. et al. Specificity, propagation, and memory of pericentric heterochromatin. _Mol. Syst. Biol._ 10, 746 (2014).
Article PubMed Google Scholar * Elbaum-Garfinkle, S. et al. The disordered P granule protein LAF-1 drives phase separation into droplets with tunable viscosity and dynamics. _Proc. Natl
Acad. Sci. USA_ 112, 7189–7194 (2015). Article ADS CAS PubMed Google Scholar * Maharana, S. et al. RNA buffers the phase separation behavior of prion-like RNA binding proteins.
_Science_ 360, 918–921 (2018). Article ADS CAS PubMed Google Scholar * Van Lindt, J. et al. F/YGG-motif is an intrinsically disordered nucleic-acid binding motif. _RNA Biol._ 19,
622–635 (2022). Article PubMed Google Scholar * Oya, E. et al. H3K14 ubiquitylation promotes H3K9 methylation for heterochromatin assembly. _EMBO Rep._ 20, e48111 (2019). Article CAS
PubMed Google Scholar * Aagaard, L. et al. Functional mammalian homologues of the Drosophila PEV-modifier Su(var)3-9 encode centromere-associated proteins which complex with the
heterochromatin component M31. _Embo J._ 18, 1923–1938 (1999). Article CAS PubMed Google Scholar * Yamamoto, K. & Sonoda, M. Self-interaction of heterochromatin protein 1 is required
for direct binding to histone methyltransferase, SUV39H1. _Biochem Biophys. Res. Commun._ 301, 287–292 (2003). Article CAS PubMed Google Scholar * Sanulli, S. et al. HP1 reshapes
nucleosome core to promote phase separation of heterochromatin. _Nature_ 575, 390–394 (2019). Article ADS CAS PubMed Google Scholar * Pidoux, A. L., Uzawa, S., Perry, P. E., Cande, W.
Z. & Allshire, R. C. Live analysis of lagging chromosomes during anaphase and their effect on spindle elongation rate in fission yeast. _J. Cell Sci._ 113, 4177–4191 (2000). Article CAS
PubMed Google Scholar * Braun, S. et al. The Cul4-Ddb1(Cdt)² ubiquitin ligase inhibits invasion of a boundary-associated antisilencing factor into heterochromatin. _Cell_ 144, 41–54
(2011). Article CAS PubMed Google Scholar * Trewick, S. C., Minc, E., Antonelli, R., Urano, T. & Allshire, R. C. The JmjC domain protein Epe1 prevents unregulated assembly and
disassembly of heterochromatin. _Embo j._ 26, 4670–4682 (2007). Article CAS PubMed Google Scholar * Zofall, M. & Grewal, S. I. Swi6/HP1 recruits a JmjC domain protein to facilitate
transcription of heterochromatic repeats. _Mol. Cell_ 22, 681–692 (2006). Article CAS PubMed Google Scholar * Keller, C., Kulasegaran-Shylini, R., Shimada, Y., Hotz, H. R. & Bühler,
M. Noncoding RNAs prevent spreading of a repressive histone mark. _Nat. Struct. Mol. Biol._ 20, 994–1000 (2013). Article CAS PubMed Google Scholar * Kim, H. S. et al. Identification of a
BET family bromodomain/casein kinase II/TAF-containing complex as a regulator of mitotic condensin function. _Cell Rep._ 6, 892–905 (2014). Article CAS PubMed Google Scholar * Wang, J.
et al. Epe1 recruits BET family bromodomain protein Bdf2 to establish heterochromatin boundaries. _Genes Dev._ 27, 1886–1902 (2013). Article CAS PubMed Google Scholar * Komander, D.,
Clague, M. J. & Urbé, S. Breaking the chains: structure and function of the deubiquitinases. _Nat. Rev. Mol. Cell Biol._ 10, 550–563 (2009). Article CAS PubMed Google Scholar *
Leznicki, P. & Kulathu, Y. Mechanisms of regulation and diversification of deubiquitylating enzyme function. _J. Cell Sci._ 130, 1997–2006 (2017). Article CAS PubMed Google Scholar *
Reyes-Turcu, F. E., Zhang, K., Zofall, M., Chen, E. & Grewal, S. I. Defects in RNA quality control factors reveal RNAi-independent nucleation of heterochromatin. _Nat. Struct. Mol.
Biol._ 18, 1132–1138 (2011). Article CAS PubMed Google Scholar * Ryan, C. J. et al. Hierarchical modularity and the evolution of genetic interactomes across species. _Mol. Cell_ 46,
691–704 (2012). Article CAS PubMed Google Scholar * Liu, Y. et al. Functional characterization of the Arabidopsis ubiquitin-specific protease gene family reveals specific role and
redundancy of individual members in development. _Plant J._ 55, 844–856 (2008). Article CAS PubMed Google Scholar * Bühler, M., Spies, N., Bartel, D. P. & Moazed, D. TRAMP-mediated
RNA surveillance prevents spurious entry of RNAs into the Schizosaccharomyces pombe siRNA pathway. _Nat. Struct. Mol. Biol._ 15, 1015–1023 (2008). Article PubMed Google Scholar * Kuzdere,
T. et al. Differential phosphorylation of Clr4(SUV39H) by Cdk1 accompanies a histone H3 methylation switch that is essential for gametogenesis. _EMBO Rep_. e55928,
https://doi.org/10.15252/embr.202255928 (2022). * Babu, M. M. The contribution of intrinsically disordered regions to protein function, cellular complexity, and human disease. _Biochem Soc.
Trans._ 44, 1185–1200 (2016). Article CAS PubMed Google Scholar * Bah, A. & Forman-Kay, J. D. Modulation of Intrinsically Disordered Protein Function by Post-translational
Modifications. _J. Biol. Chem._ 291, 6696–6705 (2016). Article CAS PubMed Google Scholar * Bah, A. et al. Folding of an intrinsically disordered protein by phosphorylation as a
regulatory switch. _Nature_ 519, 106–109 (2015). Article ADS CAS PubMed Google Scholar * Oldfield, C. J. & Dunker, A. K. Intrinsically disordered proteins and intrinsically
disordered protein regions. _Annu Rev. Biochem_ 83, 553–584 (2014). Article CAS PubMed Google Scholar * Qamar, S. et al. FUS Phase Separation Is Modulated by a Molecular Chaperone and
Methylation of Arginine Cation-π Interactions. _Cell_ 173, 720–734.e715 (2018). Article CAS PubMed Google Scholar * Stowell, J. A. W. et al. A low-complexity region in the YTH domain
protein Mmi1 enhances RNA binding. _J. Biol. Chem._ 293, 9210–9222 (2018). Article CAS PubMed Google Scholar * Yang, P. et al. G3BP1 Is a Tunable Switch that Triggers Phase Separation to
Assemble Stress Granules. _Cell_ 181, 325–345.e328 (2020). Article CAS PubMed Google Scholar * Keller, C. et al. HP1(Swi6) mediates the recognition and destruction of heterochromatic
RNA transcripts. _Mol. Cell_ 47, 215–227 (2012). Article CAS PubMed Google Scholar * Johnson, W. L. et al. RNA-dependent stabilization of SUV39H1 at constitutive heterochromatin. _Elife_
6, https://doi.org/10.7554/eLife.25299 (2017). * Shirai, A. et al. Impact of nucleic acid and methylated H3K9 binding activities of Suv39h1 on its heterochromatin assembly. _Elife_ 6,
https://doi.org/10.7554/eLife.25317 (2017). * Velazquez Camacho, O. et al. Major satellite repeat RNA stabilize heterochromatin retention of Suv39h enzymes by RNA-nucleosome association and
RNA:DNA hybrid formation. _Elife_ 6, https://doi.org/10.7554/eLife.25293 (2017). * Gerace, E. L., Halic, M. & Moazed, D. The methyltransferase activity of Clr4Suv39h triggers RNAi
independently of histone H3K9 methylation. _Mol. Cell_ 39, 360–372 (2010). Article CAS PubMed Google Scholar * Bühler, M., Verdel, A. & Moazed, D. Tethering RITS to a nascent
transcript initiates RNAi- and heterochromatin-dependent gene silencing. _Cell_ 125, 873–886 (2006). Article PubMed Google Scholar * Al-Sady, B., Madhani, H. D. & Narlikar, G. J.
Division of labor between the chromodomains of HP1 and Suv39 methylase enables coordination of heterochromatin spread. _Mol. Cell_ 51, 80–91 (2013). Article CAS PubMed Google Scholar *
Chang, A. Y., Castel, S. E., Ernst, E., Kim, H. S. & Martienssen, R. A. The Conserved RNA Binding Cyclophilin, Rct1, Regulates Small RNA Biogenesis and Splicing Independent of
Heterochromatin Assembly. _Cell Rep._ 19, 2477–2489 (2017). Article CAS PubMed Google Scholar * Bähler, J. et al. Heterologous modules for efficient and versatile PCR-based gene
targeting in Schizosaccharomyces pombe. _Yeast_ 14, 943–951 (1998). Article PubMed Google Scholar * Murray, J. M., Watson, A. T. & Carr, A. M. Transformation of Schizosaccharomyces
pombe: Lithium Acetate/ Dimethyl Sulfoxide Procedure. _Cold Spring Harb. Protoc._ 2016, https://doi.org/10.1101/pdb.prot090969 (2016). Article PubMed Google Scholar * Winston, F. EMS and
UV mutagenesis in yeast. _Curr. Protoc. Mol. Biol._ CHAPTER 13, Unit 13.13B (2008). Google Scholar * Roche, B., Arcangioli, B. & Martienssen, R. A. RNA interference is essential for
cellular quiescence. _Science_ 354, https://doi.org/10.1126/science.aah5651 (2016). * Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. _Nat. Methods_ 9, 357–359
(2012). Article CAS PubMed Google Scholar * Garrison, E. & Marth, G. Haplotype-based variant detection from short-read sequencing. _arXiv e-prints_, arXiv:1207.3907 (2012).
<https://ui.adsabs.harvard.edu/abs/2012arXiv1207.3907G>. * Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to
the human genome. _Genome Biol._ 10, R25 (2009). Article PubMed PubMed Central Google Scholar * Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing
genomic features. _Bioinformatics_ 26, 841–842 (2010). Article CAS PubMed Google Scholar * Keller, A., Nesvizhskii, A. I., Kolker, E. & Aebersold, R. Empirical statistical model to
estimate the accuracy of peptide identifications made by MS/MS and database search. _Anal. Chem._ 74, 5383–5392 (2002). Article CAS PubMed Google Scholar * Nesvizhskii, A. I., Keller,
A., Kolker, E. & Aebersold, R. A statistical model for identifying proteins by tandem mass spectrometry. _Anal. Chem._ 75, 4646–4658 (2003). Article CAS PubMed Google Scholar * Wang,
H. et al. Rubisco condensate formation by CcmM in β-carboxysome biogenesis. _Nature_ 566, 131–135 (2019). Article ADS CAS PubMed Google Scholar Download references ACKNOWLEDGEMENTS We
thank all Martienssen lab members as well as Danesh Moazed and Mo Motamedi for valuable discussions and suggestions. The authors acknowledge assistance from the Cold Spring Harbor Laboratory
Shared Resources, which are funded in part by the Cancer Center Support Grant (5PP30CA045508). This work was supported by the Howard Hughes Medical Institute, and a grant from the National
Institute of Health (R35GM144206) (to R.A.M.). AUTHOR INFORMATION Author notes * Benjamin Roche Present address: University of North Dakota, School of Medicine & Health Sciences, 1301 N
Columbia Rd. Stop 9037, Grand Forks, ND, 58202, USA AUTHORS AND AFFILIATIONS * Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, 11724, USA Hyun-Soo Kim, Benjamin Roche, Sonali
Bhattacharjee, An-Yun Chang, Christopher Hammell & Robert A. Martienssen * Howard Hughes Medical Institute, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
Hyun-Soo Kim & Robert A. Martienssen * Institute for Advanced Biosciences, UMR InsermU1209/CNRS5309/UGA, University of Grenoble Alpes, Grenoble, France Leila Todeschini & André
Verdel Authors * Hyun-Soo Kim View author publications You can also search for this author inPubMed Google Scholar * Benjamin Roche View author publications You can also search for this
author inPubMed Google Scholar * Sonali Bhattacharjee View author publications You can also search for this author inPubMed Google Scholar * Leila Todeschini View author publications You can
also search for this author inPubMed Google Scholar * An-Yun Chang View author publications You can also search for this author inPubMed Google Scholar * Christopher Hammell View author
publications You can also search for this author inPubMed Google Scholar * André Verdel View author publications You can also search for this author inPubMed Google Scholar * Robert A.
Martienssen View author publications You can also search for this author inPubMed Google Scholar CONTRIBUTIONS H.S.K. performed experiments and bioinformatics. B.R. contributed sRNA-seq
experiments and bioinformatics. A.Y.C. contributed to EMS mutagenesis, C.H. contributed to LLPS. S.B. LT and AV contributed to protein purification. H.S.K. and R.A.M. designed the
experiments and wrote the manuscript. CORRESPONDING AUTHOR Correspondence to Robert A. Martienssen. ETHICS DECLARATIONS COMPETING INTERESTS The authors declare no competing interests. PEER
REVIEW PEER REVIEW INFORMATION _Nature Communications_ thanks the anonymous reviewer(s) for their contribution to the peer review of this work. A peer review file is available. ADDITIONAL
INFORMATION PUBLISHER’S NOTE Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. SUPPLEMENTARY INFORMATION SUPPLEMENTARY
INFORMATION PEER REVIEW FILE REPORTING SUMMARY RIGHTS AND PERMISSIONS OPEN ACCESS This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use,
sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative
Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated
otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds
the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. Reprints and
permissions ABOUT THIS ARTICLE CITE THIS ARTICLE Kim, HS., Roche, B., Bhattacharjee, S. _et al._ Clr4SUV39H1 ubiquitination and non-coding RNA mediate transcriptional silencing of
heterochromatin via Swi6 phase separation. _Nat Commun_ 15, 9384 (2024). https://doi.org/10.1038/s41467-024-53417-9 Download citation * Received: 21 March 2023 * Accepted: 02 October 2024 *
Published: 30 October 2024 * DOI: https://doi.org/10.1038/s41467-024-53417-9 SHARE THIS ARTICLE Anyone you share the following link with will be able to read this content: Get shareable link
Sorry, a shareable link is not currently available for this article. Copy to clipboard Provided by the Springer Nature SharedIt content-sharing initiative
Trending News
Once a bustling hub, landmark for peace move now stands silentNEW DELHI: The Dr B R Ambedkar Bus Terminus near Delhi Gate no more resembles the once bustling hub, which generated muc...
Rates climb to 1. 35% – rba on mission to whip inflationThere was no suprise in the board of the Reserve Bank of Australia lifting interest rates at its July meeting. The only ...
Over half of americans think you must earn $1 million a year to be rich—here’s what it really takesAmericans have varying ideas of how much money you need to earn each year to be considered "rich," but most pe...
Vicarious liability: what is it and who is at fault? | zegalImagine you brought your Hermès bag to a clubhouse and the waiter spilled red wine on it, ruining your $240,000 bag. Can...
Path to paris challenge | england hockeyTeam GB are looking to get more children and families active through their ‘Path to Paris,’ and this is a perfect way to...
Latests News
Clr4suv39h1 ubiquitination and non-coding rna mediate transcriptional silencing of heterochromatin via swi6 phase separationABSTRACT Transcriptional silencing by RNAi paradoxically relies on transcription, but how the transition from transcript...
What is your experience of a day spent without Wikipedia?In protest against the Stop Online Piracy Act (SOPA) Bill, Wikipedia went black for a day, just like many other websites...
Understanding hurricane risks: 5 essential readsJune 1 marks the start of the 2018 Atlantic hurricane season, with some communities still rebuilding after last year’s l...
Take AARP's Weekly News Quiz: October 31 EditionMemorial Day Sale! Join AARP for just $11 per year with a 5-year membership Join now and get a FREE gift. Expires 6/4 G...
Bare beginnings to healthcare empireMANIPUR: Dr Thangjam Dhabali Singh’s journey from abject poverty in a small Manipuri village to becoming one of Northeas...