You are on page 1of 15

REvIEWS

Born to run: control of transcription


elongation by RNA polymerase II
Fei Xavier Chen   , Edwin R. Smith and Ali Shilatifard*
Abstract | The dynamic regulation of transcription elongation by RNA polymerase II (Pol II) is an
integral part of the implementation of gene expression programmes during development. In most
metazoans, the majority of transcribed genes exhibit transient pausing of Pol II at promoter-​
proximal regions, and the release of Pol II into gene bodies is controlled by many regulatory
factors that respond to environmental and developmental cues. Misregulation of the elongation
stage of transcription is implicated in cancer and other human diseases, suggesting that
mechanistic understanding of transcription elongation control is therapeutically relevant. In this
Review , we discuss the features, establishment and maintenance of Pol II pausing, the transition
into productive elongation, the control of transcription elongation by enhancers and by factors of
other cellular processes, such as topoisomerases and poly(ADP-​ribose) polymerases (PARPs), and
the potential of therapeutic targeting of the elongation stage of transcription by Pol II.

Super elongation complex


Genome-​wide studies have demonstrated that following that have also been identified from in vitro studies17.
(SEC). A complex containing transcription initiation, most metazoan genes undergo More recently, transcription elongation factors have
the most active form of positive a regulatory step termed promoter-​proximal pausing1,2. been identified by genome-​wide profiling of factors pre-
transcription elongation After transcribing 20–120 nucleotides downstream of the viously implicated in other cellular processes, including
factor-b; required for almost
transcription start site (TSS), RNA polymerase II (Pol II) topoisomerases and poly(ADP-​ribose) polymerases
all rapid inductions of
transcription and for release of pauses; its release into productive elongation requires (PARPs), thereby uncovering interconnectivity between
paused RNA polymerase II. the activity of positive transcription elongation factor DNA replication, DNA damage repair and transcription
b (P-​TEFb), which is a kinase composed of the catalytic elongation control.
subunit cyclin-​dependent kinase 9 (CDK9) and its regu- In this Review, we discuss recent advances in char-
latory subunit, cyclin T3–5. The activity of P-​TEFb is highly acterizing the general features of Pol II pausing and the
regulated, being part of at least three larger complexes: regulation of early and late transcription elongation
the super elongation complex (SEC)6,7, bromodomain-​ steps. We discuss the progress and remaining chal-
containing protein 4 (BRD4)-associated P-​T EFb lenges in understanding the molecular mechanisms of
(BRD4–P-​TEFb)8,9 and 7SK small nuclear ribonucleo­ elongation control by comparing the functions of sev-
protein (snRNP)-associated P-​TEFb (7SK–P-​TEFb)10,11. eral pausing and elongation factors, including negative
Pol II that is released to transcribe into gene bodies inter- elongation factor (NELF), DRB sensitivity-​inducing
acts with additional elongation factors that enhance its factor (DSIF), Pol II-​associated factor 1 (PAF1) and the
processivity, modify the chromatin landscape and regulate P-​TEFb-containing complexes (Table 1). We also discuss
co-​transcriptional RNA processing12. the roles of distal enhancers and factors such as topoi-
Many transcription elongation factors, such as somerases and PARPs, which function in other cellular
P-​TEFb, transcription factor S-​II, elongin A and eleven-​ processes, in the regulation of transcription elongation
nineteen lysine-​rich leukaemia protein (ELL), were by Pol II. On the basis of these findings, we forecast the
originally identified in vitro by biochemical methods13. potential directions in therapeutic targeting of the regu­
Simpson Querrey Center for
Epigenetics and the For example, P-​TEFb was purified from Drosophila lation of the elongation stage of transcription by Pol II
Department of Biochemistry melanogaster cell extracts as a factor sensitive to the in cancer and other human diseases.
and Molecular Genetics, transcription elongation inhibitor 5,6-dichloro-1-β-​
Feinberg School of Medicine, d-ribofuranosylbenzimidazole (DRB), whereas the ELL Features of Pol II pausing control
Northwestern University,
Chicago, IL, USA.
component of the SEC was purified from rat liver extracts One of the first genes found to undergo promoter-​
as a factor that suppresses transient pausing of Pol II proximal pausing of Pol II was the D. melanogaster heat
*e-​mail: ash@
northwestern.edu (refs14,15). Other transcription elongation factors were shock protein 70 (Hsp70) gene in non-​heat shock con-
https://doi.org/10.1038/ identified from genetic screens16, and some of these, such ditions18,19. Subsequently, promoter-​proximal pausing in
s41580-018-0010-5 as the product of the SPT genes, are orthologues of factors D. melanogaster was found to occur at highly transcribed

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Table 1 | Pausing-​related factors


Pausing-​related Subunits occupancy Function in pausing refs
factors
NELF • NELF-​A Promoter Stabilizes paused Pol II by 28,88,100,101

• NELF-​B preventing premature promoter-​


• NELF-​C or NELF-​D proximal termination
• NELF-​E
DSIF • SPT4 • Promoter Promotes the recruitment of 88,91,92

• SPT5 • Gene body NELF and capping factors


PAF1C • PAF1 • Enhancer Modulates enhancer activity 25,112,113,269

• CTR9 • Promoter and maintains paused Pol II


• LEO1 • Gene body by hindering its release into
• Parafibromin productive elongation
• WDR61
• RTF1
Gdown1a – Promoter Blocks TFIIF recruitment and 106,109,205

prevents early termination of


promoter-​proximal Pol II
PARP1 – • Enhancer ADP-​ribosylates NELF and 242

• Promoter inhibits its function in pausing


P-​TEFb • CDK9 • Enhancer Phosphorylates the Pol II CTD, 127

• CCNT1 or CCNT2 • Promoter NELF and the SPT5 CTR to


• Gene body promote release from pausing
SEC • AFF1 or AFF4 • Enhancer Most active P-​TEFb-containing 6,7,125,128

• ELL2 • Promoter complex; promotes rapid


• AF9 or ENL • Gene body release of paused Pol II into
• EAF1 or EAF2 productive elongation
• P-​TEFb
BRD4–P-​TEFb • BRD4 • Enhancer Stimulates P-​TEFb activity and 9,132

• P-​TEFb • Promoter promotes pause release


• Gene body
7SK–P-​TEFb • 7SK snRNP Promoter Sequesters P-​TEFb and prevents 10,11,127

• MEPCE pause release


• L ARP7
• HEXIM1 or HEXIM 2
• P-​TEFb
AF9, ALL1-fused gene from chromosome 9 protein; AFF1, AF4/FMR2 family member 1; BRD4, bromodomain-​containing protein 4;
CCNT1, cyclin T1; CDK9, cyclin-​dependent kinase 9; CTD, carboxy-​terminal domain; CTR , carboxy-​terminal region; CTR9, RNA
polymerase-​associated protein CTR9 homologue; DSIF, DRB sensitivity-​inducing factor ; EAF, ELL-​associated factor ELL, eleven-​
nineteen lysine-​rich leukaemia protein; ENL , eleven-​nineteen leukaemia; L ARP7 , La-​related protein 7; LEO1, PAF1/RNA
polymerase II complex component LEO1; MEPCE, methyl phosphate capping enzyme; NELF, negative elongation factor ; PAF1C,
RNA polymerase II-​associated factor 1 complex; PARP1, poly(ADP-​ribose) polymerase 1; P-​TEFb, positive transcription elongation
factor b; RTF1, PAF1/RNA polymerase II complex component RTF1 homologue; SEC, super elongation complex; snRNP, small
nuclear ribonucleoprotein; SPT4, transcription elongation factor SPT4; SPT5, transcription elongation factor SPT5; TFIIF,
transcription factor IIF; WDR61, WD repeat-​containing protein 61. aGdown1 is a subunit of the Pol II complex Pol II(G).

housekeeping genes20. Genome-​wide profiling methods of transcription stages but do not report on the stability
firmly established that pausing occurred at the major- and dynamics of pausing. Recently, the ability to meas-
ity of Pol II-​transcribed genes1,21. The importance of ure the dynamics of Pol II pausing was made possible
Pausing index
An estimate of RNA understanding promoter-​proximal pausing has long by the discovery of triptolide, a compound extracted
polymerase II (Pol II) promoter-​ been recognized, not only because of the importance from a traditional Chinese medicine that can inhibit the
proximal pausing; the ratio of of stress response pathways but also because important ATPase activity of the xeroderma pigmentosum group
Pol II occupancy at the developmental genes such as MYC and transcription B-​complementing protein (XPB), the helicase subunit
promoter-​proximal region
compared with the gene body.
of the human immunodeficiency virus (HIV) provirus of the general transcription factor (GTF) IIH (TFIIH)26.
were long known to be regulated at this step22. These Triptolide treatment prevents transcription reinitiation
Travelling ratio and other studies discussed below established sev- by Pol II and therefore enables the fate of pre-​existing
The ratio of gene-​body RNA eral features that are common to promoter-​proximal paused Pol II to be measured at different time points of
polymerase II (Pol II) to
pausing of Pol II. treatment. Such studies have shown that genes differ in
promoter-​proximal Pol II.
the time of residency of Pol II at the promoter-​proximal
Initiating Pol II The stability and dynamics of pausing. The degree of region, from a few minutes for most genes to an hour for
RNA polymerase II that is pausing is generally estimated from the pausing index a very few stably paused genes27–30. Although the half-​life
recruited by the general (or stalling index)1,21 or by the inverse travelling ratio of paused Pol II generally correlates with the pausing
transcription factors and is
involved in promoter melting
(or Pol II release ratio)23–25. Although instructive in cer- index, the former better predicts the steady-​state levels
before being released for tain situations, these ratios reflect the net residence of of mRNA23,29,31, such that genes with the longest half-​life of
transcription. the initiating Pol II and elongating Pol II during a series paused Pol II rather than the highest pausing index are

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Paused Pol II
more likely to be lowly expressed. The lack of correla- for DNA binding to sustain an open chromatin struc-
RNA polymerase II that has tion between pausing index and pausing stability was ture at promoters48,49,56,57. However, one study found
initiated non-​productive recently demonstrated using methylation footprinting that GC content, independent of transcription or paus-
transcription and is transiently with whole genome bisulfite sequencing32. Some genes ing, can explain the position of the NFR58, which could
pausing at the
with a high pausing index and low expression level have potentially involve the additional action of chromatin
promoter-​proximal region.
residency times of paused Pol II of just a few minutes, remodellers59.
Bisulfite sequencing suggesting that some genes with a high pausing index are One caveat of MNase-​based assays is the sequence
Bisulfite deaminates undergoing rounds of initiation and promoter-​proximal preference of MNase, which is biased towards AT-​
unmethylated DNA cytosine termination32. rich sequences, as well as its differential sensitivity to
into uracil, which can be used
to infer the position of
A long-​s tanding question is what causes some nucleosomes of different conformations. For exam-
methylated cytosine through genes to exhibit more pausing than other genes and ple, using mainly limited or titrated enzyme digestion,
sequencing. how gene-​specific pause–release is regulated during a class of MNase-​hypersensitive nucleosomes termed
development and in response to environmental cues. fragile nucleosomes has been reported at NFRs of pro-
TATA box
Several studies have examined the specific DNA and moters in yeast and higher eukaryotes60–66. However, it
A short AT-​rich motif found in
many RNA polymerase II chromatin features associated with promoters of genes has also been suggested that these MNase-​hypersensitive
promoters; bound by the with Pol II pausing. Over 20 years ago, the TATA box particles at promoters are non-​histone complexes67,68. As
general transcription factor at the MYC promoter was shown to be required for an alternative strategy, chemical mapping of histone
TATA-​binding protein. pausing at this gene22. Later, genome-​wide analyses in H4 was used to directly map n ­ ucleosome occupancy,
G-​quadruplex
D. melanogaster found that paused promoters that tend which confirmed the presence of fragile nucleosomes.
(G4). A highly stable nucleic to have high GC content are enriched for a promoter Notably, fragile nucleosomes were found in the mouse
acid structure comprising two element called the pause button and for the presence and Schizosaccharomyces pombe genomes, but not in
or more stacked guanine of the GAGA motif (which recruits transcription factor S. cerevisiae69, which coincides with the presence of
tetrads. Intramolecular G-​
GAGA) and tend to lack a TATA box33–35. Although paused Pol II in these species52. Indeed, nucleosome levels
quadruplexes formed in
nascent RNA can contribute to promoter elements are generally less well defined in measured by chemical mapping appear to correlate with
RNA polymerase II pausing. mammalian cells, paused promoters in mammalian the degree of pausing, opposing the interpretation of
cells also tend to have high GC content and tend to early stu­dies using MNase. The discrepancy could be
lack a TATA box2,31. due to the different sensitivities of chemical mapping
and MNase-​based assays in detecting the unique nucleo­
R-​loops contribute to Pol II pausing. R-​loops are RNA– some structure around paused promoters, which exhibit
DNA hybrids formed when the nascent RNA hybridizes a looser ­conformation and higher rate of nucleosome
with the template strand of DNA during transcription turnover (nucleosome remodelling).
(leaving the non-​template, or coding strand, looping Nucleosome composition can also influence paus-
out)36,37. RNA–DNA hybrids are more stable than double-​ ing, for example, nucleosomes with the histone variant
stranded DNA (dsDNA) and thus can block access of H2A.Z are less stable and are suggested to negatively cor-
successive Pol II to the DNA template38–40. Interestingly, relate with the establishment of p ­ ausing31,70,71. However,
the GC skew that is correlated with pausing also favours the exact content and conformation of the nucleosomes
the formation of R-​loops owing to the higher thermo- around promoters as well as their i­nterplay with the
dynamic stability of RNA–DNA hybrids bearing G-​rich dynamics of pausing remain to be fully examined.
RNA36,41–43. The capacity of the exposed coding strand to
form stable G-​quadruplex (G4) structures could further The establishment of Pol II pausing
stabilize R-loops44,45. Indeed, G4 motifs correlate with Transcription initiation by Pol II commences with the
promoter-​proximal pausing46. However, paused Pol II assembly of the preinitiation complex (PIC), which
mainly localizes 20–120 nucleotides downstream of includes the polymerase, GTFs and a large number of
the TSS whereas R-​loops can cover wider regions of the co-​regulators (reviewed in refs72,73) (Fig. 1a). This pro-
transcription unit, peaking around 1.5 kb downstream cess occurs in a stepwise manner with TFIIH, which is
of the TSS37,47. This suggests that R-​loops are more a complex comprising about ten subunits, and is the last
broadly involved in the control of the processive stage of essential component of the PIC to be recruited. TFIIH
transcription elongation. is unique among the GTFs as it is required not only for
transcription initiation but also for subsequent phos-
Pausing and nucleosome features. Nucleosome-​free phorylation of Pol II to ensure the proper establishment
regions (NFRs), as determined by micrococcal nucle- of pausing.
ase sequencing (MNase-​s eq), are found at promot-
ers of actively transcribed genes and correlate with Transient promoter melting by the helicase module
promoter-​proximal Pol II levels48–50. However, NFRs of TFIIH. The TFIIH holoenzyme is organized into
are also found by MNase-​seq in Saccharomyces cerevi- two structurally and functionally distinct subcom-
siae, which lacks promoter-​proximal pausing51–54, indi- plexes: the core module and the CDK-​activating kinase
cating that the establishment of the NFR is associated (CAK) module (reviewed in ref.74). The catalytic sub-
with but not exclusive to paused promoters. GC-​rich unit in the core module is the ATP-​dependent 3′ to 5′
sequences favour intrinsic nucleosome occupancy55,56, helicase XPB (Ssl2 in budding yeast), which opens the
although in vivo, the lowest nucleosome occupancy dsDNA (promoter melting) to allow Pol II access to the
can be seen at paused promoters with high GC content, template strand, which is referred to as open complex
­perhaps because paused Pol II competes with histones formation75 (Fig. 1a). Permanganate can react with the

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

a Initiation and pausing establishment

CTD Mediator

CDK7 GTFs
TFIIH XPB CTD Ser5 phosphorylation
TFIIF CTD Ser2 phosphorylation
Pol II NELF phosphorylation
DSIF phosphorylation

b Pausing stabilization

Mediator

TFIIF DSIF NELF –1 +1


PAF1C F
Gdown1 NEL

Pol II NELF
Transcript DSIF
SEC

PAF1C

–1 +1
c Release from pausing

DSIF NELF
PAF1C SEC
Gdown1

–1 +1

Fig. 1 | establishment, maintenance and release of rNa polymerase ii pausing. a | General transcription factors (GTFs)
recruit RNA polymerase II (Pol II) to the promoter to form the preinitiation complex. One GTF, TFIIH, has a dual role in
helping to open the double-​stranded DNA through the helicase subunit xeroderma pigmentosum group B-​
complementing protein (XPB) to allow Pol II to initiate transcription, and in establishing pausing by phosphorylating the
Ser5 residues of the Pol II carboxy-​terminal domain (CTD) through its cyclin-​dependent kinase 7 (CDK7) subunit. The
Mediator complex enables the formation of long-​range enhancer–promoter interactions; the GTF TFIIF has dual roles in
promoting transcription initiation and early elongation. b | Paused Pol II is stabilized by interacting with several pausing
factors, such as negative elongation factor (NELF), DRB sensitivity-​inducing factor (DSIF), Pol II-​associated factor 1
complex (PAF1C) and Gdown1. Depletion of NELF (top) results in Pol II destabilization before it passes through the +1
nucleosome downstream of the transcription start site and in promoter-​proximal transcription termination. PAF1C
depletion (bottom) leads to increased recruitment of the super elongation complex (SEC), which results in the release of
paused Pol II into productive elongation. Gdown1 contributes to pausing by blocking recruitment of TFIIF. c | Release from
pausing is driven by the recruitment and activation of positive transcription elongation factor b (P-​TEFb)-containing
complexes, for example, the SEC, which phosphorylates NELF, DSIF and Ser2 residues of the Pol II CTD. Phosphorylated
NELF dissociates from chromatin, whereas PAF1C and phosphorylated DSIF transition from being pausing factors to
being positive elongation factors by promoting Pol II processivity and contributing to co-​transcriptional RNA processing
(not shown).

single-​stranded DNA and has been used to demonstrate The kinase module of TFIIH prepares Pol II for pausing.
that Pol II rapidly proceeds from the site of open com- The CAK module of TFIIH contains CDK7 (Kin28 in
plex formation to the site of pausing1,21,76. The promoter budding yeast), which phosphorylates the Ser5 and
melting activity of XPB is required for each new round Ser7 residues of the Pol II carboxy-​terminal domain
of transcription initiation76. Treatment of cells with (CTD)77,78. Ser5 phosphorylation is the much more
the XPB inhibitor triptolide blocks new initiation, studied of the two modifications; it is known to occur
thereby allowing the decoupling of the study of during transcription initiation and to gradually undergo
­transcription elongation and Pol II pausing from that dephosphorylation as Pol II enters productive elon-
of transcription initiation5,27,28,32. gation79 (Fig.  1). Therefore, Ser5 phosphorylation is

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Mediator complex
commonly used as a mark of paused Pol II to distin- to the Pol II CTD, the carboxy-​terminal region (CTR)
A large multiprotein complex guish it from unphosphorylated or non-​transcriptionally of SPT5 has a repeat region (in this case a pentapep-
that interacts with transcription engaged Pol II. tide repeat) that is phosphorylated by both the kinase
factors and RNA polymerase II. TFIIH recruitment requires CDK8, which is the module of TFIIH and by P-​TEFb and is involved in the
Mediator can promote
kinase module of the Mediator complex. Once phos- recruitment and activation of the capping complex91–94.
enhancer–promoter looping to
activate transcription and can phorylated, TFIIH evicts the Mediator through CDK7- Biochemical and structural studies demonstrated that
also recruit transcription mediated phosphorylation of Ser5 of the Pol II CTD and SPT5 makes key contacts with the nascent RNA exiting
elongation factors. thus allows the release of Pol II from the PIC80,81. In bud- Pol II, which could contribute to its role in pausing95–97.
ding yeast, a direct role for Ser5 phosphorylation in this NELF is a four-​subunit complex, in which NELF-​A
Auxin-​inducible degradation
In cells expressing a plant
process was suggested by replacing wild-​type RNA poly­ and NELF-​C (or its isoform NELF-​D) form the core
auxin-​inducible E3 ubiquitin merase II subunit 1 (Rpb1) with Rpb1 bearing a CTD-​ sub-​complex, which associates with NELF-​B and NELF-​
ligase, any protein of interest S5A mutant that cannot be phosphorylated by CDK7. E98 (Table 1). In the presence of DSIF, NELF is a potent
can be tagged for rapid The CTD-​S5A mutant mimics chemical inhibition of inhibitor of transcription in vitro88. However, deplet-
degraded upon addition of
CDK7, with both the CTD mutant and CDK7 inhibi- ing NELF in cells generally leads to decreased levels of
auxin to the medium.
tion resulting in the accumulation of Mediator at core promoter-​proximal Pol II and reduced transcription at
promoters and impaired escape of Pol II from the PIC82. many paused genes99–101. One way to reconcile the find-
Pol II CTD Ser5 phosphorylation also helps to ings from in vitro transcription studies and from NELF
recruit and activate the capping enzymes83 that carry depletion studies is if NELF stabilizes paused Pol II
out a multi­step modification of the 5′ end of the nas- by preventing premature promoter-​proximal tran-
cent RNA. Sequencing of small capped RNAs indicated scription termination, a process whereby Pol II disso-
that co-​transcriptional capping is important for the stabil- ciates from promoters without passing through the +1
ity of paused Pol II (ref.28). Uncapped RNAs are subject to nucleosomes28,29 (Fig. 1b). All four subunits of NELF can
degradation by nucleases such as 5′-3′ exoribo­nuclease 2 associate with RNA, raising the possibility that NELF
(XRN2), which can mediate the termination of promoter-​ interactions with short nascent RNA could contribute
proximal Pol II pausing, suggesting that decapping factors to the inhibition of premature transcription termina-
are prime regulators of the residency time of paused Pol II tion98,102,103. Studies of NELF function in cells have been
(ref.84). This idea is supported by studies employing CDK7 performed with either depletion or deletion of its sub-
inhibition, otherwise inhibiting Ser5 phosphorylation or units and monitoring of Pol II pausing following a long
blocking nascent RNA capping, which led to defective period of selection, which could include many indirect
establishment of pausing77,85–87. Additionally, phospho- effects of NELF loss. Therefore, future studies of NELF
rylation of Ser5 stimulates the ­catalytic activity of the and its role in pausing should concentrate on the in vivo
histone-​lysine N-​methyltransferase SETD1A and thus role or roles of NELF in pausing immediately following
helps maintain at promoters the levels of trimethylation of NELF loss, as the in vitro conditions that led to current
histone H3 Lys 4 (H3K4me3), which are associated with models of NELF function could be missing many factors
transcription activation86. that regulate NELF activity.
The precise location of pausing downstream of the The Pol II subunit Gdown1 (also known as GRINL1A)
TSS, which can vary between 20 and 120 nucleotides, also appears to regulate pausing through preventing
could be governed in part by the speed of Pol II escap- promoter-​proximal transcription termination. Gdown1
ing from initiation and how quickly pausing factors competes for binding to Pol II with TFIIF, which is a
are recruited to it76. However, extending the distance GTF that boosts both transcription initiation and
between the HSP70 TSS and the normal site of paus- early elongation89,104–109 (Fig. 1b; Table 1). Gdown1 also
ing by inserting a 5 bp linker led to a concomitant shift inhibits transcription termination factor 2 (TTF2).
downstream in the site of Pol II, whereas a 10 bp inser- Therefore, Gdown1 may contribute to pausing by
tion led to a much more distal and less focused site of blocking recruitment of TFIIF and by inhibiting early
pausing, suggesting that additional mechanisms control transcription termination. Further in vivo studies of
the distance between the TSS and the pause site47. the roles of Gdown1 in transcription regulation would
be highly informative.
Maintenance of Pol II pausing
Maintencance of Pol II pausing appears to be controlledMaintaining Pol II pausing by preventing Pol II release.
by different factors that regulate distinct features ofDSIF, NELF and Gdown1 were each implicated in
the transcriptionally engaged Pol II. Current evidence pausing using in  vitro transcription systems, and
indicates that maintaining Pol II pausing requires boththeir depletion in cells leads to decreased transcrip-
factors that prevent promoter-​proximal transcription tion. By contrast, the PAF1 complex (PAF1C), which
termination and factors that prevent the release of Pol II
had been shown to promote transcription elongation
into productive elongation (Fig. 1b). using in vitro transcription systems110,111, was geneti-
cally implicated in antagonizing the release of paused
Maintaining Pol II pausing by preventing promoter-​ Pol II at erythroid genes in zebrafish112. To address
proximal transcription termination. Studies of tran- this discrepancy, genome-​wide studies in mammal­
scription elongation in  vitro have found that two ian cells found that depleting PAF1 by RNAi or by
complexes, DSIF and NELF, are required for transcrip- auxin-​inducible degradation generally leads to the release of
tion inhibition by DRB88–90. DSIF is composed of tran- Pol II into gene bodies and increased levels of mature
scription elongation factors SPT4 and SPT5. Similar transcripts at paused genes, which is in agreement

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

with the result of the genetic studies of PAF1C25,113,114 factors at distinct stages of transcription could determine
Acute protein degradation
Experimentally induced
(Fig. 1b). Genes with higher pausing indices were more whether PAF1 functions as a positive or negative regu-
degradation of proteins by likely to exhibit pause release upon PAF1 depletion, but lator of gene expression115. The recent finding that PAF1
targeting them for proteasomal the basis of this gene specificity is not yet fully under- occupies and inhibits the activity of enhancers associated
degradation. stood. Perhaps interactions with particular transcription with genes regulated by PAF1 is consistent with the idea
that transcription factors or other enhancer-​binding fac-
tors function with PAF1 in the regulation of pausing113.
a 7SK–P-TEFb complex CTD Ser5 phosphorylation As discussed below, although PAF1 generally inhibits
CTD Ser2 phosphorylation enhancer activity and the release of promoter-​proximal
P-TEFb Histone acetylaton paused Pol II, PAF1 can function as a positive trans­
HEXIM1 or
cription elongation factor following the release of Pol II
LARP7 HEXIM2
MEPCE from pausing (Fig. 1c).
Together, these studies have revealed that Pol II paus-
7SK RNA
ing is subjected to complex and multilayered regulation
by a cohort of pausing factors. However, a full under-
standing of the detailed interplay among these factors
b BRD4–P-TEFb complex on a genome-​wide scale is still missing. Accordingly,
further investigation is warranted to systematically
CTD compare different pausing factors in the same genetic
P-TEFb and epigenetic backgrounds using not only RNAi but
BRD4 also acute protein degradation to determine their direct
gene targets and their primary roles in maintaining
Pol II pausing.

c SEC and SEC-like complexes Promoting the release of paused Pol II


Target genes of SEC The release of Pol II into productive elongation requires
the activity of the Pol II CTD kinase P-​TEFb4. The mech-
EAF1 or ELL2
EAF2 anism of release is thought to involve not only phos-
AFF1 or AFF4 phorylation of the Pol II CTD Ser2 residues but also
P-TEFb
AF9 or ENL
phosphorylation of DSIF and NELF. P-​TEFb is part of
macromolecular complexes, including the SEC, which
Pol II
regulate its activity (Fig. 1c).
Target genes of SEC-like complexes P-​TEFb was discovered as a complex that allows Pol II
to efficiently synthesize long transcripts in a DRB-​
AFF2 or sensitive manner14,116. Treatment of cells with DRB or
P-TEFb AFF3 other P-​TEFb inhibitors leads to a global defect in the
AF9 or ENL
release of paused Pol II and a reduction in steady-​state
levels of transcripts3,5,117, demonstrating the essential role
Pol II
of P-​TEFb in mediating Pol II release and its require-
Fig. 2 | release of rNa polymerase ii pausing by P-​TeFb-containing complexes. ment for transcription of most Pol II-​transcribed genes.
Three major positive transcription elongation factor b (P-​TEFb)-containing complexes Well-​known substrates of P-​TEFb include NELF, DSIF
have been identified in mammals: 7SK small nuclear ribonucleoprotein–P-​TEFb and the CTD of Pol II (refs118–121). NELF dissociates from
(7SK–P-TEFb), bromodomain-​containing protein 4–P-​TEFb (BRD4–P-​TEFb) and the super chromatin upon its phosphorylation by P-​TEFb118,122,
elongation complex (SEC) or SEC-​like complexes. a | 7SK–P-​TEFb inhibits the activity of whereas DSIF switches from being a pausing factor
P-​TEFb and thus suppresses the release of paused RNA polymerase II (Pol II). The complex to being a positive elongation factor upon P-​TEFb
contains the non-​coding 7SK small nuclear RNA (snRNA), which interacts with and is phosphorylation of the SPT5 CTR119,123 (Fig. 1c).
stabilized by La-​related protein 7 (L ARP7) and 7SK snRNA methyl phosphate capping
enzyme (MEPCE), and HEXIM1 or HEXIM2, which mediate the inhibition of P-​TEFb by
binding and sequestering it. b | In the BRD4–P-​TEFb complex, BRD4 stimulates the kinase Several P-​TEFb complexes control Pol II release from
activity of P-​TEFb, which phosphorylates the Ser2 residues of the Pol II carboxy-​terminal pausing. Biochemical studies have shown that P-​TEFb is
domain (CTD) and thus promotes the release of paused Pol II. The interaction between part of several larger complexes by associating with 7SK
BRD4 and P-​TEFb is mediated by a carboxy-​terminal region of BRD4 (not shown). The snRNP, BRD4 and the SEC7–11,124,125 (Fig. 2). 7SK snRNP
bromodomains of BRD4 recognize acetylated histones and mediate BRD4 recruitment. represses transcription by sequestering and inhibiting a
c | The SEC seems to be the most active P-​TEFb complex because it is required for the large portion of cellular P-​TEFb10,11. This inhibition is
expression of rapidly induced genes. SEC assembly relies on the scaffold protein mediated through the binding of P-​TEFb by a dimer of
AF4/FMR2 family member 1 (AFF1) or AFF4, which interacts with P-​TEFb and with HEXIM1 or HEXIM2 (ref.126) (Fig. 2a). Up to 90% of P-​
ALL1-fused gene from chromosome 9 protein (AF9) or eleven-​nineteen leukaemia (ENL) TEFb is sequestered with the 7SK-​P-TEFb complex127.
and with eleven-​nineteen lysine-​rich leukaemia protein 2 (ELL2). ELL-​associated factor 1
By contrast, the SEC and BRD4 represent active forms
(EAF1) or EAF2 participate in the SEC through binding directly to ELL2. AF9 and ENL
recognize acetylated histones to facilitate SEC recruitment, and ELL2 promotes of P-​TEFb that can promote the release of Pol II from
transcription elongation by suppressing transient Pol II pausing (not shown). The SEC-​like pausing6,9,128 (Fig. 2b,c). During gene activation, especially
complexes are composed of AFF2 or AFF3, P-​TEFb, and AF9 or ENL , but lack ELL2–EAF rapid transcription induction, P-​TEFb is evicted from
modules. The SEC and the SEC-​like complexes might stimulate transcription activation 7SK snRNP127 and can incorporate into the SEC, which
of different target genes. leads to the enzymatic activation of P-​TEFb, which then

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

phosphorylates its targets, including the Ser2 residues of elongation complex named the little elong­ation com-
the Pol II CTD125,127–129. The transition of P-​TEFb from plex (LEC), which regulates Pol II transcription of small
repressive to active complexes depends on multiple nuclear RNA (snRNA) genes, suggesting that additional
factors, including KRAB-​associated protein 1 (KAP1; factors could specify the recruitment of LEC to snRNA
also known as TRIM28) and the splicing factors serine/ genes and of the SEC to protein-​coding and long non-​
arginine-​rich splicing factor 1 (SRSF1) and SRSF2, which coding RNA genes147,149–151. The use of auxin-​inducible
associate with 7SK snRNP on chromatin130,131. degradation of MED26 and other Mediator subunits
Although both BRD4–P-​TEFb and the SEC phos- should allow further dissection of their suggested con-
phorylate Ser2 residues of the Pol II CTD, it remains to tribution to SEC recruitment and other possible roles in
be determined how the two complexes work together transcription elongation control. Notably, SEC recruit-
(or separately), to promote transcription elongation. ment would be present at most genes actively transcribed
Intriguingly, a recent study reported that acute deg- by Pol II. In mammals, AF4/FMR2 family member 1
radation of BRD4 resulted in a severe defect in Ser2 (AFF1) and AFF4, which are the scaffolding proteins of
phosphorylation and transcription elongation without the canonical SEC, have two additional paralogues, AFF2
obvious change in the genomic occupancy of the P-​TEFb and AFF3. AFF2 and AFF3 can each be assembled into
catalytic subunit CDK9 (ref.132). This suggests that BRD4 active P-​TEFb-containing and ENL-​containing SEC-​like
is primarily involved in stimulating, but not recruiting, complexes, and affect the expression of distinct groups
P-​TEFb or that BRD4 can function independently of P-​ of genes compared with those affected by the full SEC6
TEFb133. Indeed, it has been reported that BRD4 is also (Fig. 2c). This suggests that generic recruitment to actively
an (atypical) kinase that can phosphorylate Ser2 of CTD transcribed genes is not sufficient to explain the unique
in vitro134 and can facilitate transcription elongation function of the SEC.
independently of P-​TEFb135.
Previous work on HIV activation indicates that Productive transcription elongation
BRD4 maintains basal transcription from the HIV long Productive elongation by Pol II affects and is also
terminal repeat (LTR), whereas transactivator of tran- affected by chromatin structure through both nucleo­
scription (TAT)-mediated recruitment of the SEC to the some turnover and co-​transcriptional modification of
HIV LTR is required for full activation of the provirus histones and DNA. A chromatin structure that allows
transcription136,137. Interestingly, TAT interacts with P-​ polymerases to move forward while preventing cryptic
TEFb, specifically comprising the cyclin T1 isoform intragenic transcription initiation requires factors that
through zinc-​dependent binding to a Cys residue that balance the disassembly and reassembly of nucleosomes
is not found in cyclin T2 (ref.138). However, the degree to (reviewed in refs152–154). At the 3′ end of genes, elon-
which cellular genes depend on cyclin T1 versus cyclin gating Pol II transitions into transcription termination,
T2 is currently unknown. which involves RNA cleavage and polyadenylation
SEC subunits are frequently found with transloca- (Box 1).
tion partners of the histone H3 Lys4 methyltransferase
myeloid/lymphoid or mixed-lineage leukaemia (MLL; Nucleosome turnover during transcription elong­
also known as KMT2A) in cancer139. Although mecha- ation. The dynamic turnover of nucleosomes during
nisms have been proposed for SEC recruitment to MLL elongation is mainly driven by the activity of chromatin
target genes through its interactions with MLL fusion remodellers and histone chaperones, which facilitate
proteins in cancer and for the co-​option of the SEC by the movement of Pol II through nucleosomes and aid in
TAT for HIV transcription139,140, it is less clear how the their reassembly. The histone octamer of a nucleosome
SEC is recruited to chromatin in normal physiological has a tripartite arrangement of one H3–H4 tetramer and
conditions. One possibility is that tissue-​specific tran- two H2A–H2B dimers155. During transcription by Pol II,
scription factors, such as the Notch pathway effector one H2A–H2B dimer is removed from the nucleo­some,
HES1, bind and recruit the SEC to activate their tran- leaving a subnucleosomal particle called the h ­ exasome156.
scription programmes141. At epidermal growth factor tar- This process can be enhanced by facilitates chroma-
get genes such as FOS and JUN, the SEC is recruited by tin transcription (FACT), which travels with Pol II,
Integrator complex the Integrator complex142. Another, nonexclusive possibil- interacts directly with the H2A–H2B dimer and regu­
A large multiprotein complex ity is that the SEC recognizes certain chromatin profiles. lates transcription-​couple disassembly and reassem-
found only in metazoans that
For instance, the YEATS (YAF9, ENL, AF9, TAF14 and bly of nucleosomes157–159. Dislodgement of H2A–H2B
can directly interact with RNA
polymerase II; facilitates SAS5) domains of the SEC subunits eleven-​nineteen leu- dimers by FACT can enhance nucleosome breathing to
transcription and 3′-end kaemia (ENL) and ALL1-fused gene from chromosome 9 facilitate the passage of Pol II (refs160,161). The dynamic
processing of enhancer RNAs protein (AF9), which are frequent translocation part- regulation of nucleosomes during transcription elong­
and small nuclear RNAs and ners of MLL, can bind acetylated H3 and are required ation by FACT can be further facilitated by other
recruitment of the super
elongation complex to
for recruiting the SEC to chromatin and driving high ­histone chaperones and chromatin remodellers, such as
immediate early-​response levels of transcription in acute myeloid leukaemia and of SPT6, chromodomain-​helicase-DNA-​binding protein 1
genes. HIV provirus143–146. (CHD1) and imitation SWI (ISWI)162,163.
Metazoan-​specific Mediator complex subunit 26 Factors implicated in the regulation of pausing can
Nucleosome breathing
(MED26) has been suggested to contribute to SEC recruit­ also facilitate transcription elongation through chroma-
A property of nucleosomes
whereby DNA at the edges of ment through its interaction with the SEC subunits ELL-​ tin. For example, after release of Pol II into productive
the histone core transiently associated factor 1 (EAF1) and EAF2 (refs147,148). However, elongation, PAF1C travels with the elongating Pol II and
unwrap and rewrap. EAF1 and EAF2 are also part of a distinct transcription acts as an organizing platform for co-​transcriptional

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Box 1 | ending transcription elongation by termination


the transition from transcription elongation to
transcription termination requires expulsion of Polyadenylation CTD R1810me2s
signal CTD Ser2 phosphorylation
elongating rNa polymerase ii (Pol ii) from chromatin
at the end of the gene. this process is coordinated DNA
with rNa cleavage and polyadenylation, which are
mediated by various protein complexes, including the XRN2
cleavage and polyadenylation specificity factor (CPsF) SMN
Senataxin DSIF
and the cleavage stimulation factor (CstF) (reviewed AAU
CTD PAF1C
A A A
in refs 259,260
). two different but not mutually exclusive Transcript R-loop
models for Pol ii transcription termination have been
CPSF Pol II
debated for decades, namely, the torpedo model and
CSTF
the allosteric model. the torpedo model posits that
cleavage of the nascent transcript allows access to 5′-3′ exoribonuclease 2 (XrN2), which chases down and promotes the
eviction of Pol ii. the allosteric model posits that a poly(a) signal-​dependent conformational change of Pol ii at the end of
genes triggers termination and dislodges Pol ii from chromatin. two recent studies, one advocating for the torpedo
model261 and the other for the allosteric model262, reported decreased, but not abolished, termination by XrN2
inhibition261,262, implying that rNa degradation following cleavage might contribute to efficient transcription termination
but that it is not absolutely required for termination at most genes263.
transcription termination of many genes is associated with Pol ii pausing downstream of the poly(a) site (reviewed in
refs259,260), which is thought to favour the cleavage of transcripts and degradation of the downstream cleaved rNa.
indeed, reduction of 3′-end Pol ii pausing by inhibition of positive transcription elongation factor b (P-​teFb) leads to
decreased cleavage and termination264,265. similar to the proposed role of r-​loops in promoter-​proximal pausing
regulation, pausing at the 3′ end could be enhanced by r-​loop formation downstream of the poly(a) site (see the figure)
and could be further enhanced by rNai-​induced formation of repressive chromatin36,266. r-​loops at the 3′ end of
genes are resolved by senataxin, which facilitates degradation of transcripts downstream of poly(a) by XrN2 (ref.267)
(see the figure). a recent study suggests that senataxin is recruited to termination sites by its interaction with survival
of motor neuron protein (sMN), which recognizes symmetrical dimethylation of arg1810 (r1810me2s) on the
carboxy-​terminal domain (CtD) of Pol ii (ref.268).
DsiF, DrB sensitivity-​inducing factor; PaF1C, Pol ii-​associated factor 1 complex.

processes by recruiting a variety of nucleosome remod- but perturbation of its deposition has a minor effect
ellers (CHD1 (ref. 164) ), histone chaperones (SPT6 on gene expression179–181. Studies in mammalian cells
(ref.165) and FACT166,167) and histone modifiers (com- have identified a role for regulators of H2B ubiqui-
plex proteins associated with SET1 (COMPASS)168, E3 tylation in regulating transcription at both enhanc-
ubiquitin-​protein ligase BRE1169,170 and histone-​lysine ers and promoters, but no direct role in promoting
N-​methyltransferase, H3K79 specific (also known as transcription elongation has been identified182,183.
DOT1L)168,169. Therefore, defects in productive elong­ Histone-​lysine N-​methyltransferase SETD2 associ-
ation owing to PAF1C ablation could result from the ates with the phosphorylated Pol II CTD and deposits
deregulation of any number of these factors. As another H3K36me3 in gene bodies, where it can affect RNA
example, DSIF transits from being a pausing factor to splicing and prevent cryptic transcription154,184 (Fig. 3c).
an elongation factor upon phosphorylation by P-​TEFb. This regulation is mediated by a variety of factors that
Upon the release of Pol II pausing, DSIF promotes can recognize H3K36me3, such as histone deacetylases
productive elongation by interacting with elongating and nucleosome remodellers185,186. In mammals, SETD2-
Pol II and reinforcing the closed conformation of the catalysed H3K36me3 is recognized by the PWWP
Pol II clamp to facilitate the passage of the template DNA domain of DNA (cytosine-5)-methyltransferase 3B,
through the central cleft of the polymerase92,171–173. which methylates DNA in gene bodies to help prevent
cryptic transcription187,188.
The chromatin landscape of productive elong­ation. Histone H3K79me2 and H3K79me3 are found
A variety of post-​translational modifications are asso- in gene bodies of actively transcribed genes, but the
ciated with distinct stages of transcription. Active function of these modifications is not well under-
promoters possess high levels of H3K4me3 and mul- stood (Fig. 3c). The methylation of H3K79 is mediated
tiple acetylated H3 and H4 Lys residues, whereas gene by DOT1L with the assistance of PAF1 and BRE1
bodies of transcribed genes are enriched with mono-​ (refs168,189–192). In metazoans, DOT1L associates with
ubiquitylated H2B (H2Bub), H3K36me3, H3K79me2 the SEC proteins AF9 and ENL, suggesting that DOT1L
Pol II clamp
and H3K79me3 (refs174–176) (Fig. 3). However, owing and the SEC share some degree of co-​regulation193. The
Part of the RNA polymerase II
(Pol II) complex comprising the to the high correlation of all these histone modi- catalytic domain of DOT1 is structurally unrelated to
amino-​terminal regions of RNA fications with gene expression and their complex the SET domain found in other Lys methyltransferases,
Pol II subunit 1 (Rpb1) and ­crosstalk177,178, it has been challenging to discriminate and the Lys that it modifies is located in the globular
Rpb2, and the carboxy-​ the direct involvement of each modification in tran- domain of H3, not in its tail194. No domains that recog-
terminal region of Rpb6, which
swings over the Pol II active-​site
scription regulation. In budding and fission yeasts, the nize H3K79me or any demethylases for these residues
cleft to encase the DNA deposition of H2Bub by the E3 ubiquitin ligase Bre1 have been identified, further hampering investigation of
template and nascent RNA. is regulated by chromatin remodelling factor Chd1, the role of H3K79me in transcription elongation.

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Cornelia de Lange syndrome


Enhancers regulate elongation transcription elongation. Heterozygous mutations in
A developmental syndrome Enhancer–promoter interactions are essential for fine- cohesin subunits cause Cornelia de Lange syndrome, prob-
caused by heterozygous tuning transcription, including transcription elong­ ably owing to defective enhancer–promoter communica-
germline loss-​of-function ation22,195 (reviewed in refs196,197). Although the topo- tion and not the chromosome cohesion role of cohesin203.
mutations in the cohesin loading
logical organization of long-​range enhancer–promoter Mutations that stabilize the SEC core component AFF4
protein nipped-​B-like protein or
in structural components of the interactions is not fully understood, the cohesin and were found in patients with CHOPS syndrome, which shares
cohesin ring. Mediator complexes have important roles in enabling many phenotypic properties with Cornelia de Lange syn-
these interactions198–200. Cohesin and Mediator physi­ drome204. The similarities in phenotypes between loss of
CHOPS syndrome cally interact with each other and co-​o ccupy both function of cohesin and gain of function of the SEC raise
A developmental syndrome with
a similar phenotype to Cornelia
active enhancers and promoters199,200. Cohesin forms a the possibility that cohesin-​dependent enhancer activity
de Lange syndrome that is ring-​like structure that is thought to hold together the modulates the role of the SEC in transcription elongation.
caused by germline gain-​ enhancer and promoter198 (Fig. 4). However, cohesin There are at least two lines of evidence that link
of-function mutations in the depletion predominantly affects the release of paused Mediator to transcription elongation control in addition
super elongation complex
Pol II rather than the establishment of pausing201,202, sug- to its interaction with cohesin80. First, Mediator allevi-
scaffolding protein AF4/FMR2
family member 4. gesting that enhancer–promoter looping directly governs ates the Gdown1-dependent stabilization of Pol II paus-
ing106,205, and second, Mediator appears to recruit the SEC
and BRD4 to promoter regions and thus stimulates Pol II
release147,206,207. Owing to the transient association of
Enhancer TSS TTS Mediator with PIC before the full establishment of pausing
as discussed above, the SEC and BRD4, which are recruited
a
TTotal Pol II early during that short time frame by Mediator, must be
Ser5P retained on chromatin through additional mechanisms,
Ser2P for example, through their binding to acetylated histones.
Active enhancers are enriched in H3K4me1 and in
acetylated H3K27 (H3K27ac) (Fig. 3), which are largely
catalysed by the COMPASS family members myeloid/
lymphoid or mixed-​lineage leukaemia protein 3 (MLL3;
also known as KMT2C) and MLL4 (also known as
KMT2D)208,209 and by p300 and CREB-​binding pro-
b
H3K4me1 tein (CREBBP)210,211, respectively (Fig. 4). In contrast to
H3K4me2 H3K4me1, which is present at both active and poised
H3K4me3 enhancers, the presence of H3K27ac correlates with
enhancer activity139,211,212. Enhancer RNAs were suggested
to interact with and facilitate the eviction of NELF from
promoters to activate the release of paused Pol II (ref.103),
providing a potential direct link between enhancer acti-
vation and transcription elongation. Recent work has sug-
c gested that components of all three P-​TEFb-containing
H3K27ac complexes bind to enhancers and regulate their activity:
H2Bub the snRNA in the inactive 7SK–P-​TEFb complex inter-
H3K36me3 acts with a chromatin remodelling complex to inhibit
H3K79me3
enhancer RNA transcription 213, whereas BRD4–P-​
TEFb and the SEC promote enhancer activation135,214–216
(Table 1). PAF1 occupies enhancers and can restrict the
hyperactivation of a subset of them113. Increased enhancer
activity upon PAF1 depletion correlates with release from
pausing of nearby genes. Furthermore, deletion of PAF1-
Fig. 3 | Post-​translational modification of rNa polymerase ii and histones at active
genes with pausing and at active enhancers. a | Average chromatin occupancy profiles
regulated enhancers resulted in loss of pause–release at
of total RNA polymerase II (Pol II), Pol II with carboxy-​terminal domain (CTD) their cognate genes without greatly affecting transcription
phosphorylated at Ser5 (Ser5P) and at Ser2 (Ser2P) on active genes that exhibit Pol II initiation113. The concordant change of enhancer activa-
pausing and at active enhancers. In general, the occupancy of Pol II is higher at promoters tion and Pol II release without increasing rates of initiation
than at enhancers. Ser5P levels are enriched at promoter regions, whereas Ser2P levels reinforces the notion that enhancers contribute directly to
increase along the gene bodies. b | Average profiles of histone H3 Lys4 (H3K4) the elongation process.
methylation on active genes and enhancers. On active genes, trimethylation of H3K4
(H3K4me3) localizes around the transcription start site (TSS) and is surrounded by Multitasking factors control elongation
H3K4me2; H3K4me1 extends furthest into gene bodies. Enhancers are generally Recent studies identified various regulators of transcrip-
enriched in H3K4me1. c | Average profiles of acetylation of H3K27 (H3K27ac), mono-​
tion elongation that have known roles in other cellular
ubiquitylation of H2B (H2Bub), H3K36me3 and H3K79me3 on active genes and
enhancers. H3K27ac peaks at active enhancers and promoters of active genes. By processes, including factors previously implicated in
contrast, H2Bub, H3K36me3 and H3K79me3 levels are highest in gene bodies. The level DNA replication and the DNA damage response. These
of H3K79me3 is relatively higher towards the 5′ end of gene bodies, and H3K36me3 is factors modulate signal-​induced transcription or directly
enriched towards the 3′ end of genes. H2Bub is more evenly distributed across gene perturb the interplay between the transcription machin-
bodies. TTS, transcription termination site. ery and chromatin. Here, we discuss recent progress in

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Lowly active or
during transcription introduces overwound DNA ahead
PAF1C of the polymerase and underwound DNA behind it, with
inactive enhancers
the former impeding the processivity of the polymer-
Pol II MLL3–COMPASS ase218. Notably, the formation of supercoiled structures
NURD or MLL4–COMPASS around promoter regions could stabilize paused Pol II,
and the activity of DNA topoisomerase IIβ (TOP2β)
can promote the release of pausing219,220. TOP2β activ-
ity can introduce DNA double strand breaks (DSBs) at
promoters of a subset of paused, neuronal-​stimulation
DSIF
NELF
early-​response genes and induces Pol II release and pro-
Gdown1 CTD Ser5 phosphorylation ductive elongation221. Upon the relaxation of supercoils,
CTD Ser2 phosphorylation TOP2β ligates the DSBs; this is facilitated by tyrosyl-​
DSIF phosphorylation DNA phosphodiesterase 2 (TDP2), which hydrolyses the
H3K4me1
–1 Transcript +1 phosphotyrosyl bond between trapped topoisomerases
H3K4me3
Promoter-proximal Signal-induced H3K27ac and DNA222,223.
pausing gene expression H4 acetylation In contrast to TOP2β, which is thought to be active
through enhancer around promoter regions, a recent study suggested
activation
that TOP1 is inactive at promoters but becomes fully
Highly active
enhancers activated by Pol II CTD phosphorylation and relieves
torsional stress to promote efficient transcription
SEC MLL3–COMPASS elongation at gene bodies224. Intriguingly, only CTD
BRD4 or MLL4–COMPASS phosphorylation induced by BRD4, but not P-​TEFb or
eRNA
p300 TFIIH, is capable of stimulating the catalytic activity of
and CBP TOP1 in vitro, suggesting that TOP1 activation by CTD
Cohesin
Mediator phosphorylation is context-​dependent or conformation-​
dependent224. The activity of TOP1 can also be regu-
lated through sumoylation by the protein inhibitor of
activated STAT protein 1 (PIAS1) E3 ligase complex225.
Consistent with a role of TOP1 in promoting produc-
tive elongation, previous work has shown that poisoning
–1 +1
or depletion of TOP1 leads to more severe defects in
transcription at longer genes226,227.
Release of Pol II from
promoter-proximal pausing Underwound DNA behind paused Pol II favours
the opening of dsDNA and thus the formation of R-​
Fig. 4 | regulation of transcription elongation by enhancers. Lowly active or inactive loops 228,229, which can promote Pol II pausing and
enhancers are marked by the presence of monomethylated histone H3 Lys4 (H3K4me1), inhibit productive elongation. Resolution of R-​loops to
which is catalysed by myeloid/lymphoid or mixed-​lineage leukaemia protein 3 promote productive elongation has been described for
(MLL3)-containing or MLL4-containing complex of proteins associated with SET1 ATP-​dependent DNA helicase Q5 (RECQ5), RNase H
(COMPASS), with or without low levels of acetylated H3K27 (H3K27ac), which is catalysed and the helicases senataxin and ATP-​dependent RNA
by p300 or CREB-​binding protein (CBP). RNA polymerase II (Pol II)-associated factor 1
helicase A (DHX9)230,231. RECQ5 is a general elongation
complex (PAF1C) and the nucleosome remodelling and deacetylase (NURD) complex can
bind less active enhancers and prevent their activation and transcription of enhancer RNAs factor that attenuates the movement of Pol II to mini-
(eRNAs). Genes neighbouring the less active enhancers tend to exhibit higher levels of mize co-​transcriptional DNA instability232. Furthermore,
pausing. Following signalling cues, p300, CBP and H4 acetyltransferases (not shown) are RECQ5 induces PIAS1-catalysed sumoylation of TOP1
recruited to enhancers by transcription factors (not shown) to catalyse histone acetylation and thus enhances the recruitment of RNA splicing fac-
at enhancers and promoters. Activation of enhancers is often associated with the tors to actively transcribed genes225. TOP3β is the only
production of eRNAs. The physical interaction between enhancers and promoters requires known RNA topoisomerase in humans, but it is also
cohesin and Mediator complexes. The increased level of histone acetylation contributes to active towards DNA233,234 and consequently can con-
the binding of bromodomain-​containing protein 4 (BRD4) and the super elongation tribute to the resolution of R-​loops235. The recruitment
complex (SEC) to chromatin through their bromodomains and YEATS (YAF9, ENL , AF9, of TOP3β to chromatin requires its interaction with
TAF14 and SAS5) domains, respectively. BRD4-containing and SEC-​containing positive
Tudor domain-​containing protein 3 (TDRD3), which
transcription elongation factor b (P-​TEFb) phosphorylate negative elongation factor (NELF),
DRB sensitivity-​inducing factor (DSIF) and Ser2 residues of the Pol II carboxy-​terminal recognizes methylated histone H3 Arg235. Additionally,
domain (CTD), which releases the paused Pol II into productive transcription elongation. binding to TDRD3 facilitates substrate recognition by
TOP3β and thus stimulates its activity in preventing
R-​loop formation236,237.
understanding how topoisomerases and PARPs regulate
transcription elongation. PARP1 ADP-​ribosylates transcription elongation
factors. PARPs are a group of enzymes that catalyse
The role of topoisomerases in transcription elongation. the transfer of ADP-​ribose derived from NAD+ to tar-
Topoisomerases are enzymes capable of creating tran- get proteins. Aside from the well-​known function of
sient DNA breaks to alleviate DNA supercoiling and thus PARPs in promoting DNA repair and thus maintaining
are essential for cellular processes that require access to genome stability238, numerous studies have implicated
chromatin (reviewed in ref.217). The progression of Pol II PARPs, especially PARP1, in regulating transcription

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

regulation and the control of promoter-​proximal paus-


MLL translocation ing, it will be important to investigate enhancer versus
in paediatric leukaemia promoter-​proximal roles of PARP1 in the regulation of
Pause MYC overexpressed Go transcription elongation.
in solid tumours
Concluding remarks
Understanding the full extent of the regulation of tran-
Fig. 5 | cancer addiction into transcription elongation. The precise regulation of scription elongation at the level of both pause release and
transcription elongation in physiological conditions is mediated by a cohort of processive elongation has been challenging owing to the
elongation factors that modulate different steps of elongation through cooperative and intertwined processes during gene expression, including
competitive mechanisms. Misregulated transcription elongation can be used to support
transcription initiation, splicing and transcription ter-
oncogenesis, as observed in paediatric leukaemia resulting from myeloid/lymphoid or
mination. Although disrupting factors by depletion or
mixed-​lineage leukaemia (MLL) translocations or in solid tumours driven by MYC
overexpression. Skewing the balance of transcription elongation control to higher rates deletion has been used as the major strategy to interro­
of Pol II pause–release and increased productive elongation leads to altered gene gate the function of proteins or protein complexes in
expression and altered splicing, which is essential for oncogenic gene expression controlling transcription elongation, dissecting the
programmes. The regulation of this process can in turn provide therapeutic targets for primary function of a factor in transcription regulation
human diseases that are caused by defects in transcription elongation control. will be greatly aided by the additional development of
specific small molecule inhibitors of different steps
of the transcription process. Also, the use of the auxin-​
elongation (Table 1). PARP1 can regulate transcription by inducible degradation system on factors involved in the
decompacting chromatin through the ADP-​ribosylation process will allow the study of transcription immediately
of histones and chromatin-​binding proteins, including following factor removal, which could distinguish indi-
transcription factors and remodellers in response to rect effects. Small molecule inhibitors are already avail-
heat shock or signal transduction pathways (reviewed able for CDK7, CDK8 and CDK9, which enables the
in refs239,240). ADP-​ribose on chromatin could also be selective targeting of the kinase modules of the TFIIH,
converted into ATP and serve as a local source of energy Mediator and P-​TEFb complexes, respectively253–256.
for chromatin remodelling to promote transcription241. More recently, protein depletion by auxin-​inducible
A recent study using a chemical genetic approach degradation and phthalimide-​based acute degradation
identified the NELF complex as one of the hundreds of has provided new insight into transcription elongation
potential PARP1 substrates242,243. Consistent with PARP1 mechanisms113,132,144,173.
regulation of transcription elongation, small molecule Disambiguating the various roles of multitasking
inhibition of PARP1 or depletion of PARP1 increased the factors in transcription elongation from the other cel-
levels of paused Pol II at a subset of genes242. Moreover, lular processes in which they participate could be rele-
PARP1 can be recruited by TOP2β to become activated vant for understanding human diseases. A recent study
by TOP2β-​introduced DNA breaks to promote tran- used an in vivo functional screening strategy and iden-
scription activation220,244. In addition, PARP1 can ADP-​ tified numerous pausing and elongation factors that are
ribosylate the FACT complex to promote its dissociation required for glioblastoma cell survival257. Mutations that
from chromatin245,246. PARP1 also ADP-​r ibosylates stabilize the SEC, which is already known to be a cofactor
enhancer-​binding factors247,248. For example, PARP1- of HIV TAT136,140 and the oncogenic MLL chimaeras139,
catalysed ADP-​ribosylation of lysine-​specific demethyl­ lead to the developmental disorder CHOPS syndrome204.
ase 5B (KDM5B), which demethylates H3K4me3 at both Therefore, progress in elucidating the molecular mecha-
enhancers and promoters249, prevents the interaction nisms underlying the regulation of transcription elong­
of KDM5B with chromatin and thus could affect the ation will not only contribute to our understanding of
enhancer chromatin landscape248. the intricate control of gene expression programmes but
Independent of its catalytic activity, PARP1 can pro- could also provide new therapeutic strategies for these
mote binding of the pioneer transcription factor and and other diseases linked to transcription elongation
pluripotency factor SOX2 to a subset of enhancers in factors. Given that devastating diseases, such as paedi-
embryonic stem cells, whereas ADP-​ribosylation of atric blood cancers resulting from MLL translocations
SOX2 promotes its degradation during differentia- or solid tumours driven by MYC overexpression, appear
tion247,250. ADP-​ribosylation by PARP1 at enhancers to be diseases of uncontrolled transcription elongation,
can serve as a platform to facilitate the recruitment of such molecular and biochemical studies could have a
chromatin factors such as the nucleosome remodelling major therapeutic impact23,258 (Fig. 5).
and deacetylase (NURD) complex during DNA dam-
Published online xx xx xxxx
age repair251,252. Given the relationship between enhancer

1. Muse, G. W. et al. RNA polymerase is poised for detection of promoter-​proximal pausing and 5. Jonkers, I., Kwak, H. & Lis, J. T. Genome-​wide
activation across the genome. Nat. Genet. 39, enhancer RNAs. dynamics of Pol II elongation and its interplay with
1507–1511 (2007). 3. Ni, Z. et al. P-​TEFb is critical for the maturation of promoter proximal pausing, chromatin, and exons.
2. Core, L. J., Waterfall, J. J. & Lis, J. T. Nascent RNA RNA polymerase II into productive elongation in vivo. eLife 3, e02407 (2014).
sequencing reveals widespread pausing and divergent Mol. Cell. Biol. 28, 1161–1170 (2008). 6. Luo, Z. et al. The super elongation complex
initiation at human promoters. Science 322, 4. Peng, J., Marshall, N. F. & Price, D. H. Identification of family of RNA polymerase II elongation
1845–1848 (2008). a cyclin subunit required for the function of Drosophila factors: gene target specificity and transcriptional
This study introduces the method of global nuclear P-​TEFb. J. Biol. Chem. 273, 13855–13860 output. Mol. Cell. Biol. 32, 2608–2617
run-​on sequencing (GRO-​seq) that allows sensitive (1998). (2012).

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

7. Lin, C. et al. AFF4, a component of the ELL/P-​TEFb 30. Nilson, K. A. et al. Oxidative stress rapidly stabilizes 53. Steinmetz, E. J. et al. Genome-​wide distribution of
elongation complex and a shared subunit of MLL promoter-​proximal paused Pol II across the human yeast RNA polymerase II and its control by Sen1
chimeras, can link transcription elongation to genome. Nucleic Acids Res. 45, 11088–11105 helicase. Mol. Cell 24, 735–746 (2006).
leukemia. Mol. Cell 37, 429–437 (2010). (2017). 54. Lee, C. K., Shibata, Y., Rao, B., Strahl, B. D. & Lieb, J. D.
This study identifies the SEC, which contains 31. Day, D. S. et al. Comprehensive analysis of Evidence for nucleosome depletion at active regulatory
translocation partners of the MLL gene in leukaemia. promoter-​proximal RNA polymerase II pausing regions genome-​wide. Nat. Genet. 36, 900–905
8. Yang, Z. et al. Recruitment of P-​TEFb for stimulation of across mammalian cell types. Genome Biol. 17, 120 (2004).
transcriptional elongation by the bromodomain (2016). 55. Tillo, D. & Hughes, T. R. G+C content dominates
protein Brd4. Mol. Cell 19, 535–545 (2005). 32. Krebs, A. R. et al. Genome-​wide single-​molecule intrinsic nucleosome occupancy. BMC Bioinformatics
9. Jang, M. K. et al. The bromodomain protein Brd4 is a footprinting reveals high RNA polymerase II turnover 10, 442 (2009).
positive regulatory component of P-​TEFb and at paused promoters. Mol. Cell 67, 411–422.e4 56. Valouev, A. et al. Determinants of nucleosome
stimulates RNA polymerase II-​dependent (2017). organization in primary human cells. Nature 474,
transcription. Mol. Cell 19, 523–534 (2005). 33. Lagha, M. et al. Paused Pol II coordinates tissue 516–520 (2011).
10. Yang, Z., Zhu, Q., Luo, K. & Zhou, Q. The 7SK small morphogenesis in the Drosophila embryo. Cell 153, 57. Gilchrist, D. A. et al. Pausing of RNA polymerase II
nuclear RNA inhibits the CDK9/cyclin T1 kinase to 976–987 (2013). disrupts DNA-​specified nucleosome organization to
control transcription. Nature 414, 317–322 (2001). 34. Hendrix, D. A., Hong, J. W., Zeitlinger, J., Rokhsar, D. S. enable precise gene regulation. Cell 143, 540–551
11. Nguyen, V. T., Kiss, T., Michels, A. A. & Bensaude, O. & Levine, M. S. Promoter elements associated with (2010).
7SK small nuclear RNA binds to and inhibits the RNA Pol II stalling in the Drosophila embryo. Proc. Natl This work reports that paused Pol II competes with
activity of CDK9/cyclin T complexes. Nature 414, Acad. Sci. USA 105, 7762–7767 (2008). nucleosomes for occupancy at paused promoters.
322–325 (2001). 35. Li, J. & Gilmour, D. S. Distinct mechanisms of 58. Fenouil, R. et al. CpG islands and GC content dictate
12. Herzel, L., Ottoz, D. S. M., Alpert, T. & transcriptional pausing orchestrated by GAGA factor nucleosome depletion in a transcription-​independent
Neugebauer, K. M. Splicing and transcription touch and M1BP, a novel transcription factor. EMBO J. 32, manner at mammalian promoters. Genome Res. 22,
base: co-transcriptional spliceosome assembly and 1829–1841 (2013). 2399–2408 (2012).
function. Nat. Rev. Mol. Cell Biol. 18, 637–650 (2017). 36. Ginno, P. A., Lim, Y. W., Lott, P. L., Korf, I. & Chedin, F. 59. de Dieuleveult, M. et al. Genome-​wide nucleosome
13. Shilatifard, A., Conaway, R. C. & Conaway, J. W. The GC skew at the 5′ and 3′ ends of human genes links specificity and function of chromatin remodellers in ES
RNA polymerase II elongation complex. Annu. Rev. R-​loop formation to epigenetic regulation and cells. Nature 530, 113–116 (2016).
Biochem. 72, 693–715 (2003). transcription termination. Genome Res. 23, 60. Kubik, S. et al. Nucleosome stability distinguishes two
14. Marshall, N. F. & Price, D. H. Purification of P-​TEFb, 1590–1600 (2013). different promoter types at all protein-​coding genes in
a transcription factor required for the transition into 37. Sanz, L. A. et al. Prevalent, dynamic, and conserved yeast. Mol. Cell 60, 422–434 (2015).
productive elongation. J. Biol. Chem. 270, R-​Loop structures associate with specific epigenomic 61. Vera, D. L. et al. Differential nuclease sensitivity
12335–12338 (1995). signatures in mammals. Mol. Cell 63, 167–178 profiling of chromatin reveals biochemical footprints
15. Shilatifard, A., Lane, W. S., Jackson, K. W., (2016). coupled to gene expression and functional DNA
Conaway, R. C. & Conaway, J. W. An RNA polymerase 38. Lesnik, E. A. & Freier, S. M. Relative thermodynamic elements in maize. Plant Cell 26, 3883–3893 (2014).
II elongation factor encoded by the human ELL gene. stability of DNA, RNA, and DNA:RNA hybrid 62. Henikoff, S., Henikoff, J. G., Sakai, A., Loeb, G. B. &
Science 271, 1873–1876 (1996). duplexes: relationship with base composition Ahmad, K. Genome-​wide profiling of salt fractions
This study implicates a bona fide transcriptional and structure. Biochemistry 34, 10807–10815 maps physical properties of chromatin. Genome Res.
elongation factor as a driver of human cancer. (1995). 19, 460–469 (2009).
16. Hartzog, G. A., Wada, T., Handa, H. & Winston, F. 39. Tous, C. & Aguilera, A. Impairment of transcription 63. Jin, C. et al. H3.3/H2A. Z double variant-​containing
Evidence that Spt4, Spt5, and Spt6 control elongation by R-​loops in vitro. Biochem. Biophys. Res. nucleosomes mark ‘nucleosome-​free regions’ of active
transcription elongation by RNA polymerase II in Commun. 360, 428–432 (2007). promoters and other regulatory regions. Nat. Genet.
Saccharomyces cerevisiae. Genes Dev. 12, 357–369 40. Belotserkovskii, B. P., Soo Shin, J. H. & Hanawalt, P. C. 41, 941–945 (2009).
(1998). Strong transcription blockage mediated by R-​loop 64. Ishii, H., Kadonaga, J. T. & Ren, B. MPE-​seq,
17. Wada, T. et al. DSIF, a novel transcription elongation formation within a G-​rich homopurine-​homopyrimidine a new method for the genome-​wide analysis of
factor that regulates RNA polymerase II processivity, sequence localized in the vicinity of the promoter. chromatin structure. Proc. Natl Acad. Sci. USA 112,
is composed of human Spt4 and Spt5 homologs. Nucleic Acids Res. 45, 6589–6599 (2017). E3457–E3465 (2015).
Genes Dev. 12, 343–356 (1998). 41. Ratmeyer, L., Vinayak, R., Zhong, Y. Y., Zon, G. & 65. Mieczkowski, J. et al. MNase titration reveals
18. Gilmour, D. S. & Lis, J. T. RNA polymerase II interacts Wilson, W. D. Sequence specific thermodynamic and differences between nucleosome occupancy and
with the promoter region of the noninduced hsp70 structural properties for DNA. RNA duplexes. chromatin accessibility. Nat. Commun. 7, 11485
gene in Drosophila melanogaster cells. Mol. Cell. Biol. Biochemistry 33, 5298–5304 (1994). (2016).
6, 3984–3989 (1986). 42. Kellner, W. A., Bell, J. S. & Vertino, P. M. GC skew This study introduces a MNase titration-​based
19. Rougvie, A. E. & Lis, J. T. The RNA polymerase II defines distinct RNA polymerase pause sites in CpG methodology to measure chromatin accessibility.
molecule at the 5′ end of the uninduced hsp70 gene island promoters. Genome Res. 25, 1600–1609 66. Xi, Y., Yao, J., Chen, R., Li, W. & He, X. Nucleosome
of D. melanogaster is transcriptionally engaged. Cell (2015). fragility reveals novel functional states of chromatin
54, 795–804 (1988). 43. Ginno, P. A., Lott, P. L., Christensen, H. C., Korf, I. & and poises genes for activation. Genome Res. 21,
20. Rougvie, A. E. & Lis, J. T. Postinitiation transcriptional Chedin, F. R-​Loop formation is a distinctive 718–724 (2011).
control in Drosophila melanogaster. Mol. Cell. Biol. characteristic of unmethylated human CpG island 67. Chereji, R. V., Ocampo, J. & Clark, D. J. MNase-​
10, 6041–6045 (1990). promoters. Mol. Cell 45, 814–825 (2012). sensitive complexes in yeast: nucleosomes and non-​
21. Zeitlinger, J. et al. RNA polymerase stalling at 44. Lam, E. Y., Beraldi, D., Tannahill, D. & histone barriers. Mol. Cell 65, 565–577.e3 (2017).
developmental control genes in the Drosophila Balasubramanian, S. G-​Quadruplex structures are 68. Kubik, S. et al. A reply to “MNase-​sensitive complexes
melanogaster embryo. Nat. Genet. 39, 1512–1516 stable and detectable in human genomic DNA. in yeast: nucleosomes and non-​histone barriers,” by
(2007). Nat. Commun. 4, 1796 (2013). Chereji et al. Mol. Cell 65, 578–580 (2017).
22. Krumm, A., Hickey, L. B. & Groudine, M. Promoter-​ 45. Shrestha, P., Xiao, S., Dhakal, S., Tan, Z. & Mao, H. 69. Voong, L. N. et al. Insights into nucleosome
proximal pausing of RNA polymerase II defines a Nascent RNA transcripts facilitate the formation of organization in mouse embryonic stem cells through
general rate-​limiting step after transcription initiation. G-​quadruplexes. Nucleic Acids Res. 42, 7236–7246 chemical mapping. Cell 167, 1555–1570 (2016).
Genes Dev. 9, 559–572 (1995). (2014). This study uses a chemical mapping approach to
23. Rahl, P. B. et al. c-​Myc regulates transcriptional pause 46. Eddy, J. et al. G4 motifs correlate with promoter-​ evaluate pausing and nucleosome occupancy at
release. Cell 141, 432–445 (2010). proximal transcriptional pausing in human genes. promoter regions.
24. Reppas, N. B., Wade, J. T., Church, G. M. & Struhl, K. Nucleic Acids Res. 39, 4975–4983 (2011). 70. Weber, C. M., Ramachandran, S. & Henikoff, S.
The transition between transcriptional initiation and 47. Kwak, H., Fuda, N. J., Core, L. J. & Lis, J. T. Precise Nucleosomes are context-​specific, H2A. Z-​modulated
elongation in E. coli is highly variable and often rate maps of RNA polymerase reveal how promoters direct barriers to RNA polymerase. Mol. Cell 53, 819–830
limiting. Mol. Cell 24, 747–757 (2006). initiation and pausing. Science 339, 950–953 (2014).
25. Chen, F. X. et al. PAF1, a molecular regulator of (2013). 71. Hu, G. et al. H2A. Z facilitates access of active and
promoter-​proximal pausing by RNA polymerase II. This study introduces precision run-​on and repressive complexes to chromatin in embryonic stem
Cell 162, 1003–1015 (2015). Ssquencing (PRO-​seq), which allows the mapping of cell self-​renewal and differentiation. Cell Stem Cell 12,
This study identifies PAF1 as a pausing factor that engaged Pol II at base-​pair resolution. 180–192 (2013).
prevents the release of paused Pol II into 48. Schones, D. E. et al. Dynamic regulation of 72. Sainsbury, S., Bernecky, C. & Cramer, P. Structural
productive elongation. nucleosome positioning in the human genome. basis of transcription initiation by RNA polymerase II.
26. Titov, D. V. et al. XPB, a subunit of TFIIH, is a target of Cell 132, 887–898 (2008). Nat. Rev. Mol. Cell Biol. 16, 129–143 (2015).
the natural product triptolide. Nat. Chem. Biol. 7, 49. Mavrich, T. N. et al. Nucleosome organization in 73. Grunberg, S. & Hahn, S. Structural insights into
182–188 (2011). the Drosophila genome. Nature 453, 358–362 transcription initiation by RNA polymerase II.
27. Chen, F., Gao, X. & Shilatifard, A. Stably paused genes (2008). Trends Biochem. Sci. 38, 603–611 (2013).
revealed through inhibition of transcription initiation 50. Ozsolak, F., Song, J. S., Liu, X. S. & Fisher, D. E. 74. Compe, E. & Egly, J. M. TFIIH: when transcription met
by the TFIIH inhibitor triptolide. Genes Dev. 29, High-​throughput mapping of the chromatin structure DNA repair. Nat. Rev. Mol. Cell Biol. 13, 343–354
39–47 (2015). of human promoters. Nat. Biotechnol. 25, 244–248 (2012).
28. Henriques, T. et al. Stable pausing by RNA polymerase (2007). 75. Kim, T. K., Ebright, R. H. & Reinberg, D. Mechanism of
II provides an opportunity to target and integrate 51. Yuan, G. C. et al. Genome-​scale identification of ATP-​dependent promoter melting by transcription
regulatory signals. Mol. Cell 52, 517–528 (2013). nucleosome positions in S. cerevisiae. Science 309, factor IIH. Science 288, 1418–1422 (2000).
29. Shao, W. & Zeitlinger, J. Paused RNA polymerase II 626–630 (2005). 76. Li, J. et al. Kinetic competition between elongation
inhibits new transcriptional initiation. Nat. Genet. 49, 52. Booth, G. T., Wang, I. X., Cheung, V. G. & Lis, J. T. rate and binding of NELF controls promoter-​proximal
1045–1051 (2017). Divergence of a conserved elongation factor and pausing. Mol. Cell 50, 711–722 (2013).
References 27–29 reveal the existence of genes transcription regulation in budding and fission yeast. 77. Glover-​Cutter, K. et al. TFIIH-​associated Cdk7 kinase
with stably paused Pol II. Genome Res. 26, 799–811 (2016). functions in phosphorylation of C-​terminal domain

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Ser7 residues, promoter-​proximal pausing, and a DRB sensitivity-​inducing factor/RNA polymerase II 125. Luo, Z., Lin, C. & Shilatifard, A. The super elongation
termination by RNA polymerase II. Mol. Cell. Biol. 29, complex and RNA. Mol. Cell. Biol. 22, 2918–2927 complex (SEC) family in transcriptional control.
5455–5464 (2009). (2002). Nat. Rev. Mol. Cell Biol. 13, 543–547 (2012).
78. Akhtar, M. S. et al. TFIIH kinase places bivalent marks 103. Schaukowitch, K. et al. Enhancer RNA facilitates NELF 126. Michels, A. A. et al. MAQ1 and 7SK RNA interact with
on the carboxy-​terminal domain of RNA polymerase II. release from immediate early genes. Mol. Cell 56, CDK9/cyclin T complexes in a transcription-​dependent
Mol. Cell 34, 387–393 (2009). 29–42 (2014). manner. Mol. Cell. Biol. 23, 4859–4869 (2003).
79. Harlen, K. M. & Churchman, L. S. The code and 104. Tan, S., Conaway, R. C. & Conaway, J. W. Dissection of 127. Zhou, Q., Li, T. & Price, D. H. RNA polymerase II
beyond: transcription regulation by the RNA transcription factor TFIIF functional domains required elongation control. Annu. Rev. Biochem. 81, 119–143
polymerase II carboxy-​terminal domain. Nat. Rev. for initiation and elongation. Proc. Natl Acad. Sci. USA (2012).
Mol. Cell Biol. 18, 263–273 (2017). 92, 6042–6046 (1995). 128. Lin, C. et al. Dynamic transcriptional events in
80. Allen, B. L. & Taatjes, D. J. The mediator complex: 105. Ishibashi, T. et al. Transcription factors IIS and IIF embryonic stem cells mediated by the super
a central integrator of transcription. Nat. Rev. Mol. enhance transcription efficiency by differentially elongation complex (SEC). Genes Dev. 25,
Cell Biol. 16, 155–166 (2015). modifying RNA polymerase pausing dynamics. 1486–1498 (2011).
81. Soutourina, J. Transcription regulation by the Proc. Natl Acad. Sci. USA 111, 3419–3424 129. Lu, H. et al. AFF1 is a ubiquitous P-​TEFb partner to
mediator complex. Nat. Rev. Mol. Cell Biol. (2014). enable Tat extraction of P-​TEFb from 7SK snRNP and
https://doi.org/10.1038/nrm.2017.115 (2017). 106. Cheng, B. et al. Functional association of Gdown1 with formation of SECs for HIV transactivation. Proc. Natl
82. Jeronimo, C. & Robert, F. Kin28 regulates the RNA polymerase II poised on human genes. Mol. Cell Acad. Sci. USA 111, E15–E24 (2014).
transient association of mediator with core promoters. 45, 38–50 (2012). 130. McNamara, R. P. et al. KAP1 recruitment of the 7SK
Nat. Struct. Mol. Biol. 21, 449–455 (2014). 107. Wu, Y. M. et al. Regulation of mammalian transcription snRNP complex to promoters enables transcription
83. Bentley, D. L. Coupling mRNA processing with by Gdown1 through a novel steric crosstalk revealed elongation by RNA polymerase II. Mol. Cell 61,
transcription in time and space. Nat. Rev. Genet. 15, by cryo-​EM. EMBO J. 31, 3575–3587 (2012). 39–53 (2016).
163–175 (2014). 108. Mullen Davis, M. A., Guo, J., Price, D. H. & Luse, D. S. 131. Ji, X. et al. SR proteins collaborate with 7SK and
84. Brannan, K. et al. mRNA decapping factors and the Functional interactions of the RNA polymerase II-​ promoter-​associated nascent RNA to release paused
exonuclease Xrn2 function in widespread premature interacting proteins Gdown1 and TFIIF. J. Biol. Chem. polymerase. Cell 153, 855–868 (2013).
termination of RNA polymerase II transcription. 289, 11143–11152 (2014). 132. Winter, G. E. et al. BET bromodomain proteins
Mol. Cell 46, 311–324 (2012). 109. DeLaney, E. & Luse, D. S. Gdown1 associates function as master transcription elongation factors
85. Nilson, K. A. et al. THZ1 reveals roles for Cdk7 in efficiently with RNA Polymerase II after promoter independent of CDK9 recruitment. Mol. Cell 67,
co-​transcriptional capping and pausing. Mol. Cell 59, clearance and displaces TFIIF during transcript 5–18.e19 (2017).
576–587 (2015). elongation. PLoS ONE 11, e0163649 (2016). This study uses acute protein degradation to study
86. Ebmeier, C. C. et al. Human TFIIH kinase CDK7 110. Kim, J., Guermah, M. & Roeder, R. G. The human the regulation of transcription elongation by BRD4.
regulates transcription-​associated chromatin PAF1 complex acts in chromatin transcription 133. Rahman, S. et al. The Brd4 extraterminal domain
modifications. Cell Rep. 20, 1173–1186 (2017). elongation both independently and cooperatively with confers transcription activation independent of pTEFb
87. Larochelle, S. et al. Cyclin-​dependent kinase control of SII/TFIIS. Cell 140, 491–503 (2010). by recruiting multiple proteins, including NSD3.
the initiation-​to-elongation switch of RNA polymerase 111. Chen, Y. et al. DSIF, the Paf1 complex, and Tat-​SF1 Mol. Cell. Biol. 31, 2641–2652 (2011).
II. Nat. Struct. Mol. Biol. 19, 1108–1115 (2012). have nonredundant, cooperative roles in RNA 134. Devaiah, B. N. et al. BRD4 is an atypical kinase that
88. Yamaguchi, Y. et al. NELF, a multisubunit complex polymerase II elongation. Genes Dev. 23, 2765–2777 phosphorylates serine2 of the RNA polymerase II
containing RD, cooperates with DSIF to repress RNA (2009). carboxy-​terminal domain. Proc. Natl Acad. Sci. USA
polymerase II elongation. Cell 97, 41–51 (1999). 112. Bai, X. et al. TIF1gamma controls erythroid cell fate by 109, 6927–6932 (2012).
89. Renner, D. B., Yamaguchi, Y., Wada, T., Handa, H. & regulating transcription elongation. Cell 142, 135. Kanno, T. et al. BRD4 assists elongation of both
Price, D. H. A highly purified RNA polymerase II 133–143 (2010). coding and enhancer RNAs by interacting with
elongation control system. J. Biol. Chem. 276, 113. Chen, F. X. et al. PAF1 regulation of promoter-​ acetylated histones. Nat. Struct. Mol. Biol. 21,
42601–42609 (2001). proximal pause release via enhancer activation. 1047–1057 (2014).
90. Palangat, M., Renner, D. B., Price, D. H. & Landick, R. Science 357, 1294–1298 (2017). 136. Sobhian, B. et al. HIV-1 Tat assembles a
A negative elongation factor for human RNA This study reveals a connection between PAF1 multifunctional transcription elongation complex and
polymerase II inhibits the anti-​arrest transcript-​ regulating enhancer activation to regulate pause stably associates with the 7SK snRNP. Mol. Cell 38,
cleavage factor TFIIS. Proc. Natl Acad. Sci. USA 102, release of nearby genes. 439–451 (2010).
15036–15041 (2005). 114. Yu, M. et al. RNA polymerase II-​associated factor 1 137. Cho, W. K. et al. Modulation of the Brd4/P-​TEFb
91. Wen, Y. & Shatkin, A. J. Transcription elongation factor regulates the release and phosphorylation of paused interaction by the human T-​lymphotropic virus type 1
hSPT5 stimulates mRNA capping. Genes Dev. 13, RNA polymerase II. Science 350, 1383–1386 tax protein. J. Virol. 81, 11179–11186 (2007).
1774–1779 (1999). (2015). 138. Garber, M. E. et al. The interaction between HIV-1 Tat
92. Doamekpor, S. K., Sanchez, A. M., Schwer, B., 115. Jaenicke, L. A. et al. Ubiquitin-​dependent turnover of and human cyclin T1 requires zinc and a critical
Shuman, S. & Lima, C. D. How an mRNA capping MYC antagonizes MYC/PAF1C complex accumulation cysteine residue that is not conserved in the murine
enzyme reads distinct RNA polymerase II and Spt5 to drive transcriptional elongation. Mol. Cell 61, CycT1 protein. Genes Dev. 12, 3512–3527 (1998).
CTD phosphorylation codes. Genes Dev. 28, 54–67 (2016). 139. Shilatifard, A. The COMPASS family of histone H3K4
1323–1336 (2014). 116. Marshall, N. F. & Price, D. H. Control of formation of methylases: mechanisms of regulation in development
93. Pei, Y. & Shuman, S. Interactions between fission two distinct classes of RNA polymerase II elongation and disease pathogenesis. Annu. Rev. Biochem. 81,
yeast mRNA capping enzymes and elongation factor complexes. Mol. Cell. Biol. 12, 2078–2090 (1992). 65–95 (2012).
Spt5. J. Biol. Chem. 277, 19639–19648 (2002). 117. Sehgal, P. B., Darnell, J. E. Jr & Tamm, I. 140. He, N. et al. HIV-1 Tat and host AFF4 recruit two
94. Mandal, S. S. et al. Functional interactions of RNA-​ The inhibition by DRB (5,6-dichloro-1-beta-D- transcription elongation factors into a bifunctional
capping enzyme with factors that positively and ribofuranosylbenzimidazole) of hnRNA and mRNA complex for coordinated activation of HIV-1
negatively regulate promoter escape by RNA production in HeLa cells. Cell 9, 473–480 (1976). transcription. Mol. Cell 38, 428–438 (2010).
polymerase II. Proc. Natl Acad. Sci. USA 101, 118. Fujinaga, K. et al. Dynamics of human 141. Liu, K. et al. The super elongation complex drives
7572–7577 (2004). immunodeficiency virus transcription: P-​TEFb neural stem cell fate commitment. Dev. Cell 40,
95. Qiu, Y. & Gilmour, D. S. Identification of regions in phosphorylates RD and dissociates negative effectors 537–551 e6 (2017).
the Spt5 Subunit of DRB Sensitivity-​inducing Factor from the transactivation response element. Mol. Cell. This study shows that SEC can physically interact
(DSIF) that are involved in promoter-​proximal pausing. Biol. 24, 787–795 (2004). with and be recruited by the RBPJ (also known as
J. Biol. Chem. 292, 5555–5570 (2017). 119. Kim, J. B. & Sharp, P. A. Positive transcription CSL) transcription factor to activate Notch
96. Bernecky, C., Plitzko, J. M. & Cramer, P. Structure of a elongation factor B phosphorylates hSPT5 and RNA signalling.
transcribing RNA polymerase II-​DSIF complex reveals polymerase II carboxyl-​terminal domain independently 142. Gardini, A. et al. Integrator regulates transcriptional
a multidentate DNA-​RNA clamp. Nat. Struct. Mol. of cyclin-​dependent kinase-​activating kinase. J. Biol. initiation and pause release following activation.
Biol. 24, 809–815 (2017). Chem. 276, 12317–12323 (2001). Mol. Cell 56, 128–139 (2014).
97. Ehara, H. et al. Structure of the complete elongation 120. Marshall, N. F., Peng, J., Xie, Z. & Price, D. H. Control 143. Wan, L. et al. ENL links histone acetylation to
complex of RNA polymerase II with basal factors. of RNA polymerase II elongation potential by a novel oncogenic gene expression in acute myeloid
Science 357, 921–924 (2017). carboxyl-​terminal domain kinase. J. Biol. Chem. 271, leukaemia. Nature 543, 265–269 (2017).
98. Vos, S. M. et al. Architecture and RNA binding of the 27176–27183 (1996). 144. Erb, M. A. et al. Transcription control by the ENL
human negative elongation factor. eLife 5, e14981 121. Sanso, M. et al. P-​TEFb regulation of transcription YEATS domain in acute leukaemia. Nature 543,
(2016). termination factor Xrn2 revealed by a chemical genetic 270–274 (2017).
99. Jadlowsky, J. K. et al. Negative elongation factor is screen for Cdk9 substrates. Genes Dev. 30, 117–131 References 143 and 144 reveal that binding of
required for the maintenance of proviral latency but (2016). ENL to acetylated histones is required for the
does not induce promoter-​proximal pausing of RNA This study uses a chemical genetic screen to recruitment of the SEC and DOT1L to their target
polymerase II on the HIV long terminal repeat. identify 100 potential substrates of CDK9 that are genes to promote MLL-​rearranged leukaemia.
Mol. Cell. Biol. 34, 1911–1928 (2014). implicated in various steps of transcription. 145. Zhang, Z. et al. Crosstalk between histone
100. Gilchrist, D. A. et al. NELF-​mediated stalling of 122. Wu, C. H. et al. NELF and DSIF cause promoter modifications indicates that inhibition of arginine
Pol II can enhance gene expression by blocking proximal pausing on the hsp70 promoter in methyltransferase CARM1 activity reverses HIV
promoter-​proximal nucleosome assembly. Genes Dev. Drosophila. Genes Dev. 17, 1402–1414 (2003). latency. Nucleic Acids Res. 45, 9348–9360 (2017).
22, 1921–1933 (2008). 123. Yamada, T. et al. P-​TEFb-mediated phosphorylation of 146. Gates, L. A. et al. Acetylation on histone H3 lysine 9
101. Core, L. J. et al. Defining the status of RNA hSpt5 C-​terminal repeats is critical for processive mediates a switch from transcription initiation to
polymerase at promoters. Cell Rep. 2, 1025–1035 transcription elongation. Mol. Cell 21, 227–237 elongation. J. Biol. Chem. 292, 14456–14472
(2012). (2006). (2017).
102. Yamaguchi, Y., Inukai, N., Narita, T., Wada, T. & 124. Smith, E., Lin, C. & Shilatifard, A. The super 147. Takahashi, H. et al. Human mediator subunit MED26
Handa, H. Evidence that negative elongation factor elongation complex (SEC) and MLL in development functions as a docking site for transcription elongation
represses transcription elongation through binding to and disease. Genes Dev. 25, 661–672 (2011). factors. Cell 146, 92–104 (2011).

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

148. Lens, Z. et al. Solution structure of the N-​terminal 172. Klein, B. J. et al. RNA polymerase and transcription enhancers. Nat. Rev. Mol. Cell Biol. 16, 144–154
domain of Mediator subunit MED26 and molecular elongation factor Spt4/5 complex structure. Proc. Natl (2015).
characterization of its interaction with EAF1 and TAF7. Acad. Sci. USA 108, 546–550 (2011). 198. Rollins, R. A., Korom, M., Aulner, N., Martens, A. &
J. Mol. Biol. 429, 3043–3055 (2017). 173. Shetty, A. et al. Spt5 plays vital roles in the control of Dorsett, D. Drosophila nipped-​B protein supports
149. Hu, D. et al. The little elongation complex functions at sense and antisense transcription elongation. sister chromatid cohesion and opposes the stromalin/
initiation and elongation phases of snRNA gene Mol. Cell 66, 77–88.e5 (2017). Scc3 cohesion factor to facilitate long-​range activation
transcription. Mol. Cell 51, 493–505 (2013). 174. Guenther, M. G., Levine, S. S., Boyer, L. A., Jaenisch, R. of the cut gene. Mol. Cell. Biol. 24, 3100–3111
150. Smith, E. R. et al. The little elongation complex & Young, R. A. A chromatin landmark and transcription (2004).
regulates small nuclear RNA transcription. Mol. Cell initiation at most promoters in human cells. Cell 130, 199. Phillips-​Cremins, J. E. et al. Architectural protein
44, 954–965 (2011). 77–88 (2007). subclasses shape 3D organization of genomes during
151. Takahashi, H. et al. MED26 regulates the transcription 175. Fuchs, G., Hollander, D., Voichek, Y., Ast, G. & Oren, M. lineage commitment. Cell 153, 1281–1295
of snRNA genes through the recruitment of little Cotranscriptional histone H2B monoubiquitylation is (2013).
elongation complex. Nat. Commun. 6, 5941 (2015). tightly coupled with RNA polymerase II elongation rate. 200. Kagey, M. H. et al. Mediator and cohesin connect gene
152. Talbert, P. B. & Henikoff, S. Histone variants on the Genome Res. 24, 1572–1583 (2014). expression and chromatin architecture. Nature 467,
move: substrates for chromatin dynamics. Nat. Rev. 176. Vakoc, C. R., Sachdeva, M. M., Wang, H. & Blobel, G. A. 430–435 (2010).
Mol. Cell Biol. 18, 115–126 (2017). Profile of histone lysine methylation across transcribed 201. Schaaf, C. A. et al. Genome-​wide control of RNA
153. Lai, W. K. M. & Pugh, B. F. Understanding nucleosome mammalian chromatin. Mol. Cell. Biol. 26, 9185–9195 polymerase II activity by cohesin. PLoS Genet. 9,
dynamics and their links to gene expression and DNA (2006). e1003382 (2013).
replication. Nat. Rev. Mol. Cell Biol. 18, 548–562 177. Lee, J. S., Smith, E. & Shilatifard, A. The language of 202. Fay, A. et al. Cohesin selectively binds and regulates
(2017). histone crosstalk. Cell 142, 682–685 (2010). genes with paused RNA polymerase. Curr. Biol. 21,
154. Venkatesh, S. & Workman, J. L. Histone exchange, 178. Barski, A. et al. High-​resolution profiling of histone 1624–1634 (2011).
chromatin structure and the regulation of methylations in the human genome. Cell 129, 203. Wu, Y. et al. Drosophila Nipped-​B mutants model
transcription. Nat. Rev. Mol. Cell Biol. 16, 178–189 823–837 (2007). cornelia de lange syndrome in growth and behavior.
(2015). 179. Batta, K., Zhang, Z., Yen, K., Goffman, D. B. & PLoS Genet. 11, e1005655 (2015).
155. Kornberg, R. D. & Lorch, Y. Twenty-​five years of the Pugh, B. F. Genome-​wide function of H2B 204. Izumi, K. et al. Germline gain-​of-function mutations in
nucleosome, fundamental particle of the eukaryote ubiquitylation in promoter and genic regions. Genes AFF4 cause a developmental syndrome functionally
chromosome. Cell 98, 285–294 (1999). Dev. 25, 2254–2265 (2011). linking the super elongation complex and cohesin.
156. Kireeva, M. L. et al. Nucleosome remodeling induced 180. Lee, J. S. et al. Codependency of H2B Nat. Genet. 47, 338–344 (2015).
by RNA polymerase II: loss of the H2A/H2B dimer monoubiquitination and nucleosome reassembly on This study identifies mutations of AFF4 that
during transcription. Mol. Cell 9, 541–552 Chd1. Genes Dev. 26, 914–919 (2012). stabilize the protein as the cause of CHOPS
(2002). 181. Tanny, J. C., Erdjument-​Bromage, H., Tempst, P. & syndrome.
157. Orphanides, G., LeRoy, G., Chang, C. H., Luse, D. S. & Allis, C. D. Ubiquitylation of histone H2B controls RNA 205. Jishage, M. et al. Transcriptional regulation by Pol II(G)
Reinberg, D. FACT, a factor that facilitates transcript polymerase II transcription elongation independently involving mediator and competitive interactions of
elongation through nucleosomes. Cell 92, 105–116 of histone H3 methylation. Genes Dev. 21, 835–847 Gdown1 and TFIIF with Pol II. Mol. Cell 45, 51–63
(1998). (2007). (2012).
158. Bondarenko, V. A. et al. Nucleosomes can form a polar 182. Segala, G., Bennesch, M. A., Pandey, D. P., Hulo, N. & 206. Galbraith, M. D. et al. HIF1A employs CDK8-mediator
barrier to transcript elongation by RNA polymerase II. Picard, D. Monoubiquitination of histone H2B blocks to stimulate RNAPII elongation in response to
Mol. Cell 24, 469–479 (2006). eviction of histone variant H2A.Z. from inducible hypoxia. Cell 153, 1327–1339 (2013).
159. Saunders, A. et al. Tracking FACT and the RNA enhancers. Mol. Cell 64, 334–346 (2016). 207. Donner, A. J., Ebmeier, C. C., Taatjes, D. J. &
polymerase II elongation complex through chromatin 183. Xie, W. et al. RNF40 regulates gene expression in an Espinosa, J. M. CDK8 is a positive regulator of
in vivo. Science 301, 1094–1096 (2003). epigenetic context-​dependent manner. Genome Biol. transcriptional elongation within the serum response
160. Hondele, M. et al. Structural basis of histone 18, 32 (2017). network. Nat. Struct. Mol. Biol. 17, 194–201 (2010).
H2A-​H2B recognition by the essential chaperone 184. McDaniel, S. L. & Strahl, B. D. Shaping the cellular 208. Herz, H. M. et al. Enhancer-​associated H3K4
FACT. Nature 499, 111–114 (2013). landscape with Set2/SETD2 methylation. Cell. Mol. monomethylation by trithorax-​related, the Drosophila
161. Kemble, D. J., McCullough, L. L., Whitby, F. G., Life Sci. 74, 3317–3334 (2017). homolog of mammalian Mll3/Mll4. Genes Dev. 26,
Formosa, T. & Hill, C. P. FACT disrupts nucleosome 185. Carrozza, M. J. et al. Histone H3 methylation by Set2 2604–2620 (2012).
structure by binding H2A-​H2B with conserved peptide directs deacetylation of coding regions by Rpd3S to 209. Hu, D. et al. The MLL3/MLL4 branches of the
motifs. Mol. Cell 60, 294–306 (2015). suppress spurious intragenic transcription. Cell 123, COMPASS family function as major histone H3K4
162. Petesch, S. J. & Lis, J. T. Overcoming the nucleosome 581–592 (2005). monomethylases at enhancers. Mol. Cell. Biol. 33,
barrier during transcript elongation. Trends Genet. 28, 186. Smolle, M. et al. Chromatin remodelers Isw1 and 4745–4754 (2013).
285–294 (2012). Chd1 maintain chromatin structure during 210. Tie, F. et al. CBP-​mediated acetylation of histone H3
163. Teves, S. S., Weber, C. M. & Henikoff, S. Transcribing transcription by preventing histone exchange. lysine 27 antagonizes Drosophila Polycomb silencing.
through the nucleosome. Trends Biochem. Sci. 39, Nat. Struct. Mol. Biol. 19, 884–892 (2012). Development 136, 3131–3141 (2009).
577–586 (2014). 187. Baubec, T. et al. Genomic profiling of DNA 211. Creyghton, M. P. et al. Histone H3K27ac separates
164. Simic, R. et al. Chromatin remodeling protein Chd1 methyltransferases reveals a role for DNMT3B in active from poised enhancers and predicts
interacts with transcription elongation factors and genic methylation. Nature 520, 243–247 (2015). developmental state. Proc. Natl Acad. Sci. USA 107,
localizes to transcribed genes. EMBO J. 22, 188. Neri, F. et al. Intragenic DNA methylation prevents 21931–21936 (2010).
1846–1856 (2003). spurious transcription initiation. Nature 543, 72–77 212. Shen, Y. et al. A map of the cis-​regulatory sequences in
165. Kaplan, C. D., Holland, M. J. & Winston, F. Interaction (2017). the mouse genome. Nature 488, 116–120 (2012).
between transcription elongation factors and mRNA References 187 and 188 demonstrate that 213. Flynn, R. A. et al. 7SK-​BAF axis controls pervasive
3′-end formation at the Saccharomyces cerevisiae DNMT3B recognizes SETD2-mediated H3K36me3 transcription at enhancers. Nat. Struct. Mol. Biol. 23,
GAL10-GAL7 locus. J. Biol. Chem. 280, 913–922 and catalyses intragenic DNA methylation to 231–238 (2016).
(2005). ensure the fidelity of gene transcription. 214. Hertweck, A. et al. T-​Bet activates Th1 genes through
166. Pruneski, J. A., Hainer, S. J., Petrov, K. O. & 189. Zhu, B. et al. The human PAF complex coordinates mediator and the super elongation complex. Cell Rep.
Martens, J. A. The Paf1 complex represses SER3 transcription with events downstream of RNA 15, 2756–2770 (2016).
transcription in Saccharomyces cerevisiae by facilitating synthesis. Genes Dev. 19, 1668–1673 (2005). 215. Loven, J. et al. Selective inhibition of tumor oncogenes
intergenic transcription-​dependent nucleosome 190. Wang, E. et al. Histone H2B ubiquitin ligase RNF20 is by disruption of super-​enhancers. Cell 153, 320–334
occupancy of the SER3 promoter. Eukaryot. Cell 10, required for MLL-​rearranged leukemia. Proc. Natl (2013).
1283–1294 (2011). Acad. Sci. USA 110, 3901–3906 (2013). 216. Lin, C., Garruss, A. S., Luo, Z., Guo, F. & Shilatifard, A.
167. Pavri, R. et al. Histone H2B monoubiquitination 191. Nakanishi, S. et al. Histone H2BK123 The RNA Pol II elongation factor Ell3 marks enhancers
functions cooperatively with FACT to regulate monoubiquitination is the critical determinant for in ES cells and primes future gene activation. Cell 152,
elongation by RNA polymerase II. Cell 125, 703–717 H3K4 and H3K79 trimethylation by COMPASS and 144–156 (2013).
(2006). Dot1. J. Cell Biol. 186, 371–377 (2009). 217. Pommier, Y., Sun, Y., Huang, S. N. & Nitiss, J. L. Roles
168. Krogan, N. J. et al. The Paf1 complex is required 192. Lee, J. S. et al. Histone crosstalk between H2B of eukaryotic topoisomerases in transcription,
for histone H3 methylation by COMPASS and monoubiquitination and H3 methylation mediated by replication and genomic stability. Nat. Rev. Mol. Cell
Dot1p: linking transcriptional elongation to COMPASS. Cell 131, 1084–1096 (2007). Biol. 17, 703–721 (2016).
histone methylation. Mol. Cell 11, 721–729 193. Mohan, M. et al. Linking H3K79 trimethylation to Wnt 218. Corless, S. & Gilbert, N. Effects of DNA supercoiling on
(2003). signaling through a novel Dot1-containing complex chromatin architecture. Biophys. Rev. 8, 51–64
169. Wood, A., Schneider, J., Dover, J., Johnston, M. & (DotCom). Genes Dev. 24, 574–589 (2010). (2016).
Shilatifard, A. The Paf1 complex is essential for 194. Dillon, S. C., Zhang, X., Trievel, R. C. & Cheng, X. The 219. Bunch, H. et al. Transcriptional elongation requires
histone monoubiquitination by the Rad6-Bre1 SET-​domain protein superfamily: protein lysine DNA break-​induced signalling. Nat. Commun. 6,
complex, which signals for histone methylation by methyltransferases. Genome Biol. 6, 227 10191 (2015).
COMPASS and Dot1p. J. Biol. Chem. 278, (2005). 220. Ju, B. G. et al. A topoisomerase IIbeta-​mediated
34739–34742 (2003). 195. Yankulov, K., Blau, J., Purton, T., Roberts, S. & dsDNA break required for regulated transcription.
170. Ng, H. H., Dole, S. & Struhl, K. The Rtf1 component of Bentley, D. L. Transcriptional elongation by RNA Science 312, 1798–1802 (2006).
the Paf1 transcriptional elongation complex is polymerase II is stimulated by transactivators. Cell 77, 221. Madabhushi, R. et al. Activity-​induced DNA breaks
required for ubiquitination of histone H2B. J. Biol. 749–759 (1994). govern the expression of neuronal early-​response
Chem. 278, 33625–33628 (2003). 196. Smith, E. & Shilatifard, A. Enhancer biology and genes. Cell 161, 1592–1605 (2015).
171. Grohmann, D. et al. The initiation factor TFE and the enhanceropathies. Nat. Struct. Mol. Biol. 21, 222. Gomez-​Herreros, F. et al. TDP2 protects transcription
elongation factor Spt4/5 compete for the RNAP clamp 210–219 (2014). from abortive topoisomerase activity and is required
during transcription initiation and elongation. 197. Heinz, S., Romanoski, C. E., Benner, C. & Glass, C. K. for normal neural function. Nat. Genet. 46, 516–521
Mol. Cell 43, 263–274 (2011). The selection and function of cell type-​specific (2014).

www.nature.com/nrm
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

223. Zeng, Z., Cortes-​Ledesma, F., El Khamisy, S. F. & 240. Gupte, R., Liu, Z. & Kraus, W. L. PARPs and ADP-​ ROCK, CDK8, and CDK11. Angew. Chem. Int. Ed Engl.
Caldecott, K. W. TDP2/TTRAP is the major 5′-tyrosyl ribosylation: recent advances linking molecular 48, 8952–8957 (2009).
DNA phosphodiesterase activity in vertebrate cells functions to biological outcomes. Genes Dev. 31, 256. Chao, S. H. et al. Flavopiridol inhibits P-​TEFb and
and is critical for cellular resistance to topoisomerase 101–126 (2017). blocks HIV-1 replication. J. Biol. Chem. 275,
II-​induced DNA damage. J. Biol. Chem. 286, 241. Wright, R. H. et al. ADP-​ribose-derived nuclear ATP 28345–28348 (2000).
403–409 (2011). synthesis by NUDIX5 is required for chromatin 257. Miller, T. E. et al. Transcription elongation factors
224. Baranello, L. et al. RNA polymerase II regulates remodeling. Science 352, 1221–1225 represent in vivo cancer dependencies in glioblastoma.
topoisomerase 1 activity to favor efficient (2016). Nature 547, 355–359 (2017).
transcription. Cell 165, 357–371 (2016). 242. Gibson, B. A. et al. Chemical genetic discovery of This study uses an in vivo functional screening
225. Li, M., Pokharel, S., Wang, J. T., Xu, X. & Liu, Y. PARP targets reveals a role for PARP-1 in transcription strategy and demonstrates the requirement of
RECQ5-dependent SUMOylation of DNA elongation. Science 353, 45–50 (2016). many pausing and elongation factors for
topoisomerase I prevents transcription-​associated This study utilizes a chemical genetic strategy and glioblastoma cell survival.
genome instability. Nat. Commun. 6, 6720 (2015). identifies hundreds of substrates of PARPs, many 258. Liang, K. et al. Therapeutic targeting of MLL
226. Solier, S. et al. Transcription poisoning by of which are involved in transcription elongation, degradation pathways in MLL-​rearranged leukemia.
topoisomerase I is controlled by gene length, splice including the NELF complex. Cell 168, 59–72.e13 (2017).
sites, and miR-142-3p. Cancer Res. 73, 4830–4839 243. Leutert, M., Pedrioli, D. M. & Hottiger, M. O. 259. Kuehner, J. N., Pearson, E. L. & Moore, C. Unravelling
(2013). Identification of PARP-​specific ADP-​ribosylation the means to an end: RNA polymerase II transcription
227. King, I. F. et al. Topoisomerases facilitate transcription targets reveals a regulatory function for ADP-​ termination. Nat. Rev. Mol. Cell Biol. 12, 283–294
of long genes linked to autism. Nature 501, 58–62 ribosylation in transcription elongation. Mol. Cell 63, (2011).
(2013). 181–183 (2016). 260. Porrua, O. & Libri, D. Transcription termination and
228. Zhang, X. et al. Attenuation of RNA polymerase II 244. Calderwood, S. K. A critical role for topoisomerase IIb the control of the transcriptome: why, where and how
pausing mitigates BRCA1-associated R-​loop and DNA double strand breaks in transcription. to stop. Nat. Rev. Mol. Cell Biol. 16, 190–202 (2015).
accumulation and tumorigenesis. Nat. Commun. 8, Transcription 7, 75–83 (2016). 261. Fong, N. et al. Effects of transcription elongation rate
15908 (2017). 245. Huang, J. Y. et al. Modulation of nucleosome-​binding and Xrn2 exonuclease activity on RNA polymerase II
229. Skourti-​Stathaki, K. & Proudfoot, N. J. A double- activity of FACT by poly(ADP-​ribosyl)ation. Nucleic termination suggest widespread kinetic competition.
edged sword: R loops as threats to genome integrity Acids Res. 34, 2398–2407 (2006). Mol. Cell 60, 256–267 (2015).
and powerful regulators of gene expression. Genes Dev. 246. Heo, K. et al. FACT-​mediated exchange of histone 262. Zhang, H., Rigo, F. & Martinson, H. G. Poly(A) signal-​
28, 1384–1396 (2014). variant H2AX regulated by phosphorylation of H2AX dependent transcription termination occurs through a
230. Tuduri, S. et al. Topoisomerase I suppresses genomic and ADP-​ribosylation of Spt16. Mol. Cell 30, 86–97 conformational change mechanism that does not
instability by preventing interference between (2008). require cleavage at the poly(A) site. Mol. Cell 59,
replication and transcription. Nat. Cell Biol. 11, 247. Gao, F., Kwon, S. W., Zhao, Y. & Jin, Y. PARP1 437–448 (2015).
1315–1324 (2009). poly(ADP-​ribosyl)ates Sox2 to control Sox2 protein 263. Libri, D. Endless quarrels at the end of genes.
231. Groh, M., Lufino, M. M., Wade-​Martins, R. & Gromak, levels and FGF4 expression during embryonic stem Mol. Cell 60, 192–194 (2015).
N. R-​Loops associated with triplet repeat expansions cell differentiation. J. Biol. Chem. 284, 264. Ni, Z., Schwartz, B. E., Werner, J., Suarez, J. R. &
promote gene silencing in Friedreich ataxia and 22263–22273 (2009). Lis, J. T. Coordination of transcription, RNA
fragile X syndrome. PLoS Genet. 10, e1004318 248. Krishnakumar, R. & Kraus, W. L. PARP-1 regulates processing, and surveillance by P-​TEFb kinase on heat
(2014). chromatin structure and transcription through a shock genes. Mol. Cell 13, 55–65 (2004).
232. Saponaro, M. et al. RECQL5 controls transcript KDM5B-​dependent pathway. Mol. Cell 39, 736–749 265. Laitem, C. et al. CDK9 inhibitors define elongation
elongation and suppresses genome instability (2010). checkpoints at both ends of RNA polymerase
associated with transcription stress. Cell 157, 249. Kidder, B. L., Hu, G. & Zhao, K. KDM5B focuses H3K4 II-​transcribed genes. Nat. Struct. Mol. Biol. 22,
1037–1049 (2014). methylation near promoters and enhancers during 396–403 (2015).
233. Wilson-​Sali, T. & Hsieh, T. S. Preferential cleavage of embryonic stem cell self-​renewal and differentiation. 266. Skourti-​Stathaki, K., Kamieniarz-​Gdula, K. &
plasmid-​based R-​loops and D-​loops by Drosophila Genome Biol. 15, R32 (2014). Proudfoot, N. J. R-​Loops induce repressive chromatin
topoisomerase IIIbeta. Proc. Natl Acad. Sci. USA 99, 250. Liu, Z. & Kraus, W. L. Catalytic-​independent functions marks over mammalian gene terminators. Nature
7974–7979 (2002). of PARP-1 determine Sox2 pioneer activity at 516, 436–439 (2014).
234. Ahmad, M. et al. Topoisomerase 3beta is the major intractable genomic loci. Mol. Cell 65, 589–603.9 267. Skourti-​Stathaki, K., Proudfoot, N. J. & Gromak, N.
topoisomerase for mRNAs and linked to (2017). Human senataxin resolves RNA/DNA hybrids formed
neurodevelopment and mental dysfunction. Nucleic 251. Chou, D. M. et al. A chromatin localization screen at transcriptional pause sites to promote Xrn2-
Acids Res. 45, 2704–2713 (2017). reveals poly (ADP ribose)-regulated recruitment of the dependent termination. Mol. Cell 42, 794–805
235. Yang, Y. et al. Arginine methylation facilitates the repressive polycomb and NuRD complexes to sites of (2011).
recruitment of TOP3B to chromatin to prevent R loop DNA damage. Proc. Natl Acad. Sci. USA 107, 268. Zhao, D. Y. et al. SMN and symmetric arginine
accumulation. Mol. Cell 53, 484–497 (2014). 18475–18480 (2010). dimethylation of RNA polymerase II C-​terminal domain
236. Siaw, G. E., Liu, I. F., Lin, P. Y., Been, M. D. & 252. Spruijt, C. G. et al. ZMYND8 co-​localizes with NuRD control termination. Nature 529, 48–53 (2016).
Hsieh, T. S. DNA and RNA topoisomerase activities of on target genes and regulates poly(ADP-​Ribose)- 269. Xu, Y. et al. Architecture of the RNA polymerase
Top3beta are promoted by mediator protein tudor dependent recruitment of GATAD2A/NuRD to sites of II-​Paf1C-​TFIIS transcription elongation complex.
domain-​containing protein 3. Proc. Natl Acad. Sci. DNA damage. Cell Rep. 17, 783–798 (2016). Nat. Commun. 8, 15741 (2017).
USA 113, E5544–E5551 (2016). 253. Kwiatkowski, N. et al. Targeting transcription
237. Goto-​Ito, S., Yamagata, A., Takahashi, T. S., Sato, Y. & regulation in cancer with a covalent CDK7 inhibitor. Author contributions
Fukai, S. Structural basis of the interaction between Nature 511, 616–620 (2014). F.X.C., E.R.S. and A.S. researched data for the article, made
topoisomerase IIIbeta and the TDRD3 auxiliary factor. 254. Pelish, H. E. et al. Mediator kinase inhibition further substantial contributions to the discussion of content, wrote
Sci. Rep. 7, 42123 (2017). activates super-​enhancer-associated genes in AML. the article and reviewed and edited the manuscript before
238. Ray Chaudhuri, A. & Nussenzweig, A. The multifaceted Nature 526, 273–276 (2015). submission.
roles of PARP1 in DNA repair and chromatin This study shows that inhibition of the Mediator-​
remodelling. Nat. Rev. Mol. Cell Biol. 18, 610–621 associated kinases CDK8 and CDK19 with Competing interests
(2017). cortistatin A leads to activation of super-​enhancers The authors declare no competing interests.
239. Hottiger, M. O. Nuclear ADP-​ribosylation and its role and upregulation of super-​enhancer-associated
in chromatin plasticity, cell differentiation, and genes. Publisher’s note
epigenetics. Annu. Rev. Biochem. 84, 227–263 255. Cee, V. J., Chen, D. Y., Lee, M. R. & Nicolaou, K. C. Springer Nature remains neutral with regard to jurisdictional
(2015). Cortistatin A is a high-​affinity ligand of protein kinases claims in published maps and institutional affiliations.

Nature Reviews | Molecular Cell Biology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

You might also like