Review Article
Regulation of Promoter Proximal Pausing of RNA Polymerase II in Metazoans

https://doi.org/10.1016/j.jmb.2021.166897Get rights and content

Highlights

  • Promoter proximal pausing of RNA Polymerase II is a critical early elongation step.

  • Pausing is governed by a variety of intrinsic and extrinsic interactions.

  • DSIF and NELF are critical for establishing and maintaining pausing.

  • TFIID regulates both promoter proximal pausing and pause release.

  • P-TEFb regulates release of promoter proximal pausing by phosphorylation of DSIF.

Abstract

Regulation of transcription is a tightly choreographed process. The establishment of RNA polymerase II promoter proximal pausing soon after transcription initiation and the release of Pol II into productive elongation are key regulatory processes that occur in early elongation. We describe the techniques and tools that have become available for the study of promoter proximal pausing and their utility for future experiments. We then provide an overview of the factors and interactions that govern a multipartite pausing process and address emerging questions surrounding the mechanism of RNA polymerase II’s subsequent advancement into the gene body. Finally, we address remaining controversies and future areas of study.

Introduction

Eukaryotic transcription is a highly regulated process that requires precise temporal and spatial coordination of a multitude of factors at the initiation, elongation, and termination stages. Promoter proximal pausing of RNA polymerase II (Pol II) has emerged as a significant step in the canonical transcription cycle. In metazoans, transcription initiation is followed by an accumulation of Pol II ~30–60 nt downstream of the transcription start site.1 This step is not present in Saccharomyces cerevisiae and Caenorhabditis elegans, likely owing to the absence of the key negative elongation factor (NELF). The first evidence of metazoan Pol II pausing in vivo emerged when the Chambon lab, using a nuclear run-on assay, observed a concentration of Pol II at the 5′end of the beta-globin gene in nuclei from mature hen erythrocytes that were anticipated to be transcriptionally silent.2 Similar phenomena were later observed on mammalian c-myc,3, 4, 5 HIV-16 and on non-induced Drosophila heat shock genes.7, 8 The study of the Drosophila hsp70 heat shock gene by Gilmour and Lis was particularly groundbreaking. Using protein-DNA crosslinking with UV light, a predecessor to the widely used chromatin immunoprecipitation techniques of today, they determined that a single Pol II molecule associates with the region between −12 and +65 on the non-induced hsp70 gene.7 Subsequent work by Rougvie and Lis determined that the Pol II at the 5′end of genes in Drosophila is transcriptionally engaged8 and subsequent permanganate footprinting analyses in living cells revealed the transcription bubble associated with these Pol II.9 The question remained, however, as to whether the paused Pol II was an idiosyncrasy of a small number of genes or a more general phenomenon. The advent of genomic methods provided resounding support for the latter, leading to the notion that promoter proximal pausing is a ubiquitous step in the transcription cycle for most if not all protein encoding genes in mammals and Drosophila.10, 11, 12, 13 Pausing has been linked to a number of regulatory functions, including developmental control 12, 14, 15, 16and the maintenance of transcriptionally permissive, nucleosome-free regions around promoters.17, 18 Indeed, it is this latter observation that can explain the seemingly contradictory finding that depletion of the pausing factor NELF results in a decrease rather than an increase in expression of some genes.18

Promoter proximal pausing requires at least two factors which function cooperatively: DRB sensitivity-inducing factor (DSIF) and NELF.19, 20, 21, 22 The cyclin dependent kinase positive transcription elongation factor b (P-TEFb) appears to facilitate pause release by phosphorylating Pol II, DSIF, and NELF, resulting in the dissociation of NELF from the elongation complex and the transition of DSIF from a negative elongation factor to a positive elongation factor.21, 23, 24 Extensive genome-wide and biochemical studies have provided a general framework for how pausing occurs, yet many of the underlying mechanisms remain an area of inquiry. First, the exact mechanism through which DSIF and NELF promote pausing, particularly with regards to the role of DSIF, has yet to be fully elucidated. Several factors in addition to NELF and DSIF have been implicated in pausing, most notable among them transcription factor II D (TFIID), GAGA factor, and RNA polymerase II-associated factor 1 complex (PAF1C), yet the exact role of each of these factors and the nature of their interactions with the paused complex requires further study. Finally, additional considerations, such as the role of DNA/RNA sequence, the duration and stability of the pause, and the relationship between Pol II pausing and chromatin architecture remain areas of continuing or emerging research and even debate.

Promoter proximal pausing of Pol II is governed by several intrinsic and extrinsic elements. Increasing evidence points to DNA and RNA sequence playing a role in regulating promoter proximal pausing. Enrichment of promoter elements and pause motifs, as well as GC content, likely influence the stability and efficiency of the pause.25, 26, 27 Experiments in Drosophila show that disrupting downstream promoter elements can shift the location of the pause on the hsp70 gene, suggesting that pause position is heavily reliant on DNA sequence, but it is unclear whether this result can be widely extrapolated for other promoters.28 Indeed, contrasting evidence has shown that the location of the pause depends on the kinetics of the elongating Pol II; lowering nucleotide concentrations or replacing the wild type Pol II with a slowly elongating mutant shifts the location of the pause upstream.29 Thus, the role of intrinsic Pol II-nucleic acid interactions in pausing remains a topic of ongoing controversy.

Extrinsic interactions, such as those between Pol II and DSIF, NELF, and P-TEFb, as well as between Pol II and accessory factors such as TFIID and others play critical roles in establishing the pause and governing the dynamics of pause stability and pause release. Interactions between the nucleic acid scaffold and DSIF and NELF are also likely significant contributors to the pausing mechanism. Moreover, since the discovery of Pol II pausing, the role of chromatin architecture has been a subject of controversy. Of particular interest has been the +1 nucleosome, which may play a significant role in enhancing Pol II pausing in humans,30 but likely affects Drosophila Pol II promoter proximal pausing in a promoter-specific manner.31

Taken together, the current body of knowledge about Pol II promoter proximal pausing suggests a multipartite mechanism that relies on the interplay between intrinsic and extrinsic interactions, as well as on the promoter and chromatin context of the pause. This review distinguishes itself from other recent reviews on Pol II elongation and promoter proximal pausing in several ways.32, 33, 34, 35, 36 First, we provide an overview of the broad range of the tools available for studying promoter proximal pausing, including in vivo, cell free, and genomic techniques. Furthermore, we describe the factors involved in promoter proximal pausing with a particular emphasis on the structural and molecular mechanisms of DSIF and NELF function. We also discuss the emerging evidence that suggests TFIID plays a significant and context-dependent role as a regulator of promoter proximal pausing. Additionally, we discuss and attempt to resolve some of the controversies regarding the role of chromatin architecture in promoter proximal pausing, with an emphasis on the differences between human and Drosophila systems. Finally, we provide a holistic model for Pol II pausing that illustrates the various protein factors implicated in pausing and their interactions and reconciles some of the apparent contradictions in the current body of literature.

Section snippets

Tools to study pausing

In the almost three decades since the discovery of promoter proximal pausing, numerous tools have emerged to facilitate its study (Table 1). Genome-wide techniques like ChIP-seq37 and the higher resolution ChIP-exo38, 39 and ChIP-nexus40, 41 have emerged as powerful tools that can be utilized to determine the location of Pol II and its binding partners. These techniques involve crosslinking proteins to DNA in vivo followed by immunoprecipitation of protein-DNA adducts. ChIP-exo and ChIP-nexus

DSIF

DSIF is a two-subunit transcription factor composed of Spt4 and Spt5 and is widely conserved across eukaryotes.75 In metazoans, DSIF acts to stimulate or repress transcription elongation, depending on its phosphorylation state. The pausing function of DSIF was first discovered by the Handa lab as an activity that rendered Pol II transcription sensitive to inhibition by DRB, a nucleoside analog.19 DSIF associates with the Pol II elongation complex after the transcription of at least eighteen

NELF

NELF was first identified as a four-subunit complex (NELF-A, -B, -C/D isoforms, -E) that works cooperatively with DSIF to repress Pol II elongation.20, 79 NELF is recruited to the elongation complex by DSIF22 and plays a multifaceted role in establishing and maintaining promoter proximal pausing.

Phosphorylation-dependent regulation of the pause

P-TEFb, predominantly composed of Cdk995 and Cyclin T in Drosophila96 and Cdk995 and Cyclin T1 or Cyclin T2 in human cells97 is broadly considered to be critical for the release of Pol II from the pause and its transition into productive elongation. P-TEFb is frequently found as a part of larger complexes. In its inhibited form, P-TEFb is associated with the 7SK noncoding RNA and the MeCPE, LARP7, and HEXIM factors.98 In its most active form, P-TEFb is part of the multi-subunit super-elongation

TFIID

TFIID is a general transcription factor that provides the foundation for assembling a preinitiation complex (PIC). The relationship between TFIID and Pol II promoter proximal pausing remains a topic of some controversy. In 2019, the Biswas lab used co-immunoprecipitation experiments to show that the TAF subunit of TFIID interacts with components of the SEC both in mammalian cells and in vitro. AF9 and EAF1, two SEC subunits, both have serine-rich domains that are required for this interaction.

GAGA factor

GAGA factor (GAF) is a sequence-specific DNA binding factor that has been shown to promote promoter proximal pausing in Drosophila. Early work from the Lis lab indicated that point mutations in the GAGA element significantly decrease pausing on the Drosophila hsp70 promoter.118, 119 However, it was unclear whether the loss of paused Pol II was due to disruption of pausing or to diminished transcription initiation, a prerequisite for pausing. The Gilmour lab later established that the GAGA

PAF1C

The multi-subunit Paf1 complex (PAF1C) has been implicated in promoter proximal pausing but its role is controversial. Initial studies by the Shilatifard group showed that PAF1C loss results in release of paused Pol II into gene bodies in both Drosophila and human HCT116 and MCF7 cell lines, with a greater effect observed at highly paused genes. PAF1 knockdown also results in increased SEC recruitment and Pol II CTD phosphorylation at the Ser2 position.124 Hence, they proposed that PAF1C helps

Additional proteins

Since the discovery of DSIF and NELF, many more proteins have been implicated in promoter proximal pausing of Pol II and it is likely that more will be discovered in the future. We discuss some of them in the following section but note that the complete picture remains to be fully resolved.

Recently, the pre-exon junction complex (pre-EJC), which regulates expression of long genes in Drosophila has also been identified as an intron length-dependent facilitator of Pol II pausing. Knockdown of the

Role of sequence in pausing

Promoter elements such as GAGA, Inverted GAGA, Inr, and DPE are enriched at paused genes in Drosophila.25, 42 Some of these, such as the Inr and DPE elements, are TFIID binding sites and are likely key players in regulating differential TFIID function, as discussed above. In Drosophila, sequence can act as a particularly strong contributor to Pol II pause stability. Reporter-ChIP-nexus experiments, which detect pausing on plasmids transfected into cells, have shown that an Inr variant with a G

Drosophila versus human and the role of the +1 nucleosome

The role of nucleosome organization, in particular the positioning of the +1 nucleosome, in establishing and maintaining Pol II promoter proximal pausing has been the subject of several contradictory studies. Early experiments by the Luse and Kingston labs established that partially purified Pol II elongation complexes from HeLa cells pause on templates containing assembled nucleosomes.134, 135 Mapping of micrococcal nuclease cuts in HeLa nuclei also determined that human hsp70 is nucleosomal

Duration of the pause

The duration of the pause and whether Pol II, upon release, transitions to productive elongation or prematurely terminates transcription impacts the level of gene expression. These dynamics were initially studied by blocking transcription initiation with the chemical triptolide, which covalently inhibits the ATPase activity of the TFIIH subunit XPB.142This ATPase activity plays a critical role in overcoming a block to initiation imposed by XPB.143, 144 The stability of the paused Pol II is then

Open questions and future directions

Promoter proximal pausing of Pol II requires NELF and DSIF and may also involve accessory factors such as TFIID, PAF1, GAF (in Drosophila), the pre-EJC, and FACT, as well as chromatin remodelers and histone modifiers (Figure 5). However, for many of these factors, how they promote pausing in the context of the Pol II-DSIF-NELF complex remains a mystery. Do they associate directly with the elongation complex or do some of them, such as FACT and other chromatin-associated factors, mediate pausing

Accession numbers

The following structures were used to generate the figures in this review: PDB ID: 6GML, structure of human Pol II-NELF-DSIF complex; PDB ID: 6GMH, structure of human Pol II-DSIF-SPT6-PAF1C; PDB ID: 5OIK, structure of Pol II-DSIF; PDB ID: 6ASX, structure of paused bacterial RNA Polymerase; PDB ID: 6ALF, structure of post-translocated bacterial RNA Polymerase.

CRediT authorship contribution statement

Roberta Dollinger: Writing - original draft, Writing - review & editing. David S. Gilmour: Writing - review & editing, Funding acquisition.

Acknowledgements

Funding for this work was provided by NIH grant GM047477 to David S. Gilmour. We thank Seychelle M. Vos for thoughtful reading of the manuscript.

Declarations of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References (148)

  • Y. Qiu et al.

    Identification of regions in the Spt5 subunit of DRB sensitivity-inducing factor (DSIF) that are involved in promoter-proximal pausing

    J. Biol. Chem.

    (2017)
  • C.B. Ball et al.

    Use of the nuclear walk-on methodology to determine sites of RNA polymerase II initiation and pausing and quantify nascent RNAs in cells

    Methods

    (2019)
  • T. Nojima et al.

    Mammalian NET-Seq reveals genome-wide nascent transcription coupled to RNA processing

    Cell

    (2015)
  • A. Mayer et al.

    Native elongating transcript sequencing reveals human transcriptional activity at nucleotide resolution

    Cell

    (2015)
  • C.M. Weber et al.

    Nucleosomes are context-specific, H2A.Z-modulated barriers to RNA polymerase

    Mol. Cell

    (2014)
  • N.B. Reppas et al.

    The transition between transcriptional initiation and elongation in E. coli is highly variable and often rate limiting

    Mol. Cell

    (2006)
  • S.H. Chao et al.

    Flavopiridol inactivates P-TEFb and blocks most RNA polymerase II transcription in vivo

    J. Biol. Chem.

    (2001)
  • T. Henriques et al.

    Stable pausing by RNA polymerase II provides an opportunity to target and integrate regulatory signals

    Mol. Cell

    (2013)
  • Z. Zhang et al.

    Negative elongation factor NELF represses human immunodeficiency virus transcription by pausing the RNA polymerase II complex

    J. Biol. Chem.

    (2007)
  • T. Kulinski et al.

    The apical loop of the HIV-1 TAR RNA hairpin is stabilized by a cross-loop base pair

    J. Biol. Chem.

    (2003)
  • D. Wang et al.

    Structural basis of transcription: role of the trigger loop in substrate specificity and catalysis

    Cell

    (2006)
  • K. Schaukowitch et al.

    Enhancer RNA facilitates NELF release from immediate early genes

    Mol. Cell

    (2014)
  • T. Narita et al.

    NELF interacts with CBC and participates in 3' end processing of replication-dependent histone mRNAs

    Mol. Cell

    (2007)
  • N.F. Marshall et al.

    Purification of P-TEFb, a transcription factor required for the transition into producitve elongation

    J. Biol. Chem.

    (1995)
  • J. Peng et al.

    Identification of a cyclin subunit required for the function of Drosophila P-TEFb

    J. Biol. Chem.

    (1998)
  • N. He et al.

    HIV-1 Tat and host AFF4 recruit two transcription elongation factors into a bifunctional complex for coordinated activation of HIV-1 transcription

    Mol. Cell

    (2010)
  • I. Jonkers et al.

    Getting up to speed with transcription elongation by RNA polymerase II

    Nature Rev. Mol. Cell Biol.

    (2015)
  • P. Gariglio et al.

    Clustering of RNA polymerase B molecules in the 5' moiety of the adult beta-globin gene of hen erythrocytes

    Nucleic Acids Res.

    (1981)
  • D.L. Bentley et al.

    A block to elongation is largely responsible for decreased transcription of c-myc in differentiated HL60 cells

    Nature

    (1986)
  • D. Eick et al.

    Transcriptional arrest within the first exon is a fast control mechanism in c-myc gene expression

    Nucleic Acids Res.

    (1986)
  • A. Nepveu et al.

    Intragenic pausing and anti-sense transcription within the murine c-myc locus

    EMBO J.

    (1986)
  • S. Kao et al.

    Anti-termination of transcription within the long terminal repeat of HIV-1 by tat gene product

    Nature

    (1987)
  • D.S. Gilmour et al.

    RNA polymerase II Interacts with the promoter region of the noninduced hsp7O Gene in Drosophila melanogaster cells

    Mol. Cell. Biol.

    (1986)
  • C. Giardina et al.

    Promoter melting and TFIID complexes on Drosophila genes in vivo

    Genes Dev.

    (1992)
  • G.W. Muse et al.

    RNA polymerase is poised for activation across the genome

    Nature Genet.

    (2007)
  • J. Zeitlinger et al.

    RNA polymerase stalling at developmental control genes in the Drosophila melanogaster embryo

    Nature Genet.

    (2007)
  • L.J. Core et al.

    Nascent RNA sequencing reveals widespread pausing and divergent initiation at human promoters

    Science

    (2008)
  • X. Wang et al.

    Transcription elongation controls cell fate specification in the Drosophila embryo

    Genes Dev.

    (2007)
  • A.N. Boettiger et al.

    Synchronous and stochastic patterns of gene activation in the Drosophila embryo

    Science

    (2009)
  • D.A. Gilchrist et al.

    NELF-mediated stalling of Pol II can enhance gene expression by blocking promoter-proximal nucleosome assembly

    Genes Dev.

    (2008)
  • T. Wada et al.

    DSIF, a novel transcription elongation factor that regulates RNA polymerase II processivity, is composed of human Spt4 and Spt5 homologs

    Genes Dev.

    (1998)
  • C.H. Wu et al.

    NELF and DSIF cause promoter proximal pausing on the hsp70 promoter in Drosophila

    Genes Dev.

    (2003)
  • A. Missra et al.

    Interactions between DSIF (DRB sensitivity inducing factor), NELF (negative elongation factor), and the Drosophila RNA polymerase II transcription elongation complex

    Proc. Natl. Acad. Sci. USA

    (2010)
  • D.A. Hendrix et al.

    Promoter elements associated with RNA Pol II stalling in the Drosophila embryo

    PNAS

    (2008)
  • S. Gressel et al.

    CDK9-dependent RNA polymerase II pausing controls transcription initiation

    Elife

    (2017)
  • W. Shao et al.

    Reporter-ChIP-nexus reveals strong contribution of the Drosophila initiator sequence to RNA polymerase pausing

    Elife

    (2019)
  • H. Kwak et al.

    Precise maps of RNA Polymerase reveal how promoters direct initiation and pausing

    Science

    (2013)
  • S. Jimeno-Gonzalez et al.

    A positioned +1 nucleosome enhances promoter-proximal pausing

    Nucleic Acids Res.

    (2015)
  • J. Li et al.

    Distinct mechanisms of transcriptional pausing orchestrated by GAGA factor and M1BP, a novel transcription factor

    EMBO J.

    (2013)
  • F.X. Chen et al.

    Born to run: control of transcription elongation by RNA polymerase II

    Nature Rev. Mol. Cell Biol.

    (2018)
  • Cited by (37)

    • New roles for elongation factors in RNA polymerase II ubiquitylation and degradation

      2023, Biochimica et Biophysica Acta - Gene Regulatory Mechanisms
    View all citing articles on Scopus
    View full text