visualizing the role of boundary elements in enhancer ...in brief yokoshi et al. employ quantitative...

29
Article Visualizing the Role of Boundary Elements in Enhancer-Promoter Communication Graphical Abstract Highlights d Intra-domain enhancer-promoter interaction occurs in the absence of TADs d Topological boundaries increase transcriptional output independently of TAD formation d TAD formation is essential for domain-skipping activity of distal enhancers Authors Moe Yokoshi, Kazuma Segawa, Takashi Fukaya Correspondence [email protected] In Brief Yokoshi et al. employ quantitative live- imaging methods to visualize impacts of TAD formation on enhancer-promoter communication. They show that intra- domain enhancer-promoter interaction occurs independently of TAD formation. In contrast, domain-skipping activity of distal enhancers is lost upon TAD disruption, suggesting that intra-domain and inter-domain enhancer-promoter interactions are differentially regulated by chromosome topology. Yokoshi et al., 2020, Molecular Cell 78, 224–235 April 16, 2020 ª 2020 Elsevier Inc. https://doi.org/10.1016/j.molcel.2020.02.007

Upload: others

Post on 08-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Article

Visualizing the Role of Bou

ndary Elements inEnhancer-Promoter Communication

Graphical Abstract

Highlights

d Intra-domain enhancer-promoter interaction occurs in the

absence of TADs

d Topological boundaries increase transcriptional output

independently of TAD formation

d TAD formation is essential for domain-skipping activity of

distal enhancers

Yokoshi et al., 2020, Molecular Cell 78, 224–235April 16, 2020 ª 2020 Elsevier Inc.https://doi.org/10.1016/j.molcel.2020.02.007

Authors

Moe Yokoshi, Kazuma Segawa,

Takashi Fukaya

[email protected]

In Brief

Yokoshi et al. employ quantitative live-

imaging methods to visualize impacts of

TAD formation on enhancer-promoter

communication. They show that intra-

domain enhancer-promoter interaction

occurs independently of TAD formation.

In contrast, domain-skipping activity of

distal enhancers is lost upon TAD

disruption, suggesting that intra-domain

and inter-domain enhancer-promoter

interactions are differentially regulated by

chromosome topology.

Page 2: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Molecular Cell

Article

Visualizing the Role of Boundary Elementsin Enhancer-Promoter CommunicationMoe Yokoshi,1 Kazuma Segawa,2 and Takashi Fukaya1,3,4,*1Laboratory of Transcription Dynamics, Research Center for Biological Visualization, Institute for Quantitative Biosciences, The University of

Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan2Department of Bioinformatics and Systems Biology, Faculty of Science, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan3Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, Bunkyo-ku, Tokyo 113-0032, Japan4Lead Contact

*Correspondence: [email protected]

https://doi.org/10.1016/j.molcel.2020.02.007

SUMMARY

Formation of self-associating loop domains is afundamental organizational feature of metazoan ge-nomes. Here, we employed quantitative live-imagingmethods to visualize impacts of higher-order chro-mosome topology on enhancer-promoter communi-cation in developing Drosophila embryos. Evidenceis provided that distal enhancers effectively producetranscriptional bursting from target promoters overdistances when they are flanked with boundary ele-ments. Importantly, neither inversion nor deletion ofa boundary element abrogates this ‘‘enhancer-as-sisting activity,’’ suggesting that they can facilitateintra-domain enhancer-promoter interaction andproduction of transcriptional bursting independentlyof topologically associating domain (TAD) formation.In contrast, domain-skipping activity of distal en-hancers was lost after disruption of topological do-mains. This observation raises a possibility thatintra-domain and inter-domain enhancer-promoterinteractions are differentially regulated by chromo-some topology.

INTRODUCTION

Enhancers are regulatory DNAs that control spatiotemporal ON/

OFF pattern of gene expression in response to developmental

timing and environmental signals. Whole-genome chromatin

immunoprecipitation (ChIP) studies estimated that the human

genome contains ~400,000 enhancers (ENCODE Project Con-

sortium, 2012), suggesting that a typical human gene is regu-

lated by approximately 20 enhancers. A number of evidences

have been provided that non-coding variations at enhancers

are a major source of phenotypic polymorphism in a population

(e.g., Guenther et al., 2014; Chan et al., 2010). Recent live-imag-

ing studies suggested that enhancers drive bursts of de novo

transcription, or transcriptional bursting, from their target core

promoters to control the level of gene activities in developing

Drosophila embryos (Fukaya et al., 2016; Bothma et al., 2014).

224 Molecular Cell 78, 224–235, April 16, 2020 ª 2020 Elsevier Inc.

Single-molecule fluorescence in situ hybridization assay also

concluded that enhancers modulate bursting parameters in

mammalian cells (Bartman et al., 2016, 2019). More recently,

allele-sensitive single-cell RNA sequencing method further sup-

ported the idea that enhancer control of transcriptional bursting

is a general mechanism of gene regulation conserved among

species (Larsson et al., 2019).

Importantly, recent chromosome conformation capture

methods revealed that the genome consists of a series of self-

associating loop domains, or topologically associating domains

(TADs) (Dixon et al., 2012; Nora et al., 2012; Sexton et al., 2012).

In vertebrates, it is thought that chromatin extruding cohesin, a

ring-shaped structural maintenance of chromosomes (SMC)

complex, stops at the convergent CCCTC-binding factor

(CTCF) sites to establish TADs (Fudenberg et al., 2016; Sanborn

et al., 2015; Rao et al., 2014). One of the key insights obtained

from these studies is that enhancers and their target genes typi-

cally reside in a same TAD, suggesting that regulatory interac-

tions mainly occur within individual topological domains (re-

viewed in Long et al., 2016). However, it remains largely

unclear how TAD formation itself influences dynamics of

enhancer-promoter communication and resulting transcriptional

bursting.

In this study, we successfully visualized impacts of genome to-

pology on transcription dynamics by using MS2/MCP live-imag-

ing method in developing Drosophila embryos (Garcia et al.,

2013; Lucas et al., 2013). Evidence is provided that large

enhancer-promoter distance significantly diminishes the level

of gene activities by affecting the timing and the size of transcrip-

tional bursting. Intriguingly, bursting profiles were recovered

when distal enhancer and target promoter were flanked with a

pair of topological boundaries. This ‘‘enhancer-assisting activ-

ity’’ was not lost even after inversion or deletion of the CTCF-

binding site at one side, giving rise to a possibility that boundary

elements can facilitate distal enhancers to induce transcriptional

bursting independently of TAD formation. This idea was further

supported by genome editing and live-imaging analysis of highly

structured endogenous fushi-tarazu (ftz) locus. Although intra-

domain enhancer-promoter interaction can take place even after

deletion of topological boundary, quantitative image analysis re-

vealed that TAD formation per se mediates subtle increase

(~10%–20%) in total RNA production, which may contribute to

robust gene expression in a natural population. In contrast,

Page 3: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

A

24x MS2

Drosophila SysntheticCore Promoter

yellowsna shadow

Enhancer-Promoter distance: 6.5 kb

enhancer

C Enhancer-Promoter distance: 9 kb

24x MS2 yellowsna shadowenhancer

Drosophila SysntheticCore Promoter

0

0.2

0.4

0.6

0.8

1.0

Cum

ulat

ive

frac

tion

0 6 8 10 122 4 x105

Total RNA produced (AU)

Distance: 9 kb (363)Distance: 6.5 kb (356)

G

P <10-71

EMid nuclear cycle 14

His2Av-mRFP Most ventral nuclei

Onset of gastrulation

5.0 min 19.8 min 39.9 min

His2Av-mRFP Cumulative RNA production

B Distance: 6.5 kb D

5.0 min 19.8 min 39.9 min

His2Av-mRFP Cumulative RNA production

Distance: 9 kb

0 10 20 30 40Time (min)

F

Mea

n cu

mul

ativ

eR

NA

pro

duct

iuon

(A

U)

0

2

3

4

5

6

1

Distance:6.5 kb

Distance:9 kb

x105

0

0.5

1.0

1.5

2.0

0 10 20 30 40Time (min)

Distance: 6.5 kb

MS

2 in

tens

ity (

AU

) x104

H

0

0.5

1.0

1.5

2.0

0 10 20 30 40Time (min)

Distance: 9 kb

MS

2 in

tens

ity (

AU

) x104

I

OFF

ON

0 10 20 30 40Time (min)

Bur

st

J

0 10 20 30 40Time (min)

OFF

ON

Bur

st

K

Distance: 6.5 kb Distance: 9 kb

Num

ber

of b

urst

Frequency

0

2

4

6

8

10

P

median 67

RN

A pr

oduc

ed p

er b

urst

(AU

)

Burst size

0

5

10

15x10

4

L

56median

Duration

Tim

e (m

in)

0

2

4

6

8

10

M

median 86

N

% of 6.5 kbreporter

Tim

e (m

in)

Onset of first burst

0

10

30

40

20

O

median 247

Amplitude

02468

141210

x103

MS

2 in

tens

ity (

AU

)

median 66

(legend on next page)

Molecular Cell 78, 224–235, April 16, 2020 225

Page 4: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

analysis of the neighboring Hox geneSex comb reduced (Scr) re-

vealed that gene-skipping activity of distal T1 enhancer is strictly

dependent on TAD formation, suggesting that intra-domain and

inter-domain enhancer-promoter interactions are differentially

regulated by chromosome topology. Our study provides a first

glimpse into previously uncharacterized roles of topological

boundaries in the control of enhancer-promoter communication

during animal development.

RESULTS

Enhancer-Promoter Distance Significantly ImpactsTranscriptional OutputTo quantitatively visualize how enhancer-promoter distance in-

fluences the dynamics of transcriptional activities, the well-

defined snail (sna) shadow enhancer was positioned in two

different locations of the synthetic MS2 reporter gene, 6.5 kb

or 9 kb downstream of the core promoter sequence (Figures

1A and 1C). The ~1.5-kb sna shadow enhancer drives expres-

sion in the presumptive mesoderm cells and plays an essential

role in the formation of ventral furrow by delineating the boundary

from neurogenic ectoderm (Dunipace et al., 2011; Perry et al.,

2010). To quantify transcription activities in living Drosophila em-

bryos, production of nascent MS2 transcripts was visualized us-

ing maternally provided MCP-GFP fusion protein during nuclear

cycle (nc) 14 (Video S1). In either configuration, sna shadow

enhancer mediated expression at the ventral region. However,

there was significant diminishment in the level of nascent RNA

production when enhancer-promoter distance became larger

(Figures 1B and 1D; Video S2). To unambiguously compare

expression profiles of two different reporter genes at the same

dorsal-ventral (DV) position, we quantified transcription activities

at themost ventral nuclei defined by the location of ventral furrow

formation during onset of gastrulation (Figure 1E). Consistent

with whole ventral view (Figures 1B and 1D), there was clear

(more than 50%) reduction in the total amount of RNA synthesis

Figure 1. Enhancer-Promoter Distance Impacts Transcriptional Outpu

(A) Schematic representation of the yellow reporter gene containing the 155-bp D

shadow enhancer, and 243 MS2 RNA stem loops (Bertrand et al., 1998) within t

(B) False coloring of yellow reporter gene expression in the embryo containing D

colored nuclei is proportional to the level of cumulative RNA production at given tim

in gray. Images are oriented with anterior to the left and ventral view facing up. S

(C) Schematic representation of the yellow reporter gene containing the 155-bpDS

50 UTR. The 2.5-kb linker sequence derived from bacterial DNA was inserted in b

(D) False coloring of yellow reporter gene expression in the embryo containingDS

colored nuclei is proportional to the level of cumulative RNA production at given tim

in gray. Images are oriented with anterior to the left and ventral view facing up. S

(E) Snapshot of a gastrulating embryo (left) and false coloring of most ventral nucle

in gray. Images are oriented with anterior to the left and ventral view facing up. S

(F) Mean cumulative RNA production per nucleus. Shades represent standard de

for 6.5-kb reporter; 363 nuclei for 9-kb reporter) from three independent embryo

(G) A cumulative plot showing fraction of most ventral nuclei (y axis) and total RN

(H and I) A representative trajectory of transcriptional activity of theMS2 reporter g

(J) Binarized burst profile obtained from the analysis of raw trajectory shown i

shown in (I).

(L–P) Boxplots showing the distribution of burst size (L), burst duration (M), ampli

lower (25%) and upper (75%) quantiles, and the solid line indicates the median. W

363 most ventral nuclei, respectively, were analyzed from three individual embr

Relative median values normalized to the 6.5-kb reporter were shown in bottom.

226 Molecular Cell 78, 224–235, April 16, 2020

when the distance became larger (Figures 1F and 1G). These

data indicate that the size of enhancer-promoter separation

has a significant impact on gene activities. Supporting this

view, transcriptional output was further diminished when dis-

tance was extended from 9 kb to 11 kb (Figure S1A).

To explore the mechanism underlying this ‘‘distance effect,’’

we obtained MS2 signal trajectories from over 300 nuclei at the

most ventral region per reporter. In both 6.5-kb and 9-kb re-

porters, sna shadow enhancer produced intermittent bursts of

de novo transcription, or transcriptional bursting (Figures 1H

and 1I), but their profiles look quite different. To characterize

quantitative differences in detail, we computationally detected

individual bursting events (Figures 1J and 1K) and analyzed func-

tional parameters, including frequency, timing, and burst size

(Figure S1B). We found that burst size (i.e., number of nascent

transcripts produced per burst) became nearly half when the dis-

tance got larger (Figure 1L). This reduction seems to be mainly

attributed to lower amplitude rather than shorter duration of indi-

vidual bursting events (Figures 1M and 1N). Moreover, there was

significant delay in the onset of transcriptional bursting (Fig-

ure 1O), which leads to overall diminishment of bursting fre-

quency (Figure 1P). Consistent with this, difference in output at

the first 15 min was much larger than difference in total output

(Figure S1C). Overall, our data suggest that enhancer-promoter

distance contributes to the control of gene activities by changing

the size and the timing of transcriptional bursting.

Boundary Elements Facilitate Enhancer-PromoterInteractionOur results showed that large enhancer-promoter distance di-

minishes the level of gene activities (Figure 1). However, regula-

tory interactions in this distance range are thought to commonly

take place in the Drosophila genome because whole-genome

enhancer survey estimated median enhancer-promoter separa-

tion to be ~10 kb (Kvon et al., 2014). Then how do enhancers

overcome large distances from their target promoters?

t

rosophila synthetic core promoter (DSCP) (Pfeiffer et al., 2008), the 1.5-kb sna

he 50 UTR.SCP-MS2-yellow-sna shadow enhancer (distance: 6.5 kb). Intensity of false-

e in a given nucleus. Themaximumprojected image of His2Av-mRFP is shown

cale bar indicates 25 mm.

CP, the 1.5-kb sna shadow enhancer, and 243MS2RNA stem loopswithin the

etween yellow and sna shadow enhancer.

CP-MS2-yellow-linker-sna shadow enhancer (distance: 9 kb). Intensity of false-

e in a given nucleus. Themaximumprojected image of His2Av-mRFP is shown

cale bar indicates 25 mm.

i at mid-nc 14 (right). Themaximum projected image of His2Av-mRFP is shown

cale bar indicates 25 mm.

viation of the mean across all analyzed nuclei at most ventral region (356 nuclei

s.

A production (x axis). The p value of Wilcoxon rank-sum test was shown.

enewith 6.5-kb (H) and 9-kb enhancer-promoter distance (I) in individual nuclei.

n (H). (K) Binarized burst profile obtained from the analysis of raw trajectory

tude (N), onset of first burst (O), and burst frequency (P). The box indicates the

hiskers extend to the most extreme, non-outlier data points. A total of 356 and

yos for the reporter gene with 6.5-kb and 9-kb enhancer-promoter distance.

See also Figure S1.

Page 5: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

0

0.4

0.8

1.2

1.6

0 10 20 30 40

Time (min)

MS

2 in

tens

ity (

AU

)

No boundary

x104

I

0

0.4

0.8

1.2

1.6

MS

2 in

tens

ity (

AU

)

0 10 20 30 40

Time (min)

Nhomie/Homie

x104

J

0

0.4

0.8

1.2

1.6

MS

2 in

tens

ity (

AU

)

0 10 20 30 40

Time (min)

Nhomie/Homieinv

x104

KN

umbe

r of

bur

st

0

2

4

6

8

10

Tim

e (m

in)

0

10

30

40

20

Tim

e (m

in)

0

2

4

6

8

10

0

5

10

15

MS

2 in

tens

ity (

AU

)

FrequencyPL Onset of first burst

ODurationM AmplitudeN

x103

Nhomie/Homie Nhomie/HomieNo boundary inv

median 125 125median 273 198 median 146 126 median 181 160 median 45 57 % ofNo boundary

RN

A pr

oduc

ed p

er b

urst

(AU

)

0

5

15

25

20

10

Burst size

x104

6,390 6,410 6,430 6,450 6,470 (kb)

6,39

06,

410

6,43

06,

450

6,47

0 (

kb)

Color Range [0-472]

A

Reporterlanding site

9,94

09,

960

9,98

010

,000

10,0

20 (

kb)

Color Range [0-474]

9,940 9,960 9,980 10,000 10,020 (kb)

Nhomie

Homie

B

eve TAD

Nhomie Homie

Rad21CTCFPol II

eve TER94CG12134

[0-1132]

[0-3]

[0-2000]

C

inv

F G

0

0.2

0.4

0.6

0.8

1.0

Cum

ulat

ive

frac

tion

0 6 8 10 122 4 x105

Total RNA produced (AU)

H

Nhomie/Homie (350)

Nhomie/Homie (359)inv

P <10-70P <10

-90No boundary (363)

P <10-12

Nhomie/Homie

01234567

0 10 20 30 40

Time (min)

x105

Mea

n cu

mul

ativ

eR

NA

pro

duct

ion

(AU

)

Nhomie/Homieinv

01234567

0 10 20 30 40

Time (min)

x105

Mea

n cu

mul

ativ

eR

NA

pro

duct

ion

(AU

)

E No boundary

x105

01234567

0 10 20 30 40

Time (min)

Mea

n cu

mul

ativ

eR

NA

pro

duct

ion

(AU

)

D

Nhomie Homie Homieor

Enhancer-Promoter distance: 9 kb

24x MS2 yellowsna shadow

enhancer

Figure 2. Boundary Elements Augment Transcriptional Output

(A and B) Hi-C map of the reporter gene landing site used in this study (A) and the endogenous eve locus (B). Hi-C data from a WT embryonic cell line (Cubenas-

Potts et al., 2017) was visualized with Juicebox browser (Durand et al., 2016).

(C) Organization of the endogenous eve locus. Rad21 ChIP sequencing (ChIP-seq) data from a WT embryonic cell line (Van Bortle et al., 2014), CTCF ChIP-chip

data from 0- to 12-h WT embryos (Roy et al., 2010), and Pol II ChIP-seq data from 2- to 3-h WT embryos (Sun et al., 2015) were visualized with Integrative

Genomics Viewer (IGV).

(D) Schematic representation of the yellow reporter gene containing the 1,328-bp Nhomie, the 155-bp DSCP, the 2.5-kb linker sequence, the 1.5-kb sna shadow

enhancer, and 243MS2RNA stem loopswithin the 50 UTR. The 367-bp Homie was placed in the same orientation as in endogenous eve locus (Homie) or inverted

orientation (Homieinv).

(legend continued on next page)

Molecular Cell 78, 224–235, April 16, 2020 227

Page 6: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Intriguingly, when we looked at Hi-C profile (Cubenas-Potts

et al., 2017), we noticed that our reporter gene landing site lacks

formation of TADs (Figure 2A). On the other hand, many of devel-

opmental patterning genes are known to be located in topolog-

ical domains (e.g., Stadler et al., 2017; Ulianov et al., 2016). A

well-characterized pair-rule gene even-skipped (eve) is also

located within a clear TAD structure (Figure 2B). Previous genetic

studies showed that the eve TAD is shaped by pairing of

well-characterized boundary elements, Nhomie and Homie

(Fujioka et al., 2009, 2013), both of which contain high level of

CTCF and Rad21 binding (Figure 2C). This interaction is thought

to be strictly orientation dependent because inversion of Nhomie

or Homie disrupts long-range interaction in cis and trans (Fujioka

et al., 2009, 2016). We have also examined orientation

dependence of Homie elements by analyzing trans-homolog

enhancer-promoter interaction or transvection (Lim et al.,

2018b). Because transvection strictly requires pairing of bound-

ary elements across homologous chromosomes, we utilized this

system to functionally characterize their interaction properties.

Our data showed that inversion of Homie is sufficient to eliminate

transvection activities, supporting the idea that they act as an

orientation-dependent boundary element in early embryos.

To explore the possibility that TAD formation may increase

an efficiency of intra-domain enhancer-promoter interaction,

Nhomie and Homie were inserted into the 50 and 30 locations of

synthetic MS2 locus as a loop anchor (Figure 2D). Live-imaging

analysis revealed substantial increase in RNA production when

these boundary elements were placed in the same orientation

as in endogenous eve locus (Figures 2E and 2F; Video S3).

Intriguingly, however, even after inversion of Homie, transcrip-

tional upregulation was still observed (Figures 2E and 2G; Video

S4), giving rise to a possibility that boundary elements can

somehow facilitate enhancer-promoter interaction indepen-

dently of TAD formation. Supporting this view, transcriptional

output of the inverted Homie reporter remained to be compara-

ble even after deletion of Homie at the 30 location (Figures S2A–

S2E). Importantly, the reporter gene containing Homie produced

even higher output than the one with inverted Homie (Figure 2H),

implicating that the Nhomie/Homie pairing can further increase

transcription activities. Overall, these observations are consis-

tent with the idea that boundary elements employ pairing-

dependent and independent mechanisms to facilitate distal

enhancers to activate target promoters from remote locations.

We also suggest that boundary elements can exert their function

in various genomic context because Nhomie/Homie facilitated

(E–G) Mean cumulative RNA production per nucleus. Embryos containing the MS2

with Nhomie and Homieinv (G) were analyzed. Shade represents standard deviatio

boundary reporter; 350 nuclei for Nhomie/Homie reporter; 359 nuclei for Nhomie/

same as the plot in Figure 1F (distance: 9 kb).

(H) A cumulative plot showing fraction of nuclei (y axis) and total RNA production (x

is the same as the plot in Figure 1G (distance: 9 kb).

(I–K) A representative trajectory of transcriptional activity of the MS2 reporter gen

and Homieinv (K) in individual nuclei.

(L–P) Boxplots showing the distribution of burst size (L), burst duration (M), ampli

lower (25%) and upper (75%) quantiles, and the solid line indicates the median. W

and 359 most ventral nuclei, respectively, were analyzed from three individual e

Nhomie/Homieinv. Plots of no boundary are the same as the plots in Figures 1L–

boundary elements were shown in bottom. See also Figure S2.

228 Molecular Cell 78, 224–235, April 16, 2020

enhancer-promoter interaction separated by a shorter genomic

distance (Figures S2F–S2I).

When we analyzed bursting profiles after placing boundary

elements, there was clear upregulation of burst production

(Figures 2I–2K). Significant changes were seen for burst size

and timing. Nhomie/Homie increased overall frequency by pro-

moting rapid induction of transcriptional bursting (Figures 2O,

2P, and S1D). In addition, when Nhomie and Homie were

aligned as in eve locus, burst size became more than 2.5-fold

bigger (Figure 2L). Burst size remained to be nearly twice

bigger even after Homie inversion (Figure 2L). In either case,

amplitude was substantially increased with slightly extended

burst duration when compared with the control (Figures 2M

and 2N). These results are consistent with the idea that pair-

ing-dependent and independent mechanisms contribute to

induction of transcriptional bursting by distal enhancers.

Because bursts are thought to occur by recruiting a series of

RNA polymerase II (Pol II) clusters through the formation of

transcription ‘‘hubs’’ or transcriptional ‘‘condensates’’ (Boija

et al., 2018; Cho et al., 2016, 2018; Chong et al., 2018; Lim

et al., 2018b; Sabari et al., 2018), it might be possible that

topological boundaries act as a scaffold to increase local

concentration of active transcription machineries at nearby

enhancers (see Discussion).

Deletion of SF1 Topological Boundary EliminatesExpression of Scr, but Not ftzTo further explore the role of boundary elements in the context of

endogenous genome, we decided to focus on another well-char-

acterized pair-rule gene fushi-tarazu (ftz) because it is located in

the highly structured Hox gene cluster, Antennapedia complex.

Recent Hi-C study revealed that ftz transcription unit and its

regulatory elements are embedded in a same topological

domain as in eve locus (Stadler et al., 2017). ftz TAD has been

shown to be bordered by well-characterized boundary elements

SF1 and SF2 (Li et al., 2015b; Belozerov et al., 2003), both of

which are enriched for CTCF and Rad21 as well as other archi-

tectural proteins, including Su(Hw), CP190, Mod(mdg4), and

TFIIIC (Figure 3A). To monitor ftz transcription in living embryos,

we first inserted 243 MS2 sequence into the 30 UTR by using

CRISPR/Cas9 (Lim et al., 2018b; Ren et al., 2013). Homozygotes

of ftz-MS2 allele were fully viable and phenotypically indistin-

guishable from wild-type (WT) (Lim et al., 2018a), suggesting

that MS2 stem loops do not impede either transcription or

translation. Subsequently, SF1 was removed by the second

reporter gene without boundary elements (E), with Nhomie and Homie (F), and

n of the mean across all analyzed nuclei at most ventral region (363 nuclei for no

Homieinv reporter) from three independent embryos. Plot of no boundary is the

axis). The p values of Wilcoxon rank-sum test were shown. Plot of no boundary

e without boundary elements (I), with Nhomie and Homie (J), and with Nhomie

tude (N), onset of first burst (O), and burst frequency (P). The box indicates the

hiskers extend to the most extreme, non-outlier data points. A total of 363, 350,

mbryos for the reporter gene with no boundary element, Nhomie/Homie, and

1P (distance: 9 kb). Relative median values normalized to the reporter without

Page 7: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

A

B C

D E F

Figure 3. SF1 Deletion Eliminates Expres-

sion of Scr, but Not ftz

(A) Organization of the endogenous Scr-ftz locus.

Rad21 and TFIIIC ChIP-seq data from a WT em-

bryonic cell line (Van Bortle et al., 2014), CTCF,

Su(Hw), CP190, Mod(mdg4), and BEAF-32 ChIP-

chip data from 0- to 12-h WT embryos (Roy et al.,

2010), and Pol II ChIP-seq data from 2- to 3-h WT

embryos (Sun et al., 2015) were visualized with IGV

browser. Approximate locations of stripe 1, stripe

2, and T1 enhancers were shown in orange.

(B and C) Double fluorescent in situ hybridization of

endogenous ftz (upper panel) and Scr (lower panel)

in WT ftz-MS2 (B) and ⊿SF1 ftz-MS2 homozygote

embryos (C). Embryos at late nc 14 were shown.

Imageswere cropped and rotated to align embryos

(anterior to the left and posterior to the right). In

⊿SF1 embryos, there was faint ectopic expression

of Scr that colocalized with anterior ftz stripes.

Scale bars indicate 50 mm.

(D) False coloring of nuclei at stripes 1 and 2

region (yellow) and inter-stripe region (blue). The

maximum projected image of His2Av-mRFP is

shown in gray. Images are oriented with anterior to

the left and ventral to the bottom. Scale bar in-

dicates 25 mm.

(E and F) A cumulative plot showing fraction of

nuclei (y axis) and total RNA production (x axis) in

stripe 1 (E) and stripe 2 nuclei (F). The p value of

Wilcoxon rank-sum test was shown. Change in

median value after SF1 deletion was shown in up-

per left. See also Figure S3.

round of genome editing. Deletion has been confirmed by PCR

analysis of genomic DNA (Figures S3A and S3B).

We then performed in situ hybridization to examine spatial

patterning of ftz expression. Even after SF1 deletion, ftz re-

mained to be expressed in seven stripes (Figures 3B and 3C),

suggesting that TAD formation is not a prerequisite for the estab-

lishment of ftz stripes. We also examined expression of the

neighboring Hox gene, Sex comb reduced (Scr). Scr is ex-

pressed at the posterior head segment whose transcription is

driven by the distal T1 enhancer located 30 of the ftz gene and

~25 kb away from the Scr promoter (Figure 3A; Gindhart et al.,

1995). Because ftz TAD is located in between T1 enhancer and

Scr promoter, this interaction is thought to require skipping of

an intervening topological domain. Indeed, recent Hi-C analysis

captured inter-domain T1-Scr interaction in earlyDrosophila em-

bryos (Stadler et al., 2017). Contrary to stripe enhancers, T1

enhancer failed to mediate correct Scr expression in the

absence of SF1 (Figures 3B and 3C), suggesting that pairing be-

tween SF1 and SF2 is essential for the domain-skipping activity

of distal T1 enhancer. Although Scr expression in nc 14 was lost

Mol

in ⊿SF1 mutant embryos, there was no

reduction in the number of sex comb teeth

(Figure S3C), suggesting that the remain-

ing intronic enhancer compensates Scr

expression in later development (Eksi

et al., 2018). Indeed, we detected Scr

expression in both WT and SF1 deletion

mutant after germband extension (Figures S3D and S3E). Impor-

tantly, we reproducibly detected weak ectopic Scr expression at

anterior region of mutant embryos (Figure 3C). Double fluores-

cent in situ hybridization revealed that ectopic Scr pattern over-

laps with anterior ftz stripes (Figures S3F and S3G), suggesting

that loss of SF1 leads to crosstalk between Scr promoter and

neighboring stripe enhancers. Staining of ectopicScr expression

in ⊿SF1 mutant embryos was much weaker than in WT (Figures

3B and 3C), which is consistent with previous observation that ftz

enhancers exhibit intrinsic preference for TATA-containing ftz

promoter over TATA-less Scr promoter (Calhoun and Levine,

2003; Calhoun et al., 2002; Ohtsuki et al., 1998). Overall, our

data suggest that intra-domain and inter-domain enhancer-pro-

moter interactions have differential dependence on chromo-

some topology.

Topological Regulation of ftz Stripe FormationNext, we performed live-imaging analysis to explore quantitative

differences during ftz stripe formation in WT and ⊿SF1 embryos

(Video S5). We particularly focused on stripes 1 and 2 because

ecular Cell 78, 224–235, April 16, 2020 229

Page 8: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

0

0.5

1.0

1.5

2.0

0 10 20 30 40

Time (min)

MS

2 in

tens

ity (

AU

)

2.5

WT Stripe 1A

0

0.5

1.0

1.5

2.0

0 10 20 30 40

Time (min)

MS

2 in

tens

ity (

AU

)

2.5

WT Stripe 2E

0

0.5

1.0

1.5

2.0

0 10 20 30 40

Time (min)

MS

2 in

tens

ity (

AU

)

2.5

⊿SF1 Stripe 1B

0

0.5

1.0

1.5

2.0

0 10 20 30 40

Time (min)

MS

2 in

tens

ity (

AU

)2.5

⊿SF1 Stripe 2F

0 10 20 30 40

Time (min)

Mea

n in

tens

ity (

AU

)

0

0.4

0.8

1.2

1.6

2.0 ⊿SF1WTStripe 1D

⊿SF1WTStripe 2

Mea

n in

tens

ity (

AU

)

0

0.4

0.8

1.2

1.6

2.0

H

0 10 20 30 40

Time (min)

0 10 20 30 40

Total ON duration (min)

0 10 20 30 40

Total ON duration (min)

0

0.2

0.4

0.6

0.8

1.0

Cum

ulat

ive

frac

tion

0

0.2

0.4

0.6

0.8

1.0

Cum

ulat

ive

frac

tion

C

G

Stripe 1

Stripe 2

WT (249)⊿SF1 (236)

P < 10-8

WT (232)⊿SF1 (246)

P < 10-4

x104

x104

x104

x104

27.5 min23.1 min

25.6 min23.0 min

x104

x104

Figure 4. Dynamics of ftz Stripe Formation in SF1 Deletion Mutant

(A and B) A representative trajectory of transcriptional activity of ftz-MS2 in WT (A) and ⊿SF1 allele (B) in individual nuclei at the stripe 1 region.

(C) A cumulative plot showing fraction of nuclei (y axis) and total active duration (x axis) in nuclei at stripe 1 region. The p value of Wilcoxon rank-sum test was

shown. Median values were shown in upper left.

(D) Mean MS2 spot intensity per nucleus. Shades represent standard deviation of the mean across all analyzed nuclei at stripe 1 region (249 nuclei for WT; 236

nuclei for ⊿SF1) from three independent embryos.

(E and F) A representative trajectory of transcriptional activity of ftz-MS2 in WT (E) and ⊿SF1 allele (F) in individual nuclei at the stripe 2 region.

(G) A cumulative plot showing fraction of nuclei (y axis) and total active duration (x axis) in nuclei at stripe 2 region. The p value of Wilcoxon rank-sum test was

shown. Median values were shown in upper left.

(H) Mean MS2 spot intensity per nucleus. Shades represent standard deviation of the mean across all analyzed nuclei at stripe 2 region (232 nuclei for WT; 246

nuclei for ⊿SF1) from three independent embryos. See also Figure S4.

enhancers responsible for each stripe were separately posi-

tioned at downstream and upstream of the ftz transcription unit

(Figure 3A; Schroeder et al., 2011; Calhoun and Levine, 2003), al-

lowing us to monitor impacts of SF1 deletion on enhancers at

different locations. In addition, it provides a nice system to visu-

alize transcriptional repression because formation of inter-stripe

region requires silencing by the product of gap gene repressor

Kr€uppel (Kr) (Calhoun and Levine, 2003). We observed slight

diminishment (~10%–13%) in the level of total RNA production

at stripes 1 and 2 after SF1 deletion (Figures 3D–3F), suggest-

ing that TAD formation can facilitate both 50 and 30 enhancers to

efficiently interact with ftz promoter. Importantly, however,

reduction was not as drastic as seen for double deletion of

Nhomie and Homie in our reporter system (more than 60%

change; Figures 2E and 2F), implicating that the remaining

SF2 facilitates enhancer-promoter interaction independently

of TAD formation as in the case of Homie inversion or single

deletion at synthetic locus (Figures S2A–S2E). We suggest

that, although TAD formation per se can increase overall tran-

scriptional output, it has a limited contribution on intra-domain

enhancer-promoter communication and spatial patterning of

gene expression. Supporting this view, recent whole-genome

analysis of Drosophila balancer chromosome showed that

expression for the majority of genes is not altered even after

large-scale chromosome rearrangements and misregulation of

TAD structures (Ghavi-Helm et al., 2019). We also suggest

that transcriptional repression is not dependent on the higher-

order chromosome topology because nuclei at inter-stripe re-

230 Molecular Cell 78, 224–235, April 16, 2020

gion did not exhibit derepression after SF1 deletion (Fig-

ure S4A). This result is consistent with previous observation

that Kr acts as a short-range repressor that mediates direct

repression of promoters and transcriptional activators in a close

linkage (Gray and Levine, 1996).

To examine how SF1 deletion influences transcription dy-

namics, we next analyzed individual MS2 trajectories. In WT em-

bryos, transcription generally occurs in burst both at stripe 1 and

stripe 2 (Figures 4A and 4E; Video S5), but individual bursting

events were hard to be discerned due to continuity of bursting

activities. In contrast, each burst was clearly separated at in-

ter-stripe region (Figure S4B). We speculate that ftz locus is opti-

mized to achieve a high level of gene expression at stripe regions

by producing next burst before clearance of previous bursts.

Intriguingly, ftz bursts became less continuous when SF1 was

deleted (Figures 4B and 4F). When we quantified total active

duration to avoid ambiguity to define individual bursting event,

it turned out that this trend was true for entire nuclei both at stripe

1 and stripe 2 (Figures 4C and 4G). In addition, there was slight

reduction in the mean fluorescent intensity of ftz transcription

foci (Figures 4D and 4H). These data suggest that, although

TADs are not a prerequisite for transcriptional activation, they

can facilitate intra-domain enhancer-promoter interaction to

achieve highest level of RNA production. We speculate that sub-

tle increase mediated by TAD formation can contribute to robust

gene expression under various stress conditions in natural envi-

ronment, which may give selective advantages in a population.

At the inter-stripe region, there appears to be no repression lag

Page 9: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Inter-domaininteraction

Enhancer

Boundaryelement Boundary

element

Intra-domaininteraction

Enhancer

Increase local concentration oftranscription machinaries

B

EnhancerBoundaryelement Boundary

element

Assembly of transcription hub

A

TADformation

Figure 5. Roles of Boundary Elements in

Enhancer-Promoter Communication

(A) Intra-domain enhancer-promoter interaction

can take place in the absence of TADs (left). In

contrast, TAD formation is required for inter-

domain enhancer-promoter interaction (right).

(B) Boundary elements promote distal enhancers

to activate target promoters independently of TAD

formation. Not only cohesin and CTCF but also

many other regulatory proteins are highly enriched

at TAD boundaries. They may contribute to as-

sembly of transcription ‘‘hub’’ by facilitating

recruitment of transcription factors (yellow and

orange ovals) and co-activators (pink oval) at

nearby enhancers and Pol II complexes (blue

ovals) at core promoters. See also Figure S5.

even after SF1 deletion (Figures S4B–S4D), supporting the idea

that short-range repressors can inhibit burst production inde-

pendently of TAD formation. Finally, we examined activity of

SF1 and SF2 boundary elements in the synthetic reporter

system (Figure S4E). They increased the level of total RNA pro-

duction (Figures S4F and S4G) but with different degree from

Nhomie and Homie (Figures 2E–2H), suggesting that each pair

of boundary elements has distinct capability of facilitating

enhancer-promoter communication to differentially control tran-

scriptional activities at individual loci.

DISCUSSION

In this study, we presented evidence that boundary elements

help distal enhancers to induce transcriptional bursting from

their target promoters. Our data suggest that TAD formation is

not a prerequisite for facilitating intra-domain enhancer-pro-

moter interaction because inversion or deletion of Homie did

not abrogate this ‘‘enhancer-assisting activity.’’ Supporting this

view, deletion of SF1 boundary element from the endogenous lo-

cus did not eliminate ftz stripe formation. In contrast, analysis of

the neighboring Hox geneScr revealed that domain-skipping ac-

tivity of distal T1 enhancer relies on pairing between SF1 and SF2

boundary elements, suggesting that intra-domain and inter-

domain enhancer-promoter interactions have differential depen-

dence on TAD formation (Figure 5A).

Roles of TADs in Transcriptional RegulationIt is well established that TAD formation is mainly regulated by

a Zn-finger DNA-binding protein CTCF and the ring-shaped

SMC complex cohesin in vertebrates. When CTCF or Rad21,

a kleisin subunit of cohesin, is acutely removed by auxin-induc-

ible degron (Natsume et al., 2016), essentially all TADs are lost

in mammalian cultured cells (Nora et al., 2017; Rao et al., 2017).

Consistent with this, knockout of cohesin-loading factor Nipbl

leads to loss of TADs in mouse liver (Schwarzer et al., 2017).

Importantly, these studies agree well in a point that elimination

Mol

of TADs has a modest influence on over-

all transcription activities. Thus, topolog-

ical boundaries and resulting TAD struc-

tures have been implicated to play an

auxiliary role in enhancer-promoter communication and gene

expression. In this study, we combined quantitative live imag-

ing and genome-editing methods to directly visualize impacts

of higher-order genome topology on enhancer-promoter inter-

action. Our data suggest that TAD formation itself has minor in-

fluence on intra-domain enhancer-promoter interaction and

spatial patterning of gene expression because we observed

only ~20% reduction in the total output after Homie inversion

(Figure 2; Nhomie/Homie versus Nhomie/Homieinv) and

~10%–13% reduction after SF1 deletion (Figure 3). Consistent

with this, recent whole-genome sequencing study of balancer

chromosomes that contain massive rearrangements of genome

topology revealed that alternation of TAD structures has minor

impacts on overall level and specificity of gene expression in

Drosophila (Ghavi-Helm et al., 2019). Similarly, in mammalian

system, genome-editing analysis of Sox9 locus showed that

TAD disruption results in only ~10%–15% reduction of Sox9

expression and no detectable phenotypes in developing mouse

limb buds (Despang et al., 2019), again suggesting non-essen-

tial role of TADs in gene expression. Supporting this view, it

was recently reported that Drosophila CTCF-null mutant can

progress through embryogenesis (Gambetta and Furlong,

2018). Moreover, it is known that C. elegans and Arabidopsis

lack CTCF and TAD-like structures (Crane et al., 2015; Feng

et al., 2014; Grob et al., 2014), yet they achieve productive

gene expression during development. We speculate that

TAD-mediated intra-domain interactions confer robustness of

transcriptional program under environmental fluctuations,

such as temperature change and nutrient availability, thereby

providing selective advantages in a population. Contrary to

intra-domain regulatory interactions, our data clearly showed

that inter-domain T1-Scr interaction was compromised in

⊿SF1 embryos (Figure 3), suggesting that pairing of boundary

elements is required for gene-skipping activity of distal en-

hancers (Figure 5A). Given an essential role of SF1 in correct

Scr expression, it might be possible that ftz TAD is formed as

a consequence of bringing distal T1 enhancer and Scr

ecular Cell 78, 224–235, April 16, 2020 231

Page 10: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

promoter together by looping out an intervening ftz transcrip-

tion unit. Importantly, ⊿SF1 mutant embryos developed nor-

mally even without early Scr expression (Figures 3B, 3C, and

S3C–S3E), suggesting that enhancer redundancy acts as an

alternative mechanism to achieve reliable gene expression un-

der various genetic perturbations, including misregulation of

TAD formation.

Enhancer-Assisting Activity of Boundary ElementsOur data suggest that boundary elements are capable of facili-

tating enhancer-promoter communication independently of

TAD formation because the level of gene activities remained to

be substantially higher even after Homie inversion or deletion

(Figures 2 andS2A–S2E). As inmammalian genome, TADbound-

aries are enriched for CTCF and Rad21 in Drosophila (Van Bortle

et al., 2014). In past studies, much effort has been focused on

functional analysis of these proteins because CTCF and cohesin

have been long known to play a fundamentally important role in

genome organization (reviewed in Ong and Corces, 2014). How-

ever, these are not the only proteins enriched at boundaries.

Whole-genome ChIP assays revealed that so-called insulator

proteins, such as Su(Hw),Mod(mdg4), CP190, andBEAF-32, co-

localize with CTCF-binding sites in the Drosophila genome (Ne-

gre et al., 2010). Moreover, other regulatory proteins, such as

Fs(1)h, DREF, Chromator, L3mbt, Z4 (also known as Putzig),

TFIIIC, and condensin subunits (CAP-H2 and Barren), are shown

to be a major constituent of the complex at TAD boundaries (Cu-

benas-Potts et al., 2017; Van Bortle et al., 2014; Kellner et al.,

2013). Indeed, Nhomie and Homie used in this study are highly

enriched for these components (Figure S5). Among these, Fs(1)

h is known as aDrosophila homolog of bromodomain-containing

protein Brd4 (Kellner et al., 2013). Recent studies suggested that

Brd4 undergoes dynamic liquid-liquid phase separation through

the conserved intrinsically disordered region at C-terminal

domain, producing transcriptional condensates or transcription

hubs that colocalize with the Mediator and Pol II clusters (Cho

et al., 2018; Sabari et al., 2018; Shin et al., 2018). In addition,

Brd4 is known to interact with the positive transcription elonga-

tion factor P-TEFb (Bisgrove et al., 2007), a complex of Cdk9

and its regulatory subunit cyclin T1. Intriguingly, P-TEFb is also

suggested to undergo liquid-liquid phase separation though the

conserved histidine-rich domain of cyclin T1 to increase proces-

sivity of Ser2 phosphorylation at Pol II CTD (Guo et al., 2019; Lu

et al., 2018). Thus, it might be possible that boundary-enriched

Fs(1)h acts as a scaffold to increase local concentration of tran-

scription factors, co-activators, elongation factors, and Pol II

complexes (Figure 5B), thereby affecting the timing and the size

of transcriptional bursting independently of TAD formation.

Importantly, other boundary-enriched proteins DREF, Chroma-

tor, and Z4 are also suggested to be involved in recruitment of

Pol II into asubset of genes (Haberle et al., 2019; Zabidi andStark,

2016), giving rise to a possibility that these proteins further

contribute to assembly of transcription hubs at nearby enhancers

(Figure 5B). It appears that this mechanism is particularly

important for distal enhancers to overcome large genomic

separation because extension of enhancer-promoter distance

from 6.5 kb to 9 kb leads to significant (more than 50%) diminish-

ment in the level of total output when boundary elements

232 Molecular Cell 78, 224–235, April 16, 2020

are not present (Figure 1). We speculate that this is a common

regulatory mechanism in the Drosophila genome because

typical enhancer-promoter separation is estimated to be ~10

kb (Kvon et al., 2014). Future functional studies should elucidate

the molecular mechanism underlying ‘‘enhancer-assisting

activity’’ of boundary elements.

STAR+METHODS

Detailed methods are provided in the online version of this paper

and include the following:

d KEY RESOURCES TABLE

d LEAD CONTACT AND MATERIALS AVAILABILITY

d EXPERIMENTAL MODEL AND SUBJECT DETAILS

d METHOD DETAILS

B Site specific transgenesis by phiC31 system

B Genome editing by CRISPR/Cas9

B Fly strain

B Genomic DNA extraction and PCR analysis

B cDNA synthesis

B In situ hybridization

B Quantification of sex comb teeth numbers

B MS2 Live imaging

B Plasmids

B Visualization of genomics data

B Image analysis

B Nuclei segmentation and tracking

B Recording MS2 signal

B Detection of transcriptional bursting

B Description of bursting properties

B Quantification of total active duration

B False-coloring by cumulative RNA production

B False-coloring by instantaneous MS2 signals

d DATA AND CODE AVAILABILITY

SUPPLEMENTAL INFORMATION

Supplemental Information can be found online at https://doi.org/10.1016/j.

molcel.2020.02.007.

ACKNOWLEDGMENTS

We thank Bomyi Lim and Tyler Heist for sharing nuclei segmentation and

tracking code; Hitomi Takishita, Mai Nakabayashi, Yuko Maeyama, and Mis-

ako Sato for fly husbandry; University of Tokyo Olympus Bioimaging Center

(TOBIC) for helping acquisition of preliminary imaging data not used in this

study; the Tabata laboratory for use of P-97 needle puller and help for quanti-

fication of sex comb teeth; and the Bloomington Drosophila Stock Center for

fly strains. We are also grateful to members of the Fukaya laboratory for dis-

cussions. M.Y. is supported by the Grants-in-Aid for Young Scientists (B)

(JP17K17834) from the Japan Society for the Promotion of Science and the

Employment Stability Support for Young Researchers from the University of

Tokyo. This study was funded by the Grants-in-Aid for Research Activity

Start-up (JP18H06040), the Grants-in-Aid for Scientific Research (B)

(JP19H03154), and the Grants-in-Aid for Challenging Research (Exploratory)

(JP19K22378) from the Japan Society for the Promotion of Science; the

Grants-in-Aid for Leading Initiative for Excellent Young Researchers from the

Ministry of Education, Culture, Sports, Science and Technology in Japan;

and research grants from the Molecular Biology Society Japan, the Mochida

Memorial Foundation for Medical and Pharmaceutical Research, the Nakajima

Page 11: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Foundation, the Inamori Foundation, the Takeda Science Foundation, and the

Sumitomo Foundation.

AUTHOR CONTRIBUTIONS

M.Y. and T.F. designed and performed the experiments. K.S. produced SF1

deletion mutant with help of T.F. T.F. performed image analysis and wrote

the manuscript. All the authors discussed the results.

DECLARATION OF INTERESTS

The authors declare no competing interests.

Received: August 23, 2019

Revised: December 19, 2019

Accepted: February 5, 2020

Published: February 27, 2020

SUPPORTING CITATIONS

The following reference appears in the Supplemental Information: Li

et al. (2015a).

REFERENCES

Bartman, C.R., Hsu, S.C., Hsiung, C.C., Raj, A., and Blobel, G.A. (2016).

Enhancer regulation of transcriptional bursting parameters revealed by forced

chromatin looping. Mol. Cell 62, 237–247.

Bartman, C.R., Hamagami, N., Keller, C.A., Giardine, B., Hardison, R.C.,

Blobel, G.A., and Raj, A. (2019). Transcriptional burst initiation and polymerase

pause release are key control points of transcriptional regulation. Mol. Cell 73,

519–532.e4.

Belozerov, V.E., Majumder, P., Shen, P., and Cai, H.N. (2003). A novel bound-

ary element may facilitate independent gene regulation in the Antennapedia

complex of Drosophila. EMBO J. 22, 3113–3121.

Bertrand, E., Chartrand, P., Schaefer, M., Shenoy, S.M., Singer, R.H., and

Long, R.M. (1998). Localization of ASH1 mRNA particles in living yeast. Mol.

Cell 2, 437–445.

Bischof, J., Maeda, R.K., Hediger, M., Karch, F., and Basler, K. (2007). An opti-

mized transgenesis system for Drosophila using germ-line-specific phiC31 in-

tegrases. Proc. Natl. Acad. Sci. USA 104, 3312–3317.

Bisgrove, D.A., Mahmoudi, T., Henklein, P., and Verdin, E. (2007). Conserved

P-TEFb-interacting domain of BRD4 inhibits HIV transcription. Proc. Natl.

Acad. Sci. USA 104, 13690–13695.

Boija, A., Klein, I.A., Sabari, B.R., Dall’Agnese, A., Coffey, E.L., Zamudio, A.V.,

Li, C.H., Shrinivas, K., Manteiga, J.C., Hannett, N.M., et al. (2018).

Transcription factors activate genes through the phase-separation capacity

of their activation domains. Cell 175, 1842–1855.e16.

Bothma, J.P., Garcia, H.G., Esposito, E., Schlissel, G., Gregor, T., and Levine,

M. (2014). Dynamic regulation of eve stripe 2 expression reveals transcriptional

bursts in living Drosophila embryos. Proc. Natl. Acad. Sci. USA 111,

10598–10603.

Calhoun, V.C., and Levine, M. (2003). Long-range enhancer-promoter interac-

tions in the Scr-Antp interval of the Drosophila Antennapedia complex. Proc.

Natl. Acad. Sci. USA 100, 9878–9883.

Calhoun, V.C., Stathopoulos, A., and Levine, M. (2002). Promoter-proximal

tethering elements regulate enhancer-promoter specificity in the Drosophila

Antennapedia complex. Proc. Natl. Acad. Sci. USA 99, 9243–9247.

Chan, Y.F., Marks, M.E., Jones, F.C., Villarreal, G., Jr., Shapiro, M.D., Brady,

S.D., Southwick, A.M., Absher, D.M., Grimwood, J., Schmutz, J., et al.

(2010). Adaptive evolution of pelvic reduction in sticklebacks by recurrent

deletion of a Pitx1 enhancer. Science 327, 302–305.

Cho, W.K., Jayanth, N., English, B.P., Inoue, T., Andrews, J.O., Conway, W.,

Grimm, J.B., Spille, J.H., Lavis, L.D., Lionnet, T., and Cisse, I.I. (2016). RNA po-

lymerase II cluster dynamics predict mRNA output in living cells. eLife 5,

e13617.

Cho, W.K., Spille, J.H., Hecht, M., Lee, C., Li, C., Grube, V., and Cisse, I.I.

(2018). Mediator and RNA polymerase II clusters associate in transcription-

dependent condensates. Science 361, 412–415.

Chong, S., Dugast-Darzacq, C., Liu, Z., Dong, P., Dailey, G.M., Cattoglio, C.,

Heckert, A., Banala, S., Lavis, L., Darzacq, X., and Tjian, R. (2018). Imaging dy-

namic and selective low-complexity domain interactions that control gene

transcription. Science 361, eaar2555.

Crane, E., Bian, Q., McCord, R.P., Lajoie, B.R., Wheeler, B.S., Ralston, E.J.,

Uzawa, S., Dekker, J., and Meyer, B.J. (2015). Condensin-driven remodelling

of X chromosome topology during dosage compensation. Nature 523,

240–244.

Cubenas-Potts, C., Rowley, M.J., Lyu, X., Li, G., Lei, E.P., and Corces, V.G.

(2017). Different enhancer classes inDrosophila bind distinct architectural pro-

teins and mediate unique chromatin interactions and 3D architecture. Nucleic

Acids Res. 45, 1714–1730.

Despang, A., Schopflin, R., Franke, M., Ali, S., Jerkovi�c, I., Paliou, C., Chan,

W.L., Timmermann, B., Wittler, L., Vingron, M., et al. (2019). Functional dissec-

tion of the Sox9-Kcnj2 locus identifies nonessential and instructive roles of

TAD architecture. Nat. Genet. 51, 1263–1271.

Dixon, J.R., Selvaraj, S., Yue, F., Kim, A., Li, Y., Shen, Y., Hu, M., Liu, J.S., and

Ren, B. (2012). Topological domains in mammalian genomes identified by

analysis of chromatin interactions. Nature 485, 376–380.

Dunipace, L., Ozdemir, A., and Stathopoulos, A. (2011). Complex interactions

between cis-regulatory modules in native conformation are critical for

Drosophila snail expression. Development 138, 4075–4084.

Durand, N.C., Robinson, J.T., Shamim,M.S., Machol, I., Mesirov, J.P., Lander,

E.S., and Aiden, E.L. (2016). Juicebox provides a visualization system for Hi-C

contact maps with unlimited zoom. Cell Syst. 3, 99–101.

Eksi, S.E., Barmina, O., McCallough, C.L., Kopp, A., and Orenic, T.V. (2018). A

Distalless-responsive enhancer of the Hox gene Sex combs reduced is

required for segment- and sex-specific sensory organ development in

Drosophila. PLoS Genet. 14, e1007320.

ENCODE Project Consortium (2012). An integrated encyclopedia of DNA ele-

ments in the human genome. Nature 489, 57–74.

Feng, S., Cokus, S.J., Schubert, V., Zhai, J., Pellegrini, M., and Jacobsen,

S.E. (2014). Genome-wide Hi-C analyses in wild-type and mutants

reveal high-resolution chromatin interactions in Arabidopsis. Mol. Cell 55,

694–707.

Fudenberg, G., Imakaev, M., Lu, C., Goloborodko, A., Abdennur, N., and

Mirny, L.A. (2016). Formation of chromosomal domains by loop extrusion.

Cell Rep. 15, 2038–2049.

Fujioka, M., Wu, X., and Jaynes, J.B. (2009). A chromatin insulator mediates

transgene homing and very long-range enhancer-promoter communication.

Development 136, 3077–3087.

Fujioka, M., Sun, G., and Jaynes, J.B. (2013). The Drosophila eve insulator

Homie promotes eve expression and protects the adjacent gene from repres-

sion by polycomb spreading. PLoS Genet. 9, e1003883.

Fujioka, M., Mistry, H., Schedl, P., and Jaynes, J.B. (2016). Determinants of

chromosome architecture: insulator pairing in cis and in trans. PLoS Genet.

12, e1005889.

Fukaya, T., Lim, B., and Levine, M. (2016). Enhancer control of transcriptional

bursting. Cell 166, 358–368.

Gambetta, M.C., and Furlong, E.E.M. (2018). The insulator protein CTCF is

required for correct Hox gene expression, but not for embryonic development

in Drosophila. Genetics 210, 129–136.

Garcia, H.G., Tikhonov, M., Lin, A., and Gregor, T. (2013). Quantitative imaging

of transcription in living Drosophila embryos links polymerase activity to

patterning. Curr. Biol. 23, 2140–2145.

Ghavi-Helm, Y., Jankowski, A., Meiers, S., Viales, R.R., Korbel, J.O., and

Furlong, E.E.M. (2019). Highly rearranged chromosomes reveal uncoupling

Molecular Cell 78, 224–235, April 16, 2020 233

Page 12: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

between genome topology and gene expression. Nat. Genet. 51,

1272–1282.

Gindhart, J.G., Jr., King, A.N., and Kaufman, T.C. (1995). Characterization of

the cis-regulatory region of theDrosophila homeotic geneSex combs reduced.

Genetics 139, 781–795.

Gray, S., and Levine, M. (1996). Short-range transcriptional repressors

mediate both quenching and direct repression within complex loci in

Drosophila. Genes Dev. 10, 700–710.

Grob, S., Schmid, M.W., and Grossniklaus, U. (2014). Hi-C analysis in

Arabidopsis identifies the KNOT, a structure with similarities to the flamenco

locus of Drosophila. Mol. Cell 55, 678–693.

Guenther, C.A., Tasic, B., Luo, L., Bedell, M.A., and Kingsley, D.M. (2014). A

molecular basis for classic blond hair color in Europeans. Nat. Genet. 46,

748–752.

Guo, Y.E., Manteiga, J.C., Henninger, J.E., Sabari, B.R., Dall’Agnese, A.,

Hannett, N.M., Spille, J.H., Afeyan, L.K., Zamudio, A.V., Shrinivas, K., et al.

(2019). Pol II phosphorylation regulates a switch between transcriptional and

splicing condensates. Nature 572, 543–548.

Haberle, V., Arnold, C.D., Pagani, M., Rath, M., Schernhuber, K., and Stark, A.

(2019). Transcriptional cofactors display specificity for distinct types of core

promoters. Nature 570, 122–126.

Kellner, W.A., Van Bortle, K., Li, L., Ramos, E., Takenaka, N., and Corces, V.G.

(2013). Distinct isoforms of the Drosophila Brd4 homologue are present at en-

hancers, promoters and insulator sites. Nucleic Acids Res. 41, 9274–9283.

Kvon, E.Z., Kazmar, T., Stampfel, G., Yanez-Cuna, J.O., Pagani, M.,

Schernhuber, K., Dickson, B.J., and Stark, A. (2014). Genome-scale functional

characterization of Drosophila developmental enhancers in vivo. Nature

512, 91–95.

Larsson, A.J.M., Johnsson, P., Hagemann-Jensen, M., Hartmanis, L.,

Faridani, O.R., Reinius, B., Segerstolpe, A., Rivera, C.M., Ren, B., and

Sandberg, R. (2019). Genomic encoding of transcriptional burst kinetics.

Nature 565, 251–254.

Li, L., Lyu, X., Hou, C., Takenaka, N., Nguyen, H.Q., Ong, C.T., Cubenas-Potts,

C., Hu, M., Lei, E.P., Bosco, G., et al. (2015a). Widespread rearrangement of

3D chromatin organization underlies polycomb-mediated stress-induced

silencing. Mol. Cell 58, 216–231.

Li, M., Ma, Z., Liu, J.K., Roy, S., Patel, S.K., Lane, D.C., and Cai, H.N.N.

(2015b). An organizational hub of developmentally regulated chromatin loops

in the Drosophila Antennapedia complex. Mol. Cell. Biol. 35, 4018–4029.

Lim, B., Fukaya, T., Heist, T., and Levine, M. (2018a). Temporal dynamics of

pair-rule stripes in living Drosophila embryos. Proc. Natl. Acad. Sci. USA

115, 8376–8381.

Lim, B., Heist, T., Levine, M., and Fukaya, T. (2018b). Visualization of transvec-

tion in living Drosophila embryos. Mol. Cell 70, 287–296.e6.

Long, H.K., Prescott, S.L., andWysocka, J. (2016). Ever-changing landscapes:

transcriptional enhancers in development and evolution. Cell 167, 1170–1187.

Lu, H., Yu, D., Hansen, A.S., Ganguly, S., Liu, R., Heckert, A., Darzacq, X., and

Zhou, Q. (2018). Phase-separation mechanism for C-terminal hyperphosphor-

ylation of RNA polymerase II. Nature 558, 318–323.

Lucas, T., Ferraro, T., Roelens, B., De Las Heras Chanes, J., Walczak, A.M.,

Coppey, M., and Dostatni, N. (2013). Live imaging of bicoid-dependent tran-

scription in Drosophila embryos. Curr. Biol. 23, 2135–2139.

Natsume, T., Kiyomitsu, T., Saga, Y., and Kanemaki, M.T. (2016). Rapid protein

depletion in human cells by auxin-inducible degron tagging with short homol-

ogy donors. Cell Rep. 15, 210–218.

Negre, N., Brown, C.D., Shah, P.K., Kheradpour, P., Morrison, C.A., Henikoff,

J.G., Feng, X., Ahmad, K., Russell, S., White, R.A.H., et al. (2010). A compre-

hensive map of insulator elements for the Drosophila genome. PLoS Genet. 6,

e1000814.

Nora, E.P., Lajoie, B.R., Schulz, E.G., Giorgetti, L., Okamoto, I., Servant, N.,

Piolot, T., van Berkum, N.L., Meisig, J., Sedat, J., et al. (2012). Spatial partition-

ing of the regulatory landscape of the X-inactivation centre. Nature 485,

381–385.

234 Molecular Cell 78, 224–235, April 16, 2020

Nora, E.P., Goloborodko, A., Valton, A.L., Gibcus, J.H., Uebersohn, A.,

Abdennur, N., Dekker, J., Mirny, L.A., and Bruneau, B.G. (2017). Targeted

degradation of CTCF decouples local insulation of chromosome domains

from genomic compartmentalization. Cell 169, 930–944.e22.

Ohtsuki, S., Levine, M., and Cai, H.N. (1998). Different core promoters possess

distinct regulatory activities in the Drosophila embryo. Genes Dev. 12,

547–556.

Ong, C.T., and Corces, V.G. (2014). CTCF: an architectural protein bridging

genome topology and function. Nat. Rev. Genet. 15, 234–246.

Perry, M.W., Boettiger, A.N., Bothma, J.P., and Levine, M. (2010). Shadow en-

hancers foster robustness of Drosophila gastrulation. Curr. Biol. 20,

1562–1567.

Pfeiffer, B.D., Jenett, A., Hammonds, A.S., Ngo, T.T., Misra, S., Murphy, C.,

Scully, A., Carlson, J.W.,Wan, K.H., Laverty, T.R., et al. (2008). Tools for neuro-

anatomy and neurogenetics in Drosophila. Proc. Natl. Acad. Sci. USA 105,

9715–9720.

Rao, S.S.P., Huntley, M.H., Durand, N.C., Stamenova, E.K., Bochkov, I.D.,

Robinson, J.T., Sanborn, A.L., Machol, I., Omer, A.D., Lander, E.S., and

Aiden, E.L. (2014). A 3D map of the human genome at kilobase resolution re-

veals principles of chromatin looping. Cell 159, 1665–1680.

Rao, S.S.P., Huang, S.C., Glenn St Hilaire, B., Engreitz, J.M., Perez, E.M.,

Kieffer-Kwon, K.R., Sanborn, A.L., Johnstone, S.E., Bascom, G.D.,

Bochkov, I.D., et al. (2017). Cohesin loss eliminates all loop domains. Cell

171, 305–320.e24.

Ren, X., Sun, J., Housden, B.E., Hu, Y., Roesel, C., Lin, S., Liu, L.P., Yang, Z.,

Mao, D., Sun, L., et al. (2013). Optimized gene editing technology for

Drosophila melanogaster using germ line-specific Cas9. Proc. Natl. Acad.

Sci. USA 110, 19012–19017.

Ringrose, L. (2009). Transgenesis in Drosophila melanogaster. Methods Mol.

Biol. 561, 3–19.

Roy, S., Ernst, J., Kharchenko, P.V., Kheradpour, P., Negre, N., Eaton, M.L.,

Landolin, J.M., Bristow, C.A., Ma, L., Lin, M.F., et al.; modENCODE

Consortium (2010). Identification of functional elements and regulatory circuits

by Drosophila modENCODE. Science 330, 1787–1797.

Sabari, B.R., Dall’Agnese, A., Boija, A., Klein, I.A., Coffey, E.L., Shrinivas, K.,

Abraham, B.J., Hannett, N.M., Zamudio, A.V., Manteiga, J.C., et al. (2018).

Coactivator condensation at super-enhancers links phase separation and

gene control. Science 361, eaar3958.

Sanborn, A.L., Rao, S.S., Huang, S.C., Durand, N.C., Huntley, M.H., Jewett,

A.I., Bochkov, I.D., Chinnappan, D., Cutkosky, A., Li, J., et al. (2015).

Chromatin extrusion explains key features of loop and domain formation in

wild-type and engineered genomes. Proc. Natl. Acad. Sci. USA 112,

E6456–E6465.

Schindelin, J., Arganda-Carreras, I., and Frise, E. (2012). Fiji: an open-source

platform for biological-image analysis. Nat. Methods 9, 676–682.

Schroeder, M.D., Greer, C., and Gaul, U. (2011). How to make stripes: deci-

phering the transition from non-periodic to periodic patterns inDrosophila seg-

mentation. Development 138, 3067–3078.

Schwarzer, W., Abdennur, N., Goloborodko, A., Pekowska, A., Fudenberg, G.,

Loe-Mie, Y., Fonseca, N.A., Huber, W., Haering, C.H., Mirny, L., and Spitz, F.

(2017). Two independent modes of chromatin organization revealed by cohe-

sin removal. Nature 551, 51–56.

Sexton, T., Yaffe, E., Kenigsberg, E., Bantignies, F., Leblanc, B., Hoichman,

M., Parrinello, H., Tanay, A., and Cavalli, G. (2012). Three-dimensional folding

and functional organization principles of the Drosophila genome. Cell 148,

458–472.

Shin, Y., Chang, Y.C., Lee, D.S.W., Berry, J., Sanders, D.W., Ronceray, P.,

Wingreen, N.S., Haataja,M., and Brangwynne, C.P. (2018). Liquid nuclear con-

densates mechanically sense and restructure the genome. Cell 175, 1481–

1491.e13.

Stadler, M.R., Haines, J.E., and Eisen, M.B. (2017). Convergence of topologi-

cal domain boundaries, insulators, and polytene interbands revealed by high-

Page 13: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

resolution mapping of chromatin contacts in the early Drosophila mela-

nogaster embryo. eLife 6, e29550.

Sun, Y., Nien, C.Y., Chen, K., Liu, H.Y., Johnston, J., Zeitlinger, J., and

Rushlow, C. (2015). Zelda overcomes the high intrinsic nucleosome barrier

at enhancers during Drosophila zygotic genome activation. Genome Res.

25, 1703–1714.

Ulianov, S.V., Khrameeva, E.E., Gavrilov, A.A., Flyamer, I.M., Kos, P.,

Mikhaleva, E.A., Penin, A.A., Logacheva, M.D., Imakaev, M.V., Chertovich,

A., et al. (2016). Active chromatin and transcription play a key role in chromo-

some partitioning into topologically associating domains. Genome Res.

26, 70–84.

Van Bortle, K., Nichols, M.H., Li, L., Ong, C.T., Takenaka, N., Qin, Z.S.,

and Corces, V.G. (2014). Insulator function and topological domain

border strength scale with architectural protein occupancy. Genome

Biol. 15, R82.

Venken, K.J.T., He, Y., Hoskins, R.A., and Bellen, H.J. (2006). P[acman]: a BAC

transgenic platform for targeted insertion of large DNA fragments in

D. melanogaster. Science 314, 1747–1751.

Zabidi, M.A., and Stark, A. (2016). Regulatory enhancer-core-promoter

communication via transcription factors and cofactors. Trends Genet. 32,

801–814.

Molecular Cell 78, 224–235, April 16, 2020 235

Page 14: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

STAR+METHODS

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER

Antibodies

Sheep Anti-Digoxigenin-AP Fab fragments Roche Cat# 11093274910; RRID: AB_514497

Mouse Anti-Biotin Invitrogen Cat# 03-3700; RRID: AB_2532265

Alexa Fluor 555 donkey anti-sheep IgG Invitrogen Cat# A21436; RRID: AB_2535857

Alexa Fluor 488 donkey anti-mouse IgG Invitrogen Cat# A21202; RRID: AB_141607

Bacterial and Virus Strains

DH5a Competent Cells This study N/A

Chemicals, Peptides, and Recombinant Proteins

DIG RNA Labeling MIx, 10x conc. Roche Cat# 11277073910

Biotin RNA Labeling MIx, 10x conc. Roche Cat# 11685597910

ProLong Gold antifade reagent Thermo Fisher Cat# P36930

Western Blocking Buffer Roche Cat# 11921681001

Modified Silicone Fluids Shin-Etsu Silicone FL-100-1000CS

Modified Silicone Fluids Shin-Etsu Silicone FL-100-450CS

polylap (polyethylene membrane) Ube Film N/A

KOD One PCR Master Mix Takara Cat# KMM-101

TRIzol Reagent Invitrogen Cat# 15596026

PrimeScript 1st strand cDNA Synthesis Kit Takara Cat# 6110A

in vitro Transcription T7 Kit Takara Cat# 6140

Experimental Models: Organisms/Strains

D. melanogaster nos > MCP-GFP, His2Av-mRFP/CyO This study N/A

D. melanogaster DSCP-MS2-yellow-sna shadow enhancer This study N/A

D. melanogaster DSCP-MS2-yellow-linker-sna shadow enhancer This study N/A

D. melanogaster DSCP-MS2-yellow-long linker-sna shadow

enhancer

This study N/A

D. melanogaster Nhomie-DSCP-MS2-yellow-linker-sna shadow

enhancer-Homie

This study N/A

D. melanogaster Nhomie-DSCP-MS2-yellow-linker-sna shadow

enhancer-HomieinvThis study N/A

D. melanogaster Nhomie-DSCP-MS2-yellow-linker-sna shadow

enhancer

This study N/A

D. melanogaster Nhomie-DSCP-MS2-yellow-sna shadow

enhancer-Homie

This study N/A

D. melanogaster ftz-MS2 Lim et al., 2018b N/A

D. melanogaster ⊿SF1 ftz-MS2 This study N/A

D. melanogaster SF1-DSCP-MS2-yellow-linker-sna shadow

enhancer-SF2

This study N/A

D. melanogaster y[1] M{vas-int.Dm}ZH-2A w[*]; PBac{y[+]-attP-

3B}VK00033

Venken et al., 2006 BDSC

Bischof et al., 2007 # 24871

D. melanogaster y[1] M{vas-int.Dm}ZH-2A w[*]; PBac{y[+]-attP-

3B}VK00001

Venken et al., 2006 BDSC

Bischof et al., 2007 # 24861

Oligonucleotides

Primers are listed in STAR Methods. This study N/A

Recombinant DNA

Plasmids are listed in STAR Methods. N/A N/A

(Continued on next page)

e1 Molecular Cell 78, 224–235.e1–e5, April 16, 2020

Page 15: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Continued

REAGENT or RESOURCE SOURCE IDENTIFIER

Software and Algorithms

MATLAB 2019a MathWorks https://www.mathworks.com

Fiji Schindelin et al., 2012 https://fiji.sc

IGV Broad Institute https://software.broadinstitute.org/

software/igv/

Juicebox Durand et al., 2016 https://github.com/aidenlab/Juicebox/

wiki/Download

Deposited Data

Original images of fluorescent in situ hybridization This study https://dx.doi.org/10.17632/yypbkpxkx3.1

LEAD CONTACT AND MATERIALS AVAILABILITY

Further information and requests for resources and reagents should be directed to and will be fulfilled by the Lead Contact, Takashi

Fukaya ([email protected]).

EXPERIMENTAL MODEL AND SUBJECT DETAILS

In all imaging experiments, we studied Drosophila melanogaster embryos at nuclear cycle 14 unless otherwise noted. The following

fly lines were used in this study: nos > MCP-GFP, His2Av-mRFP/CyO (this study), DSCP-MS2-yellow-sna shadow enhancer (this

study),DSCP-MS2-yellow-linker-sna shadow enhancer (this study),DSCP-MS2-yellow-long linker-sna shadow enhancer (this study),

Nhomie-DSCP-MS2-yellow-linker-sna shadow enhancer-Homie (this study), Nhomie-DSCP-MS2-yellow-linker-sna shadow

enhancer-Homieinv (this study), Nhomie-DSCP-MS2-yellow-linker-sna shadow enhancer (this study), Nhomie-DSCP-MS2-yellow-

sna shadow enhancer-Homie (this study), ftz-MS2 (Lim et al., 2018b), ⊿SF1 ftz-MS2 (this study), SF1-DSCP-MS2-yellow-linker-

sna shadow enhancer-SF2 (this study), y[1] M{vas-int.Dm}ZH-2A w[*]; PBac{y[+]-attP-3B}VK00033 (Bloomington Drosophila Stock

Center #24871), y[1] M{vas-int.Dm}ZH-2A w[*]; PBac{y[+]-attP-3B}VK00001 (Bloomington Drosophila Stock Center #24861).

METHOD DETAILS

Site specific transgenesis by phiC31 systemAll reporter plasmids were integrated into a unique landing site on the third chromosome using VK00033 strain (Venken et al., 2006).

PhiC31wasmaternally provided using vas-phiC31 strain (Bischof et al., 2007). Microinjection was performed as previously described

(Ringrose, 2009). In brief, 0-1 hour embryoswere collected and dechorionatedwith bleach. Aligned embryoswere dried with silica gel

for ~7 min and covered with FL-100-1000CS silicone oil (Shin-Etsu Silicone). Subsequently, microinjection was performed using

FemtoJet 5247 (Eppendorf) and DM IL LED invertedmicroscope (Leica) equipped withM-152Micromanipulator (Narishige). Injection

needle was prepared with P-97 or P-1000 needle puller (Shutter). Injection mixture contains ~500 ng/ml plasmid DNA, 5 mM KCl,

0.1 mM phosphate buffer, pH 6.8. mini-White marker was used for subsequent screening.

Genome editing by CRISPR/Cas9pCFD3 gRNA expression plasmids, pBS-3xP3-GFP donor plasmid and pBS-hsp70-Cas9 plasmid (addgene #46294) were co-in-

jected to homozygote ftz-MS2 embryos (Lim et al., 2018a; Lim et al., 2018b). Microinjection was performed as described in previous

section. Injection mixture contains 450 ng/ml pCFD3 gRNA expression plasmids, 450 ng/ml pBS-3xP3-GFP donor plasmid, 450 ng/ml

pBS-hsp70-Cas9 plasmid, 5 mM KCl, 0.1 mM phosphate buffer, pH 6.8. 3xP3-GFP marker was used for subsequent screening.

Fly strainThe nanos >MCP-GFP, His2Av-mRFP expression plasmid (Lim et al., 2018b) was integrated into a unique landing site on the second

chromosome using VK00001 strain (Venken et al., 2006) to obtain maternal expression of the MCP-GFP and His2Av-mRFP fusion

proteins. The resulting transgenic allele was balanced over CyO.

Genomic DNA extraction and PCR analysisGenomic DNAwas extracted from 20 adults of WT ftz-MS2 and ⊿SF1 ftz-MS2 homozygotes. Flies were homogenized with 250 mL of

DNA extraction buffer (0.1 M Tris-HCl pH 9.0, 0.1 M EDTA, 1% SDS), and then incubated at 70 �C for 30 min. Subsequently, 35 mL of

5 M KOAc was added and incubated for 30 min on ice. After centrifugation at 20,000 g for 30 min at 4 �C, supernatant was collected.

Genomic DNA was further purified by phenol-chloroform extraction and recovered by isopropanol precipitation. PCR analysis of

Molecular Cell 78, 224–235.e1–e5, April 16, 2020 e2

Page 16: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

purified DNA was done by using following primers: primer A (50-TAC AAC AGC CAC AAC GTC TAT ATC-30), primer B (50-ACT TAG

CTT CCC ACA AAC CAT AAC C-30), primer C (50-AAG TGG GAG AAT GAG ACT CG-30), primer D (50-TAG CGG CTG AAG CAC TGC

AC-30), primer E (50-ACGGAA TCC TTC TAT TGCAGT TCCG-30) and primer F (50-GCA TGT ACTCAA ATA AGGCAAG-30). KODOne

PCR Master Mix (Takara) was used for the reaction. All PCR products were then verified by sanger sequencing.

cDNA synthesisTotal RNA was extracted from 40 adults of Oregon-R using TRIzol reagent (Thermo Fisher) followed by chloroform purification and

isopropanol precipitation. Three mg of total RNAwas subjected to reverse transcription using PrimeScript 1st strand cDNA Synthesis

Kit (Takara).

In situ hybridizationEmbryos were dechorionated and fixed in fixation buffer (0.5x PBS, 4% formaldehyde and 50% Heptane) for ~25 min at room tem-

perature. Antisense RNA probes labeled with digoxigenin (DIG RNA Labeling Mix 103 conc, Roche) and biotin (Biotin RNA Labeling

Mix 10 3 conc, Roche) were used. Template DNA for ftz probe was PCR amplified from genomic DNA using primers (50-CGT AAT

ACG ACT CAC TAT AGG GTG GGG AAG AGA GTA ACT GAG CAT CGC-30) and (50-ATT CGC AAA CTC ACC AGC GT-30). Template

DNA for Scr probe was PCR amplified from cDNA using primers (50-CAG TCAAAG TGAGACATTGGCGC-30) and (50-CGT AAT ACG

ACT CAC TAT AGG GCC GAC CAA TTG CAT TGA ACA CAC-30). Hybridization was performed at 55 �C overnight in hybridization

buffer (50% formamide, 5x SSC, 50 mg/ml Heparin, 100 mg/ml salmon sperm DNA, 0.1% Tween-20). Subsequently, embryos

were washed with hybridization buffer at 55 �C and incubated with Western Blocking Buffer (Roche) at room temperature for one

hour. Then, embryos were incubated with sheep anti-digoxigenin (Roche) and mouse anti-biotin primary antibodies (Invitrogen) at

4 �C for overnight, followed by incubation with Alexa Fluor 488 donkey anti-sheep (Invitrogen) and Alexa Flour 555 goat anti-mouse

(Invitrogen) fluorescent secondary antibodies at room temperature for one hour. DNA was stained with DAPI, and embryos were

mounted in ProLong Gold Antifade Mountant (Thermo Fisher Scientific). Imaging was performed on a Zeiss LSM 900 confocal mi-

croscope. Plan-Apochromat 20x / 0.8 N.A. objective was used. Images were captured in 16-bit. Maximum projections were obtained

for all z sections, and resulting images were shown. Brightness of images was linearly adjusted using Fiji (https://fiji.sc).

Quantification of sex comb teeth numbersAdult males of WT ftz-MS2 and ⊿SF1 ftz-MS2 homozygotes were used for counting numbers of sex comb teeth. Forelegs were

viewed with a Leica MZ16F stereomicroscope and images were acquired with a LUMIX DMC-GH3 camera (Panasonic). Sex

comb teeth were then manually quantified.

MS2 Live imagingMCP-GFP, His2Av-mRFP/CyO virgin females were mated with homozygous males carrying the MS2 reporter gene. The resulting em-

bryoswere dechorinated andmounted between a polyethylenemembrane (Ube Film) and a coverslip (18mmx18mm), and embedded

in FL-100-450CS (Shin-Etsu Silicone). Embryos were imaged using a Zeiss LSM 800 (Figures 1, 2, and S1B–S1D) or Zeiss LSM

900 (Figures 3, 4, S1A, S2, S3, and S4). Temperature was kept in between 24.0 to 25.0 �C during imaging. Plan-Apochromat 40x /

1.4 N.A. oil immersion objective was used. At each time point, a stack of 26 images separated by 0.5 mm was acquired. Typical

time resolution of resulting maximum projection was 16.5 s. In subsequent image analysis, all movies were considered to have

same time resolution (16.5 s/frame). Images were captured in 16-bit. Images have been typically taken from the end of nc 13 to the

onset of gastrulation at nc 14. During imaging, data acquisition was occasionally stopped for seconds to correct z-position, and

data were concatenated afterward. For each cross shown inmain figures, three biological replicateswere taken. The same laser power

and microscope setting were used for a set of experiments.

Plasmidspbphi-DSCP-MS2-yellow

Two DNA oligos (50-TTT CCC TCG AGG AGC TCG CCC GGG GAT CGA GCG CAG CGG TAT AAA AGG GCG CGG GGT GGC TGA

GAG CAT CAG TTG TGA ATG AAT GTT CGA GCC GAG C-30) and (50-GGA AAG GAT CCG TTT GGT ATG CGT CTT GTG ATT CAA

AGT TGG CTT ATT CAA AGG ATA TTA ACG AAG GCA GCG GCA CGT CTG CTC GGC TCG AAC AT-30) were annealed and blunt-

ended by PCR. Resulting DNA fragment was inserted between XhoI and BamHI sites in pbphi-snaPr-MS2-yellow plasmid devoid of

intronic modification (Fukaya et al., 2016).

pbphi-DSCP-MS2-yellow-sna shadow enhancer

A DNA fragment containing snail shadow enhancer was purified from pbphi-snail shadow enhancer (Lim et al., 2018b) by digesting

with HindIII and NheI. The resulting fragment was inserted between HindIII and NheI sites in pbphi-DSCP-MS2-yellow.

pbphi-DSCP-MS2-yellow-linker-sna shadow enhancer

A DNA fragment containing partial sequence of LacZ was amplified using primers (50-TTT CCA AGC TTC GGT TAC GAT GCG CCC

ATC T-30) and (50-GGA AAA AGC TTC AAT GGC AGA TCC CAG CGG T-30) and digested with HindIII. The resultant fragment was

inserted into the unique HindIII site in the pbphi-DSCP-MS2-yellow-sna shadow enhancer as a linker DNA.

e3 Molecular Cell 78, 224–235.e1–e5, April 16, 2020

Page 17: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

pbphi-DSCP-MS2-yellow-long linker-sna shadow enhancer

A DNA fragment containing partial sequence of LacZ was amplified using primers (50-AAA TTG ACG TCT CGT TGC TGC ATA AAC

CGA C-30) and (50-AAT TTG ACG TCT CGC TCG CCA CTT CAA CAT C-30) and digested with AatII. The resultant fragment was in-

serted into the unique AatII site in the pbphi-DSCP-MS2-yellow-linker sna shadow enhancer to extend the linker length.

pbphi-Nhomie-DSCP-MS2-yellow-linker-sna shadow enhancer

ADNA fragment containing Nhomie was amplified from genomic DNA using primers (50-TTT AAGCGGCCGCGA TAT CACCTCGTT

GCGGTT CC-30) and (50-TTA AAG CGGCCGCGC TAGCTT GTGGGA TGGCCAGGGGG-30) and digested with NotI. The resultant

fragment was inserted into the unique NotI site in the pbphi-DSCP-MS2-yellow-linker-sna shadow enhancer. Orientation of Nhomie

insertion was confirmed by sequencing.

pbphi-Nhomie-DSCP-MS2-yellow-linker-sna shadow enhancer-Homie

A DNA fragment containing Homie was amplified from genomic DNA using primers (50-TTT AAT CTA GAA ATA CTA AAA AGT TTT

TAC G-30) and (50-TTA AAT CTA GAG ATT ACA CGC TGC GAT GGT TTG-30) and digested with XbaI. The resultant fragment was

inserted into the unique XbaI site in the pbphi-Nhomie-DSCP-MS2-yellow-linker-sna shadow enhancer. Orientation of Homie inser-

tion was confirmed by sequencing.

pbphi-Nhomie-DSCP-MS2-yellow-linker-sna shadow enhancer-Homieinv

A DNA fragment containing Homie was amplified from genomic DNA using primers (50-TTT AAT CTA GAA ATA CTA AAA AGT TTT

TAC G-30) and (50-TTA AAT CTA GAG ATT ACA CGC TGC GAT GGT TTG-30) and digested with XbaI. The resultant fragment was

inserted into the unique XbaI site in the pbphi-Nhomie-DSCP-MS2-yellow-linker-sna shadow enhancer. Orientation of Homie inser-

tion was confirmed by sequencing.

pbphi-Nhomie-DSCP-MS2-yellow-sna shadow enhancer-Homie

The pbphi-Nhomie-DSCP-MS2-yellow-linker-sna shadow enhancer-Homie was digestedwith HindIII to remove linker DNA, followed

by self-ligation of resulting linearized plasmid.

pbphi-SF1-DSCP-MS2-yellow-linker-sna shadow enhancer

A DNA fragment containing SF1 was amplified from genomic DNA using primers (50-ACA TGG CGG CCG CGA ATT CGG TTT TCG

AAG CCG GAG C-30) and (50-AAT TTG CGG CCG CGG ATT CCC CAT CCT ATA CCC TTC-30) and digested with NotI. The resultant

fragment was inserted into the unique NotI site in the pbphi-DSCP-MS2-yellow-linker-sna shadow enhancer. Orientation of SF1

insertion was confirmed by sequencing.

pbphi-SF1-DSCP-MS2-yellow-linker-sna shadow enhancer-SF2

A DNA fragment containing SF2 was amplified from genomic DNA using primers (50-ACA TGT CTA GAC CAT CCT CTT GTG AGG

CTG GG-30) and (50-AAT TTT CTA GAC TGA TTG ACG AAT TGC GTG CG-30) and digested with XbaI. The resultant fragment was

inserted into the unique XbaI site in the pbphi-SF1-DSCP-MS2-yellow-linker-sna shadow enhancer. Orientation of SF2 insertion

was confirmed by sequencing.

pCFD3-dU6-SF1 gRNA-1

Two DNA oligos (50-GTC GCA ATA GAA GGA TTC CGT TGG-30) and (50-AAA CCC AAC GGA ATC CTT CTA TTG-30) were annealed

and inserted into the pCFD3-dU6:3gRNA vector (addgene # 49410) using BbsI sites.

pCFD3-dU6-SF1 gRNA-2

Two DNA oligos (50-GTCGGAGGGCAA AAT CTG TGAGG-30) and (50-AAA CCC TCA CAG ATT TTG CCC TC-30) were annealed and

inserted into the pCFD3-dU6:3gRNA vector (addgene # 49410) using BbsI sites.

pBS-50SF1-loxP-3xP3-GFP-loxP-30SF1ADNA fragment containing 50SF1was amplified from genomic DNA using primers (50-GGGGGCTCGAGAAGGAGCAGTAAGGCA

CGA G-30) and (50-AAA AAG ATA TCT GGC GGC GCG CCG CAG GAC G-30) and digested with XhoI and EcoRV. The resultant frag-

ment was inserted between XhoI and EcoRV sites in pBS-loxP-3xP3-GFP-loxP (Lim et al., 2018a). Subsequently, a DNA fragment

containing 30SF1 was amplified from genomic DNA using primers (50-GGG GGA CTA GTA GGC GGG GAT GTT GGC GCC T-30)and (50-AAA AAG CGG CCG CCG GTG ATG GGT GAA ACG AAG-30) and digested with SpeI and NotI. The resultant fragment

was inserted between SpeI and NotI sites in pBS-50SF1-loxP-3xP3-GFP-loxP.

Visualization of genomics dataFor visualization of Hi-C data, Juicebox (version 1.11.08; Durand et al., 2016) was used (https://github.com/aidenlab/Juicebox/wiki/

Download). Hi-C data pre-implemented in the software (Kc167 DpnII HinfI; Cubenas-Potts et al., 2017) was shown. For visualization

of chromatin immunoprecipitation profile, previously reported ChIP-seq data was downloaded from the Gene Expression Omnibus

(GEO) database. Processed files were obtained from the following sources: Fs(1)h-L (GEO: GSE42086), DREF (GEO: GSE63518),

L3mbt (GEO: GSE63518), Z4 (GEO: GSE63518), Chromator (GEO: GSE54529), TFIIIC (GEO: GSE54529), Rad21 (GEO:

GSE54529), CAP-H2 (GEO: GSE54529), Barren (GEO: GSE54529), and Pol II (GEO: GSE65441). ChIP-chip data (CTCF, Su(Hw),

Mod(mdg4), CP190, BEAF-32) was obtained from the modENCODE database (http://data.modencode.org). All data was visualized

using IGV (version 2.4.19).

Image analysisAll the image processing methods and analysis were implemented in MATLAB (R2019a, MathWorks).

Molecular Cell 78, 224–235.e1–e5, April 16, 2020 e4

Page 18: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Nuclei segmentation and trackingFor each time point, maximum projections were obtained for all 26 z sections per image. His2Av-mRFP was used to segment nuclei.

Nuclei-labeled channel images were pre-processed with Gaussian filtering, top-hap filtering, and adaptive histogram equalization, in

order to enhance the signal-to-noise contrast. Processed imageswere converted into binary images using a threshold value obtained

from Otsu’s method. Nuclei were then watershedded to further separate and distinguish from neighboring nuclei. Subsequently, bi-

nary images were manually corrected by inspecting all individual time frames using Fiji (https://fiji.sc). The number and the position of

separate components within a frame was obtained, where each component serves as a mask for individual nuclei. Nuclei tracking

was done by finding the object with minimal movement across the frames of interest. In Figures S1A, S2, S4F, and S4G, 512 3 512

maximum projection images were initially cropped into 430 3 430 to remove nuclei at the edge, and used for subsequent analysis.

Recording MS2 signalMaximum projections of MCP-GFP images were used to record fluorescent intensities. Using nuclei segmentation files as a mask for

each nucleus, fluorescence intensities within each nucleus were extracted. the signal of MS2 foci was determined by taking an

average of the top three pixels (Figures 1, 2, S1, S2, S4F, and S4G) or top two pixels (Figures 3, 4, and S4A–S4D) with the highest

fluorescence intensity within each nucleus. After obtaining MS2 trajectories, median fluorescence intensities within a nucleus were

subtracted as a nuclear background. Subsequently, minimum MS2 intensity was determined for individual trajectories and sub-

tracted to make the baseline zero.

Detection of transcriptional burstingA transcriptional burst was defined as a local change in fluorescence intensity. First, MS2 trajectories were smoothed by the moving

averagemethod.When a nucleus had above-threshold transcriptional activity, burst was considered to beON. Burst was considered

to be ended when the intensity dropped below 55% of the local peak value. When the burst duration is less than 5 time frames, it was

considered as a false-positive derived from detection noise. When signal trace exhibits continuous decreasing at the beginning of

burst detection, it was also considered as a false-positive. Location of defined burst was then moved two time frames afterward

to better capture the center of individual bursting event. Same method and threshold value were used throughout the analysis.

Description of bursting propertiesFrom each trajectory, number of bursts, amplitude and duration of each burst, integrated signal of each burst (burst size), onset of first

burst, cumulative integrated signal (cumulative RNA production) and total integrated signal (total RNA production) produced by each

nucleus were measured. To determine amplitude, the peak value during the burst was measured using trajectories after moving

average. The duration was determined by measuring the length of each burst. Burst size was measured by taking the area under

a single burst using rawMS2 intensities. Cumulative and total RNA production weremeasured by taking the area under the trajectory

using rawMS2 intensities. The amplitude, duration, and burst sizewere determined by taking average of all analyzed bursts in a single

nucleus.

Quantification of total active durationTo determine threshold, the maximum MS2 signal throughout all trajectories was determined for each embryo. The 10% of the

maximum was used as a threshold. Total duration above the threshold was measured for each nucleus.

False-coloring by cumulative RNA productionCumulative RNA production at each nucleus was measured as described in the previous section. Using segmentation mask, individ-

ual nuclei were false-colored with the pixel intensity proportional to the level of cumulative RNA production at given time in a given

nucleus. Resulting image was then colored and layered over the maximum projected image of His2Av-mRFP.

False-coloring by instantaneous MS2 signalsMS2 signal intensity at each nucleus wasmeasured as described in the previous section. Using segmentation mask, individual nuclei

were false-colored with the pixel intensity proportional to the instantaneous MS2 signal at given time in a given nucleus. Resulting

image was then colored and layered over the maximum projected image of His2Av-mRFP.

DATA AND CODE AVAILABILITY

Original imaging data of fluorescent in situ hybridization has been deposited to Mendeley Data (https://dx.doi.org/10.17632/

yypbkpxkx3.1)

e5 Molecular Cell 78, 224–235.e1–e5, April 16, 2020

Page 19: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Molecular Cell, Volume 78

Supplemental Information

Visualizing the Role of Boundary Elements

in Enhancer-Promoter Communication

Moe Yokoshi, Kazuma Segawa, and Takashi Fukaya

Page 20: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

0 10 20 30 40Time (min)

6.5 kb

enhancerM

ean

cum

ulativ

eRN

A pr

oduc

tiuon

(AU)

0

468

10

2

x105

0 10 20 30 40Time (min)

9 kb

0

468

10

2

x105

Mea

n cu

mula

tive

RNA

prod

uctiu

on (A

U)

0 10 20 30 40Time (min)

11 kb

0

468

10

2

x105

Mea

n cu

mula

tive

RNA

prod

uctiu

on (A

U)

A

Figure S1

00.51.01.52.0

0 10 20 30 40Time (min)

MS2

inte

nsity

(AU) x104

size

duration

ampli

tude

onset offirst burst

frequency

B

Total

median 20 46median % of6.5 kb reporter

RN

A pr

oduc

tion

(AU

)

0

2

4

6

8

10 x105 First 15 min Total

median 441 334 median 276 223 % ofNo boundary

RN

A pr

oduc

tion

(AU

)

0

2

4

6

8

10 x105C DFirst 15 min

Distance: 6.5 kb Distance: 9 kb Nhomie/Homie Nhomie/HomieNo boundary inv

Page 21: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Figure S1. Delayed burst induction leads to lower output, related to Figure 1. (A) Mean cumulative RNA production per nucleus. Shade represents standard deviation

of the mean across all analyzed nuclei at most ventral region (84 nuclei for the 6.5-kb

reporter, 87 nuclei for the 9-kb reporter, 84 nuclei for the 11-kb reporter).

(B) Trajectory of transcriptional activity shown in Figure 1H. Amplitude, duration and

burst size were measured for every single bursting event. Frequency was determined by

counting the total number of bursts during analysis. Onset of first burst was defined as a

time when the first burst started.

(C, D) Boxplots showing the distribution of RNA production at first 15min (left), and

total RNA production (right). The box indicates the lower (25%) and upper (75%)

quantile and the solid line indicates the median.�Whiskers extend to the most extreme,

non-outlier data points. In (C), a total of 356 and 363 most ventral nuclei, respectively,

were analyzed from three individual embryos for the reporter gene with 6.5-kb and 9-kb

enhancer-promoter distance. In (D), a total of 363, 350 and 359 most ventral nuclei,

respectively, were analyzed from three individual embryos for the reporter gene with no

boundary element, Nhomie/Homie and Nhomie/Homieinv. Plots of no boundary in (D) are

the same as the plots in (C) (distance: 9 kb).

Page 22: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Nhomie alone (101)Nhomie/Homie (80)inv

P > 0.5No boundary (96) P < 10-9

0

0.2

0.4

0.6

0.8

1.0

Cum

ulat

ive

fract

ion

0 6 8 10 122 4 x105

Total RNA produced (AU)

10

32

4

65

Mea

n cu

mul

ativ

eR

NA

prod

uctio

n (A

U)

0 10 20 30 40Time (min)

x105

Nhomie/Homieinv

10

32

4

65

Mea

n cu

mul

ativ

eR

NA

prod

uctio

n (A

U)

0 10 20 30 40Time (min)

x105

Nhomie alone

10

32

4

65

Mea

n cu

mul

ativ

eR

NA

prod

uctio

n (A

U)

0 10 20 30 40Time (min)

x105

No boundary

inv

A

Nhomie Homie

Enhancer-Promoter distance: 9 kb

24x MS2 yellowsna shadow

enhancer

24x MS2 yellowsna shadow

enhancer

Deletion

B C D

E

Figure S2

F

Nhomie Homie

Enhancer-Promoter distance: 6.5 kb

24x MS2 yellowsna shadow

enhancer

G

Mea

n cu

mul

ativ

eR

NA

prod

uctiu

on (A

U)

0

468

10

2

0 10 20 30 40Time (min)

12

Distance: 6.5 kbNo boundary

x105

II

0 6 12 180

0.2

0.4

0.6

0.8

1.0

Total RNA produced (AU)

Cum

ulat

ive

fract

ion

No boundary (84)

Nhomie/Homie (91)P < 10-12

x1050 10 20 30 40Time (min)

Mea

n cu

mul

ativ

eR

NA

prod

uctiu

on (A

U)

0

468

10

2

12

Distance: 6.5 kbNhomie/Homie

x105

H

Page 23: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Figure S2. A single boundary element can increase transcriptional output, related to Figure 2. (A) Homie was removed from the 3´ location of the yellow reporter gene.

(B-D) Mean cumulative RNA production per nucleus. Shade represents standard

deviation of the mean across all analyzed nuclei at most ventral region (80 nuclei for the

Nhomie/Homieinv reporter, 101 nuclei for the Homie-deletion reporter, 96 nuclei for the

no boundary reporter).

(E) A cumulative plot showing fraction of most ventral nuclei (y-axis) and total RNA

production (x-axis). The p values of Wilcoxon Rank-Sum test were shown.

(F) Schematic representation of the 6.5-kb reporter gene containing Nhomie and Homie.

(G, H) Mean cumulative RNA production per nucleus. Shade represents standard

deviation of the mean across all analyzed nuclei at most ventral region (84 nuclei for the

the no boundary reporter, 91 nuclei for the Nhomie/Homie reporter). Plot of no boundary

is the same as the plot in Figure S1A (distance: 6.5 kb).

(I) A cumulative plot showing fraction of most ventral nuclei (y-axis) and total RNA

production (x-axis). The p value of Wilcoxon Rank-Sum test was shown.

Page 24: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

A

homologyarm

3xP3-GFP

SF1

gRNA gRNA

homologyarm

WTScr-ftz locus

Donor plasmid

⊿SF1Scr-ftz locus 3xP3-GFP

Homology-directedrepair

Primer F

Primer E

Primer A

Primer BPrimer D

Primer C

Scr ftz-MS2

Scr ftz-MS2

4000—

1500—2000—

1000—

(bp) WT⊿SF1

Primer A+B Primer C+D Primer E+F

WT⊿SF1 WT⊿SF1

B

D

E

WT

⊿SF1

12

10

8

6

4

2

0Num

ber

of s

ex c

omb

teet

hpe

r leg

WT ⊿SF1

n=58 n=48

C

Scr

1 2 3 4 5ftz

stripe1 2 3 4 5ftz

stripe

ectopicScr expression

WT ⊿SF1F G

Figure S3

Page 25: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Figure S3. Characterization of SF1 deletion mutant, related to Figure 3. (A) Schematic representation of SF1 deletion from the endogenous Scr-ftz locus. Two gRNA expressing plasmids were co-injected with the Cas9 expressing plasmid and the

3xP3-GFP donor plasmid. Approximate annealing sites for primers used in (B) were shown. (B) PCR analysis of genomic DNA purified from WT ftz-MS2 and ⊿SF1 ftz-MS2

homozygotes. SF1 deletion was further confirmed by sanger sequencing of resulting PCR products. (C) Boxplots showing the distribution of numbers of sex comb teeth per first leg for WT

and homozygote ⊿SF1 mutant males. The box indicates the lower (25%) and upper (75%) quantile and the solid line indicates the median. Whiskers extend to the most extreme, non-outlier data points. A total of 58 and 48 legs, respectively, were analyzed for WT and

⊿SF1. Number of sex comb teeth was not reduced in homozygote ⊿SF1 mutant adults. (D, E) Fluorescent in situ hybridization of endogenous Scr in WT ftz-MS2 (D) and homozygote ⊿SF1 ftz-MS2 embryos (E). Embryos after germ band extension were shown.

Images were cropped and rotated to align embryos (anterior to the left and posterior to the right). Scale bar indicate 50 μm. (F, G) Merged images of endogenous ftz (blue) and Scr (gray) in WT ftz-MS2 (F) and

⊿SF1 ftz-MS2 homozygote embryos (G) shown in Figure 3B and C. Anterior part of embryo was zoomed in. Images were cropped and rotated to align embryos (anterior to the left and posterior to the right).

Page 26: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

D⊿SF1

0

0.8

1.2

1.6

0.4

Mea

n in

tens

ity (A

U)

0 10 20 30 40Time (min)

x104 WT

Figure S4

00.51.01.52.0

MS2

inte

nsity

(AU

) 2.5

B

0 10 20 30 40Time (min)

WT interstripex104

Repression

00.51.01.52.0

MS2

inte

nsity

(AU

) 2.5

C

0 10 20 30 40Time (min)

⊿SF1 interstripex104

Repression

Interstripe

0

0.2

0.4

0.6

0.8

1.0

Cum

ulat

ive

fract

ion

A

WT (241)⊿SF1 (235) P > 0.8

0 8 12 164 x105

Total RNA produced (AU)

median 102% of WT

Interstripe

E

SF1 SF2

Enhancer-Promoter distance: 9 kb

24x MS2 yellowsna shadow

enhancer

F G

0 4 8 12Total RNA produced (AU)

Mea

n cu

mul

ativ

eR

NA

prod

uctiu

on (A

U)

0

468

10

2

Mea

n cu

mul

ativ

eR

NA

prod

uctiu

on (A

U)

0

468

10

2

0 10 20 30 40Time (min)

0 10 20 30 40Time (min)

160

0.20.40.60.81.0

Cum

ulat

ive

fract

ion

No boundary (87)SF1/SF2 (96)

P < 10-4

No boundary SF1/SF2

median 139% of No boundary

x105 x105

x105

Page 27: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Figure S4. Dynamics of transcriptional repression at inter-stripe region, related to Figure 4. (A) A cumulative plot showing fraction of nuclei (y-axis) and total RNA production (x-axis) in inter-stripe nuclei. The p value of Wilcoxon Rank-Sum test was shown. Change in median value after SF1 deletion was shown in upper right. (B, C) A representative trajectory of transcriptional activity of WT ftz-MS2 (B) and ⊿SF1 ftz-MS2 (C) in individual nuclei at the inter-stripe region. (D) Mean MS2 spot intensity per nucleus. Shades represent standard deviation of the mean across all analyzed nuclei at inter-stripe region (241 nuclei for WT, 235 nuclei for ⊿SF1) from three independent embryos. (E) Schematic representation of the yellow reporter gene containing the 2231-bp SF1 and the 1993-bp SF2. Orientation of SF1 and SF2 is same as in endogenous Scr-ftz locus. (F) Mean cumulative RNA production per nucleus. Shade represents standard deviation of the mean across all analyzed nuclei at most ventral region (87 nuclei for no boundary reporter, 96 nuclei for SF1/SF2 reporter). Plot of no boundary is the same as the plot in Figure S1A (distance: 9 kb). (G) A cumulative plot showing fraction of most ventral nuclei (y-axis) and total RNA production (x-axis). The p value of Wilcoxon Rank-Sum test was shown.

Page 28: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Pol IIBEAF-32

CP190Mod(mdg4)

Su(Hw)CTCF

BarrenCAP-H2

Rad21

Z4L3mbt

ChromatorDREF

Fs(1)h-L

TFIIIC

eveCG12134 TER94

[0-2000]

[0-5]

[0-5]

[0-4]

[0-5]

[0-3]

[0-60]

[0-1226]

[0-1132]

[0-335]

[0-400]

[0-166]

[0-132]

[0-269]

[0-253]

Figure S5

Nhomie Homie

Page 29: Visualizing the Role of Boundary Elements in Enhancer ...In Brief Yokoshi et al. employ quantitative live-imaging methods to visualize impacts of TAD formation on enhancer-promoter

Figure S5. ChIP-seq and ChIP-chip profiles of endogenous eve locus, related to Figure 5. Not only CTCF and Rad21, but also other regulatory proteins are highly enriched at

topological boundaries. Fs(1)h-L ChIP-seq data from a WT embryonic cell line (Kellner, et al., 2013), DREF, L3mbt and Z4 ChIP-seq data from a WT embryonic cell line (Li, et al., 2015a), Chromator, TFIIIC, Rad21, CAP-H2 and Barren ChIP-seq data from a WT

embryonic cell line (Van Bortle, et al., 2014), CTCF, Su(Hw), Mod(mdg4), CP190 and BEAF-32 ChIP-chip data from 0-12 h WT embryos (Roy, et al., 2010), and Pol II ChIP-seq data from 2-3 h WT embryos (Sun, et al., 2015) were visualized with IGV browser.