You are on page 1of 14

ARTICLE

DOI: 10.1038/s41467-017-02089-9 OPEN

KSHV episomes reveal dynamic chromatin loop


formation with domain-specific gene regulation
Mel Campbell1, Tadashi Watanabe 2, Kazushi Nakano 1, Ryan R. Davis 3, Yuanzhi Lyu 1,
Clifford G. Tepper 4, Blythe Durbin-Johnson5, Masahiro Fujimuro2 & Yoshihiro Izumiya 1,4,6

The three-dimensional structure of chromatin organized by genomic loops facilitates RNA


1234567890

polymerase II access to distal promoters. The Kaposi’s sarcoma-associated herpesvirus


(KSHV) lytic transcriptional program is initiated by a single viral transactivator, K-Rta. Here
we report the KSHV genomic structure and its relationship with K-Rta recruitment sites using
Capture Hi–C analyses. High-resolution 3D viral genomic maps identify a number of direct
physical, long-range, and dynamic genomic interactions. Mutant KSHV chromosomes har-
boring point mutations in the K-Rta responsive elements (RE) significantly attenuate not only
the directly proximate downstream gene, but also distal gene expression in a domain-specific
manner. Genomic loops increase in the presence of K-Rta, while abrogation of K-Rta binding
impairs the formation of inducible genomic loops, decreases the expression of genes net-
worked through the looping, and diminishes KSHV replication. Our study demonstrates that
genomic architectural dynamics plays an essential role in herpesvirus gene expression.

1 Department of Dermatology, School of Medicine, University of California Davis (UC Davis), Sacramento, CA 95817, USA. 2 Department of Cell Biology,

Kyoto Pharmaceutical University, Kyoto 607-8412, Japan. 3 Department of Pathology and Laboratory Medicine, School of Medicine, UC Davis, Sacramento,
CA 95817, USA. 4 Department of Biochemistry and Molecular Medicine, UC Davis School of Medicine, Sacramento, CA 95817, USA. 5 Division of
Biostatistics, Department of Public Health Sciences, UC Davis School of Medicine, Sacramento, CA 95817, USA. 6 UC Davis Comprehensive Cancer Center,
Sacramento, CA 95817, USA. Mel Campbell, Tadashi Watanabe, and Kazushi Nakano contributed equally to this work. Correspondence and requests for
materials should be addressed to Y.I. (email: yizumiya@ucdavis.edu)

NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications 1


ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02089-9

T
issue-specific cellular gene expression is regulated by the highlight the vital significance of higher-order viral genomic
formation of active chromatin hubs (ACHs) at enhancer structure to the viral life cycle.
regions of the genome, where many tissue specific-gene Like all herpesviruses, KSHV can exhibit two alternative life-
promoters are brought into proximity1. As reviewed by Palstra cycles, known as latency and lytic replication. In latency, the viral
et al.2, the concept of ACHs originated, in part, from the fact that genome persists in the host as low copy-number nuclear epi-
the protein concentration of many nuclear factors is below the somes, and its expression is largely silenced with the exception of
dissociation constant of protein-protein or protein-DNA inter- several genes24. KSHV lytic replication is initiated by expression
actions. Accordingly, it is necessary to have mechanisms to of a single viral protein, K-Rta. K-Rta is both essential and suf-
increase the local concentration of nuclear factors at a given ficient to induce lytic reactivation of the latent KSHV genome in
chromatin site. Transcription factors pinpoint their binding sites the BCBL-1 cell line model, as well as in the de novo infection
by three-dimensional scrutiny of nuclear space, and the formation model25.
of productive transcription complexes on DNA is intrinsically In this study, we performed an unbiased chromosome con-
dynamic3,4. A higher concentration of factors favors efficient formation analysis by Capture Hi–C analysis, which is 3C com-
binding to DNA templates by facilitating rapid re-association of bined with next-generation sequencing (NGS)26. We questioned
dissociating factors at the same or abutting sequences. Thus, the if the KSHV genome is structured to maximize effects of K-Rta
concentration of transcription factors and co-factors near tran- recruitment to achieve robust viral gene expression and also took
scription initiation sites is a sensitive limiting component deter- advantage of an established reactivation system27 to analyze
mining the number of transcripts produced. Therefore, spatial spatiotemporal gene regulation in high resolution. We identified
and temporal clustering of cognate binding sites is proposed to be several KSHV genomic loops that encompassed K-Rta direct
an important means to boost the local concentration of factors binding sites, and induction of K-Rta expression further induced
and thus is indispensable for the regulation of the transcriptional genomic loop formation. These studies complement our recent
rate of genes5. report utilizing immuno-FISH approaches to study KSHV tran-
Development of chromosome conformation capture (3C) scription in situ and examine KSHV gene expression in the 3D
techniques has permitted the examination of ACH formation, nuclear space of an infected cell28. The study showed that epi-
and numerous studies have indeed demonstrated widespread somes responding to stimulation aggregated and recruited
occurrence of stimulus-responsive enhancer-promoter and molecules that formed large “transcription factories”. With the
promoter-promoter interactions between co-regulated genes6–8. results reported here, we show that KSHV has evolved spatio-
It is important to note that core promoters typically only support temporal gene regulatory mechanisms to assemble viral genomic
low-level basal transcription; cis-regulatory modules or enhancers domains and recruit RNA polymerase II, which results in effec-
actually carry most of the regulatory information for gene tive viral gene expression from uniquely structured viral
expression9. These elements can be located near the core pro- episomes.
moter or distal to a target gene which is typically found within
accessible chromatin10,11. The combined input of activating and
Results
repressing transcriptional factors localized to an ACH then
K-Rta binding sites on the KSHV genome. The KSHV
determines its overall regulatory output, which dictates tissue-
immediate early gene, K-Rta, is both necessary and sufficient to
specific gene expression patterns12–14. Transcriptional factors,
switch latent KSHV into the lytic infection cycle27,29,30. We
communication proteins, as well as non-coding RNA are sug-
speculated that robust gene activation by K-Rta associates with a
gested to be involved in the assembly of distant regulatory ele-
unique structure formed by the KSHV genome, which is pre-
ments11,15–18.
fabricated with an architecture designed for K-Rta recruitment.
Gamma-herpesvirus replication takes place in specific tissues
To examine this hypothesis, the K-Rta binding sites in the KSHV
and viral gene expression occurs robustly in a highly-organized
genome were precisely mapped with ChIP-seq analysis in the
fashion. More than eighty viral genes are encoded by a relatively
context of the BCBL-1 cell line model. BCBL-1 is derived from a
small viral genome, thus gene regulatory information such as the
KSHV-infected human primary effusion (body cavity-based)
formation of ACHs is likely to be needed and embedded within
lymphoma (PEL) and contains latent KSHV genomes. The TREx-
the viral lifecycle as an accompaniment to DNA sequences
(F3H3)-K-Rta BCBL-1 (hereafter called TREx-K-Rta BCBL-1)
required for efficient viral replication. Furthermore, such small
subline containing a Tet-inducible Flagx3 and HAx3-tagged K-
viral episomes provide a unique experimental model to study 3D
Rta expression cassette was generated from TREx-BCBL-1 cells27.
genetic interactions and their association with gene expression.
Doxycycline (Dox)-induced K-Rta expression (Fig. 1a, top panel),
Kaposi’s sarcoma-associated herpesvirus (KSHV) episomes fur-
which consequently triggers KSHV reactivation, was confirmed
nish a superb system for defining these mechanisms due to the
by probing for the viral lytic protein K-bZIP (Fig. 1a, middle
combination of benefits derived from prior knowledge of defined
panel). The K-Rta recruitment sites were then examined by ChIP-
viral transcriptional factors necessary to initiate viral gene
seq analysis at 24 h post doxycycline treatment (Fig. 1b). The
expression, smaller genomic size, simpler genome organization,
results showed multiple recruitment sites of K-Rta, including the
and a tractable genomic sequence using a bacterial artificial
promoter regions for ORF6, 7, 8, K2, 70, PAN RNA, 31, 45, K-
chromosome (BAC) system. Similar to cellular chromosomes, the
Rta, K-bZIP, ORF57, K12, 73, and 75, in addition to both ori-Lyt
viral genome is wrapped upon post-translationally modified
loci (Supplementary Table 1). These results were in good agree-
histones and interacts with multiple histone modifying
ment with previous promoter analyses that identified K-Rta
enzymes19–21. Such histone modifications correlate with tran-
responsive promoters in reporter assays conducted with 293
scriptionally “active”, “poised”, and “repressed” domains of viral
cells31. Among those promoters, the K-Rta response elements
chromatin in a manner equivalent to cellular chromosomes22. In
(RE) in the PAN and K12 promoters have been studied in
addition, viral genomic loops were previously identified in
detail32,33, which allowed us to prepare mutant KSHV genomic
KSHV-infected cells, in which CCCTC-binding factor (CTCF)-
episomes (described below).
cohesin complexes establish physical looping of the KSHV gen-
ome18. Disruption of such genetic interactions by ablation of
components of the cohesin complex deregulated the KSHV KSHV chromatin interactions. Next, we examined the rela-
latency-lytic switch leading to viral reactivation18,23. These results tionship between KSHV genomic structure and K-Rta

2 NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02089-9 ARTICLE

a TREx-F3-H3-K-Rta
BCBL-1
Dox
– +
(kDa)

100 K-Rta

37
K-bZIP

50
β-actin

b ChIP-seq (K-Rta) K12/Ori-R


30
Ori-L
Sequence reads

K5/PAN
K2 31/32 45/50p
6 7 8 70 K-bZIP
41 57 73 75

30 Input DNA
Sequence reads

0
K2 2 K3 70 K4 ori-Lyt K5 K6 PAN RNA17 19 20 23 24 29b 29a 39 42 43 45 46 47 48 49 52 53 55 K9 K10 K10.5 K11 58 59 60 61 62 65 66 67 K12 K13 72 73 75 K15

K1 4 6 7 8 9 10 11 K7 16 18 21 22 25 26 27 28 303132 33 34 35 36 37 38 40 41 44 50 K8 K8.1 54 56 57 63 64 68 69 T0.7 K14 74

0 10 20 30 40 50 60 70 80 90 100 110 120 130 138 kb

Fig. 1 K-Rta recruitment sites on the KSHV genome KSHV reactivation. a KSHV lytic protein expression in TREx-K-Rta BCBL−1 cells during latency (−) and
at 24 h post-dox treatment (+) was examined by immunoblotting. Blots were probed with the indicated antibodies. b K-Rta recruitment sites were
examined by ChIP-sequencing analysis using K-Rta ChIP DNA prepared from TREx-K-Rta BCBL−1 cells 24 h post-dox treatment. Peak finding was
performed with MACS2 program using the default parameter settings. Peaks mapped to the KSHV genome are plotted and the location of the each peak is
indicated by the name of the nearest ORF name. Peaks are also summarized in Supplementary Table 1

recruitment sites before and during KSHV reactivation. The cluster region (Fig. 2a). The frequency of several of these latent
possible occurrence of long-range interactions was investigated by interactions were highly significant above the empirical dis-
comprehensively examining all KSHV intra-genomic interactions tribution of the population of detected ligation junctions (p <
using Capture Hi–C, a chromosome conformation capture (3C) 0.001) (Table 2 outlier test, part A). The relative genomic loop
approach coupled with target sequence capture and NGS26. The frequencies following reactivation are also depicted as a heatmap
capture step was achieved using a custom library of biotinylated to visualize inducible genomic loop formation (Fig. 2b). When
oligonucleotide baits (xGen Lockdown probes; IDT) that target reactivation was triggered by induction of K-Rta expression,
the entire KSHV genome, including terminal repeats. This step genomic loops among regions encoding early and immediate-
was incorporated in order to enhance the sensitivity of Hi–C early lytic genes, especially at the K-Rta promoter loci, were
analysis to detect chromatin interactions of the KSHV genome by increased (Fig. 2b, BamHI position #14). On the other hand, the
enabling substantial enrichment of KSHV-derived sequences number of genomic loops associated with the late and latent gene
(Supplementary Fig. 1). For this experiment, we used the same cluster regions (ORF63–ORF75; BamHI sites #19–29 in Fig. 2a)
design as described above with TREx-K-Rta BCBL-1 cells and did not increase significantly at this time point (24 h). In the
performed the capture Hi–C analysis from cells during latency presence of K-Rta, frequencies of genomic interactions were
and induced into the lytic cycle. We obtained 4656 (latent library) further increased between the PAN promoter (BamHI position
and 5520 (lytic library) sequence reads that were successfully #5) and the K-Rta promoter locus (BamHI position #14) (Fig. 2b
ligated and mapped within the KSHV genome at the 29 BamHI and Supplementary Fig. 2), as well as additional loops engendered
sites distributed along the ~ 150-kb KSHV episome. These liga- between the PAN promoter region and the downstream lytic gene
tion junction partners (BamHI sites) were then visualized with cluster region. Genomic loops formed by contacts between the K2
Circos34. The number of sequence reads and identified genomic promoter region (BamHI position #2), which is another major K-
interactions are summarized in Table 1. KSHV genomic inter- Rta recruitment site (Fig. 1b), to the K-Rta promoter region
actome maps depicting intra-genomic connections both before (BamHI, position #14), as well as to the PAN promoter region
(latency) and after K-Rta induction (reactivation) are shown in (BamHI position #5) were also increased (Fig. 2b and Supple-
Fig. 2a. The results showed that there is a trend for loop for- mentary Fig. 2). Statistical analysis showed, with the exception of
mation within similar functional genomic domains (topology the K2/K-Rta promoter (#2/#14) interaction, that all of the
domains), such as the latency cluster region and early-lytic gene highlighted induced pairs in Fig. 2b were confirmed to be highly

NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications 3


ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02089-9

Table 1 Capture Hi–C sequencing reads and genomic links of ligation products

BCBL-1 cells/iSLKr.219 cells Total read Valid genome Total genomic loops with Genomic loops between KSHV-KSHV loops/total
pairs loops (Total) KSHV DNA (host + viral) KSHV DNA fragments KSHV loops (%)
TREx K-Rta BCBL Dox(−) 23,230,497 20,345 19,531 4656 23.8
TREx K-Rta BCBL Dox(+) 25,593,592 12,739 12,203 5520 45.2
TREx K-Rta BCBL Dox (−)a 51,479,787 920,102 656,969 122,956 18.7
TREx K-Rta BCBL Dox(+)a 49,486,847 977,802 698,569 236,110 33.8
iSLK r.219 Dox (−)#1 30,807,748 58,035 51,779 5196 10.1
iSLK r.219 Dox (+)#1 25,005,026 41,918 38,812 6224 16.0
iSLK r.219 Dox (−)#2 24,713,145 13,799 9937 2569 25.9
iSLK r.219 Dox (+)#2 24,903,367 15,414 11,215 3621 32.3
iSLK r.219 Dox (−)#3 21,885,721 11,970 8857 2207 24.9
iSLK r.219 Dox (+)#3 25,208,803 12,161 9056 3075 34.0
aDerived from DpnII. All other datasets are derived from BamHI ligation products.

significant above the background distribution of the data used to CTCF binding sites, a factor associated with loop formation and
generate the heatmap (p < 0.001) (Table 2 outlier test, part B). which is known to recognize the CCA(C/G)(C/T)AG(A/G)(G/T)
Capture Hi–C interaction frequencies highlighted in Fig. 2b were GGC core sequence37. Using the mutant episomes described
also validated by qPCR (Fig. 2c). above, K-Rta mediated viral gene expression was examined by
RT-qPCR to define the importance of a single K-Rta direct
binding site to the entire KSHV gene expression program. KSHV
Inducible genomic loops and K-Rta recruitment sites. To RT-qPCR arrays provided an expression read-out for every
complement and confirm our Capture Hi–C results obtained with KSHV ORF over 72 h of reactivation (Fig. 4c and Supplementary
BamHI digestions, we further generated a higher resolution Fig. 3). As expected, PAN RNA decreased dramatically in the
genomic interaction map by performing Capture Hi–C analyses PAN Mu cells. Surprisingly, disruption of the K-Rta-K12 binding
using the DpnII restriction enzyme. The enzyme digests KSHV site in the K12 Mu genome also diminished PAN expression,
genomic DNA at 350 different sites and the frequent digestion despite the presence of exogenous K-Rta and an intact K-Rta RE
allowed us to generate a linear genomic map by quantifying the at the PAN promoter (Fig. 4c, inset). Similarly, K12 expression
Capture Hi–C sequencing reads encompassing the region at 3-kb was reduced not only in K12 Mu cells but also in the PAN Mu
intervals (Fig. 3). The linear heatmap of induced genomic looping cells (Fig. 4c, inset). As expected, the DKO virus was the most
formation was overlaid with the K-Rta ChIP-seq results (Fig. 1), defective for expression of these two lytic genes (Fig. 4c and
which allowed visualization of the relationship between K-Rta Supplementary Fig. 3). Interestingly, there were clusters of
recruitment sites and inducible genomic loop formation. gene loci that were responsive to each particular K-Rta binding
A number of genomic interactions among early and site mutation (Fig. 4d). The expression of a contiguous stretch of
immediate-early lytic encoding regions, especially near the genes from K6-ORF43 was particularly sensitive in the PAN
K-Rta recruitment sites, were again elevated (Fig. 3 heatmap, red), K-Rta RE mutation (Fig. 4d, upper and middle panels), ORFs
and genomic links with late gene and latent gene clusters were marked in red and purple in the genome map); whereas for the
either reduced or unchanged by K-Rta expression at 24 h post K12 K-Rta RE mutation, the expression of several discrete clusters
induction (Fig. 3 heatmap, blue). The results suggested that K-Rta of genes that include ORF11-17, ORF37-40, ORF50-52, ORF54-
binding sites are brought into proximity and aggregated in three- 55, and a latent gene cluster region were decreased (Fig. 4d, upper
dimensional space via the formation of genomic loops during and lower panels), ORFs marked blue and purple in the genome
active transcription. Although only 2 of 13, 3-kb interval pairs map). Importantly, the affected ORF clusters encompassed
that were evaluated attained statistical significance over the genomic regions that are physically involved with the K-Rta-
background distribution (p < 0.001) (Table 2 outlier test, part C), induced genomic looping events identified (Fig. 3), thereby pro-
the overall frequency of genomic loops within the KSHV genome viding validation of both the findings from the Capture Hi–C
consistently increased during reactivation, while genomic links studies and their functional significance. In the iSLK cells, K-Rta
between KSHV and host chromosomes simultaneously decreased is supplied exogenously in a dox-inducible manner, thus K-Rta
(Table 1). Similar loop frequency patterns were also observed in expression is largely equivalent across the different recombinant
Capture Hi–C experiments using libraries prepared from KSHV-infected cell lines (Fig. 4b, c). However, endogenous K-Rta
recombinant KSHV-infected iSLK cells in three biological repli- expression was also sensitive to both the PAN promoter–K-Rta
cates (Table 1). interaction and the K12 promoter–K-Rta interaction (Fig. 4e). As
expected, viral replication was diminished in mutant viruses,
Single K-Rta binding sites regulate distal gene expression. The exhibiting reductions in encapsidated viral DNA copy number
significance of K-Rta recruitment and its association with the relative to BAC16 WT at 120 h post reactivation with Dox
genomic loops was next examined after preparing recombinant (Fig. 4f).
viral chromatin, which harbors two known K-Rta direct binding
sites32,33. Using a two-step marker-less Red recombination
approach with KSHV BAC1635,36, we created a series of point Clustering of K-Rta binding sites at the KSHV ACH. As cap-
mutations in either the PAN K-Rta response element (RE; PAN ture Hi–C data (physical evidence) and KSHV PCR array results
Mu), the K12 RE (K12 Mu), or both REs (double-knockout, (functional evidence) indicated loop formation between regions
DKO, Fig. 4a). A panel of iSLK cell lines infected with recom- of the KSHV genome containing the PAN RE and K12 RE, we
binant KSHV harboring K-Rta binding site mutations were asked whether binding of K-Rta at one RE would influence
subsequently established. Our mutations do not involve putative binding at the other, presumably proximal RE. K-Rta ChIP

4 NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02089-9 ARTICLE

a b

on
ORF4
ORF7

ORF6
K15.

TR

.K1

gi
OR

F7
....

.
...
.

re
..
.

.
5

OR
F74 .

er
.
K1

F
.

#1

on
OR

ot
.
4
OR

gi
om
.

#5
F9
F7

re
O
.

OR

#2
3

pr
OR K1

R
PA
(29) BamH I

10

10
BamHI (1)
(28) Ba mHI

F4
(27) Ba HI
F7 3

. RF

ta
K2
(26) Ba mHI
.

/K
.

11
2.

(25

R
. RF

re

K9
K-
Bam

re
re
O
) Ba

mH
(24

gi
or 2
K. 2 RF #2 K2 region
.

gi
gi
mH
i-

8
4
on
I
K1 Lyt-

) Ba

on
on

#1
O.

#1
(2
2/T R K3

I
3)

mH
0
OR 0.7. . F7

29
Ba
OR

1
I
F6

m
OR 9 . 4

28
.

HI

2
OR F6 2) K. .1
8 ( . 4K4.2

27
K
F
OR 67A . . HI

3
.
am t

26
F6 -Ly
7. B

4
OR
F ) ori

25
OR 66 I (3

5
F65 . mH .

24
. Ba I

6
mH I K5
#5 PAN region

23
BaamH I (4) .

7
B amH I K6

22
(22 B amH I .

8
)Ba B amH
mH

21
I B amHI .

9
B amHI ) K7 N
PA

20
ORF6 B mHI (5

11 0
4 .
Ba F16

1
OR

9
.

on 1
(21) Ba ORF17. .5

iti 18
.ORF17

13 2
mHI

os 17
ORF18
.

Ip 6
14
. BamHI (6)

H 51
(20) BamHI ORF19

Ba
.

15
Value

am 14 1
.ORF20

m
ORF63

17 6
1
H
BamHI (7) ORF21

B 3
. (19) BamHI

Ip

1
.

12
ORF62.

os

18
ORF22 9

11
iti

20 9
1
o
1 .
ORF6

10
n
. OR
. F23 6

22 1

9
0

2
ORF6 .

8
ORF2
59
ORF . 4 3

23
.

7
F58 . B
Ba amH

24
OR

6
.. mH I(8

25
I ) 0

5
OR
1 Ba F2
K1 5

27 6

4
. m

2
HI
I

.. .
H

(9

3
m

) .ORF
Ba

28
0.5 .

2
K1 . OR 26
8)

Ba

O F2
I
(1

..

29
mH

1
O RF 7
mH
Ba

R 2
a

O F 8.
HI

mH

I(1
)B

0
Bam

O RF 29
K1
)Bam

0)
(17

RF 3
I

BamHI(13
(15)BamH

I(1
(14) Bam HI

31 0.
.

O.
HI(1

1)

.
. RF 9.a
R
(16

F3
O
K9

.2
2)
F5 .

OR F34 5.

33
7

.O ORF 36
..

F2
)

R 3
OR
OR

.
6
F5

OR F38.

F
F5 .

. OR 9
5.
OR

OR
OR F54

F3
.
OR 53.

#18 K9/K10 region


OR
...
OR

F52

. F41

7
..

F3
O
.R 2
K8.1
K8
. OR

O
F

c 6.00E-01
. R
ORF4

F40
0.

.
F49

ORF44
.
F5

ORF48

.ORF45
ORF46
.
ORF47.
.

F4

KSHV ORF sense No Dox


OR

OR

5.1x
3
.

KSHV ORF antisense * + Dox/24 h


5.00E-01
#14 K-Rta promoter 2.4x
containing region *

Relative to Bac16
4.00E-01 3.3x
**
3.00E-01 4.1x
*
2.00E-01 2.8x
**
1.00E-01

0.00E+00

0/ A
A

-1 TA

TA
K2 TA

1/ A
K2 TA
PA 2

2
/K

PA N/K

T
PA RT

F4 T

F4 RT

K9 /pR
K9 pR

pR
R

R /pR
R
N

/p

/p

O 1/p
/p
PA

0
N

-1
R
O
Fig. 2 Comprehensive mapping of KSHV genomic loop formation in TREx-K-Rta BCBL-1 cells with Capture Hi–C. a Circos diagrams depicting KSHV
genomic links detected by Capture Hi–C. Each arc connects two BamHI fragments and represents a distinct interaction. Blue- and Red-colored lines indicate
genomic links occurring before or after of K-Rta induction, respectively. Black outer hatches mark genomic positions. ORF names are indicated in the outer
circle. Positions of non-coding RNA are shown by gray shading in the outer circle and position of BamHI sites are also indicated. b Regulation of genomic
loops by KSHV reactivation in TREx-K-Rta BCBL-1. The relative increase/decrease of genomic links by induction of K-Rta was obtained by subtracting the
relative sequence reads of non-reactivated TREx-K-Rta BCBL-1 cells from those in reactivated TREx-K-Rta BCBL-1 cells, and visualized in a heatmap by using
R package ggplot2. Values are sequence reads at each position divided by the total sequence reads contained within the KSHV genome and multiplied by
103. The numbers indicate positions of the BamHI sites, which correspond to the BamHI site numbering shown with in the Circos plot in panel a. Several
BamHI sites and their corresponding column in the heatmap are labeled, and genomic loops that increased during reactivation are marked by black squares.
P-values for selected loops are listed in Table 2 part A (constitutive) and part B (induced). c Enriched Capture-C DNA (BamHI ligation products) was
analyzed by qPCR for the contacts listed using DNA from TREx-K-Rta BCBL-1 cells±dox. Values (mean±SD, n = 3) represent signals obtained relative to a
BAC16 BamHI random ligation matrix. Fold increase (+dox/−dox) is listed. (*p < 0.05; **p < 0.01, Student’s t-test)

analysis was performed using chromatin from dox-induced of PAN RNA 3000-fold above the level present in reactivated
(0 and 8 h) iSLK BAC16 WT, PAN Mu, and K12 Mu cells and PAN Mu cells transfected with empty vector (Fig. 5b, left panel).
the degree of K-Rta recruitment to each RE was examined. In Despite this level of PAN complementation, gene expression
WT-infected cells, K-Rta binding was very robust at the K12 RE, analysis demonstrated that the PAN Mu virus was still dramati-
but reduced binding at this RE was observed with either the K12 cally defective in lytic KSHV gene expression (Fig. 5b, right
Mu or PAN Mu virus (Fig. 5a, right panel). A similar reduction in panel), indicating that activation of the PAN promoter and/or
PAN RE binding was observed in cell lines containing either of active transcription of PAN RNA is important for the transacti-
the mutant viruses, although the overall binding of K-Rta was vation of many other viral genes. These results are consistent with
lower than that observed at the K12 RE (Fig. 5a, left panel). Taken the fact that K12 Mu also impaired PAN RNA expression ~ 16-
together, these results suggest that the binding of K-Rta to its fold, yet the pattern of gene cluster expression is significantly
target promoters is dependent, at least in part, through clustering different between the two mutants (Fig. 4d). If the presence of
of the sites via genomic looping and the formation of ACHs. PAN RNA is the key, we should have expected to see similar gene
expression profiles between the two mutants.

Ectopic PAN RNA does not rescue the PAN RE mutant. One
explanation for the global defect in gene expression for the K-Rta K-Rta binding and inducible genomic loops. Next, we examined
RE site mutant viruses is that each binding site mutation results effects of K-Rta binding on inducible genomic loop formation.
in reduction in the amount of PAN RNA and/or K12 proteins Capture Hi–C analyses with iSLK/r.219 cells were first performed
produced and needed for efficient reactivation, rather than a to probe differences of genomic loop formation by cell line, and
genomic binding site/looping effect, per se. To address this, PAN we also generated a heatmap of inducible KSHV genomic loop
Mu iSLK cells were first transfected with a PAN RNA expression formation in iSLK/r.219 cells (Fig. 6a, b). We have performed the
vector 48 h prior to dox-induced reactivation. This raised the level Hi–C analyses with three biological replicates and confirmed

NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications 5


ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02089-9

Table 2 Statistical analysis of contacts

Contact paira Contact description P-value


A Constitutive Loop Formation
2/5 K2/PAN <0.001
2/18 K2/K9-10 0.871
5/14 PAN/K-Rta promoter 0.985
5/18 PAN/K9-10 <0.001
5/12 PAN/ORF41 0.998
5/16 PAN/K8.1 >0.999
12/18 ORF41/K9-10 <0.001
14/12 K-Rta promoter/ORF41 >0.999
14/18 K-Rta promoter/K9-10 <0.001
18/23 K9-10/ORF72 <0.001
18/26 K9-10/ORF74 0.985
B Induced Loop Formation
2/5 K2/PAN <0.001
2/14 K2/ K-Rta promoter 0.8658
5/14 PAN/K-Rta promoter <0.001
14/12 ORF46/ORF41 <0.001
14/18 ORF46/K9-10 <0.001
2/5 K2/PAN <0.001
C Induced Loop formation DpnII 3 kb interval
n/ab 18001 to 21000 and 117001 to 120000 >0.999
n/a 27001 to 30000 and 15001 to 18000 <0.001
n/a 27001 to 30000 and 18001 to 21000 >0.999
n/a 27001 to 30000 and 21001 to 24000 <0.001
n/a 27001 to 30000 and 51001 to 54000 >0.999
n/a 27001 to 30000 and 60001 to 63000 0.999
n/a 27001 to 30000 and 72001 to 75000 >0.999
n/a 33001 to 36000 and 117001 to 120000 >0.999
n/a 51001 to 54000 and 117001 to 120000 >0.999
n/a 63001 to 66000 and 117001 to 120000 >0.999
n/a 72001 to 75000 and 117001 to 120000 >0.999
n/a 75001 to 78000 and 117001 to 120000 >0.999
n/a 81001 to 84000 and 117001 to 120000 >0.999
D Induced Loop Formation iSLK/r.219 cells
2/5 K2/PAN 0.091
2/14 K2/K-Rta promoter 0.352
5/14 PAN/K-Rta promoter 0.200
12/14 ORF41/K-Rta promoter 0.171
14/18 K-Rta promoter /K9-10 0.588
aBamHI site numbering; bnot applicable. Statistically significant interactions are listed in bold
A Select potentially outlying pairs of positions were compared to the empirical distribution of data from the remaining pairs of positions Fn(x), with P-values calculated based on extreme value theory as 1
−Fn(x) . BCBL-1 TREx-F3-H3-K-RTA_BamHI_3C_No Dox Genomic loop numbers (Fig. 2a Circos plot, blue interactions) data were analyzed
n
B Potentially outlying pairs of positions (#2–#5, #2–#14, #5–#14, #14–#12, #14–#18) were compared to the empirical distribution of data from the remaining pairs of positions Fn(x), with p-values
calculated based on extreme value theory as 1–Fn(x)n. The genomic loop data analyzed were that presented as the heatmap in Fig. 2b
C The data analyzed were that presented as the heatmap in Fig. 3. Statistical methods were the same as described for (A)
D Potentially outlying pairs of positions (#2–#5, #2–#14, #5–#14, #12–#14, #14–#18) were analyzed. The data analyzed were presented in the heatmap of Fig. 6b. Two sample t-tests performed on log-
transformed differences were used to test if the change for each pair of positions of interest (2–5, 2–14, 5–14, 12–14, 14–18) differed from 0

dynamics of genomic looping formation to be very similar ChIP-seq results, we selected four BamHI fragments that harbor
(Table 1). Representative figures from one of the triplicates are K-Rta recruitment sites and contain regions where increased
shown in Fig. 6. Similar to TREx-K-Rta BCBL-1 cells, genomic genomic looping formation with the PAN promoter region (#5
loops within lytic genes were increased compared to those of with bright red arrow) were detected in the presence of K-Rta
latent gene cluster regions (Fig. 6b, and Supplementary Fig. 4a). (Fig. 6c red arrows). BamHI fragments that do not contain a K-
For example, genomic loops among PAN loci, Ori-Lyt, and K-Rta Rta binding site were also included as a comparison (Fig. 6c blue
promoter loci were again elevated in the presence of K-Rta pro- arrows). KSHV episome copy numbers in the iSLK cells were also
tein, indicating that the gene regulatory mechanism is conserved measured by qPCR and found that PAN Mu contained 50% more
between the two cell lines (Fig. 6b, heatmap). The frequency of KSHV episomes relative to WT in the iSLK cells (Fig. 6d). Effects
induced loops detected as significant in BCBL-1 TREx-K-Rta cells of K-Rta binding at the PAN promoter on genomic loop for-
(Table 2, part B) did not attain statistical significance in iSLK/ mation was then examined with 3C analyses and the frequencies
r.219 cells (Table 2, part D); this may be attributed to the lower of induced genomic loops in PAN Mu and WT were compared.
proportion of fully reactivating cells in the population of the The results indicated that K-Rta recruitment to the PAN pro-
iSLK/r.219 cells. However, these Capture Hi–C results were moter is important for bringing other genomic loci into proximity
indeed verified by qPCR analysis (Supplementary Fig. 4b). Next, of the PAN promoter region; however, BamHI fragments that do
we examined the relationship between K-Rta binding and not possess K-Rta binding sites were also brought into proximity
induced genomic loops. Based on the Capture Hi–C and K-Rta of PAN promoter region in a K-Rta PAN RE dependent manner

6 NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02089-9 ARTICLE
PAN RNA
K2 2 K3 70 K4 K5 K6 17 1920 23 24 29b 29a 39 42 43 45 46 47 4849 52 53 55 K9 K10 K10.5 K11 58 59 60 61 62 65 66 67 K12 K13 72 73 75 K15 TR

27 30 32 35 37
TR K1 4 6 7 8 9 10 11 K716 18 21 22 25 26 28 31 33 34 36 38 40 41 44 50 K8 K8.1 54 56 57 63 64 68 69 T0.7 K14 74

0 10 20 30 40 50 60 70 80 90 100 110 120 130 140 (kb)


K-Rta recruitment

IE & E gene cluster


sites

Late gene cluster Late gene cluster Latent genes


E rse
in

ge cti
te

ne on
s s

55-K12
K8-K12 Value
50-K12 75

37/40-K12 50

25
IE ter

27/30-K12
& sec
in

E ti

0
ge on
ne s
s

–25
17-K12

K3/K4-K12

Putative function
DNA replication Tegument proteins Modification proteins

Transcription Capsid proteins Transactivators/regulators

Exonuclease Packaging Unknown/viral homologs

Glycoproteins Cellular homologs Constitutive

Fig. 3 Dynamics of KSHV genomic loop formation. Total sequence reads in 3 kb intervals were counted and divided by the number of DpnII sites within the
3 kb-sequence. Sequence read count per DpnII site was then divided by total sequence reads containing KSHV genome to obtain relative number of
genomic loops at the respective genomic region. Relative increase/decrease of genomic links by induction of K-Rta was then obtained by subtracting the
relative sequence reads of non-reactivated TREx-K-Rta BCBL-1 cells from those of reactivated TREx-K-Rta BCBL-1 cells, and visualized by heatmap by using
R-program. The ChIP-seq results presented in Fig. 1b is superimposed above the heatmap to visualize the relationship between loop dynamics and K-Rta
recruitment sites. Genomic clusters representing IE, E, and late genes are marked. Loci where genomic loops that increased during reactivation are marked
by block borders, and increased genomic loops with the fragment containing K12 K-Rta RE are also identified with relevance to the Fig. 4

(Fig. 6e, #5/#22). The results imply that activation of promoters is formed from direct binding sites. We have been searching for a
important for looping and that K-Rta is not required to be suitable tissue culture system in which KSHV replicates to mea-
simultaneously bound to both fragments to form a genomic loop. surable degrees after de novo infection. To the end, we could
Genomic loops formed between non-K-Rta enriched regions were demonstrate that lytic infection was robust in primary human
also examined by qPCR (Fig. 6c, f, g. Two classes of contacts were gingival epithelial cells (HGEP) grown in trans-well culture with
observed in the non-K-Rta enriched loci: (i) contacts that serum free media (Supplementary Fig. 5). Encapsidated viral
appeared to be similar to K-Rta-enriched regions which were genomic copy numbers were measured by qPCR and the DNA
responsive to K-Rta induction, and were greatly reduced in PAN copy number was used to adjust for the de novo infection study.
Mu cells (Fig. 6c, f), and (ii) non-K-Rta-enriched regions that We infected primary HGEP cells with the maximal amounts of
were relatively non-responsive to K-Rta induction. This second PAN Mu virus that was feasible to concentrate (M.O.I. of 50 viral
type of interaction was detected at a similar level in both WT and copies/cell) and compared this with an equivalent amount of
PAN Mu cell lines (Fig. 6c, g), and showed no significant changes BAC16 WT virus. The results showed very weak eGFP signals in
in gene expression at the genomic region by PAN Mu (Fig. 4d). PAN Mu-infected cells (Fig. 7a). In contrast, cells receiving the
Even though PAN-Mu iSLK/r.219 cells harbor 50% more KSHV WT virus were essentially ~ 100% positive for eGFP expression,
episomes, the relative amount of pre-existing genomic loops in and the signal intensity of infected cells was significantly brighter
PAN Mu was lower than in WT, especially where gene expression than that from PAN Mu-infected HGEP. KSHV gene expression
by K-Rta induction was diminished (Figs. 4d and 6c). These array analysis further confirmed that viral gene expression was
results indicate that PAN promoter activation at the initial burst diminished with PAN promoter mutation, at levels which were
of transcription may be important to prepare latent chromatin from ~104 to 106-fold less than WT BAC16 (Fig. 7b). Although it
structures for reactivation. is currently not clear that the defect is due to lack of infectivity or
impaired gene expression, the results clearly demonstrate the
Viral ACH and de novo infection. KSHV reactivation initiated significance of activity of a viral ACH for the KSHV lytic
by supplementation of exogenous K-Rta in KSHV/Mu-infected replication.
iSLK cells allowed us to examine effects on specific viral promoter
activation on the KSHV gene expression program. However, this Discussion
masks the effects of initial activation of endogenous K-Rta Chromosomal loop formation between distal enhancers, silencers,
expression by cellular factors and pre-deposited genomic loops and core promoters has emerged as a common feature among

NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications 7


ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02089-9

a K12 K13
b c PAN RNA
WT
KSHV BAC16 1.00E+01

wild-type (WT) iSLK BAC 16 WT iSLK BAC 16 PAN Mu 7.50E+00


PAN Mu
K2 K5 ORF17 ORF24
2.00E-01 6.00E-02 8.00E-02 1.00E-03 K12 Mu
5.00E+00
Dox 0 24 48 72 (h) Dox 0 24 48 72 (h) 1.50E-01 4.00E-02
2.50E+00
6.00E-02 7.50E-04
1.00E-01 4.00E-02 5.00E-04 DKO
K12/K-Rta (kDa) (kDa) 2.00E-02
2.00E-02 2.50E-04
5.00E-02
interaction site 170 170 0 24 48 72
0 24 48 72
0 24 48 72
0 24 48 72 0 24 48 72
··· TGTAGGGGTTAGCCAC···
119,278 LANA LANA
119,293
K2 2 K3 70 K4 ori-Lyt K5 K6 PAN RNA 17 19 20 23 24 29b 29a 39 42 43 45 46 47
130 130
C G A TR K1 4 6 7 8 9 10 11 K7 16 18 21 22 25 26 27 28 30 31 32 33 34 35 36 37 38 40 41 44

Two-step 100 100


Red recombination K-Rta 0 10 20 30 40 50 60 70
K-Rta
ORF6 ORF8 ORF21 ORF25 ORF28 ORF31
KanR 6.00E-03 4.00E-03 2.00E-03 2.00E-03 8.00E-04 6.00E-04

I-SceI 40
GAPDH 40 GAPDH 4.00E-03 3.00E-03 1.50E-03 1.50E-03 6.00E-04 4.00E-04
2.00E-03 1.00E-03 1.00E-03 4.00E-04
2.00E-03 2.00E-04
1.00E-03 5.00E-04 5.00E-04 2.00E-04
Two-step
Red recombination 0 24 48 72 0 24 48 72 0 24 48 72 0 24 48 72 0 24 48 72 0 24 48 72

K12/K-Rta K12
4.00E-01
interaction site
mutant (K12 Mu) iSLK BAC 16 K12 Mu iSLK BAC 16 DKO ORF52 K9 ORF58 ORF61
3.00E-01
K13 ORF72 ORF73
2.50E-02 4.00E-03 8.00E-02 5.00E-03 6.00E-05 6.00E-03 6.00E-03
···TGTCGGGGGTAGACAC··· Dox 0 24 48 72 (h) Dox 0 24 48 72 (h) 1.88E-02 3.00E-03 6.00E-02 3.75E-03
2.00E-01
4.00E-05 4.00E-03 4.00E-03
119,278 119,293
1.25E-02 2.00E-03 4.00E-02 2.50E-03 1.00E-01
(kDa) (kDa) 6.25E-03 1.00E-03 2.00E-02 1.25E-03
2.00E-05 2.00E-03 2.00E-03

170 170 0 24 48 72 0 24 48 72 0 24 48 72 0 24 48 72 0 24 48 72 0 24 48 72 0 24 48 72 0 24 48 72

LANA LANA 48 49 5253 55 K9 K10 K10.5 K11 58 59 60 61 62 65 66 67 K12 K13 72 73 75 K15 TR


K6 K7/PAN
KSHV BAC16 130 130
50 K8 K8.1 54 56 57 63 64 68 69 T0.7 K14 74
wild-type (WT)
100 100 70 80 90 100 110 120 130 140 kb
K-Rta K-Rta ORF50/K-Rta K8 ORF57 ORF69
5.00E-02 2.00E-02 5.00E-02 4.00E-02
PAN/K-Rta Putative function
3.75E-02 1.50E-02 3.75E-02 3.00E-02
interaction site AATGGGTGGCTAACCTGTCCAAA·· GAPDH GAPDH 2.50E-02 1.00E-02 2.50E-02
DNA replication Tegument proteins Modification proteins
2.00E-02
40 40 1.25E-02 5.00E-03 1.25E-02 Transcription Capsid proteins Transactivators/ regulators 1.00E-02

PAN/K-Rta 0 24 48 72 0 24 48 72 0 24 48 72 Exonuclease Packaging Unknown/viral homologs 0 24 48 72


interaction site AATGGGTGTCTACCCGTGCCAAA·· Glycoproteins Cellular homologs Constitutive
28,547 28,568
mutant (PAN Mu)

d K-Rta-RE responsive map (Red: PAN RE responsive, Blue: K12 RE responsive, Purple: Both) e f
K2 2 K3 70 K4 ori-Lyt K5 K6 PAN RNA17 19 20 23 24 29b 29a 39 42 43 45 4647 48 49 52 53 55 K9 K10 K10.5 K11 58 59 60 61 62 65 66 67 K12 K13 72 73 75 K15 2.50E-02 1.60E+08
K1 4 6 7 8 9 10 11 K7 16 18 21 22 25 26 27 28303132 33 34 3536 37 38 40 41 44 50 K8 K8.1 54 56 57 63 64 68 69 T0.7 K14 74

0 10 20 30 40 50 60 70 80 90 100 110 120 130 138 kb

24 hr 48 hr 72 hr
2.00E-02
1 1.20E+08

Viral DNA copy number/ml


MNE (PAN Mu/WT)

1.50E-02
0.5

MNE
8.00E+07
0 1.00E-02
ori-Lyt
K1
ORF4
ORF6
ORF7
ORF8
ORF9
ORF10
ORF11
K2
ORF2
K3

K6
ORF70
K4

K5

K7
ORF16
ORF17
ORF18
ORF19
ORF20
ORF21
ORF22
ORF23
ORF24
ORF25
ORF26
ORF27
ORF28
ORF29

ORF33
ORF34
ORF35
ORF36
ORF37
ORF38
ORF39
ORF40
ORF41
ORF42
PAN RNA

ORF30
ORF31
ORF32

ORF43
ORF44
ORF45
ORF46
ORF47
ORF48
ORF49
ORF50
K8
K8.1
ORF52
ORF53
ORF54
ORF55
ORF56
ORF57
K9
K10
K 10.5
K11
ORF58
ORF59
ORF60
ORF61
ORF62
ORF63
ORF64
K12
ORF66
ORF67
ORF68
ORF69
K13
ORF72
ORF73
K14
ORF74
ORF75
K15

1
MNE (K12 Mu/WT)

5.00E-03 4.00E+07

0.5

0.00E+00
0h
24 h
48 h
72 h

0h
24 h
48 h
72 h

0h
24 h
48 h
72 h

0h
24 h
48 h
72 h
0.00E+00
0 WT PAN K12 DKO
K1

ori-Lyt
ORF4
ORF6
ORF7
ORF8
ORF9
ORF10
ORF11
K2
ORF2
K3
ORF70
K4

K5
K6

K7
ORF16
ORF17

ORF19
ORF20
ORF21
ORF22
ORF23
ORF24
ORF25

ORF39
ORF40
ORF41
ORF42

K9

ORF74
PAN RNA

ORF18

ORF26
ORF27
ORF28
ORF29
ORF30
ORF31
ORF32
ORF33
ORF34
ORF35
ORF36
ORF37
ORF38

ORF43
ORF44
ORF45
ORF46
ORF47
ORF48
ORF49
ORF50
K8
K8.1
ORF52
ORF53
ORF54
ORF55
ORF56
ORF57

K10
K 10.5
K11
ORF58
ORF59
ORF60
ORF61
ORF62
ORF63
ORF64
K12
ORF66
ORF67
ORF68
ORF69
K13
ORF72
ORF73
K14

ORF75
K15

WT PAN Mu K12 Mu DKO Mu Mu

Fig. 4 A K-Rta direct response element (RE) essential to coordinating regulation of KSHV gene clusters and reactivation. a Bacmid construction. Scheme of
two-step red recombination for the construction of the KSHV K-Rta RE mutants used in this study: PAN Mu, K12 Mu, and PAN + K12 DKO. b Immunoblot
analysis for KSHV gene products in iSLK BAC16 cell lines containing wild type and mutant KSHV bacmid clones. c Time course of KSHV gene expression in
iSLK BAC16 cell lines. A map of the KSHV ORFs and their putative functions is shown. RT-qPCR expression analysis was performed for the indicated ORFs
in WT (blue diamond) and PAN Mu (red circle), K12 Mu (purple triangle), and DKO (brown square) mutant lines. Values are mean normalized expression
(MNE) using ACTB as reference. PAN RNA and K12 expression is presented as insets. Complete KSHV gene expression signatures are presented in
Supplementary Fig. 3. d Mapping of genomic domains by viral gene expression. Normalized viral gene expression in each mutant KSHV (PAN Mu, K12 Mu)
was compared with that of wild type at each time point to reveal dependency on K-Rta direct binding to the PAN RE (middle panel) and K12 RE (bottom
panel). A gene exhibiting >50% reduction in expression at all time points (24 h, blue; 48 h, magenta; 72 h, green) during reactivation was considered as
responsive. Values represent MNE relative to BAC16 WT (1 = unchanged). The upper panel summarizes genes regulated by PAN RE (red), K12 (blue), or
both (purple). Genes unaffected by the effects of the mutations are marked in gray. Gene expression not evaluated was marked in black (ORF65). e
Endogenous K-Rta gene expression. Endogenous K-Rta expression in iSLK BAC16 lines was measured using a gene-specific primer for reverse transcription,
followed by qPCR. Values represent MNE±SD (n = 3). All 72 h means differ significantly (p < 0.01; one-way ANOVA with Tukey’s post-hoc HSD test). f
KSHV replication. Encapsidated KSHV DNA in culture supernatant 5 days post-reactivation was measured by qPCR. Values represent copy number/ml
(mean±SD, n = 3). All means differ significantly in pairwise comparisons (p < 0.01; one-way ANOVA with Tukey’s post-hoc HSD test), except PAN Mu vs.
DKO

genomes of different organisms38–40. Our studies have implicated suggest that among cells harboring latent KSHV, a non-uniform
KSHV chromosomal looping as a mechanism to promote the population of viral episomes exist that may yield different outputs
KSHV latent-to-lytic switch. Previous studies by the Lieberman following the receipt of a reactivation signal. Our recent study
group have identified CTCF-cohesin complex-mediated KSHV confirmed this idea experimentally using RNA-FISH approaches
genomic looping and its role in latency and reactivation18,23,41,42. to study KSHV transcription in situ, while examining KSHV gene
We report here on new KSHV genomic contacts that may (a) expression in the 3D nuclear space of an infected cell28. A high
prepare the latent genome for direct K-Rta binding events and (b) degree of heterogeneity in the response of individual KSHV
facilitate the formation of inducible genomic loops that are episomes to reactivation stimuli within a single cell was observed,
associated with distal promoter activation (Fig. 8). and not all episomes in a reactivating cell express viral RNA.
The intricate nature of KSHV reactivation is exemplified by However, those episomes responding to stimulation appeared to
previous findings uncovered when this event is quantified on a recruit molecules that formed large “transcription factories”28.
single-cell basis; K-Rta-mediated reactivation appears highly These outcomes are analogous to the emerging picture of
stochastic with a variety of outcomes observed. Cells that are eukaryotic gene expression when viewed on a single-cell basis;
K-Rta positive undergo one of several fates, including complete expression occurs in bursts, is inherently noisy, and is exemplified
reactivation, expression of a subset of delay-early genes, or by highly stochastic expression levels (reviewed in refs. 45,46). The
abortive reactivation43,44. Only a small percentage of K-Rta- underlying causes of this variation are still not entirely clear.
positive cells also express late proteins such as K8.144. The studies Although our 3C results were obtained from cell populations,

8 NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02089-9 ARTICLE

A
N u
ct u

N
PA M
Ve M

R
or

+ AN
+ AN
a b

T
P
Color key

W
PAN RNA
K-Rta recruitment at PAN RE K-Rta recruitment at K12 RE K7
K12
ORF50
ORF57
ORF11
40 250 ORF73
ORF69
–10 –6 –2 2 ORF72
ORF40
ORF56
ORF7
ORF9
K1
1.0E+05 ORF10
ORF6
K10
200 ORF4
ORF8
K8
30 ORF61
ORF62
ORF66
1.0E+04 ORF68
ORF25
ORF75
Fold change

Fold change
K9
ORF53
150 ORF44
K10.5
ORF46
ORF63

20 1.0E+03 K11
ORF64

MNE
ORF74
K14
ORF58
K3
100 K4
K2
K5
1.0E+02 ORF38
ORF17
K8.1
ORF22
ORF52
10 K6

* 50
ORF37
ORF70
ORF26

* 1.0E+01 ORF29
ORF45
ORF43

* * ORF31
ORF27
ORF33
ORF16
ORF35
ORF28

0 0 1.0E+00 ORF55
ORF34
ORF36
ORF39
ORF21

PAN Mu
+ Vec
+ Dox
PAN Mu
+ PAN RNA
+ Dox

i SLK WT
+ Dox
TA

TA

TA

TA

TA

TA
α- G

α- G

α- G

α- G

α- G

α- G
ORF18
lg

lg

lg

lg

lg

lg
ORF32
R

R
K13
ORF67
ORF48
ORF30
ORF41
ORF47
WT PAN Mu K12 Mu WT PAN Mu K12 Mu ORF49
ORF42
ORF24
ORF59
ORF60
ORF54

Fig. 5 Lytic gene expression involves clustering of RE sites and requires PAN RNA in cis. a Cross-talk between the PAN RE and K12 K-Rta RE iSLK cell lines
harboring BAC16WT, PAN Mu, or K12 Mu bacmids were left uninduced or induced with dox for 8 h. Cells were fixed and K-Rta ChIP was performed.
Purified ChIP DNA was quantified by qPCR using primers that bracket the PAN RE (left panel) and the K12 RE (right panel) Values represent the mean±SD
of three replicates. Fold change is shown relative to the ChIP/input DNA signal in the uninduced cells. One K-Rta RE mutation impaired recruitment of K-
Rta to the other RE. All α-RTA means differ significantly in pairwise comparisons to WT (p < 0.01; one-way ANOVA with Tukey’s post-hoc HSD test). Fold
changes between PAN Mu vs. K12 Mu α-RTA means are non-significant. b PAN RNA complementation of PAN RE mutant does not rescue defective KSHV
gene expression. (Left panel) PAN RNA expression. The expression of PAN RNA with/without complementation was quantitated by RT-qPCR and
compared to the expression of BAC16 WT. MNEs are derived from RNA harvested 48 h after dox-induction. Values represent MNE±SD (n = 3). ACTB
expression was used as the reference. (Right panel) KSHV PCR microarray. Individual viral gene expression was examined by KSHV PCR array and
visualized as a heatmap. The heatmap was generated using Version 3.2.2 of R software with heatmap.3 package

individual episomes may vary in their looping architecture, as is several loci examined (Supplementary Fig. 6); this is also con-
the case with mammalian genomes47, and loop diversity may sistent with the fact that PAA usually does not down-modulate IE
contribute to variable reactivation scenarios described above. In and E gene expression. Thus, the looping formation changes
this regard, we hypothesize that correct genomic KSHV loop appear to be more than simply a reflection of genomic con-
formation and subsequent direct K-Rta binding events are formation changes induced by viral DNA replication, although a
additional factors to spur a subset of K-Rta-expressing cells complete Hi–C analysis will be needed to fully resolve this
and/or episomes towards full reactivation. Although the question.
regulation of loop formation is unclear, examples in support By performing capture Hi–C analyses with two different cell
of cells containing either preformed (hardwired) loops48 or lines, TREx-K-Rta-BCBL-1 and iSLK/r.219 cells, we noticed that
induced loop formation49,50 have been reported. The loops we there is a difference in the hardwired loop formation between
detected in KSHV-positive cells during latency suggest one class these two cell lines. A large number of genomic loops were found
of KSHV genomic loops are hardwired. The observed induction at the K9-K10 loci (BamHI position #18) in BCBL-1 cells (Fig. 2a
of genomic links upon K-Rta expression also indicates the and Supplementary Fig. 2), but not in iSLK/r.219 cells (Fig. 6b).
presence of inducible loop formation. Our study with mutant The KSHV K10.5 (LANA2) transcript has been shown to be
KSHV further suggests that activation of specific promoters, and expressed in KSHV-infected hematopoietic cells but not KS
presumably recruitment of RNA polymerase II complex, is lesions51. Accordingly, KSHV may utilize additional ACHs in PEL
important for inducible genomic loop formation (Fig. 6c). cells to take advantage of transcriptionally active sites for effective
Although it is mostly speculative at this point, inducible viral reactivation, which may be established by active transcription
genomic loops formed during the initial burst of lytic gene at the corresponding genomic regions. We noticed that “poised”
expression following de novo infection may dictate the 3D RNA polymerase II binding sites on the KSHV genome52 sig-
genomic structure as latency is subsequently established. This nificantly overlapped with the major K-Rta direct binding sites
might explain why PAN Mu infected cells showed a lower during reactivation. These results suggested that the KSHV gen-
number of genomic loops during latency, resulting in impaired ome is “poised” to be activated and stimulation of the K6-K7/PAN
gene expression during reactivation. It will be interesting to and/or K12/T0.7 regions through direct binding of the potent
examine if manipulation of the viral gene expression program for transactivator may lead to other viral promoter activation by
lytic replication results in a different landscape of epigenetic releasing RNA polymerase II to traverse the KSHV genome.
marks during latency and leads to a different 3D genomic To gain insight into how K-Rta binding to the KSHV genome
structure of latent chromatin. leads to changes in chromosome conformation, we examined the
In our studies, cells were fixed at 24 h post-reactivation in an relationship between genomic K-Rta binding sites (Fig. 1b) and
attempt to minimize the contribution of viral DNA replication to previously published CTCF binding profiles42. The CTCF and
contact formation. We also performed analyses using a 3C library cohesion complexes have been previously implicated in KSHV
prepared from cells reactivated in the presence of the viral DNA genomic looping18,23. Alignment of the profiles shows a partial
polymerase inhibitor, phosphonoacetic acid (PAA). The results overlap between CTCF and K-Rta enrichment peaks at regions
showed there were little changes in the contact frequency of encompassing PAN, K-Rta and K12 regulatory regions. This

NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications 9


ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02089-9

result suggests that K-Rta is recruited to regions participating in knockdown (KD) of CTCF (Supplementary Fig. 8b) had a dele-
genomic loop formation, and K-Rta may bring other enzymes to terious effect on the degree of K-Rta recruitment to ~ 90% of the
these critical regions and alter the KSHV genome structure peaks listed in Supplementary Table 1. Rad21 KD (Supplemen-
(Supplementary Fig. 7a, b). Consistent with this, partial tary Fig. 8a) also had a weak, but opposing, effect on K-Rta

a b

ORF4
ORF7

ORF6
K15

TR

K1
OR
BamHI position

F7
5
F74

OR

F8
K1

OR
OR

F9
F7
1 2 3 4 5 6 7 8 9 10 1112 13 14 15 16 17 18 19 20 21 22 23 24 25 26 2728 29
i SLK/r.219

OR
3
OR K1

10
BamHI (1)
(29) BamH
(28) Ba HI
F7 3
1

(27)

RF
(26) Bam HI

11
2

O
(25)
Dox – +

RF
Bam
2

Bam HI

O
mHI
B
(24
2
K2 RF

am

I
HI
)B
O 3
(kDa) K1

am
(2
2/T K3 Value

3)
7 0

HI
0.7
4

Ba
OR
ORF

m
F6

HI
9
OR K4 4.1 5 6
K-Rta OR
F
OR 67A
F6
8
m
HI
(2
)
K 4.2
K
t 6
100 OR
F6
F66
7 Ba
I (3
) ori
-Ly
7
OR mH 4
F65 Ba I K5
mH I 8
37 BaamHHI (4)
B am I
B amH I
K6
9
K-bZIP (22)
Bam
HI
B amH I
B amH (5)
B HI
K7 N
PA 10
2
ORF64
Bam 6
ORF1
ORF17 .5 11
ORF17 0

BamHI position
(21) Bam
HI 12
ORF18
β-actin (20) BamHI
BamHI (6) ORF19
13
ORF20 14
37 ORF63 BamHI (7) ORF21 15
(19)Bam
HI
ORF62
16
ORF22
17
1
ORF6 ORF2
3 18
60
ORF ORF2
4
19
9
ORF5 B 20
F58 Ba amH
OR mH I(8
Ba
I ) OR
F2
21
1 5
K1
m
HI
(9
22
I
H

) OR
23
m

OR F26
Ba

0.5 O F2
8)

Ba
K1 24
(1

OR RF2 7
HI

mH
Ba
am

F 8
O OR 29

I
mH
25

(1
HI

Bam
)B

0 RF F3

0)
K1
Bam

HI

BamHI
(17

I
BamHI (14)

31 0

(11

O
26
(15) Bam

HI(1

R
)

F3 3
(16)

O RF
O

2
K9

RF 29
2)
27
(13)

3 a
OR RF3
7

O F36
F5

F3 5
OR
28
OR

4
6

OR F38
F5

OR 39
5

OR
29

F3
OR

F5
OR F54

OR
OR F53

7
F
F52
OR

ORF4 2
ORF4
K8.1
K8
OR

ORF4

F40
ORF50

ORF44
ORF49

ORF48

ORF46
ORF45
ORF47

1
3

c
86

48
,8

,4

d
26

33

B B Relative intracellular
PAN RE
(28,560)
KSHV DNA copies (WT = 1)
20 kb 40 kb 60 kb
137 kb
80 kb 100 kb 120 kb
vDNA Copy/GAPDH2

#2 #4 #12 #14
PAN Mu
#5 1.5
WT
#7 #10 #22 #23 #25 #27 1

0.5

e Between #2 and #5 Between #4 and #5 Between #5 and #12 Between #5 and #14 Between #5 and #22
WT PAN Mu WT PAN Mu WT PAN Mu WT PAN Mu WT PAN Mu
– + – + – + – + – + – + – + – + – + – +
4.0E-02 6.0E-01 1.0E-01 4.0E-02 3.0E-02
Relative to Bac16

Relative to Bac16
Relative to Bac16

Relative to Bac16

Relative to Bac16

8.0E-02
3.0E-02 3.0E-02
4.0E-01 2.0E-02
6.0E-02
2.0E-02 2.0E-02
4.0E-02
2.0E-01 1.0E-02
1.0E-02 1.0E-02
2.0E-02

0.0E+00 0.0E+00 0.0E+00 0.0E+00 0.0E+00

f Between Between Between g Between Between Between


#7 and #10 #7 and #22 #7 and #23 #22 and #23 #22 and #27 #22 and #25
WT PAN Mu WT PAN Mu WT PAN Mu WT PAN Mu WT PAN Mu WT PAN Mu
5.0E-02 3.0E+00 1.0E+01
– + – + – + – + – + – + – + – + – + – + – + – +
Relative to Bac16
Relative to Bac16
Relative to Bac16

4.0E-02 7.5E+00
2.0E+00
3.0E-02
5.0E+00
2.0E-02
1.0E+00
2.5E+00
1.0E-02

0.0E+00 0.0E+00 0.0E+00

Fig. 6 KSHV genomic loop formation in KSHV infected iSLK cells. a KSHV lytic protein expression. Immunoblotting analysis of K-Rta or K-bZIP expression
in iSLK/r.219 cells during latency (−) and at 24 h post-dox treatment (+). b (Left panel) Circos diagram depicting KSHV genomic links detected by Capture
Hi–C. Arcs indicate genomic links before (blue) or after (red) K-Rta induction in iSLK/r.219 cells. Black outer hatches mark genomic positions and ORF
names are indicated in the outer circle. Position of BamHI sites are indicated and numbered. (Right panel) Movement of genomic links during KSHV
reactivation in iSLK cells. Increase/decrease of KSHV genomic loops was calculated similarly to that described in Fig. 2, and visualized by heatmap by using
R package ggplot2. c Effect of K-Rta binding to PAN RE on loop formation. K-Rta ChIP seq plot and interaction sites are shown. The BamHI fragment
encompassing PAN is shown with anchor primers depicted. Target sites are indicated with red (K-Rta enriched) or blue (K-Rta valley) arrowheads.
Arrowhead numbers refer to the BamHI sites listed in the Circos plot in Fig. 6b. d Relative intracellular KSHV copy number WT = 1. Values were determined
by qPCR on isolated total DNA from each latent cell line. e Real-time PCR analysis of 3C ligation products using anchor primers located on the BamHI
fragment containing PAN K-Rta RE (#5) with BamHI acceptor primers as listed. PCR products were quantified relative to products from a BAC16 control
BamHI re-ligation matrix. qPCR results using a forward anchor/acceptor primer pair18 are shown. Non-K-Rta enriched regions. f Induced contacts. Real-time
PCR analysis of 3C ligation products (±dox 24 h) using an anchor primer located on the BamHI fragment (#7) with BamHI acceptor primers listed (#10, nt
36,700; #22, nt 111,299; #23, nt 123,970) and PCR products were quantified relative to products from a BAC16 control BamHI re-ligation matrix. g
Constitutive contacts. Real-time PCR analysis of 3C ligation products using an anchor primer located on the BamHI fragment (#22) with BamHI acceptor
primers listed (#23, nt 123,970; #27 nt 134,049, #25 nt 129,537). Values (mean±SD, n = 3) represent signals obtained relative to a BAC16 BamHI random
ligation matrix

10 NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02089-9 ARTICLE

a BAC WT 72 hr post infection b


4

KSHV gene expression [MNE (log10)]


2

–2
BAC PAN Mu 72 hr post infection
–4

–6

–8
PAN Mu WT

Fig. 7 Effects on de novo infection. a Primary HGEP cells were cultured in trans-well and infected with concentrated viral supernatants from BAC16 WT or
PAN Mu cultures at 50 viral DNA copies/cell. eGFP expression was captured 72 h post-infection. Phase contrast (left panels) and eGFP (right panels) are
shown. Exposure time for image capture for eGFP was set to 3.11 s. Scale = 100 μm. b KSHV gene expression 72 h post-infection. KSHV gene expression
was measured by KSHV PCR arrays and results were summarized as a box plot. Each point in the plot represents a KSHV ORF (PAN Mu, blue circles; WT,
red squares)

binding, which exhibited modestly increased K-Rta occupancy at and the primer pair sSalI-RTA(pENTR), CAT GTC GAC ATG GCG CAA GAT
~ 30% of the binding sites. A recent report has suggested that GAC AAG GG and asRTA-EcoRI(pENTR), CTG AGA ATT CTC AGT CTC GGA
AGT AAT TAC GCC ATT G. To construct pCW57.1-KSHV K-Rta/ORF50, the
cleavage of Rad21 by cellular factors plays a role in KSHV KSHV K-Rta/ORF50 fragment was transferred from pENTR4-KSHV K-Rta/
chromosome conformational changes that accompany lytic ORF50 to the Tet-ON lentiviral vector pCW57.1 (Addgene plasmid # 41393), by
reactivation, perhaps through opening of the cohesion ring53. using the GATEWAY system (Invitrogen, CA, USA).
Together, these results suggest that CTCF and genomic hubs
prepared by the cohesin complex may have a role in K-Rta Mutagenesis of KSHV BAC16. The KSHV bacmid BAC16 was kindly provided by
chromatin recognition. However, how K-Rta chromatin binding Dr. Jae U. Jung (University of Southern California) and mutagenesis performed
and/or activation of transcription regulates genomic looping according to previously described methods35,36. Point mutations were introduced
at the K-Rta direct binding sites within the response elements of the PAN and K12
requires further investigation. promoters32,33.
In summary, we demonstrated here that the herpesvirus epi- The primer sequences for mutagenesis were as follows
somal genome and the latent-lytic infection switch represent a (S-K12 RE Mut: ggtccacgctcacctctggcgcggccccgggaaatgggtgTctaCccccGacat
unique and effective experimental tool to study spatial and aagcagtttgtcctacTAGGGATAACAGGGTAATCGATTT,
As-K12 RE Mut: cctgcgaagggggcgtaaccgtaggacaaactgcttatgtCggggGtagAcacc
temporal gene regulation. Demonstration of KSHV replication in catttcccggggccgcGCCAGTGTTACAACCAATTAACC
the HGEP cell model and the dynamic formation of viral ACH set S-PAN RE Mut: tgttaatgacataaaggggcgtggcttccaaaaatgggtgTctaCccGTGccaaaat
a new stage for future KSHV gene regulatory studies. Identifica- atgggaacactggTAGGGATAACAGGGTAATCGATTT
tion of regulatory mechanisms of viral ACHs should lead to As-PAN RE Mut: caagctggccccttttatctccagtgttcccatattttggCACggGtagAcaccca
therapeutic intervention for KSHV-mediated malignancies. tttttggaagccacGCCAGTGTTACAACCAATTAACC,
Lowercase letters indicate identical sequence to KSHV BAC16, underlined
uppercase indicates mutagenesis sites, and uppercase indicates the pEP-KanS
Methods sequence). Whole genome sequences of each BAC clone were confirmed by next-
Cells. iSLK cells54 were maintained in DMEM medium containing 10% fetal generation DNA sequencing as described below.
bovine serum (FBS; complete DMEM), 50 μg/ml G418, and 100 μg/ml hygromycin
B in the presence of 5% CO2. iSLK cells containing BAC16 WT or BAC16 mutants KSHV BAC16 sequencing. BAC16 DNA constructs were isolated from 2 l of LB
were cultured in complete DMEM containing 250 μg/ml G418, and 1000 μg/ml culture with the Qiagen MAXI kit (Qiagen). Genomic DNA preparations were
hygromycin and 1 μg/ml puromycin. TREx-(F3H3)-K-Rta BCBL-1 cells that quantified using the Qubit Fluorometer (Life Technologies) and sheared to
expresses Flag × 3 and HA × 3 tags at the N-terminal region of K-Rta were gen- ~ 180 bp using the Covaris E220 Focused-ultrasonicator (Covaris, Inc.). Sequencing
erated and cultured in complete RPMI 1640 containing 50 μg/ml blasticidin and libraries were prepared using the NEBNext DNA Library Prep Kit (New England
100 μg/ml hygromycin B. Vero (ATCC CCL-81) and iVero (dox-inducible K-Rta BioLabs, Inc.) and multiplexed sequenced (75 bp paired-end, 5 libraries/run) on the
expression) cells were cultured in the presence of 1 µg/ml puromycin. Human Illumina MiSeq platform (Illumina, Inc.) per manufacturer’s standard protocols.
gingival epithelial cells (HGEP), obtained from pooled donors, were purchased Raw sequence reads (FASTQ format) were aligned back to the BAC16 KSHV
from CellnTec (#HGEPp). The cells were cultured in CnT-prime epithelial cell reference (Human herpesvirus 8 strain JSC-1 clone BAC16, GQ994935.1) using
medium (CellnTec #CnT-PR). BWA-mem57 with default settings. Using the BAM alignment files as input,
SAMtools mpileup58 was then used to perform variant detection. Ultra-high
Virus production. iSLK cell lines containing each recombinant BAC16 were cul- coverage was obtained for most samples (~1200×) and allowed for comparative
tured in selection media in 150 mm culture dishes until 80% confluent. For reac- whole genome sequence analysis. The point mutations in each BAC16 were vali-
tivation, selection media was removed and cultures were re-fed with complete dated with unbiased variant calling (i.e., relative to the reference human herpes-
DMEM (w/o selection drugs) containing 1 mM sodium butyrate and 1 µg/ml virus 8 strain JSC-1 clone BAC16, GQ994935). The results also demonstrated a
doxycycline. The cultures were reactivated for 4 days. The cells were removed by 24,710 C > T variant originating from the parental BAC16, and an additional
centrifugation (180×g, 10 min) and the supernatants were passed through a 0.8-µm mutation within the K12 promoter region of BAC16 K12Mu (position 119,316 A
filter, and virus particles were concentrated by ultra-centrifugation at 25,000 rpm > G) (Supplementary Table 2).
(112,000×g), for 2 h at 4 °C (Beckman SW28 rotor). Viruses were re-suspended in
500 µl of DMEM along with the residual ~500 µl media in the centrifuge tube and
Generation of stable iVero cell lines. The lentivirus vector was produced by
stored at –80 °C until use. Viral genomic copy number in the medium was mea-
tripartite-transfection of pCMV-VSV-G-RSV-Rev, pCAG-HIVgp and pCW57.1-
sured by qPCR, as described previously55.
KSHVK-Rta/ORF50 into 293 T cells (ATCC CRL-3216)59,60. The packaging
plasmids (pCMV-VSV-G-RSV-Rev, pCAG-HIVgp) were a kind gift from and Dr.
Plasmids. To construct pENTR4-KSHV K-Rta/ORF50, K-Rta/ORF50 fragments Hiroyuki Miyoshi (RIKEN, Japan). To establish Vero cells with inducible expres-
were obtained by PCR and inserted into pENTR4 no ccDB (686-1) (Addgene sion of K-Rta/ORF50 (iVero cell), Vero cells were infected with the lentiviral
plasmid # 17424)56. The PCR utilized pCIneo-3xFLAG-ORF50/RTA as template particles, and transduced cells were selected and maintained by culture in growth

NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications 11


ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02089-9

K12 loci re-ligated under ultra-dilute conditions. DNA-protein crosslinks were reversed by
incubation at 65 °C overnight with 0.2% SDS and the DNA purified with the QIA-
quick PCR Purification Kit (Qiagen). DNA samples were submitted to the UC Davis
Comprehensive Cancer Center Genomics Shared Resource for library preparation,
target enrichment, and NGS. The DNA preparations were sheared to a mean frag-
PolII ment length of 250 bp using a Covaris E220 Focused-ultrasonicator (Covaris, Inc.)
K-Rta RE
and sequencing libraries were then prepared from the DNA chimerae using the
E NEBNext DNA Library Prep Kit (New England BioLabs, Inc.). Target enrichment for
K-Rta ta R
II K-R KSHV genomic content was performed using a custom-designed KSHV genomic
Pol PolII capture probe library (described above) and according to the manufacturer’s standard
r E
ote PolIIK-Rta RE -Rta R protocols (IDT, Coralville, Iowa). Briefly, in-solution hybrid capture was performed
rom K
ta p by hybridizing libraries (500 ng) with the KSHV genomic capture probe pool (3 pmol)
K-R
in a mixture containing xGen 1X Hybridization Buffer, Cot-1 (5 µg), and xGen
Universal Blocking Oligos at a temperature of 65 °C for 4 h. Hybridized targets were
then bound to streptavidin-coupled magnetic beads (Dynabeads M-270 Streptavidin
PAN loci beads; Thermo Fisher Scientific) by incubation at 65 °C for 45 min, and unbound
DNA was removed by a series of high-stringency (65 °C) and low-stringency (room
K-Rta expression further induces the formation of viral ACH temperature) washes. KSHV genome-enriched Capture Hi–C library DNA was eluted
by clustering K-Rta RE and post-capture PCR enrichment performed with high-fidelity KAPA HiFi HotStart
DNA Polymerase (Kapa Biosystems, Inc., Wilmington, MA) and purified with
Agencourt AMPure XP magnetic beads. KSHV-enriched libraries were then validated
with an Agilent 2100 Bioanalyzer, and quantitated with a Qubit fluorometer (Invi-
Increases K-Rta & RNA polymerase II local concentration
trogen) and by qPCR-based quantification (KAPA Library Quantification Kit).
Libraries were submitted for sequencing (100-bp paired-end) on an Illumina HiSeq
2500 sequencing system. Junction sites (BamHI or DpnII sites) were mapped and
Efficient lytic gene expression visualized with Circos. Because of the relatively small size of the KSHV genome,
exclusion of non-digested fragments with an additional capture step by the incor-
Fig. 8 Proposed model of KSHV ACH. Multiple genomic regions containing poration of biotin labeled nucleotide at the ends of successfully digested fragments
K-Rta response elements (REs), including the locate the K-Rta promoter was not performed. Consequently, we have had a relatively larger fraction of non-
itself as well as other K-Rta responsive promoters, are positioned in close digested KSHV genomes that might be inaccessible for digestion by restriction
proximity to each other via 3D KSHV genomic structure. This mechanism enzyme, BamHI. In addition, there were a number of BamHI containing fragments
that were unable to be mapped to BamHI sites within the short illumina sequence
facilitates K-Rta to activate promoters efficiently. Activation of “poised” reads.
RNA polymerase II at the K12 and PAN RNA loci then allows traveling to
other viral promoters that are brought to in proximity during reactivation.
Conversely, inhibition of a single ACH-associated promoter activation thus Capture Hi–C NGS data analysis. Paired-end sequence data (FASTQ format) was
processed to identify putative Capture Hi–C interaction products (di-tags) using
impairs multiple linearly distant viral gene expression
the Capture Hi–C User Pipeline (HiCUP) (Babraham Bioinformatics; http://www.
bioinformatics.babraham.ac.uk/projects/hicup/). By executing a series of Perl
medium containing 2.5 µg/mL of puromycin (InvivoGen). KSHV BAC16 wild-type scripts, this performed identification of the Hi–C ligation junctions within the
and mutants were transfected into iVero cells by the calcium-phosphate method, reads (i.e., BamHI or DpnII sites), read alignment, and removal of PCR duplicates
and stably transfected cells selected by culture in complete medium in the presence and common Capture Hi–C artifacts. Subsequently, read pairs derived from the
of 1,000 μg/mL of hygromycin B and 2.5 μg/mL of puromycin. interacting fragments were output in SAM format. Within HiCUP, Bowtie 261 was
used to align FASTQ data to the reference KSHV genome sequence (Human
herpesvirus 8 strain JSC-1 clone BAC16, GenBank: GQ994935.1) and an in silico
Generation of stable iSLK cells. The iSLK BAC16 lines were prepared by BamHI-digested reference genome. Hi–C output was then analyzed with HOMER
infection with viral supernatants produced in iVero cell lines described above. Each (Hypergeometric Optimization of Motif EnRichment; http://homer.salk.edu/
iVero BAC16 line was induced with 1 μg/ml doxycycline + 1 mM sodium butyrate homer/interactions/) to generate a Capture Hi–C tag directory and for identifica-
for 4 days. Supernatants were collected, concentrated, and used to infect iSLK cells. tion of significant interactions. Junction sites (BamHI or DpnII sites) were mapped
For infection, iSLK cells were seeded into a 6-well plate (2 × 105 cells/well), and and visualized with Circos plotting (described below). Frequencies of chromatin
viral supernatants were added to the cells on the following day. Two days later, the looping sites are presented as the number of reads obtained with 3C of KSHV-
transduced cells were trypsinized and transferred to 10-cm dishes and cultured in infected chromatin.
complete DMEM containing 1 µg/ml puromycin, 250 µg/ml Geneticin and 1,200
µg/ml hygromycin B.
Circos plotting. 3C sequence reads and genomic features of TREx-K-Rta BCBL-1
Chromosome conformation capture. The chromosome conformation capture and iSLK r.219 cells infected with KSHV were visualized by using Circos plotting34.
(3C) procedure and analysis was carried out using chromatin derived from BAC16- Circos software was selected for this analysis based on its ability to clearly display
WT or BAC-mutant infected iSLK cells. Latent or dox-induced iSLK cells were positional relationships among data derived from the KSHV genome. The Circos
fixed with formaldehyde, digested with restriction enzyme (BamHI), diluted, and software program and companion tools were downloaded from its website (http://
re-ligated. The primer design and protocol used has been described previously by circos.ca.) Sequence reads were mapped to the KSHV reference genome
Kang et al.18. A diagrammatic representation of the 3C PAN BamHI anchor pri- (NC_009333.1). The number of sequence tags that mapped to a given BamHI or
mers are shown in Fig. 6c. Interaction frequencies between two KSHV fragments DpnII fragment is indicated in the Circos display and represent the number of
are based on the qPCR Ct values obtained using 3C chromatin template and a unique interactions involving the fragment.
given anchor/acceptor primer pair relative to the value obtained using purified
BAC16 DNA subjected to the 3C procedure.
Data visualization and interpretation. The R package ver. 3.2.4 was used to
visualize genomic links and gene expression. Heatmaps of genomic links were
KSHV genomic capture probe library. A custom KSHV genomic capture probe
plotted with R package ggplot2 ver. 2.1.0. The gene expression profiles of iSLK-Wt
library using xGen Lockdown Probe technology (Integrated DNA Technologies,
and PAN-Mu cells presented in Fig. 5 were visualized with the R package Heat-
Inc.) was designed to target the entire KSHV genome sequence (human herpesvirus
map3 version 1.1.1. Those R packages were downloaded from the R Project website
8, NCBI RefSeq NC_009333.1) and manufactured by IDT. The library consists of a
(http://www.R-project.org).
pool of 1,152 individually synthesized, 5′-biotinylated, 120mer oligonucleotide
baits. A stock solution (0.75 µM) of the pool was prepared in TE (pH 8.0).
De novo infection. Human gingival epithelial precursor cells obtained from pooled
Capture Hi–C analysis. Long-range chromatin interactions within the KSHV gen- donors were purchased from CellnTec. The cells were cultured in epithelial
ome were determined in an unbiased, comprehensive manner with Capture Hi–C, by medium (CellnTec CnT-PR) as submerged cultures on polystyrene lifts in 12-well
utilizing 3C in conjunction with KSHV genome enrichment and next-generation plates. Cells were bathed with 800 µl media in the upper chamber of the lift and
sequencing (NGS)26. The chromatin prepared from KSHV-infected cells for the 3C with 1 ml medium in the lower well. When the cells were confluent, viral stocks
procedure described above was used as input for Capture Hi–C. Briefly, infected were added to the upper chamber and the infection was allowed to proceed for 24
TREx-K-Rta BCBL-1 or iSLK/r.219 cells (1 × 108) were fixed with formaldehyde, and h. Cells were monitored for eGFP expression and RNA was purified using RNeasy
the chromatin was digested with either BamHI or DpnII restriction enzyme, and then columns (Qiagen).

12 NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications


NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02089-9 ARTICLE

Chromatin immunoprecipitation assay. Chromatin immunoprecipitation assay 11. Rajarajan, P., Gil, S. E., Brennand, K. J. & Akbarian, S. Spatial genome
(ChIP) assays were performed, as described previously62. The antibodies used were organization and cognition. Nat. Rev. Neurosci. 17, 681–691 (2016).
rabbit anti-K-Rta IgG63, mouse anti-K-Rta monoclonal antibody (a gift from Dr. 12. Swanson, C. I., Evans, N. C. & Barolo, S. Structural rules and complex
Wood, University of Nebraska), anti-Flag M2 (Sigma F1804), and rabbit and regulatory circuitry constrain expression of a Notch- and EGFR-regulated eye
mouse control IgGs (Cell Signaling Technology #2729 and #5415). Immunopre- enhancer. Dev. Cell 18, 359–370 (2010).
cipitated chromatin DNA was analyzed by SYBR Green-based quantitative PCR 13. Thanos, D. & Maniatis, T. Virus induction of human IFN beta gene expression
(qPCR) (Bio-Rad) with primers for each RE as follows: PAN RE-S (agcttgaaggat- requires the assembly of an enhanceosome. Cell 83, 1091–1100 (1995).
gatgttaatg), PAN RE-As (gaagcggcagccaaggtgactg), K12 RE-S (ccaaga- 14. Buecker, C. & Wysocka, J. Enhancers as information integration hubs in
gatccgtcctccgtgcc), and K12 RE-As (agcgggatgctaggtccacgc). development: lessons from genomics. Trends Genet. 28, 276–284 (2012).
15. Vakoc, C. R. et al. Proximity among distant regulatory elements at the beta-
ChIP-sequencing. Briefly, chromatin DNA from 1 × 108 TREx- K-Rta BCBL- globin locus requires GATA-1 and FOG-1. Mol. Cell 17, 453–462 (2005).
1 cells were used per assay and immunoprecipitated with 20 µg mouse anti-FLAG 16. Heinz, S., Romanoski, C. E., Benner, C. & Glass, C. K. The selection and
M2 (Sigma-Aldrich, F1804) or mouse control IgG (Santa Cruz Biotechnology, function of cell type-specific enhancers. Nat. Rev. Mol. Cell Biol. 16, 144–154
sc2025). 1 ng ChIP-enriched or input DNA was used to generate Illumina- (2015).
compatible libraries with the KAPA LTP Library Preparation Kit (Kapa Biosys- 17. Ing-Simmons, E. et al. Spatial enhancer clustering and regulation of enhancer-
tems, KR0453) according to the manufacturer’s recommendations. Libraries were proximal genes by cohesin. Genome Res. 25, 504–513 (2015).
submitted for sequencing (50-bp single read) on an Illumina HiSeq 2500 sequen- 18. Kang, H., Wiedmer, A., Yuan, Y., Robertson, E. & Lieberman, P. M.
cing system. The ChIP-Seq data was aligned to the human hg19 reference genome Coordination of KSHV latent and lytic gene control by CTCF-cohesin
assembly and reference KSHV genome sequence (Human herpesvirus 8 strain JSC- mediated chromosome conformation. PLoS Pathog. 7, e1002140 (2011).
1 clone BAC16, GenBank: GQ994935.1) with Bowtie 261. Peak finding was per- 19. Kim, K. Y. et al. Kaposi’s sarcoma-associated herpesvirus (KSHV) latency-
formed with the MACS2 (Model-based Analysis of ChIP-Seq 2) program64 associated nuclear antigen regulates the KSHV epigenome by association with
according to the standard parameters described in the developer’s manual. We the histone demethylase KDM3A. J. Virol. 87, 6782–6793 (2013).
used the default settings with a minimum FDR (q-value) cutoff of 0.05. The peaks 20. Toth, Z. et al. LANA-mediated recruitment of host polycomb repressive
and reads alignments were visualized using the Integrative Genomics Viewer (IGV) complexes onto the KSHV genome during de novo infection. PLoS Pathog. 12,
genome browser from the Broad Institute.
e1005878 (2016).
21. Chang, P. C. et al. Histone demethylase JMJD2A regulates Kaposi’s sarcoma-
Western blot analysis. Cells were collected in RIPA buffer (50 mM Tris–HCl, pH associated herpesvirus replication and is targeted by a viral transcriptional
6.7, 1% NP-40, 0.25% sodium deoxycholate, 150 mM NaCl, 1 mM EDTA) sup- factor. J. Virol. 85, 3283–3293 (2011).
plemented with 1 mM PMSF and 1× protease inhibitor cocktail (Roche). Antibodies 22. Toth, Z., Brulois, K. & Jung, J. U. The chromatin landscape of Kaposi’s
used for immunoblotting were anti-K-Rta63, anti-LANA (Advanced Biotechnolo- sarcoma-associated herpesvirus. Viruses 5, 1346–1373 (2013).
gies, Inc. #13-210-100, 1:1000) and anti-GAPDH (Millipore ABS16, 1:2000). The 23. Stedman, W. et al. Cohesins localize with CTCF at the KSHV latency control
uncropped western blotting images are shown in Supplementary Fig. 9. region and at cellular c-myc and H19/Igf2 insulators. EMBO J. 27, 654–666
(2008).
RT-qPCR. iSLK BAC16 WT and BAC16 mutant cells were reactivated with dox- 24. Mesri, E. A., Cesarman, E. & Boshoff, C. Kaposi’s sarcoma and its associated
ycycline (1 μg/ml) and RNA was isolated from these cells 0, 24, 48, and 72 h post- herpesvirus. Nat. Rev. Cancer 10, 707–719 (2010).
induction using RNeasy columns (Qiagen). cDNAs were synthesized using 25. Aneja, K. K. & Yuan, Y. Reactivation and lytic replication of Kaposi’s sarcoma-
SuperScript III first-strand synthesis reagents (Invitrogen). Gene expression was associated herpesvirus: an update. Front. Microbiol. 8, 613 (2017).
analyzed by qPCR using specific primers designed by Fakhari and Dittmer65. 26. Hughes, J. R. et al. Analysis of hundreds of cis-regulatory landscapes at high
KSHV gene expression was normalized to the cellular ACTB signal. RNA harvested resolution in a single, high-throughput experiment. Nat. Genet. 46, 205–212
from PAN RNA transfected cells included a DNase I treatment step. (2014).
27. Nakamura, H. et al. Global changes in Kaposi’s sarcoma-associated virus gene
expression patterns following expression of a tetracycline-inducible Rta
Data availability. The data discussed in this publication have been deposited in
transactivator. J. Virol. 77, 4205–4220 (2003).
NCBI’s GEO Database under accession number GSE99950. The authors declare
28. Chen, C. P. et al. Kaposi’s sarcoma-associated herpesvirus hijacks RNA
that all other data supporting the findings of this study are available within the
article and its Supplementary Information files, or are available from the authors polymerase II to create a viral transcriptional factory. J. Virol. 91, e02491–16
upon request. (2017).
29. Sun, R. et al. A viral gene that activates lytic cycle expression of Kaposi’s
sarcoma-associated herpesvirus. Proc. Natl Acad. Sci. USA 95, 10866–10871
Received: 12 June 2017 Accepted: 3 November 2017 (1998).
30. Lukac, D. M., Renne, R., Kirshner, J. R. & Ganem, D. Reactivation of Kaposi’s
sarcoma-associated herpesvirus infection from latency by expression of the ORF
50 transactivator, a homolog of the EBV R protein. Virology 252, 304–312 (1998).
31. Ellison, T. J., Izumiya, Y., Izumiya, C., Luciw, P. A. & Kung, H. J. A
comprehensive analysis of recruitment and transactivation potential of K-Rta
References and K-bZIP during reactivation of Kaposi’s sarcoma-associated herpesvirus.
1. de Laat, W. & Grosveld, F. Spatial organization of gene expression: the active Virology 387, 76–88 (2009).
chromatin hub. Chromosome Res.: Int. J. Mol., Supramol. Evolut. Asp. 32. Chang, P. J. et al. Open reading frame 50 protein of Kaposi’s sarcoma-
Chromosome Biol. 11, 447–459 (2003). associated herpesvirus directly activates the viral PAN and K12 genes by
2. Palstra, R. J., de Laat, W. & Grosveld, F. Beta-globin regulation and long-range binding to related response elements. J. Virol. 76, 3168–3178 (2002).
interactions. Adv. Genet. 61, 107–142 (2008). 33. Song, M. J., Li, X., Brown, H. J. & Sun, R. Characterization of interactions
3. Misteli, T. Protein dynamics: implications for nuclear architecture and gene between RTA and the promoter of polyadenylated nuclear RNA in Kaposi’s
expression. Science 291, 843–847 (2001). sarcoma-associated herpesvirus/human herpesvirus 8. J. Virol. 76, 5000–5013
4. Hager, G. L., McNally, J. G. & Misteli, T. Transcription dynamics. Mol. Cell 35, (2002).
741–753 (2009). 34. Krzywinski, M. et al. Circos: an information aesthetic for comparative
5. Droge, P. & Muller-Hill, B. High local protein concentrations at promoters: genomics. Genome Res. 19, 1639–1645 (2009).
strategies in prokaryotic and eukaryotic cells. Bioessay: News Rev. Mol. Cell. 35. Tischer, B. K., Smith, G. A. & Osterrieder, N. En passant mutagenesis: a two
Dev. Biol. 23, 179–183 (2001). step markerless red recombination system. Methods Mol. Biol. 634, 421–430
6. Li, G. et al. Extensive promoter-centered chromatin interactions provide a (2010).
topological basis for transcription regulation. Cell 148, 84–98 (2012). 36. Brulois, K. F. et al. Construction and manipulation of a new Kaposi’s sarcoma-
7. Razin, S. V., Gavrilov, A. A., Ioudinkova, E. S. & Iarovaia, O. V. associated herpesvirus bacterial artificial chromosome clone. J. Virol. 86,
Communication of genome regulatory elements in a folded chromosome. FEBS 9708–9720 (2012).
Lett. 587, 1840–1847 (2013). 37. Kim, T. H. et al. Analysis of the vertebrate insulator protein CTCF-binding sites
8. Brown, J. D. et al. NF-kappaB directs dynamic super enhancer formation in in the human genome. Cell 128, 1231–1245 (2007).
inflammation and atherogenesis. Mol. Cell 56, 219–231 (2014). 38. Furlan-Magaril, M., Varnai, C., Nagano, T. & Fraser, P. 3D genome architecture
9. Banerji, J., Rusconi, S. & Schaffner, W. Expression of a beta-globin gene is from populations to single cells. Curr. Opin. Genet. Dev. 31, 36–41 (2015).
enhanced by remote SV40 DNA sequences. Cell 27, 299–308 (1981). 39. Cattoni, D. I., Valeri, A., Le Gall, A. & Nollmann, M. A matter of scale: how
10. Levine, M., Cattoglio, C. & Tjian, R. Looping back to leap forward: emerging technologies are redefining our view of chromosome architecture.
transcription enters a new era. Cell 157, 13–25 (2014). Trends Genet. 31, 454–464 (2015).

NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications 13


ARTICLE NATURE COMMUNICATIONS | DOI: 10.1038/s41467-017-02089-9

40. Guo, Y. et al. CRISPR inversion of CTCF sites alters genome topology and 63. Izumiya, Y. et al. Kaposi’s sarcoma-associated herpesvirus-encoded protein
enhancer/promoter function. Cell 162, 900–910 (2015). kinase and its interaction with K-bZIP. J. Virol. 81, 1072–1082 (2007).
41. Kang, H., Cho, H., Sung, G. H. & Lieberman, P. M. CTCF regulates Kaposi’s 64. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9,
sarcoma-associated herpesvirus latency transcription by nucleosome R137 (2008).
displacement and RNA polymerase programming. J. Virol. 87, 1789–1799 65. Fakhari, F. D. & Dittmer, D. P. Charting latency transcripts in Kaposi’s
(2013). sarcoma-associated herpesvirus by whole-genome real-time quantitative PCR.
42. Chen, H. S., Wikramasinghe, P., Showe, L. & Lieberman, P. M. Cohesins J. Virol. 76, 6213–6223 (2002).
repress Kaposi’s sarcoma-associated herpesvirus immediate early gene
transcription during latency. J. Virol. 86, 9454–9464 (2012).
43. Guito, J. & Lukac, D. M. KSHV Rta promoter specification and viral
Acknowledgements
We are grateful to Dr. Kenichi Nakajima for assistance in HGEP tissue culture, Dr.
reactivation. Front. Microbiol. 3, 30 (2012).
Matthew L. Settles for advice in bioinformatics analyses, and Drs. Pei-Ching Chang,
44. Palmeri, D., Spadavecchia, S., Carroll, K. D. & Lukac, D. M. Promoter- and cell-
Jinjong Myoung, Charles Wood, and Jae U. Jung for providing reagents. We also thank
specific transcriptional transactivation by the Kaposi’s sarcoma-associated
Drs. Chie Izumiya, Feng Zhou and Mr. Christopher P. Chen for technical assistance. This
herpesvirus ORF57/Mta protein. J. Virol. 81, 13299–13314 (2007).
research was supported by National Institutes of Health grants (DE025985) and by an
45. Raj, A. & van Oudenaarden, A. Nature, nurture, or chance: stochastic gene
American Cancer Society Research Scholar Grant (RSG-13-383-MPC). This work was
expression and its consequences. Cell 135, 216–226 (2008).
also partially supported by grants from the U.S. Department of Agriculture (2015-67015-
46. Sanchez, A. & Golding, I. Genetic determinants and cellular constraints in noisy
23268 and 2014-67015-21787). The UC Davis Comprehensive Cancer Center Genomics
gene expression. Science 342, 1188–1193 (2013).
Shared Resource is supported by the NCI Cancer Center Support Grant (CCSG; NCI
47. Nagano, T. et al. Single-cell Hi–C reveals cell-to-cell variability in chromosome
P30CA093373).
structure. Nature 502, 59–64 (2013).
48. Jin, F. et al. A high-resolution map of the three-dimensional chromatin
interactome in human cells. Nature 503, 290–294 (2013). Author contributions
49. Apostolou, E. & Thanos, D. Virus Infection Induces NF-kappaB-dependent M.C., C.G.T., and Y.I. designed research. M.C., T.W., K.N., R.R.D., Y.L., and Y.I. per-
interchromosomal associations mediating monoallelic IFN-beta gene formed experiments. B.D.-J., M.C., K.N., Y.L., R.R.D., C.G.T., and Y.I. analyzed data. Y.
expression. Cell 134, 85–96 (2008). L., R.D., and C.G.T. prepared sequence library and performed initial sequencing analyses.
50. Deng, W. et al. Controlling long-range genomic interactions at a native locus by T.W. and M.F. prepared recombinant KSHVs and generated iVero cells. K.N. utilized the
targeted tethering of a looping factor. Cell 149, 1233–1244 (2012). computer programs Circos and R to visualize Hi–C and qPCR data, respectively. M.C.
51. Rivas, C., Thlick, A. E., Parravicini, C., Moore, P. S. & Chang, Y. Kaposi’s and Y.I. wrote the manuscript and all authors edited the manuscript.
sarcoma-associated herpesvirus LANA2 is a B-cell-specific latent viral protein
that inhibits p53. J. Virol. 75, 429–438 (2001).
52. Hu, J. et al. LANA binds to multiple active viral and cellular promoters and Additional information
associates with the H3K4methyltransferase hSET1 complex. PLoS Pathog. 10, Supplementary Information accompanies this paper at https://doi.org/10.1038/s41467-
e1004240 (2014). 017-02089-9.
53. De Leo, A., Chen, H. S., Hu, C. A. & Lieberman, P. M. Deregulation of KSHV
latency conformation by ER-stress and caspase-dependent RAD21-cleavage. Competing interests: The authors declare no competing financial interests.
PLoS Pathog. 13, e1006596 (2017).
54. Myoung, J. & Ganem, D. Infection of lymphoblastoid cell lines by Kaposi’s Reprints and permission information is available online at http://npg.nature.com/
sarcoma-associated herpesvirus: critical role of cell-associated virus. J. Virol. 85, reprintsandpermissions/
9767–9777 (2011).
55. Campbell, M. et al. A lytic viral long noncoding RNA modulates the function of Publisher's note: Springer Nature remains neutral with regard to jurisdictional claims in
a latent protein. J. Virol. 88, 1843–1848 (2014). published maps and institutional affiliations.
56. Campeau, E. et al. A versatile viral system for expression and depletion of
proteins in mammalian cells. PLoS ONE 4, e6529 (2009).
57. Li H. Aligning sequence reads, clone sequences and assembly contigs with
BWA-MEM. arXiv preprint arXiv:13033997 (2013). Open Access This article is licensed under a Creative Commons
58. Li, H. et al. The sequence alignment/map format and SAMtools. Bioinformatics Attribution 4.0 International License, which permits use, sharing,
25, 2078–2079 (2009). adaptation, distribution and reproduction in any medium or format, as long as you give
59. Komano, J., Miyauchi, K., Matsuda, Z. & Yamamoto, N. Inhibiting the Arp2/3 appropriate credit to the original author(s) and the source, provide a link to the Creative
complex limits infection of both intracellular mature vaccinia virus and primate Commons license, and indicate if changes were made. The images or other third party
lentiviruses. Mol. Biol. Cell 15, 5197–5207 (2004). material in this article are included in the article’s Creative Commons license, unless
60. Watanabe, T. et al. The hematopoietic cell-specific Rho GTPase inhibitor indicated otherwise in a credit line to the material. If material is not included in the
ARHGDIB/D4GDI limits HIV type 1 replication. AIDS Res. Hum. Retrovir. 28, article’s Creative Commons license and your intended use is not permitted by statutory
913–922 (2012). regulation or exceeds the permitted use, you will need to obtain permission directly from
61. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. the copyright holder. To view a copy of this license, visit http://creativecommons.org/
Methods 9, 357–359 (2012). licenses/by/4.0/.
62. Izumiya, Y. et al. NF-kappaB serves as a cellular sensor of Kaposi’s sarcoma-
associated herpesvirus latency and negatively regulates K-Rta by antagonizing
the RBP-Jkappa coactivator. J. Virol. 83, 4435–4446 (2009). © The Author(s) 2017

14 NATURE COMMUNICATIONS | (2018)9:49 | DOI: 10.1038/s41467-017-02089-9 | www.nature.com/naturecommunications

You might also like