Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using...

46
1 Satellite repeat transcripts modulate heterochromatin condensates 1 and safeguard chromosome stability in mouse embryonic stem cells 2 3 4 Clara Lopes Novo 1,2,* , Emily Wong 3 , Colin Hockings 4 , Chetan Poudel 4 , Eleanor Sheekey 1 , Simon Walker 5 , 5 Gabriele S. Kaminski Schierle 4 , Geeta J. Narlikar 3 and Peter J. Rugg-Gunn 1,6,7,* 6 7 8 1 Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK. 9 2 Tommy’s National Miscarriage Research Centre at Imperial College London, W12 0NN, UK. 10 3 Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, 11 USA. 12 4 Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, 13 UK. 14 5 Imaging Facility, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK. 15 6 Wellcome Trust – Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, 16 Cambridge CB2 1QR, UK. 17 *Correspondence and requests for materials should be addressed to C.L.N. (email: [email protected]) and 18 P.J.R.-G. (email: [email protected]) 19 . CC-BY-NC-ND 4.0 International license available under a was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made The copyright holder for this preprint (which this version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642 doi: bioRxiv preprint

Transcript of Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using...

Page 1: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

1

Satellite repeat transcripts modulate heterochromatin condensates 1

and safeguard chromosome stability in mouse embryonic stem cells 2

3

4

Clara Lopes Novo1,2,*, Emily Wong3, Colin Hockings4, Chetan Poudel4, Eleanor Sheekey1, Simon Walker5, 5

Gabriele S. Kaminski Schierle4 , Geeta J. Narlikar3 and Peter J. Rugg-Gunn1,6,7,* 6

7

8

1 Epigenetics Programme, Babraham Institute, Cambridge CB22 3AT, UK. 9

2 Tommy’s National Miscarriage Research Centre at Imperial College London, W12 0NN, UK. 10

3 Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, 11

USA. 12

4 Department of Chemical Engineering and Biotechnology, University of Cambridge, Cambridge CB3 0AS, 13

UK. 14

5 Imaging Facility, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK. 15

6 Wellcome Trust – Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, 16

Cambridge CB2 1QR, UK. 17

*Correspondence and requests for materials should be addressed to C.L.N. (email: [email protected]) and 18

P.J.R.-G. (email: [email protected]) 19

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 2: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

2

Summary 1

Heterochromatin maintains genome integrity and function, and is organised into distinct nuclear domains. 2

Some of these domains are proposed to form by phase separation through the accumulation of HP1ɑ. 3

Mammalian heterochromatin contains noncoding major satellite repeats (MSR), which are highly transcribed 4

in mouse embryonic stem cells (ESCs). Here, we report that MSR transcripts can drive the formation of HP1ɑ 5

droplets in vitro, and scaffold heterochromatin into dynamic condensates in ESCs, leading to the formation 6

of large nuclear domains that are characteristic of pluripotent cells. Depleting MSR transcripts causes 7

heterochromatin to transition into a more compact and static state. Unexpectedly, changing heterochromatin’s 8

biophysical properties has severe consequences for ESCs, including chromosome instability and mitotic 9

defects. These findings uncover an essential role for MSR transcripts in modulating the organisation and 10

properties of heterochromatin to preserve genome stability. They also provide new insights into the processes 11

that could regulate phase separation and the functional consequences of disrupting the properties of 12

heterochromatin condensates. 13

14

15

16

17

18

Keywords 19

Heterochromatin; phase-separation; nuclear organisation; noncoding RNA; genome stability; embryonic 20

stem cells; pluripotency 21

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 3: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

3

Introduction 1

Functional compartmentalisation of the genome segregates repetitive, gene-poor regions into constitutive 2

heterochromatin 1. Establishing and maintaining the appropriate regulation of heterochromatin is essential 3

for preserving nuclear architecture, genome stability and DNA repair, and for silencing transposon expression 4

2,3. In interphase cells, constitutive heterochromatin from different chromosomes cluster in cytologically 5

defined nuclear bodies called chromocenters. These structures are characterised by the presence of histone 6

H3 lysine 9 trimethylation (H3K9me3) and of heterochromatin protein 1ɑ (HP1ɑ) 4–6. In the context of 7

purified components, phosphorylation or DNA binding drives soluble HP1ɑ into phase-separated droplets 7,8. 8

Furthermore, earlier studies showed that human HP1 proteins display dynamics on the order of seconds 9

within heterochromatin puncta 9,10. These and other observations in cells support the possibility that 10

chromocenters could form by phase separation through the localised accumulation of HP1ɑ. A phase-11

separation based model has important implications for how heterochromatin is assembled and regulated. 12

However, the underlying biological processes that could promote HP1ɑ-mediated phase separation in vivo, 13

and the functional consequences of disrupting the phase separation of heterochromatin domains, remain as 14

yet unknown. 15

The regulation of constitutive heterochromatin is developmentally controlled 11 and this regulation is 16

perturbed during cell stress and ageing 12. Interestingly, in mouse, constitutive heterochromatin adopts an 17

unusual configuration in both embryonic stem cells (ESCs) and early embryonic cells, compared to most 18

other cell types, as it has reduced levels of H3K9me3, dynamically binds heterochromatin-associated 19

proteins, and has an uncompacted chromatin structure 13–21. Such distinct properties also extend to cytological 20

differences, for example, constitutive heterochromatin is structured into much larger and fewer 21

chromocenters in ESCs compared to most somatic cells 14,15,22,23. How and why heterochromatin in ESCs 22

adopts this unusual configuration is unknown, although one clue could be that ESCs transcribe high levels of 23

noncoding major satellite repeat (MSR) elements, which constitute the predominant DNA sequences within 24

chromocenters 13,15,19,24. MSR transcripts remain close to their sites of transcription at chromocenters, and 25

biochemical data indicate that MSR RNA can interact with heterochromatin-associated proteins, such as 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 4: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

4

HP1ɑ and SUV39H2 25–27. Importantly, the control of MSR transcription is integrated into the transcription 1

factor networks that sustain ESCs, which suggests that the regulation of heterochromatin in ESCs is an active 2

process that is tightly controlled and coupled to cell state 15. Evidence that MSR transcripts potentially have 3

a functional role in ESCs comes from studies of early mouse development, which demonstrate that MSR 4

RNA is required to establish chromocenter formation and for embryo development to proceed 28–30. An 5

important goal, therefore, is to understand the role that repetitive, noncoding RNAs, such as MSR transcripts, 6

play in promoting the biophysical properties of heterochromatin formation and stability. 7

Here, we report that noncoding satellite RNA alters the physical properties and nuclear organisation 8

of heterochromatin in mouse ESCs. We found that MSR transcripts catalyse a permissive and dynamic 9

environment within individual chromocenters and promote the formation of large heterochromatin foci that 10

are a hallmark of pluripotent cells. We further show that MSR RNA can drive the formation of HP1ɑ droplets 11

in vitro and is required for HP1ɑ organisation within heterochromatin domains in vivo. The reduction of MSR 12

transcripts prompted chromocenter properties to transition to a less dynamic and more stable heterochromatin 13

state, and triggered the reorganisation of chromocenters into more numerous, smaller and compact foci. This 14

aberrant chromocenter reorganisation led to rapid chromosome instability, as reflected by the appearance of 15

chromosome end-fusions and pericentric gaps. These findings thus uncover an unexpected protective 16

function for heterochromatin condensates in preserving genome stability. 17

18

19

Results 20

Constitutive heterochromatin forms liquid-like condensates in mouse embryonic stem cells 21

Studies of HP1ɑ behaviour in Drosophila and in mammalian cells have proposed that heterochromatin is 22

possibly held within liquid-like, phase-separated compartments 7,8. To define the biophysical properties of 23

constitutive heterochromatin in mouse ESCs, we used time-lapse imaging to track the movement and 24

dynamics of individual chromocenters in live cells. For that, we generated a stable ESC line that expressed a 25

doxycycline-inducible, monomeric GFP fusion protein that has a transcription activator-like effector (TALE) 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 5: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

5

targeting the MSR sequence (Figures 1A and B) 31. Fluorescent microscopy confirmed that the induced 1

TALE-MSR-GFP signal localised to chromocenters throughout the cell-cycle (Supplementary Figure 1A and 2

Supplementary Video 1), similar to previous observations 31. As expected, the targeting of TALE-MSR-GFP 3

to MSR elements did not affect chromocenter organisation in ESCs (their number and size, Supplementary 4

Figures 1B and 1C, respectively). Importantly, our time-lapse imaging of TALE-MSR-GFP revealed that 5

individual chromocenters separated and coalesced rapidly, with the movement of chromocenter extrusions 6

occurring within minutes (Figure 1C and Supplementary Video 2). The ability to undergo such dynamic 7

processes are features attributed to liquid-like membraneless condensates 32. Liquid-like condensates are also 8

characterised by the rapid, internal movement of molecules 33,34. And so to further assess the dynamics of 9

molecules within chromocenters of ESCs, we used fluorescence recovery after photo-bleaching (FRAP) to 10

measure the mobile (liquid) and immobile (static) TALE-MSR-GFP-bound chromatin fractions. Because 11

TALEs can bind with strong affinity (low nM Kd values) to their target DNA, both in vitro and in vivo 35,36, 12

this assay allows us to measure the physical properties of chromocenters, including both the movement of 13

heterochromatin and the dynamics of chromatin-associated proteins. One chromocenter per nuclei was 14

bleached, and the recovery of the GFP signal in this region over time quantified. The fluorescent signal 15

recovered quickly at photo-bleached chromocenters, reaching 50% of the initial intensity within ~30s after 16

bleaching (Figure 1D and Supplementary Video 3), with comparable dynamics to other molecules that were 17

previously reported to be within liquid-like condensates 37–39. In addition, an immobile, stable component of 18

∼25% remained after photo-bleaching (Figure 1E), which is in line with measurements in other cell types 19

8,36. The recovery could arise from the dissociation of bleached TALE-MSR-GFP molecules and their 20

replacement with unbleached TALE-MSR-GFP molecules or from movement of the chromatin region itself. 21

Compared to the time-scales that we observe, previous studies in ESCs have reported substantially slower 22

recovery rates of bleached histones (t1/2 = ~100s) and faster recovery of soluble heterochromatin proteins, 23

such as HP1ɑ-GFP (t1/2 = ~3.5s; 14). Thus, we favour the interpretation that we are measuring a combination 24

of both replacement and movement. Based on these results, we conclude that DNA binding factors, such as 25

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 6: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

6

TALE-MSR-GFP, bound to constitutive heterochromatin in ESCs display behaviours that are consistent with 1

being part of a phase-separated compartment that consists of both dynamic and stable components. 2

3

Satellite transcripts regulate constitutive heterochromatin behaviour in embryonic stem cells 4

Chromocenters are distinctively large and less numerous in ESCs 14,22,23, which suggests that an as-yet 5

unknown component acts to promote the fusion of these liquid-like heterochromatin condensates. Because 6

MSR RNA levels are substantially higher in ESCs relative to most other cell types 13,15,24, and because pull-7

down experiments indicate that MSR transcripts can interact with HP1ɑ 25,27, we hypothesised that MSR 8

transcripts might contribute to the formation of heterochromatin condensates. To directly test this hypothesis, 9

we applied a commonly used approach for noncoding RNA studies; we depleted both strands of MSR 10

transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete 11

target transcripts by acting at transcriptional 40 and post-transcriptional steps 41. After transfecting ESCs with 12

LNA DNA gapmers that target the MSR transcripts (MSR gapmers), MSR RNA levels were reduced by 13

~50%, compared to cells transfected with control gapmers, as shown by RT-qPCR analysis (Figure 2A). The 14

decreased level of MSR transcripts after LNA DNA gapmer transfection is comparable to that of MSR 15

transcriptional levels in more differentiated cell types (Supplementary Figure 2A) 15. The levels of other 16

noncoding transcripts, such as of minor satellites or LINEs, were unchanged after MSR gapmer treatment 17

(Figure 2A). In addition, we verified that the depletion of MSR RNA in ESCs did not affect the expression 18

of undifferentiated cell markers or promote cell differentiation (Supplementary Figures 2B and 2C). 19

We next used live-cell imaging of TALE-MSR-GFP to measure chromocenter dynamics in ESCs 20

after depleting MSR transcripts. We confirmed that following the depletion of MSR transcripts, TALE-MSR-21

GFP still localised to chromocenters, as expected (Supplementary Figure 2D). Excitingly, FRAP experiments 22

revealed that the fluorescence recovery time at photo-bleached chromocenters doubled in MSR RNA-23

depleted cells compared to controls, indicating that the dynamic movement of TALE-MSR-GFP molecules 24

within chromocenters was reduced (Figures 2B, 2C and Supplementary Video 4). Notably, the recovery 25

immediately after photo-bleaching was substantially slower in the MSR RNA depleted cells (Figure 2B) and 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 7: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

7

this reduction in recovery is consistent with the TALE proteins being bound within a less dynamic chromatin 1

14. Furthermore, the proportion of mobile GFP signal decreased slightly in the MSR RNA depleted cells 2

(Figure 2D), suggesting that the combination of dynamic and stable components within chromocenters is also 3

affected by MSR RNA levels. Together, these data lead us to propose that MSR RNA catalyses a dynamic, 4

physical environment within individual chromocenters. 5

6

Satellite RNA promotes phase-separation of HP1ɑ 7

We next investigated a potential mechanism by which MSR RNA might contribute to the biophysical 8

properties of heterochromatin. As MSR RNA binds HP1ɑ 25,27, we hypothesised that MSR RNA might affect 9

the ability of HP1ɑ to phase-separate. Excitingly, adding in vitro transcribed MSR RNA (272 nucleotides 10

containing a single repeat sequence) to recombinantly purified human HP1ɑ induced droplet formation in an 11

HP1ɑ concentration-dependent manner (Figure 3A). Furthermore, MSR RNAs containing multiple repeat 12

sequences promoted droplet formation at progressively lower HP1ɑ concentrations, comparable to, and in 13

some cases better than, enzymatically synthesised polyuridylic acid (polyU) at identical nucleotide 14

concentrations (Figure 3A and Supplementary Figure 3A). While both strands of MSR RNA exhibited the 15

ability to form droplets, transcripts corresponding to the forward strand consistently yielded larger droplets 16

as compared to the reverse strand (Figure 3A and Supplementary Figure 3A). These results suggest that MSR 17

RNAs are capable of binding to HP1ɑ and promote thematically similar behaviour as has been observed for 18

HP1ɑ with DNA in vitro 7,8. This model provides a molecular basis for understanding why HP1ɑ-associated 19

heterochromatin disperses into smaller condensates after MSR RNA levels are depleted (Figure 3B). And, 20

conversely, it offers an explanation as to why the induction of MSR transcription in differentiated cell types 21

can drive the fusion of chromocenters into larger aggregates 15. 22

We next used structured illumination microscopy (SIM) to test whether a reduction in MSR RNA 23

levels affects HP1ɑ structure at chromocenters. Upon MSR transcript depletion, HP1ɑ formed a smaller and 24

more compact core at chromocenters, concomitant with a stronger overlap between HP1ɑ and H3K9me3 25

marked volumes (Figures 3B, 3C and Supplementary Figure 3B). Consistent with this finding, we also 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 8: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

8

detected increased HP1ɑ signal at chromocenters by immunofluorescence microscopy (Figure 3D) and at the 1

underlying satellite DNA by chromatin immunoprecipitation (ChIP) (Figure 3E), following MSR RNA 2

depletion. We next investigated whether the increase of chromatin-associated HP1ɑ at chromocenters 3

affected chromatin compaction by using SiR-DNA fluorescence lifetime methodology that measures 4

chromatin compaction levels in defined genomic compartments 42. Following MSR RNA depletion, we 5

observed using this approach more compacted chromatin was present in heterochromatin domains but not in 6

euchromatic regions of the nucleus (Figure 3F). Together, these results lead us to conclude that high levels 7

of MSR RNA in ESCs promote the fusion and aggregation of chromocenters into large foci, and also prevent 8

premature heterochromatinisation and compaction of chromatin within these domains. 9

10

Satellite RNA scaffolds heterochromatin organisation in embryonic stem cells 11

Having established the effect of MSR RNA in promoting rapid chromatin dynamics within constitutive 12

heterochromatin, we next investigated the role of MSR transcripts in chromocenter organisation. Line-scan 13

microscopy analysis revealed that MSR RNA depletion triggered the rapid reorganisation of heterochromatin 14

into more numerous, brighter and smaller chromocenters (Figures 4A-C), an organisational pattern that is 15

characteristic of differentiated cell types. In addition, immunofluorescence microscopy and ChIP revealed 16

the increased accumulation of H3K9me3 at heterochromatin domains after MSR RNA depletion (Figures 4D 17

and 4E), relative to controls. These results further support that changes occur to the state of heterochromatin 18

at chromocenters upon MSR RNA depletion. We conclude, therefore, that MSR transcripts are required to 19

maintain the highly dynamic and distinctive organisation of heterochromatin domains in ESCs. 20

21

Chromocenter architecture protects chromosome stability in embryonic stem cells 22

Disrupted heterochromatin maintenance is often associated with the onset of chromosome instability, 23

elevated DNA damage, and with defective mitosis. In somatic cells, this type of heterochromatin perturbation 24

is typically triggered by the weakening of heterochromatin-associated processes 3. ESCs are unusual, 25

however, in that they can seemingly tolerate a permissive and uncompacted heterochromatin state without 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 9: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

9

adverse consequences 43,44. We therefore investigated whether a distinct heterochromatin state is not only 1

tolerated in ESCs but perhaps also required to maintain proper chromosome segregation. Interestingly, we 2

observed - shortly after MSR RNA depletion - that a large proportion of metaphase-stage nuclei showed clear 3

hallmarks of chromosome instability (Figures 5A and 5B). Specifically, ~20% of nuclei had chromosome 4

fusions, ~4% contained chromosome breaks, and ~30% showed signs of fragility at pericentromeric regions; 5

defects that were rarely observed in control samples. Furthermore, immunofluorescence microscopy revealed 6

a significant increase in the number of ɣH2AX foci in chromocenters after gapmer treatment (Figure 5C), 7

which indicates the presence of elevated damage at satellite DNA following MSR RNA depletion. 8

We then tested this association using a different system, this time by forcing an increase in 9

chromocenter heterochromatinisation using a doxycycline-inducible TALE-MSR-KRAB protein 45. 10

Consistent with our above observations, the activation of TALE-MSR-KRAB in ESCs substantially impaired 11

chromosome segregation, with ~20% of metaphase-stage nuclei containing fused chromosomes, ~10% with 12

chromatid breaks, ~50% with pericentromeric gaps and >50% of analysed mitotic chromosomes showing 13

defects in sister-chromatid cohesion (Figures 5D and 5E). In contrast, samples taken from the same ESC line 14

but without doxycycline induction were normal. These results establish that heterochromatin organisation 15

plays an important protective role in ESCs to safeguard chromosomal stability. 16

17

18

Discussion 19

Our study demonstrates that noncoding satellite RNA modulates the physical properties and nuclear 20

organisation of heterochromatin. Satellite repeat elements are highly transcribed in ESCs, but it remains 21

unknown whether such transcripts are simply a by-product of the permissive chromatin environment that 22

defines pluripotent cells or whether they themselves fulfil a functional role in genome regulation 46. We now 23

demonstrate that satellite repeat transcripts are required to maintain constitutive heterochromatin in a more 24

dynamic state that organises into large chromocenters in vivo. At the molecular level, modelling and 25

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 10: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

10

biochemical studies have proposed that RNA molecules can promote liquid-liquid phase-separation by 1

lowering the concentration required for proteins to phase separate and/or to modulate the physical properties 2

of phase-separated condensates 33,47. Additionally, RNA molecules themselves can phase-separate into either 3

liquid- or gel-like states, forming compartments that can sequester proteins and other RNAs 37,48. Although 4

noncoding transcripts have diverse sequences in different species, there are likely to be universal and 5

sequence-independent principles for how RNA molecules promote the formation of nuclear domains. 6

Our findings show that depleting MSR RNA disrupts the dynamics of heterochromatin, increases 7

heterochromatinisation and alters chromocenter organisation. These results also provide an interpretation for 8

a previous observation that an unknown nuclear RNA component can sustain the higher-order structure of 9

pericentric heterochromatin 49, and presents evidence that the ability to maintain chromocenter stability 10

resides at least in part with the transcript itself. As satellite repeats are under the transcriptional control of 11

pluripotency factors in ESCs 15, these new findings provide a direct connection between cell state and 12

heterochromatin regulation. Thus, a key developmental switch is controlled by the decline in pluripotency 13

factor availability as cells differentiate, which triggers the downregulation of MSR transcripts and the 14

reorganisation of chromocenters towards a configuration that is typical of somatic cells. Mechanistically, we 15

show that MSR RNA is a ligand that promotes the ability of HP1α to phase separate in vitro, likely by 16

modulating a similar closed-to-open equilibrium as has been hypothesised to be modulated by DNA 7. 17

General modeling of protein-nucleic acid condensation suggests that the addition of nucleic acids reduces the 18

thermodynamic and kinetic barriers of phase separation 50, consistent with our observations here. An 19

important implication arising from our work is that MSR RNA levels can be titrated through their 20

transcriptional control and by their degradation and clearance. This could enable the fine tuning of 21

heterochromatin properties into states that range from soluble to liquid to gel-like, perhaps to regulate 22

chromatin accessibility, genome compartmentalisation and/or chromosome structure. An important future 23

direction for research will be to investigate how this pathway integrates with other processes, including 24

protein phosphorylation and alternative ligands, to direct heterochromatin properties and dynamics 51. 25

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 11: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

11

In addition to scaffolding HP1ɑ, our work raises the possibility that MSR transcripts have a second 1

key function: to prevent the untimely and deleterious compaction of chromatin within heterochromatin 2

domains. It remains unexplained how heterochromatin in ESCs remains permissive and uncompacted, despite 3

residing within H3K9me3 and HP1ɑ marked territories 16,17,52,53. We propose that high levels of MSR RNA 4

prevent HP1ɑ from binding to satellite DNA, thereby sequestering HP1ɑ away from chromatin and unable 5

to compact it. This model is supported by biochemical experiments, which have shown that HP1ɑ has a 6

stronger preference for RNA than DNA 27. As RNA and DNA bind to the same hinge region on HP1ɑ, MSR 7

RNA can effectively out-compete satellite DNA for binding to HP1ɑ 27,54,55. A reduction in MSR RNA levels 8

would, therefore, enable HP1ɑ to engage with and compact chromatin. We tested this prediction and found 9

that the depletion of MSR RNA triggered the increased binding of HP1ɑ at satellite DNA and increased 10

chromatin compaction within chromocenters. These data also agree with previous findings that the 11

transcriptional downregulation of MSR RNA upon ESC differentiation is closely coupled to the progressive 12

compaction of chromatin 15,19,56 and, conversely, that activating MSR transcription in fibroblasts results in the 13

decompaction of chromocenters 15,19,56. 14

Taken together, we propose that our findings support a model in which heterochromatin-associated 15

MSR transcripts act near their sites of synthesis to hold HP1ɑ molecules in close proximity to chromatin, but 16

prevents full engagement and compaction. This increase leads to the formation of large condensates and 17

keeps HP1ɑ away from chromatin, in order to sustain a permissive heterochromatin structure in ESCs (Figure 18

6). Upon MSR RNA depletion, either experimentally or after ESC differentiation, HP1ɑ is then released and 19

can bind to satellite DNA, leading to the observed changes in heterochromatin compaction and organisation. 20

It remains important to determine whether a reduction in MSR RNA concentration within heterochromatin 21

condensates can trigger specific transitions in the material properties of heterochromatin condensates, with 22

parallels to enhancer condensates 57, or whether the relocalisation of HP1ɑ and other processes is also 23

required. Interestingly, a recent study showed that HP1 molecules rapidly exchange in and out of 24

chromocenters in fully differentiated mouse cells 56. Such rapid exchange is consistent with the low surface 25

tension of phase-separated condensates that arise from electrostatically driven multi-valent interactions, such 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 12: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

12

as those between DNA and HP1 51,58. Excitingly, the results of this study raise the potential for a dose-1

dependent effect of other charged ligands of HP1, such as MSR transcripts, in regulating heterochromatin by 2

modulating the ability of HP1ɑ to form multivalent interactions. These interactions perhaps transition 3

chromocenters from being liquid-like aggregates, such as those described in early embryonic development 8 4

and in pluripotent stem cells (as reported in this study), to being ‘ordered collapsed globules’ in differentiated 5

cells 56. It remains important to define whether HP1ɑ alone could be the chromatin-binding factor that bridges 6

and collapses chromatin into globules or if other factors, possibly cell-specific, are also involved. 7

Pericentromeric heterochromatin forms one of the subdomains of centromeres, and preserving 8

heterochromatin stability, including HP1ɑ function is required for appropriate chromosome condensation and 9

segregation in most cell types 59–63. Curiously, ESCs can tolerate mutations that weaken heterochromatin 10

pathways, while the same perturbation in somatic cells causes significant karyotypic defects 43,44. 11

Unexpectedly, we found in ESCs that MSR transcript depletion and strengthened heterochromatinisation in 12

ESCs led to the rapid appearance of mitotic defects, highlighting the functional differences of 13

heterochromatin regulation that exists between pluripotent and somatic cells. It is currently unclear why ESCs 14

require this unusual form of heterochromatin regulation to maintain chromosome stability. One possibility is 15

that this requirement arose concomitantly with the very short G1 phase of the cell cycle in ESCs 64,65, perhaps 16

to accommodate the rapid reassembly of chromocenters before the start of the next replication round. Changes 17

to the biophysical properties of chromocenters could perturb the correct timing of DNA replication and/or 18

transcription, which could quickly lead to the observed genetic instability defects. More generally and 19

importantly, our findings imply that an association exists between the biophysical state of heterochromatin 20

and the preservation of chromosome stability. Notably, noncoding transcripts are accumulated in other 21

processes in which the genome is remodelled, such as in the formation of senescence-associated 22

heterochromatin foci, cellular stress, embryo development and in human ageing and disease 29,66–70. Based on 23

the findings reported here, further work is needed in these other contexts to investigate the role of RNA 24

molecules in controlling heterochromatin properties and genome function. 25

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 13: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

13

Online Methods 1

2

Additional reagent and resource details are provided in Supplementary Table 1. 3

4

Cell culture 5

Male, E14Tg2a mouse ESCs (129P2/OlaHsdl passages 19-28) 71 were cultured in ESC media containing 6

DMEM supplemented with 15% FBS, 1mM sodium pyruvate, 0.1mM 2-mercaptoethanol, 0.1mM non-7

essential amino acids, 2mM Glutamax and 1000 U/ml LIF. Cells were maintained at 37℃ on mitotically 8

inactivated mouse embryonic fibroblasts and transferred for two passages onto gelatin-coated surfaces prior 9

to collection. ESCs were routinely verified as being mycoplasma-free using a PCR-based assay (Sigma). The 10

E14Tg2a line is not on the list of commonly misidentified cell lines (International Cell Line Authentication 11

Committee). 12

Stable, doxycycline-inducible ESC lines were made by transfecting E14Tg2a cells using 13

Lipofectamine 2000 (ThermoFisher) with 1ug PB-TET-TALE-MSR-mClover-ires-mCherry 15 or with PB-14

TET-TALE-MSR-KRAB-ires-GFP plasmids, together with 1ug pCAG-rtTA-Puro and 2ug pCyL43 15

piggyBac transposase, followed by selection with 1.2ug/ml puromycin. Some ESC lines were flow sorted to 16

purity based on mCherry or GFP expression following transient doxycycline induction, and were expanded 17

for >10 passages in the absence of doxycycline. Doxycycline was applied at 0.5-1ug/ml for 24h to induce 18

transgene expression. 19

20

Depletion of major satellite repeat transcripts 21

ESCs were grown to 70% confluency on gelatin-coated plates and were transfected with LNA gapmer oligos 22

(Exiqon) at a concentration of 100nM, using Lipofectamine RNAiMaX Reagent (ThermoFisher), following 23

the manufacturer's instructions. To ensure the robust depletion of target transcripts, the LNA gapmer 24

transfections were repeated at 48h and 72h after the first transfection. Cells were collected for further analysis 25

24h after the final transfection. The LNA gapmer sequences are: LNA DNA gapmer GFP 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 14: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

14

(gagaAAGTGTGACAagtg), LNA DNA gapmer Major Satellite 1 (acatCCACTTGACGActtg) and LNA 1

DNA gapmer Major Satellite 2 (tattTCACGTCCTAAagtg), where the lowercase letters denote the LNA 2

nucleotides 28. 3

4

RT-qPCR 5

For most RT-qPCR experiments, RNA was extracted using the RNeasy Mini Kit (Qiagen), reverse 6

transcribed using Superscript II (Life Technologies) and random primers (Promega), and subjected to qPCR 7

analysis, as previously described 72. To analyse major satellite and other repeat classes, total RNA was 8

extracted using TRIzol (Life Technologies) and treated with two rounds of 1U DNase I (Fermentas) per 1µg 9

RNA in the presence of RiboLock RNase inhibitor (Fermentas) to remove genomic DNA. RNA (1µg) was 10

reverse transcribed using Superscript II (Life Technologies) and random primers (Promega), in the presence 11

of RiboLock RNase inhibitor. cDNA was amplified with SYBR Green Jumpstart Taq Ready Mix (Sigma) 12

using primers from 73,74. Primer sequenced; Major Satellite For, GACGACTTGAAAAATGACGAAATC; 13

Major Satellite Rev, CATATTCCAGGTCCTTCAGTGTGC; Minor Satellite For, 14

TGATATACACTGTTCTACAAATCCCGTTTC; Minor Satellite Rev, 15

ATCAATGAGTTACAATGAGAAACATGGAAA; LINE L1 For, CTGGCGAGGATGTGGAGAA; LINE 16

L1 Rev, CCTGCAATCCCACCAACAAT; Nanog For, ATGCCTGCAGTTTTTCATCC; Nanog Rev, 17

GAGGCAGGTCTTCAGAGGAA; Klf4 For, ACACTTGTGACTATGCAGGCTGTG; Klf4 Rev, 18

TCCCAGTCACAGTGGTAAGGTTTC ; Hmbs For, CGTGGGAACCAGCTCTCTGA ; Hmbs Rev, 19

GAGGCGGGTGTTGAGGTTTC ; T For, TCAGCAAAGTCAAACTCACCAACA; T Rev, 20

CCGAGGTTCATACTTATGCAAGGA. 21

22

Chromatin immunoprecipitation 23

Crosslinked ChIP experiments were performed as previously described 75. Briefly, cells were fixed with 2mM 24

DSG (Sigma) for 45min and then with 1% formaldehyde for 12min. Sonicated chromatin (250µg; 200-500bp 25

fragments) was pre-cleared with blocked beads for 2h at 4°C and incubated at 4°C overnight with either 5µg 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 15: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

15

HP1ɑ antibody (ab77256, Abcam) or 1µg rabbit IgG (Jackson ImmunoResearch). Chromatin-antibody 1

complexes were incubated for 6-8 hours at 4°C with protein A/G magnetic Dynabeads (Life Technologies), 2

washed and the crosslinks were reversed. Native ChIP experiments for H3K9me3 (ab8898 Abcam) were 3

performed as previously described 72. ChIP DNA was analysed by qPCR using primers from 73,74. 4

5

In vitro transcription of major satellite repeats 6

MegaScript T3 and T7 kits (ThermoScientific) were used to transcribe forward and reverse transcripts of 7

differing lengths from major satellite repeat DNA, according to the manufacturer’s instructions. To obtain 8

transcripts with one single consensus repeat, oligos containing the forward or the reverse major satellite 9

consensus sequences were synthesised with the T7 or the T3 sequences, respectively (GenScript) and 10

hybridised. Two repeats were transcribed using the pCR4 Maj9-2 template (73; a gift from Thomas Jenuwein), 11

which was digested with SpeI or NotI for T7 or T3 Megascript kits, respectively. Eight repeats were 12

transcribed from the pySat template (76; a gift from from Niall Dillon, Addgene plasmid # 39238), which was 13

digested with NotI or SalI for T7 or T3 Megascript kits, respectively. Prior to in vitro transcription, linearised 14

plasmids were cleaned with 3M Sodium acetate and 100% ethanol. Linear templates (1µg) were in vitro 15

transcribed according to manufacturer’s instructions, and RNAs were clean by LiCl precipitation. 16

17

HP1ɑ droplet-formation assay 18

Polyuridylic acid (polyU, Santa Cruz Biotechnology, cat no. sc-215733A) was dissolved in MilliQ water, 19

then dialysed extensively against additional MilliQ water in a 3 kDa cutoff dialysis membrane (SpectraPor) 20

to remove co-purified salts, ethanol precipitated and washed prior to final resuspension in MilliQ water. In 21

vitro transcribed MSR RNAs and commercial polyU were quantitated using the total base hydrolysis method 22

of RNA degradation 77 and UV-absorbance quantitation at 260 nm 78 to yield concentration in units of 23

nucleotides. All RNAs were diluted in assay buffer (20 mM HEPES, pH 7.2, 75 mM KCl, 1 mM DTT) prior 24

to use. Recombinant human HP1α protein was purified from E. coli as previously described 7. Prior to use in 25

droplet assays, protein was dialysed in the assay buffer (20 mM HEPES, pH 7.2, 75 mM KCl, 1 mM DTT). 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 16: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

16

Droplet assays were performed using a 384 Greiner Sensoplate (no. 781892) according to the protocol by 1

Keenen and colleagues 79 with a Nikon TiEclipse microscope fitted with a 20x DIC objective. Briefly, 2x 2

solutions of HP1α (two-fold serial dilutions from 200 µM down to 1.6 µM, and 0 µM) were mixed in equal 3

volume with a 2x solutions of RNA (150 µM nucleotide). The HP1α-RNA mixture was then vigorously pipet-4

mixed before transferring into the 384 well plate. Mixtures were allowed to settle for an hour prior to droplet 5

imaging. 6

7

Microscopy imaging and analysis 8

ESCs were cultured on glass coverslips precoated with 0.1% gelatin. For immunofluorescence experiments, 9

cells were fixed with 3% paraformaldehyde in PBS for 10 min at room temperature (RT), washed three times 10

with PBS for 5min, permeabilised with 0.1% Triton X-100 in PBS for 10 min, and washed three times with 11

PBS for 5min. Cells were incubated with primary antibody (H3K9me3, 39161 Active Motif and HP1ɑ, 12

ab77256 Abcam) in blocking buffer (5% milk in PBS) and incubated either for 1h at RT or overnight at 4°C. 13

They were then washed three times with PBS for 5min and incubated with secondary antibodies for two hours 14

at RT. Images were collected on an Olympus FV1000 or Nikon A1-R confocal microscope. Coverslips were 15

mounted onto slides with media containing DAPI (Vectashield H-1200, Vector Laboratories) and were 16

imaged using a Nikon A1-R confocal microscope and a 60X oil objective. When indicated, a Z-stack of 17

images was collected with 0.2 µm spacing and projected using maximum intensity. Image files were prepared 18

using Fiji software. ImageJ software was used to quantify H3K9me3 foci, size and intensity using the 19

‘Analyze Particles’ tool (Novo et al., 2016). Linescan analysis was performed as previously described 17. 20

Colocalisation analysis was performed in Imaris (BitPlane, Oxford). 21

Super-resolution structured illumination microscopy images were acquired with a Nikon dual mode 22

super-resolution microscope with structured illumination (SIM). The resulting reconstructed images were 23

analysed in 3D in Imaris (BitPlane, Oxford). 24

25

26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 17: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

17

Live-cell imaging 1

TALE-MSR-GFP ESCs were grown on glass coverslips coated with 0.1% gelatin. Cells were imaged using 2

an Andor Revolution spinning disk confocal microscope, which was equipped with a thermostatically 3

controlled stage maintained at 37°C with a 60X oil immersion objective. A z-stack of images was collected 4

with 0.5 µm spacing and projected using maximum intensity. Three biological replicates per cell line and 5

doxycycline concentration were included in each experiment. At least 500 cells were analysed for each 6

experimental group. 7

8

Fluorescence recovery after photo-bleaching 9

FRAP experiments were performed on an Andor Revolution spinning disk confocal microscope. Images were 10

acquired every second for 300 seconds (310 frames). The first ten frames were collected before the bleach 11

pulse for baseline fluorescence. A 10x10 region surrounding a single chromocenter per cell was selected for 12

bleaching puncta using 2% laser power (488 nm). Fluorescent intensities and image analysis were performed 13

using the TrackMate plugin (v3.8.0) in the Fiji software 80. After normalisation of the intensity signal, FRAP 14

curves were generated from the fluorescence intensity of each chromocenter, at each timepoint (every 15

second). To calculate the half-time [ln(2)/K] of fluorescence recovery after photo-bleaching, data was fitted 16

to a one-phase association equation [Y=Y0 + (Plateau-Y0)*(1-exp(-K*x))], where Y0 is the fluorescence 17

after photo-bleaching (GraphPad software). To correct for loss of fluorescence due to the bleaching pulse the 18

mobile fraction was calculated as [Fm= (Plateau - First post-bleach) / (Pre-bleach - First post-bleach)] and 19

the immobile fraction was measured as remaining fluorescence intensity unrecovered at plateau phase. FRAP 20

experiments were repeated five times and data were acquired from one bleached and from at least one 21

unbleached chromocenter per cell. 22

23

Chromocenter organisation and compaction analysis 24

DAPI linescan analyses were performed using ImageJ on optical sections, where DAPI foci were at optimal 25

focal planes. Fluorescence intensity histograms were generated with a linescan across the nucleus, and the 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 18: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

18

background (outside of the nucleus) was subtracted from the baseline fluorescence within the nucleus and 1

the chromocenter signal. Differences in chromocenter intensity are shown as the ratio of chromocenter peak 2

height to nucleoplasmic signal. 3

For SiR-DNA-based analysis of chromatin compaction by FLIM 42, cells were plated in 35 mm glass 4

bottom dishes (P35-1.5-14-C, Mattek Corporation, MA, USA) the day before imaging. Cells were stained 5

with 1 µM SiR-DNA (Spirochrome Ltd., Stein am Rhein, Switzerland) and with 10 µM verapamil 6

(Spirochrome Ltd.) in cell culture medium for 1 hour, before changing to cell culture medium that contained 7

10 µM verapamil and 20 mM HEPES pH 7.4 for imaging. Fluorescence lifetime imaging (FLIM) was 8

performed on a home-built confocal platform (Olympus Fluoview FV300), which was integrated with time-9

correlated single photon counting (TCSPC) to measure fluorescence lifetime in every image pixel. Output 10

from a pulsed supercontinuum source (WL-SC-400-15, Fianium Ltd., UK, repetition rate 40MHz) was 11

filtered using a bandpass filter FF01-635/18 to excite SiR-DNA. Fluorescence emission from the sample was 12

filtered using 700/70nm (Comar Optics, UK) before passing onto a photomultiplier tube (PMC-100, Becker 13

& Hickl GmbH, Berlin, Germany). Photons were recorded in time-tagged, time-resolved mode that permits 14

the sorting of photons from each pixel into a histogram according to their arrival times. The data was recorded 15

by a TCSPC module (SPC-830, Becker and Hickl GmBH). Photons were acquired for two minutes to make 16

a single 256 X 256 FLIM image. Photon count rates were always kept below 1% of the laser repetition rate 17

to prevent pulse pile-up. Photobleaching was verified to be negligible during acquisition. Approximately 10 18

representative images with several nuclei per field of view were acquired for each condition. FLIM images 19

were analysed using FLIMfit v4.12.1 and fitted with a monoexponential decay function. Whole nuclei were 20

segmented based on intensity (debris and mitotic nuclei were manually removed). A two-level mask that 21

separated heterochromatin spots and euchromatin was created with Icy software spot detection tool 81,82. 22

Pixels in these two chromatin regions were binned and fitted separately to obtain two lifetime values for each 23

image. Statistical analysis was carried out using an unpaired two-samples Mann-Whitney test. 24

25 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 19: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

19

Quantification and statistical analysis 1

Statistical parameters including the exact value of n, SD, SEM and statistical significance are reported in the 2

figures and the figure legends. For the majority of experiments, statistical significance is determined by the 3

value of p < 0.05 by unpaired two-samples Wilcoxon test (Mann-Whitney test). 4

5

Data availability statement 6

The data that support the findings of this study are available from the corresponding authors upon reasonable 7

request. 8

9

Code availability statement 10

No custom code was used. 11

12

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 20: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

20

Acknowledgements 1

We are grateful to Niall Dillon, Maria Elena Torres-Padilla, Thomas Jenuwein and the Wellcome Trust 2

Sanger Institute for kindly providing plasmids. We thank several Babraham Institute Facilities for their 3

assistance, including the Flow Core, the Bioinformatics Group, and particularly Simon Andrews and Anne 4

Segonds-Pichon, and also Hanneke Okkenhaug in the Imaging Facility. We thank members of P.J.R.-G.’s 5

group for helpful discussions, Gavin Kelsey and Wolf Reik for comments on the manuscript, and the 6

Babraham Institute Science Policy and Oversight Committee for their support. C.N. and P.J.R.-G. were 7

funded by grants from the Biotechnology and Biological Sciences Research Council (BBS/E/B/000C0421, 8

BBS/E/B/000C0422, BB/M022285/1 and the Core Capability Grant), and European Commission Network 9

of Excellence EpiGeneSys (HEALTH-F4-2010-257082). E.W. was supported by an F32 fellowship from the 10

NIH. C.H., C.P. and G.S.K.S. were supported by the Wellcome Trust, Alzheimer’s Research UK, the Michael 11

J Fox Foundation, Infinitus China Ltd, and by the European Union’s Horizon 2020 Research and Innovation 12

Programme under grant agreement number 722380. E.S. was supported by a British Society for 13

Developmental Biology Gurdon / The Company of Biologists Summer Studentship. G.J.N. was supported by 14

an MIRA grant from the NIH (R35 GM127020). C.N. is grateful to the Tommys National Miscarriage Centre 15

for funding support. 16

17

Author Contributions 18

Conceptualisation, C.L.N. and P.J.R-G.; Methodology, C.L.N., C.H., E.W., C.P., S.W.; Investigation, 19

C.L.N., E.W., C.H., E.S.; Visualisation, C.L.N.; Writing – Original Draft, C.L.N. and P.J.R-G.; Writing – 20

Review & Editing, C.L.N., E.W., G.J.N. and P.J.R-G.; Funding Acquisition, C.L.N., G.S.K.S., G.J.N., P.J.R-21

G.; Supervision, G.S.K.S., G.J.N. and P.J.R-G. 22

23

Competing Interests: The authors declare no competing financial interests. 24

25

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 21: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

21

References 1

1. Bickmore, W. A. & van Steensel, B. Genome architecture: domain organization of interphase 2

chromosomes. Cell 152, 1270–1284 (2013). 3

2. Allshire, R. C. & Madhani, H. D. Ten principles of heterochromatin formation and function. Nat. Rev. 4

Mol. Cell Biol. 19, 229–244 (2018). 5

3. Janssen, A., Colmenares, S. U. & Karpen, G. H. Heterochromatin: Guardian of the Genome. Annu. 6

Rev. Cell Dev. Biol. 34, 265–288 (2018). 7

4. Rea, S. et al. Regulation of chromatin structure by site-specific histone H3 methyltransferases. Nature 8

vol. 406 593–599 (2000). 9

5. Lachner, M., O’Carroll, D., Rea, S., Mechtler, K. & Jenuwein, T. Methylation of histone H3 lysine 9 10

creates a binding site for HP1 proteins. Nature 410, 116–120 (2001). 11

6. Bannister, A. J. et al. Selective recognition of methylated lysine 9 on histone H3 by the HP1 chromo 12

domain. Nature 410, 120–124 (2001). 13

7. Larson, A. G. et al. Liquid droplet formation by HP1α suggests a role for phase separation in 14

heterochromatin. Nature 547, 236–240 (2017). 15

8. Strom, A. R. et al. Phase separation drives heterochromatin domain formation. Nature 547, 241–245 16

(2017). 17

9. Cheutin, T. Maintenance of Stable Heterochromatin Domains by Dynamic HP1 Binding. Science vol. 18

299 721–725 (2003). 19

10. Festenstein, R. Modulation of Heterochromatin Protein 1 Dynamics in Primary Mammalian Cells. 20

Science vol. 299 719–721 (2003). 21

11. Santenard, A., Jachoviwcz, J. & Torres-Padilla, M.-E. Establishment of pericentromeric 22

heterochromatin in development. Epigenetics & Chromatin vol. 6 (2013). 23

12. Sidler, C., Kovalchuk, O. & Kovalchuk, I. Epigenetic Regulation of Cellular Senescence and Aging. 24

Front. Genet. 8, 138 (2017). 25

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 22: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

22

13. Efroni, S. et al. Global transcription in pluripotent embryonic stem cells. Cell Stem Cell 2, 437–447 1

(2008). 2

14. Meshorer, E. et al. Hyperdynamic plasticity of chromatin proteins in pluripotent embryonic stem cells. 3

Dev. Cell 10, 105–116 (2006). 4

15. Novo, C. L. et al. The pluripotency factor Nanog regulates pericentromeric heterochromatin 5

organization in mouse embryonic stem cells. Genes Dev. 30, 1101–1115 (2016). 6

16. Ahmed, K. et al. Global chromatin architecture reflects pluripotency and lineage commitment in the 7

early mouse embryo. PLoS One 5, e10531 (2010). 8

17. Fussner, E. et al. Constitutive heterochromatin reorganization during somatic cell reprogramming. 9

EMBO J. 30, 1778–1789 (2011). 10

18. Bošković, A. et al. Higher chromatin mobility supports totipotency and precedes pluripotency in vivo. 11

Genes Dev. 28, 1042–1047 (2014). 12

19. Tosolini, M. et al. Contrasting epigenetic states of heterochromatin in the different types of mouse 13

pluripotent stem cells. Sci. Rep. 8, 5776 (2018). 14

20. Mattout, A., Biran, A. & Meshorer, E. Global epigenetic changes during somatic cell reprogramming 15

to iPS cells. J. Mol. Cell Biol. 3, 341–350 (2011). 16

21. Ricci, M. A., Manzo, C., García-Parajo, M. F., Lakadamyali, M. & Cosma, M. P. Chromatin fibers are 17

formed by heterogeneous groups of nucleosomes in vivo. Cell 160, 1145–1158 (2015). 18

22. Aoto, T., Saitoh, N., Ichimura, T., Niwa, H. & Nakao, M. Nuclear and chromatin reorganization in the 19

MHC-Oct3/4 locus at developmental phases of embryonic stem cell differentiation. Dev. Biol. 298, 20

354–367 (2006). 21

23. Kobayakawa, S., Miike, K., Nakao, M. & Abe, K. Dynamic changes in the epigenomic state and 22

nuclear organization of differentiating mouse embryonic stem cells. Genes Cells 12, 447–460 (2007). 23

24. Martens, J. H. A. et al. The profile of repeat-associated histone lysine methylation states in the mouse 24

epigenome. EMBO J. 24, 800–812 (2005). 25

25. Maison, C. et al. SUMOylation promotes de novo targeting of HP1α to pericentric heterochromatin. 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 23: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

23

Nat. Genet. 43, 220–227 (2011). 1

26. Camacho, O. V. et al. Major satellite repeat RNA stabilize heterochromatin retention of Suv39h 2

enzymes by RNA-nucleosome association and RNA:DNA hybrid formation. eLife vol. 6 (2017). 3

27. Muchardt, C. et al. Coordinated methyl and RNA binding is required for heterochromatin localization 4

of mammalian HP1alpha. EMBO Rep. 3, 975–981 (2002). 5

28. Probst, A. V. et al. A strand-specific burst in transcription of pericentric satellites is required for 6

chromocenter formation and early mouse development. Dev. Cell 19, 625–638 (2010). 7

29. Casanova, M. et al. Heterochromatin reorganization during early mouse development requires a 8

single-stranded noncoding transcript. Cell Rep. 4, 1156–1167 (2013). 9

30. Burton, A. & Torres-Padilla, M.-E. Chromatin dynamics in the regulation of cell fate allocation during 10

early embryogenesis. Nat. Rev. Mol. Cell Biol. 15, 723–734 (2014). 11

31. Miyanari, Y., Ziegler-Birling, C. & Torres-Padilla, M.-E. Live visualization of chromatin dynamics 12

with fluorescent TALEs. Nat. Struct. Mol. Biol. 20, 1321–1324 (2013). 13

32. Wang, Z., Zhang, G. & Zhang, H. Protocol for analyzing protein liquid–liquid phase separation. 14

Biophysics Reports vol. 5 1–9 (2019). 15

33. Banani, S. F., Lee, H. O., Hyman, A. A. & Rosen, M. K. Biomolecular condensates: organizers of 16

cellular biochemistry. Nat. Rev. Mol. Cell Biol. 18, 285–298 (2017). 17

34. Shin, Y. & Brangwynne, C. P. Liquid phase condensation in cell physiology and disease. Science 357, 18

(2017). 19

35. Bultmann, S. et al. Targeted transcriptional activation of silent oct4 pluripotency gene by combining 20

designer TALEs and inhibition of epigenetic modifiers. Nucleic Acids Research vol. 40 5368–5377 21

(2012). 22

36. Thanisch, K. et al. Targeting and tracing of specific DNA sequences with dTALEs in living cells. 23

Nucleic Acids Res. 42, e38 (2014). 24

37. Jain, A. & Vale, R. D. RNA phase transitions in repeat expansion disorders. Nature 546, 243–247 25

(2017). 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 24: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

24

38. Sabari, B. R. et al. Coactivator condensation at super-enhancers links phase separation and gene 1

control. Science 361, (2018). 2

39. Cho, W.-K. et al. Mediator and RNA polymerase II clusters associate in transcription-dependent 3

condensates. Science 361, 412–415 (2018). 4

40. Lee, J.-S. & Mendell, J. T. Antisense-Mediated Transcript Knockdown Triggers Premature 5

Transcription Termination. Mol. Cell 77, 1044–1054.e3 (2020). 6

41. Zamecnik, P. C. & Stephenson, M. L. Inhibition of Rous sarcoma virus replication and cell 7

transformation by a specific oligodeoxynucleotide. Proceedings of the National Academy of Sciences 8

vol. 75 280–284 (1978). 9

42. Hockings, C. et al. Illuminating chromatin compaction in live cells and fixed tissues using SiR-DNA 10

fluorescence lifetime. Cell Biology (2020). 11

43. Peters, A. H. et al. Loss of the Suv39h histone methyltransferases impairs mammalian 12

heterochromatin and genome stability. Cell 107, 323–337 (2001). 13

44. Kanellopoulou, C. et al. Dicer-deficient mouse embryonic stem cells are defective in differentiation 14

and centromeric silencing. Genes Dev. 19, 489–501 (2005). 15

45. Groner, A. C. et al. KRAB–Zinc Finger Proteins and KAP1 Can Mediate Long-Range Transcriptional 16

Repression through Heterochromatin Spreading. PLoS Genetics vol. 6 e1000869 (2010). 17

46. Schlesinger, S. & Meshorer, E. Open Chromatin, Epigenetic Plasticity, and Nuclear Organization in 18

Pluripotency. Dev. Cell 48, 135–150 (2019). 19

47. Zhang, H. et al. RNA Controls PolyQ Protein Phase Transitions. Mol. Cell 60, 220–230 (2015). 20

48. Huo, X. et al. The Nuclear Matrix Protein SAFB Cooperates with Major Satellite RNAs to Stabilize 21

Heterochromatin Architecture Partially through Phase Separation. Mol. Cell 77, 368–383.e7 (2020). 22

49. Maison, C. et al. Higher-order structure in pericentric heterochromatin involves a distinct pattern of 23

histone modification and an RNA component. Nat. Genet. 30, 329–334 (2002). 24

50. Joseph, J. A. et al. Oligonucleotides can act as superscaffolds that enhance liquid-liquid phase 25

separation of intracellular mixtures. Biophysics 1228 (2020). 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 25: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

25

51. Larson, A. G. & Narlikar, G. J. The Role of Phase Separation in Heterochromatin Formation, 1

Function, and Regulation. Biochemistry 57, 2540–2548 (2018). 2

52. Canzio, D. et al. Chromodomain-mediated oligomerization of HP1 suggests a nucleosome-bridging 3

mechanism for heterochromatin assembly. Mol. Cell 41, 67–81 (2011). 4

53. Kilic, S., Bachmann, A. L., Bryan, L. C. & Fierz, B. Multivalency governs HP1α association dynamics 5

with the silent chromatin state. Nature Communications vol. 6 (2015). 6

54. Sugimoto, K., Yamada, T., Muro, Y. & Himeno, M. Human homolog of Drosophila heterochromatin-7

associated protein 1 (HP1) is a DNA-binding protein which possesses a DNA-binding motif with weak 8

similarity to that of human centromere protein C (CENP-C). J. Biochem. 120, 153–159 (1996). 9

55. Zhao, T., Heyduk, T., Allis, C. D. & Eissenberg, J. C. Heterochromatin protein 1 binds to nucleosomes 10

and DNA in vitro. J. Biol. Chem. 275, 28332–28338 (2000). 11

56. Erdel, F. et al. Mouse Heterochromatin Adopts Digital Compaction States without Showing Hallmarks 12

of HP1-Driven Liquid-Liquid Phase Separation. Molecular Cell (2020) 13

doi:10.1016/j.molcel.2020.02.005. 14

57. Nair, S. J. et al. Phase separation of ligand-activated enhancers licenses cooperative chromosomal 15

enhancer assembly. Nat. Struct. Mol. Biol. 26, 193–203 (2019). 16

58. Jho, Y., Yoo, H. Y., Lin, Y., Han, S. & Hwang, D. S. Molecular and structural basis of low interfacial 17

energy of complex coacervates in water. Advances in Colloid and Interface Science vol. 239 61–73 18

(2017). 19

59. Lindsley, D. L. & Novitski, E. Localization of the Genetic Factors Responsible for the Kinetic 20

Activity of X Chromosomes of Drosophila Melanogaster. Genetics 43, 790–798 (1958). 21

60. Bernard, P. et al. Requirement of heterochromatin for cohesion at centromeres. Science 294, 2539–22

2542 (2001). 23

61. Taddei, A., Maison, C., Roche, D. & Almouzni, G. Reversible disruption of pericentric 24

heterochromatin and centromere function by inhibiting deacetylases. Nat. Cell Biol. 3, 114–120 25

(2001). 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 26: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

26

62. Wines, D. R. & Henikoff, S. Somatic instability of a Drosophila chromosome. Genetics 131, 683–691 1

(1992). 2

63. Bernat, R. L., Delannoy, M. R., Rothfield, N. F. & Earnshaw, W. C. Disruption of centromere 3

assembly during interphase inhibits kinetochore morphogenesis and function in mitosis. Cell 66, 4

1229–1238 (1991). 5

64. Ahuja, A. K. et al. A short G1 phase imposes constitutive replication stress and fork remodelling in 6

mouse embryonic stem cells. Nature Communications vol. 7 (2016). 7

65. Savatier, P., Huang, S., Szekely, L., Wiman, K. G. & Samarut, J. Contrasting patterns of 8

retinoblastoma protein expression in mouse embryonic stem cells and embryonic fibroblasts. 9

Oncogene 9, 809–818 (1994). 10

66. Ting, D. T. et al. Aberrant overexpression of satellite repeats in pancreatic and other epithelial cancers. 11

Science 331, 593–596 (2011). 12

67. De Cecco, M. et al. Transposable elements become active and mobile in the genomes of aging 13

mammalian somatic tissues. Aging 5, 867–883 (2013). 14

68. Gaubatz, J. W. & Cutler, R. G. Mouse satellite DNA is transcribed in senescent cardiac muscle. J. 15

Biol. Chem. 265, 17753–17758 (1990). 16

69. Tasselli, L. et al. SIRT6 deacetylates H3K18ac at pericentric chromatin to prevent mitotic errors and 17

cellular senescence. Nat. Struct. Mol. Biol. 23, 434–440 (2016). 18

70. Zhu, Q. et al. BRCA1 tumour suppression occurs via heterochromatin-mediated silencing. Nature 477, 19

179–184 (2011). 20

71. Hooper, M., Hardy, K., Handyside, A., Hunter, S. & Monk, M. HPRT-deficient (Lesch-Nyhan) mouse 21

embryos derived from germline colonization by cultured cells. Nature 326, 292–295 (1987). 22

72. Rugg-Gunn, P. J., Cox, B. J., Ralston, A. & Rossant, J. Distinct histone modifications in stem cell 23

lines and tissue lineages from the early mouse embryo. Proc. Natl. Acad. Sci. U. S. A. 107, 10783–24

10790 (2010). 25

73. Lehnertz, B. et al. Suv39h-mediated histone H3 lysine 9 methylation directs DNA methylation to 26

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 27: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

27

major satellite repeats at pericentric heterochromatin. Curr. Biol. 13, 1192–1200 (2003). 1

74. Bulut-Karslioglu, A. et al. A transcription factor-based mechanism for mouse heterochromatin 2

formation. Nat. Struct. Mol. Biol. 19, 1023–1030 (2012). 3

75. Tavares, L. et al. RYBP-PRC1 complexes mediate H2A ubiquitylation at polycomb target sites 4

independently of PRC2 and H3K27me3. Cell 148, 664–678 (2012). 5

76. Lundgren, M. et al. Transcription Factor Dosage Affects Changes in Higher Order Chromatin 6

Structure Associated with Activation of a Heterochromatic Gene. Cell vol. 103 733–743 (2000). 7

77. Katz, S. & Comb, D. G. A new method for the determination of the base composition of ribonucleic 8

acid. J. Biol. Chem. 238, 3065–3067 (1963). 9

78. Cavaluzzi, M. J. & Borer, P. N. Revised UV extinction coefficients for nucleoside-5’-monophosphates 10

and unpaired DNA and RNA. Nucleic Acids Res. 32, e13 (2004). 11

79. Keenen, M. M., Larson, A. G. & Narlikar, G. J. Visualization and Quantitation of Phase-Separated 12

Droplet Formation by Human HP1α. Methods Enzymol. 611, 51–66 (2018). 13

80. Tinevez, J.-Y. et al. TrackMate: An open and extensible platform for single-particle tracking. Methods 14

115, 80–90 (2017). 15

81. de Chaumont, F. et al. Icy: an open bioimage informatics platform for extended reproducible research. 16

Nat. Methods 9, 690–696 (2012). 17

82. Olivo-Marin, J.-C. Extraction of spots in biological images using multiscale products. Pattern 18

Recognition vol. 35 1989–1996 (2002). 19

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 28: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

28

Figure Legends 1

2

Figure 1: Constitutive heterochromatin forms liquid-like condensates in mouse embryonic stem cells 3

A) A schematic showing how doxycycline-inducible TALE-GFP is recruited to MSR DNA to enable live-4

cell visualisation of chromocenter dynamics. 5

B) Representative images of nuclei showing the TALE-MSR-GFP signal and DAPI counterstain in ESCs 6

without and with doxycycline induction. Scale bars, 5µm. 7

C) Stills from a time-lapse microscopy experiment in doxycycline-induced TALE-MSR-GFP ESCs. These 8

images of an ESC nucleus over time reveal the ability of GFP-labelled chromocenters to separate and coalesce 9

(red arrow) and to form protrusions (blue arrow). Scale bars, 5µm. 10

D) Quantification of fluorescence recovery after photo-bleaching individual chromocenters. Top panel shows 11

representative images of the FRAP experiment; the dashed squares indicate the photo-bleached region. Lower 12

panel shows the recovery of the GFP signal (log2 mean intensity ± SEM) over time at either bleached (left) 13

or unbleached (right) chromocenters. Data were collected from at least three independent photo-bleaching 14

experiments and show a summary of 49 photo-bleached chromocenters. Scale bars, 10µm. 15

E) Histogram depicting the percentage of mobile and immobile components of chromocenters, as calculated 16

from the FRAP data. 17

See also Supplementary Figure 1 and Supplementary Videos 1, 2 and 3. 18

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 29: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

A)

Figure 1

C)

D) E)

0%

20%

40%

60%

80%

100%

Recoveredfluorescence

Per

cent

age

Immobile

Mobile

Major satellite repeats

TTTCTTGCCATATTCCACGTCCTACAGTGG

TAL effector

GFP-DOX +DOX 24h

TALE-MSR-GFP in ESCsB)

DAPI GFP DAPI GFP

2’0’ 4’ 6’ 8’ 10’ 12’ 14’ 16’Time

Pre-bleach

GFP

GFP

0’’ 50’’ 100’’ 150’’ 200’’

Chr

omoc

ente

r int

enis

ty (a

.u.)

Bleached chromocenters

Time (s)0 50 100 200 300

Chromocenters without bleaching

0 50 100 200 300 Time (s)

6.5

7.0

7.5

8.0

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 30: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

29

Figure 2: Major satellite repeat transcripts regulate constitutive heterochromatin behaviour in mouse 1

embryonic stem cells 2

A) Bar chart showing depleted MSR transcript levels following the transfection of ESCs with LNA-DNA 3

gapmers that target MSR but not control sequences (GFP). The transcripts of other repetitive sequences, such 4

as minor satellites and LINE RNA, were not affected by the LNA-DNA gapmers. Individual data points show 5

the mean values for three independent experiments and were compared using an unpaired, two-sided t-test. 6

B) Top panel shows representative images of timelapse FRAP experiments in GFP or MSR gapmer-7

transfected ESCs. The dashed squares indicate the photo-bleached areas at 0 s; note that the size of the 8

bleached area was the same in all experiments. The lower panel shows GFP intensity (normalised mean 9

intensity ± SEM) of photo-bleached chromocenters (red dotted line shows moment of photo-bleaching) over 10

time after GFP or MSR gapmer transfection. Data were collected from five independent photo-bleaching 11

experiments. Scale bars, 10µm. 12

C) Chart showing the increased time taken to recover 50% of the TALE-MSR-GFP signal at bleached 13

chromocenters in ESCs with depleted MSR RNA levels following MSR-targeting gapmers. Each circle 14

represents one photo-bleached chromocenter. Data collected from five independent experiments (n=49 for 15

GFP and n=45 for MSR) and were compared using an unpaired two-sample Mann-Whitney test. 16

D) Histogram depicting the mobile and immobile components of chromocenters, as calculated from the FRAP 17

data. Data were collected from at least five independent photo-bleaching experiments (n=49 for GFP and 18

n=45 for MSR) and the mobile/immobile proportions between the two experiments were compared using an 19

unpaired two-sample Mann-Whitney test. 20

See also Supplementary Figure 2 and Supplementary Video 4. 21

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 31: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

A)

Figure 2

B)

0.0

0.5

1.0

1.5

Fold

cha

nge

rela

tive

to H

mbs

GFP MSR

MajorSatellite

GFP MSR GFP MSR

MinorSatellite

LINEs

D)

C)p=2E-4 n.s. n.s.

Recovered fluorescence

Per

cent

age

Immobile Mobile

GFPgapmers

MSRgapmers

0%

20%

40%

60%

80%

100%Ti

me

take

n to

reco

ver 5

0%flu

ores

cent

sig

nal (

s)

Pre-bleach 0’’ 50’’ 100’’ 150’’ 200’’

GFP

MS

RGap

mer

s

Fluorescence recovery after photobleaching

0

100

150

200

250

50

p=1.6E-3

p=0.03

GFP gapmers MSR gapmers

Nor

mal

ized

chr

omoc

ente

r int

enis

ty (a

.u.)

Time (s)0 50 100 150 200 250

0.25

0.5

0.75

1.0

0

GFP gapmersMSR gapmers

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 32: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

30

Figure 3: Satellite RNA promotes phase-separation of the heterochromatin protein, HP1ɑ 1

A) In vitro transcribed MSR RNAs, or synthetic polyU RNA, were added to increasing concentrations of 2

recombinant HP1ɑ protein. Images framed in blue show the conditions that allowed for the formation of 3

HP1ɑ droplets. All images show the same magnification; scale bar, 100µm. 4

B) Representative images of 3D reconstructions acquired with structured illumination microscopy (SIM), 5

showing details of H3K9me3 and HP1ɑ organisation at chromocenters in ESCs that were transfected with 6

GFP or MSR gapmers (scale bars, 2µm). White boxes indicate zoomed-in areas shown to the right (scale 7

bars, 0.5µm). Linescan analyses of the boxed chromocenters are shown below and reveal the overlap of 8

heterochromatin marks at chromocenters (HP1ɑ, red line; H3K9me3, green line). 9

C) Bar chart showing the percentage of H3K9me3 and HP1ɑ 3D volumes that colocalise with either HP1ɑ- 10

or H3K9me3-marked foci following the GFP or MSR gapmer transfection of ESCs. Foci volumes were 11

defined from at least three 3D-reconstructions of SIM for each gapmer condition (individual data points are 12

shown). 13

D) Upper, representative images of immunofluorescence microscopy showing H3K9me3, HP1ɑ and DAPI 14

signals upon gapmer transfection of ESCs with GFP or MSR gapmers. Scale bars, 20µm. Lower panel, 15

quantification of HP1ɑ immunofluorescence intensity at chromocenters that were defined by DAPI. A total 16

of 2849 and 3209 chromocenters were quantified after two independent GFP and MSR gapmer transfection 17

experiments, respectively, and were compared with a two-sample Mann-Whitney test. 18

E) Histograms of ChIP results show the fold-change over IgG of HP1ɑ bound at MSR DNA or LINE DNA 19

upon transfection of ESCs with GFP or MSR gapmers. Two biological replicates are shown side by side. 20

F) Plot shows the fluorescence lifetime of the far-red dye SiR-DNA in both euchromatin and heterochromatin 21

regions of nuclei following the transfection of ESCs with GFP or MSR gapmers. Each point represents the 22

mean lifetime intensity per image from three independent experiments, each with >8 images per sample. 23

Samples were compared with a two-sample Mann-Whitney test; error bars show mean ± SEM. 24

See also Supplementary Figure 3. 25

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 33: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

Figure 3

B)

D)

E)

F)

A)po

lyU

MS

RFo

rwar

dM

SR

Rev

erse

HP1a (μM)

0 6.3 12.5 25 50 100

RN

A (7

5 µM

)

H3K

9me3

GFP gapmers MSR gapmers

HP

1αHP1α

Mer

ge

200

100

00.0 0.00.5 0.50.25 0.751.0 1.01.5 2.0

Pix

el v

alue

Distance (μm) Distance (μm)

GFP

Linescan analysis across boxed chromocenters

MSR

H3K9me3

GFP

gapm

ers

MS

Rga

pmer

s

MergeDAPI

012345678

GFP MSR GFP MSRMajor Satellite DNA LINE DNA

Fold

cha

nge

over

IgG HP1α ChIP

C)

010

20

30

40

50

60

70

80

90

100

Per

cent

age

of

volu

me

colo

caliz

atio

n

Foci defined by:

IF signal: H3K9me3

HP1α

HP1α

H3K9me3

GFP gapmers

MSR gapmers

GFPgapmers

MSRgapmers

0

1000

2000

3000

4000

HP

1α fl

uore

scen

cein

tens

ity a

t chr

omoc

entre

s (a

.u.) p=2E-16

SiR

-DN

A lif

etim

e (p

s)

3175

3200

3225

3150

3125

GFP MSR GFP MSR

Euchromatin Heterochromatinp=0.02p=0.17

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 34: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

31

Figure 4: Degradation of satellite transcripts induces chromatin remodeling in embryonic stem cells. 1

A) Left: Chromocenter organisation as revealed by microscopy and linescan analysis of DAPI signal from a 2

single stack (2D) after ESCs were transfected with GFP or MSR gapmers. Scale bars, 5µm. Histogram (right) 3

shows the number of chromocenters per nucleus. Data were collected from three independent experiments 4

and compared using a two-sample Mann-Whitney test. 5

B, C) Plots show DAPI intensity (B) and diameter (C) of chromocenters per nucleus. A total of 100 and 105 6

chromocenters were quantified following the transfection of ESCs with GFP and MSR gapmers, respectively. 7

Data were collected from three independent experiments and compared using a two-sample Mann-Whitney 8

test. 9

(D) Quantification of H3K9me3 immunofluorescence intensity at chromocenters that were defined by DAPI. 10

A total of 2849 and 3209 chromocenters were quantified following two independent GFP and MSR gapmer 11

transfection experiments, respectively, and were compared with a two-sample Mann-Whitney test. 12

E) Histograms of ChIP results show the fold-change over IgG of the H3K9me3 signal at MSR DNA or LINE 13

DNA following GFP or MSR gapmer ESC transfections. Two biological replicates are shown side by side. 14

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 35: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

p=3E-13

Chr

omoc

entre

dia

met

er (p

ixel

s)

p=1.4E-07

2 4 6

0

20

40

60

Per

cent

age

of n

ucle

i (%

)

p=5.3E-06

GFP gapmers MSR gapmers

DA

PI i

nten

sity

of

chr

omoc

entre

s (a

.u.)

Figure 4

A)

B) C)

00

50

50Distance (pixels)

100

100 150Gre

y va

lue

00

50

Distance (pixels)

100150

100 200

Gre

y va

lue

GFP gapmers

GFP gapmers

MSR gapmers

MSR gapmers

0.4

0.620

30

10

0.8

GFP gapmers MSR gapmers

D) E)

012345678

Fold

cha

nge

over

IgG

GFP MSR GFP MSRMajor Satellite DNA LINE DNA

0

1000

2000

3000

4000

H3K

9me3

fluo

resc

ence

inte

nsity

at c

hrom

ocen

tres

(a.u

.)

H3K9me3 ChIP

GFPgapmers

MSRgapmers

p=2.5E-16

Number of chromocenters per nucleus

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 36: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

32

Figure 5: Chromocenter architecture protects chromosome stability in embryonic stem cells 1

A) Histogram showing that ESCs transfected with LNA-DNA gapmers that target MSR transcripts have a 2

high proportion of metaphases with hallmarks of genetic instability, such as chromosome end-fusions and 3

pericentric gaps. At least 31 metaphases from three independent gapmer transfections. Data points show 4

percentages for each biological replicate. 5

B) Representative images of DAPI-stained metaphases, exemplifying the cytogenetic defects observed. Scale 6

bars, 10µm. 7

C) Left: histogram showing the percentage of gamma-H2AX foci at chromocenters. Each point represents 8

the percentage in each image analysed. Right, representative images of gamma-H2AX at chromocenters 9

(white arrow). Scale bars, 5µm. A total of 317 and 420 ESCs were scored for GFP and MSR gapmer 10

transfection, respectively, and the two samples were compared with an unpaired Mann-Whitney test. 11

D) Histogram showing the genetic defects observed after TALE-MSR-KRAB induction in ESCs. At least 30 12

metaphases from three independent replicates were scored. Data points show percentages for each replicate. 13

E) Representative images of DAPI-stained metaphases exemplifying the cytogenetic defects observed after 14

TALE-MSR-KRAB induction. Scale bars, 5µm. 15

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 37: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

Figure 5

B)A)Chromosome fusion Chromosome Breaks

5

10

Per

cent

age

of c

hrom

ocen

ters

γH2AX at chromocentres

Multiple spindleCohesion defects

0

10

20

30

40

50

60

70

Met

apha

ses

with

>1

defe

ct (%

)

Hallmarks of genome instability E)

C)

D)

MS

Rga

pmer

sG

FPga

pmer

s0

10

20

30

40

Chromosomefusion

Chromosomebreaks

Pericentricgaps

Hallmarks of genome instability

Met

apha

ses

with

>1

defe

ct (%

) GFP gapmers

MSR gapmers

GFPgapmers

MSRgapmers

DAPI γH2AX Mergep=0.03

Fusions Chromatidbreaks

Pericentricgaps

Cohesindefects

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 38: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

33

Figure 6: Model of the proposed role for MSR transcripts in chromocenter organisation. 1

MSR transcripts contribute to chromocenter organisation, by: i) catalysing weak interactions between HP1ɑ 2

molecules; and ii) preventing HP1ɑ from binding to chromatin to sustain a permissive heterochromatin 3

structure in ESCs. Upon MSR RNA depletion, which normally occurs during ESC differentiation, the 4

substrate for weak interactions is reduced and more HP1ɑ molecules bind to satellite DNA, leading to the 5

observed changes in heterochromatin compaction and organisation. 6

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 39: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

HP1αHeterochromatin

with H3K9me3MSR RNA“Liquid-like”

chromocenter“Gel-like”

chromocenter

Reduced levels of MSR RNANormal, high levels of

MSR RNA in ESCs

Figure 6

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 40: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

34

Supplementary Figures 1

2

Supplementary Figure 1: Inducible targeting of GFP to satellite DNA with a TALE does not affect the 3

nuclear organisation of chromocenters, Related to Figure 1. 4

A) Representative images of ESC nuclei from a time-lapse experiment, which show that TALE-MSR-GFP 5

binds to major satellite sequences throughout the cell cycle. Scale bar, 5µm. 6

B, C) Plots showing the number (B) and diameter (C) of chromocenter foci per ESC nucleus upon 24h 7

doxycycline induction of TALE-MSR-GFP. Data were collected from three independent experiments and 8

compared using a two-sample Wilcoxon test. 9

See also Supplementary Videos 1–3. 10

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 41: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

Number of chromocentres per cell

Freq

uenc

y (%

)

p=0.29

20

10

0

TALE-MSR-GFP

1 2 3 4 5 6 1 2 3 4 5 6

TALE-MSR-GFP

DoxNo dox

Supplementary Figure 1

A)

B)

Chr

omoc

entre

dia

met

er (p

ixel

s)

p=0.22

25

20

15

10

0No dox Dox

C)

1h 1h30’ 2h 2h30’ 3h

3h30’ 4h 4h30’ 6h

6h30’ 7h 7h30’

5h 5h30’

9h8h 8h30’

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 42: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

35

Supplementary Figure 2: Embryonic stem cells do not differentiate when MSR transcript levels are 1

reduced, Related to Figure 2. 2

A) Histogram of MSR transcripts levels in ESCs that were either untransfected or transfected with GFP or 3

MSR gapmers, and in mouse embryonic fibroblasts (MEFs). Expression levels are shown as fold-change 4

relative to Hmbs. Two independent biological replicates are shown. 5

B) Immunofluorescence microscopy images of OCT4 in ESCs, following transfection with GFP or MSR 6

gapmers. DNA was counterstained with DAPI. Scale bars, 20µm. 7

C) Histogram showing the transcript levels of undifferentiated ESC marker genes (Nanog; Klf4) and 8

differentiated cells (T), as measured by RT-qPCR. Data were acquired from three independent experiments 9

and each dot represents individual biological replicates. 10

D) Representative images of live ESC nuclei showing TALE-MSR-GFP localisation at chromocenters is 11

retained following transfection with MSR gapmers. Scale bar, 10µm. 12

See also Supplementary Video 4. 13

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 43: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

Supplementary Figure 2

A)

0

0.2

0.4

0.6

0.8

1.0

1.2

T Nanog Klf4

Fold

cha

nge

rela

tive

to G

FP g

apm

er

GFP gapmers MSR gapmers

GFP gapmers MSR gapmers

MS

R g

apm

ers

B)

C) D)

Fold

cha

nge

norm

aliz

edto

Hm

bs re

lativ

e to

ES

C

0.0

0.2

0.4

0.6

0.8

1.0

1 2ESC

1 2MEF

1 2ESC + GFP

gapmers

1 2ESC + MSR

gapmers

MSR transcript levels

DAPI

Nuclei stain TALE-MSR-GFP Merge

OCT4

Merge

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 44: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

36

Supplementary Figure 3: Satellite RNA promotes phase-separation of the heterochromatin protein 1

HP1ɑ, Related to Figure 3. 2

A) Different lengths of forward MSR RNAs contribute to HP1ɑ droplet formation in vitro. Images framed in 3

blue show the conditions that allow for the formation of HP1ɑ droplets. All images show the same 4

magnification; scale bar, 100µm. 5

B) Representative 3D-SIM images after LNA-DNA gapmer transfection. For each experiment, the top panel 6

shows the 3D-SIM reconstruction of H3K9me3 (red), HP1ɑ (green) and merged, and the lower panel shows 7

the equivalent projected volumes. 8

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 45: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

A)

Supplementary Figure 3

B)

0 6.33.21.60.8 12.5 25 50 100

Recombinant HP1α (μM)

GFP

gap

mer

s 3D-S

IM

H3K9me3 HP1α Merge

3D-v

olum

e3D

-SIM

3D-v

olum

e

MS

R g

apm

ers

Forw

ard

Rev

erse

MS

R (1

repe

at)

Forw

ard

Rev

erse

MS

R (2

repe

ats)

Forw

ard

Rev

erse

MS

R (8

repe

ats)

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint

Page 46: Satellite repeat transcripts modulate heterochromatin ......2020/06/08  · 11 transcripts using sequence-specific locked nucleic acid (LNA) DNA gapmers 28. LNA DNA gapmers deplete

37

Supplementary Video 1: Visualising chromocenters throughout the cell cycle, Related to Figure 1. 1

Time lapse imaging of ESCs after doxycycline-induced expression of TALE-GFP against major satellite 2

repeats (MSR). Images were acquired every 15 min over 72 time frames (18 hrs). 3

4

Supplementary Video 2: Rapid movement of chromocenters, Related to Figure 1. 5

Time lapse imaging (compiled five frames per second) of a representative ESC expressing doxycycline-6

induced TALE-MSR-GFP. Images were acquired every 2 min for 60 frames (2h). 7

8

Supplementary Video 3: FRAP analysis of chromocenters, Related to Figure 1. 9

Time lapse imaging (compiled five frames per second) of a representative ESC expressing doxycycline-10

induced TALE-MSR-GFP and after GFP gapmer transfection. One chromocenter was photobleached and 11

images acquired every second for 310 seconds. 12

13

Supplementary Video 4: FRAP analysis of chromocenters after MSR RNA depletion, Related to Figure 14

2. 15

Time lapse imaging (compiled five frames per second) of a representative ESC expressing doxycycline-16

induced TALE-MSR-GFP and after MSR gapmer transfection. One chromocenter was photobleached and 17

images acquired every second for 310 seconds. 18

19

Supplementary Table 1: Additional information related to the Online Methods. 20

.CC-BY-NC-ND 4.0 International licenseavailable under awas not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made

The copyright holder for this preprint (whichthis version posted June 8, 2020. ; https://doi.org/10.1101/2020.06.08.139642doi: bioRxiv preprint