initial transcribed region sequences influence the composition and...

13
Initial transcribed region sequences influence the composition and functional properties of the bacterial elongation complex Padraig Deighan, 1 Chirangini Pukhrambam, 2 Bryce E. Nickels, 2 and Ann Hochschild 1,3 1 Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA; 2 Department of Genetics, Waksman Institute, Rutgers University, Piscataway, New Jersey 08854, USA The bacterial RNA polymerase (RNAP) holoenzyme consists of a catalytic core enzyme (a 2 bb'v) in complex with a s factor that is essential for promoter recognition and transcription initiation. During early elongation, the stability of interactions between s and the remainder of the transcription complex decreases. Nevertheless, there is no mechanistic requirement for release of s upon the transition to elongation. Furthermore, s can remain associated with RNAP during transcription elongation and influence regulatory events that occur during transcription elongation. Here we demonstrate that promoter-like DNA sequence elements within the initial transcribed region that are known to induce early elongation pausing through sequence-specific interactions with s also function to increase the s content of downstream elongation complexes. Our findings establish s-dependent pausing as a mechanism by which initial transcribed region sequences can influence the composition and functional properties of the transcription elongation complex over distances of at least 700 base pairs. [Keywords: RNA polymerase; s factor; transcription initiation; promoter-proximal pausing; transcription elongation complex] Supplemental material is available for this article. Received September 10, 2010; revised version accepted November 5, 2010. The s subunit of bacterial RNA polymerase (RNAP) is required for promoter-specific transcription initiation (Gross et al. 1998). When complexed with the RNAP core enzyme (subunit structure a 2 bb9v), different s factors specify the recognition of different classes of promoters (Gruber and Gross 2003). The primary s factor in Escher- ichia coli, s 70 , typically directs transcription initiation from promoters defined by two conserved hexameric DNA sequence elements, termed the 10 and 35 ele- ments for their relationship to the transcription start site (position +1). During the transition from transcrip- tion initiation to transcription elongation, the growth of the nascent RNA destabilizes the interaction between s 70 and the RNAP core enzyme (Mekler et al. 2002; Murakami et al. 2002; Vassylyev et al. 2002; Nickels et al. 2005), but the complete release of s 70 is not required for entry into the elongation phase of transcription (Ring et al. 1996; Bar-Nahum and Nudler 2001; Mukhopadhyay et al. 2001; Mooney et al. 2005). Thus, although histor- ically defined as an initiation factor, s 70 can also remain associated with the transcription elongation complex and influence the transcription process during elongation. A functional role for s 70 during elongation was first established in the context of bacteriophage l late gene transcription (for review, see Roberts et al. 1998). Specif- ically, the expression of the phage late genes under the control of the lQ anti-terminator protein depends on a s 70 -dependent pause that occurs during early elonga- tion, shortly after the s 70 -containing RNAP holoenzyme has escaped the late promoter P R9 . Formation of this paused early elongation complex, which contains a stably bound nascent RNA that is 16 or 17 nucleotides (nt) in length, depends on an interaction between s 70 and a DNA sequence element in the initial transcribed region posi- tioned from +1 to +6 that resembles a promoter 10 ele- ment (Ring et al. 1996). Furthermore, the same module of s 70 that contacts the promoter 10 element during transcription initiation (s 70 region 2) contacts the 10- like element in the initial transcribed region during early elongation pausing (Ring et al. 1996). Similar 10-like initial transcribed region DNA sequence elements have been shown to induce s 70 -dependent pausing in the context of the late promoters of other lambdoid phages (Ring et al. 1996) and in the context of several E. coli 3 Corresponding author. E-MAIL [email protected]; FAX (617) 738-7664. Article is online at http://www.genesdev.org/cgi/doi/10.1101/gad.1991811. GENES & DEVELOPMENT 25:77–88 Ó 2011 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/11; www.genesdev.org 77 Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.org Downloaded from

Upload: lyminh

Post on 28-Jun-2019

237 views

Category:

Documents


0 download

TRANSCRIPT

Initial transcribed region sequencesinfluence the composition andfunctional properties of the bacterialelongation complex

Padraig Deighan,1 Chirangini Pukhrambam,2 Bryce E. Nickels,2 and Ann Hochschild1,3

1Department of Microbiology and Molecular Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA;2Department of Genetics, Waksman Institute, Rutgers University, Piscataway, New Jersey 08854, USA

The bacterial RNA polymerase (RNAP) holoenzyme consists of a catalytic core enzyme (a2bb'v) in complex witha s factor that is essential for promoter recognition and transcription initiation. During early elongation, thestability of interactions between s and the remainder of the transcription complex decreases. Nevertheless, thereis no mechanistic requirement for release of s upon the transition to elongation. Furthermore, s can remainassociated with RNAP during transcription elongation and influence regulatory events that occur duringtranscription elongation. Here we demonstrate that promoter-like DNA sequence elements within the initialtranscribed region that are known to induce early elongation pausing through sequence-specific interactions withs also function to increase the s content of downstream elongation complexes. Our findings establishs-dependent pausing as a mechanism by which initial transcribed region sequences can influence the compositionand functional properties of the transcription elongation complex over distances of at least 700 base pairs.

[Keywords: RNA polymerase; s factor; transcription initiation; promoter-proximal pausing; transcription elongationcomplex]

Supplemental material is available for this article.

Received September 10, 2010; revised version accepted November 5, 2010.

The s subunit of bacterial RNA polymerase (RNAP) isrequired for promoter-specific transcription initiation(Gross et al. 1998). When complexed with the RNAP coreenzyme (subunit structure a2bb9v), different s factorsspecify the recognition of different classes of promoters(Gruber and Gross 2003). The primary s factor in Escher-ichia coli, s70, typically directs transcription initiationfrom promoters defined by two conserved hexamericDNA sequence elements, termed the �10 and �35 ele-ments for their relationship to the transcription startsite (position +1). During the transition from transcrip-tion initiation to transcription elongation, the growthof the nascent RNA destabilizes the interaction betweens70 and the RNAP core enzyme (Mekler et al. 2002;Murakami et al. 2002; Vassylyev et al. 2002; Nickels et al.2005), but the complete release of s70 is not required forentry into the elongation phase of transcription (Ringet al. 1996; Bar-Nahum and Nudler 2001; Mukhopadhyayet al. 2001; Mooney et al. 2005). Thus, although histor-ically defined as an initiation factor, s70 can also remain

associated with the transcription elongation complex andinfluence the transcription process during elongation.

A functional role for s70 during elongation was firstestablished in the context of bacteriophage l late genetranscription (for review, see Roberts et al. 1998). Specif-ically, the expression of the phage late genes under thecontrol of the lQ anti-terminator protein depends ona s70-dependent pause that occurs during early elonga-tion, shortly after the s70-containing RNAP holoenzymehas escaped the late promoter PR9. Formation of thispaused early elongation complex, which contains a stablybound nascent RNA that is 16 or 17 nucleotides (nt) inlength, depends on an interaction between s70 and a DNAsequence element in the initial transcribed region posi-tioned from +1 to +6 that resembles a promoter �10 ele-ment (Ring et al. 1996). Furthermore, the same moduleof s70 that contacts the promoter �10 element duringtranscription initiation (s70 region 2) contacts the �10-like element in the initial transcribed region during earlyelongation pausing (Ring et al. 1996). Similar �10-likeinitial transcribed region DNA sequence elements havebeen shown to induce s70-dependent pausing in thecontext of the late promoters of other lambdoid phages(Ring et al. 1996) and in the context of several E. coli

3Corresponding author.E-MAIL [email protected]; FAX (617) 738-7664.Article is online at http://www.genesdev.org/cgi/doi/10.1101/gad.1991811.

GENES & DEVELOPMENT 25:77–88 � 2011 by Cold Spring Harbor Laboratory Press ISSN 0890-9369/11; www.genesdev.org 77

Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.orgDownloaded from

promoters, including the lac, tnaA, and cspD promoters(Brodolin et al. 2004; Nickels et al. 2004; Hatoum andRoberts 2008). Furthermore, both bioinformatics analysisand a candidate-based approach suggest that ;20% of allE. coli promoters may carry s70-dependent pause-induc-ing sequence elements in their initial transcribed regions(Nickels et al. 2004; Hatoum and Roberts 2008).

Here we establish a new function for initial transcribedregion s70-dependent pause elements: to modify the func-tional properties of downstream elongation complexes byincreasing their s70 content. In particular, we show thatan elongation complex that encounters a s70-dependentpause element in the initial transcribed region is morelikely to retain s70, and thus is more likely to pause ata second s70-dependent pause element positioned down-stream. We further demonstrate that the effect of a pro-moter-proximal s70-dependent pause element on the s70

content of downstream elongation complexes can be de-tected over distances of at least 700 base pairs (bp). Ourfindings establish s70-dependent pausing as a mechanismwhereby initial transcribed region sequences can influ-ence the composition and functional properties of theelongation complex throughout a transcription unit. Un-like cis-acting sequences that function to recruit trans-acting factors to the elongation complex, these promoter-like initial transcribed region sequences functionallyalter the transcription complex by inhibiting loss of anRNAP subunit. We propose that engagement of a pro-moter-proximal s70-dependent pause element increasesthe s70 content of downstream elongation complexes bystabilizing the association of s70 with the early elonga-tion complex during a series of critical nucleotide addi-tion steps when the probability of s70 release is relativelyhigh.

Results

Preliminary considerations

Biophysical characterization of artificially stalled tran-scription complexes indicates that the association of s70

with the early elongation complex is stabilized by inter-actions between s70 and a pause-inducing sequence ele-ment in the initial transcribed region. Specifically, ensem-ble and single-molecule fluorescence resonance energytransfer (FRET) measurements of the s70 content of elon-gation complexes halted during early elongation indicatethat a pause-inducing sequence element positioned from+1 to +6 (like that at lPR9) functions to stabilize the asso-ciation of s70 with stalled elongation complexes contain-ing nascent RNAs 11–15 nt in length (Nickels et al. 2004;Kapanidis et al. 2005). However, conflicting results havebeen obtained regarding whether the presence of a s70-dependent pause-inducing sequence element in the ini-tial transcribed region increases the proportion of elonga-tion complexes that contain s70 at positions downstreamfrom the pause site. Ensemble FRET results suggest thata s70-dependent pause-inducing sequence element in theinitial transcribed region increases the s70 content of elon-gation complexes halted at position +50 (Mukhopadhyay

et al. 2001; Nickels et al. 2004), but single-molecule FRETresults do not (Kapanidis et al. 2005). Given the conflictingevidence obtained from these prior biophysical studiesof halted elongation complexes, we sought to investigatewhether the presence of a s70-dependent pause-inducingsequence element in the initial transcribed region influ-ences the s70 content of downstream elongation com-plexes using approaches that would enable us to monitorthe s70 content of actively transcribing complexes.

Presence of s70-dependent pause element in the initialtranscribed region increases the s70 contentof downstream elongation complexes in vitro

To determine whether the presence of a pause-inducingpromoter �10-like element in the initial transcribed re-gion increases the proportion of elongation complexesthat contain s70 at downstream positions, we took ad-vantage of prior in vitro observations indicating that, incontrast with promoter-proximal �10-like elements (i.e.,those that extend no further than approximately +15relative to the transcription start site), promoter-distal�10-like elements can induce efficient pausing only whenhigh concentrations (1 mM) of free s70 are present (Mooneyand Landick 2003; Mooney et al. 2005; Sevostyanovaet al. 2008). These findings suggest that efficient engage-ment of promoter-distal s70-dependent pause elementsrequires that free s70 rebind the elongation complex,likely because the majority of complexes lose s70 duringthe earliest phase of elongation (Mooney and Landick2003). We therefore sought to determine whether thepresence of a promoter �10-like element in the initialtranscribed region could inhibit s70 loss during the earliestphase of elongation and enable s70-dependent pausing ata promoter-distal site in the absence of high concentra-tions of free s70.

We performed a series of in vitro transcription assaysusing three templates (Fig. 1). The first template (tem-plate #1) carried both a promoter-proximal s70-dependent

Figure 1. Schematic depiction and relevant sequence of the invitro transcription templates. The �10 element of the lPR9 pro-moter is highlighted in gray. The promoter-proximal lPR9 �10-like pause-inducing element (positioned between +1 and +6)and the promoter-distal pause-inducing element (consensus �10hexamer positioned between +19 and +24) are highlighted inblack. The distal pause element also includes a TG dinucleotideupstream of the �10 hexamer, and thus is a consensus extended�10 element. The mutations that disrupt the promoter-proxi-mal pause element on template #2 and the mutations that dis-rupt the promoter-distal pause element on template #3 areunderlined. Terminator tR9 (black lollipop) is positioned to ter-minate transcription at ;116 nt.

Deighan et al.

78 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.orgDownloaded from

pause-inducing sequence element and a promoter-distals70-dependent pause-inducing sequence element. In par-ticular, template #1 carried the well-characterized pauseelement associated with the l late promoter (lPR9) posi-tioned between +1 and +6, as well as a consensus pro-moter �10 element positioned between +19 and +24.The second template (template #2) carried two base pairsubstitutions in the promoter-proximal pause element(A+2G/T+6G) that destabilize interactions with s70 andeliminate s70-dependent pausing (Ring and Roberts 1994).The third template (template #3) carried two base pairsubstitutions in the promoter-distal pause element(A+20C/T+24C); these substitutions were predicted todestabilize interactions with s70 and eliminate s70-de-pendent pausing at the promoter-distal site.

When single-round transcription time-course reactionswere performed with low concentrations of s70 (30 nM,a twofold molar excess over RNAP core) using template#1, the expected 16- and 17-nt lPR9 pause species, whichappeared early in the time course and decayed over time,were observed (Fig. 2A). In addition to these 16- and 17-ntRNA transcripts, less-prominent 35- to 37-nt RNA tran-scripts were observed throughout the time course (Fig.2A). Strikingly, when the transcription reactions wereperformed using template #2, which carries mutations inthe promoter-proximal pause element, these 35- to 37-ntRNA transcripts were not detected, consistent with theidea that the presence of a s70-dependent pause elementin the initial transcribed region can facilitate s70-depen-dent pausing at promoter-distal sites. Several lines ofevidence confirmed that the 35- to 37-nt RNA transcriptsare, in fact, the result of s70-dependent pausing at thepromoter-distal site. First, the promoter-distal pauseelement is displaced by 18 bp relative to the positionof the promoter-proximal pause element (register +19compared with register +1), and the 35- to 37-nt RNAtranscripts are ;18 nt longer than the paused species thatresult from engagement of the promoter-proximal pauseelement. Second, the 35- to 37-nt RNA species are ab-sent when reactions are performed using template #3,which carries mutations in the promoter-distal pauseelement (Fig. 2A). Third, production of the 35- to 37-ntRNA species (like production of the 16- and 17-nt RNAspecies) is reduced when reactions are done using tem-plate #1 in the presence of RNAP holoenzyme reconsti-tuted with a well-characterized s70 mutant (s70 L402F)that is deficient in s70-dependent pausing (Fig. 2B; Koet al. 1998). Fourth, the 35- to 37-nt RNA species aresensitive to the transcript cleavage factor GreB (Fig. 2B),a hallmark of previously characterized s70-dependentpause species (Marr and Roberts 2000). We therefore con-clude that the presence of a promoter-proximal s70-de-pendent pause element increases the likelihood thats70-dependent pausing will occur at a promoter-distalsite in vitro.

To test the hypothesis that the presence of a promoter-proximal pause element enables promoter-distal pausingby inhibiting the loss of s70 from the early elongationcomplex, we asked whether the addition of excess s70

could restore promoter-distal s70-dependent pausing on

template #2. Specifically, we performed the transcriptionreactions in the presence of 1 mM s70 (a 67-fold molarexcess over RNAP core) and found that the abundanceof the 35- to 37-nt RNA species generated using template#2 increased over their abundance when the reactionswere performed with low concentrations of s70 (Fig. 2C,D);thus, the abundance of the 35- to 37-nt RNAs producedfrom template #2 at high concentrations of s70 and fromtemplate #1 at low concentrations of s70 were approxi-mately the same (Fig. 2D, right). Furthermore, the addi-tion of high concentrations of s70 also increased theabundance of the 35- to 37-nt RNA species generated inreactions performed with template #1, but did not resultin the appearance of these species with template #3 (Fig.2C,D). We conclude that the presence of a s70-dependentpause element in the initial transcribed region facilitatess70-dependent pausing at the promoter-distal site in vitroby inhibiting loss of s70 during the earliest phase ofelongation.

Presence of s70-dependent pause element in the initialtranscribed region increases the s70 contentof downstream elongation complexes in vivo

Next, we sought to determine whether the presence ofa promoter �10-like element in the initial transcribedregion could inhibit s70 loss during the earliest phase ofelongation and enable s70-dependent pausing at a pro-moter-distal site in vivo. To do this, we introduced amulticopy plasmid vector carrying template #1, #2, or #3into E. coli cells. We then isolated total RNA from thecells and detected RNA transcripts generated in vivofrom these templates using hybridization with radiola-beled locked nucleic acid (LNA) probes (Valoczi et al.2004). We identified transcripts associated with s70-de-pendent pausing in vivo based on a comparison with tran-scripts generated during in vitro transcription reactions.

We first sought to identify RNA transcripts producedfrom templates #1 and #3 in vivo using an LNA probecomplementary in sequence to nucleotides +1 to +15(Fig. 3A), which enabled the detection of RNA transcriptsassociated with promoter-proximal s70-dependent paus-ing (the 16- and 17-nt species) and RNA transcripts as-sociated with promoter-distal s70-dependent pausing (the35- to 37-nt species) (Fig. 3B, lane 1). (Note that the +1 to+15 probe is not fully complementary in sequence totranscripts produced from template #2, which bearsmutations at positions +2 and +6, within the promoter-proximal pause element.) With the +1 to +15 probe, wedetected RNA transcripts associated with promoter-prox-imal s70-dependent pausing in RNA samples isolatedfrom cells carrying either template #1 or template #3(Fig. 3B). In contrast, RNA transcripts associated withpromoter-distal s70-dependent pausing were detected onlyin RNA samples isolated from cells carrying template#1, which carries an intact promoter-distal pause ele-ment (Fig. 3B).

We next determined the effect of mutating the pro-moter-proximal pause element on promoter-distal s70-dependent pausing in vivo. Thus, we compared the

Retention of s70 in elongation complex

GENES & DEVELOPMENT 79

Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.orgDownloaded from

abundance of transcripts associated with promoter-distalpausing produced from template #1 (which carries anintact promoter-proximal pause element) with the abun-dance of transcripts associated with promoter-distal paus-ing produced from template #2 (which carries an inacti-vated promoter-proximal pause element). Because of themutations present on template #2 (at positions +2 and+6), we used an LNA probe complementary in sequenceto nucleotides +7 to +19 for this analysis (Fig. 3A). In

contrast to the +1 to +15 probe, which enabled detec-tion of transcripts associated with promoter-proximaland promoter-distal pausing, the +7 to +19 probe enableddetection only of transcripts associated with promoter-distal s70-dependent pausing (the 35- to 37-nt species)(Fig. 3C). With the +7 to +19 probe, we again detected thepromoter-distal 35- to 37-nt pause species in RNA sam-ples isolated from cells carrying template #1 and did notdetect these 35- to 37-nt species when the promoter-distal

Figure 2. A promoter �10-like element in the initial transcribed region inhibits s70 loss during early elongation and enables s70-dependent pausing at a promoter-distal site in vitro. (A) Single-round in vitro transcription reactions performed with RNAPholoenzyme (15 nM core reconstituted with 30 nM s70) and three different templates (#1, #2, and #3, as depicted in Fig. 1) bearingthe indicated wild-type (WT) or mutant (Mut) promoter-proximal and promoter-distal pause elements. The reactions were performed asa time course with samples taken 0.5, 1, 3, 6, 18, and 54 min after transcription was initiated. RNA species that reflect s70-dependentpausing at the promoter-proximal element (+16 and +17) or the promoter-distal element (+35, +36, and +37) are indicated. The 35- to 37-nt RNA species, which were annotated by comparison with a reaction done in the presence of a chain-terminating nucleotide, were alsodetected when the RNA was end-labeled with [g-32P]ATP, indicating that they are not cleavage products derived from an internalportion of the RNA (Supplemental Fig. S1). We note the accumulation of abortive RNA products in reactions performed with template#2, which we attribute to the two base pair substitutions in the initial transcribed region; initial transcribed region alterations can havedramatic effects on abortive transcript production (Hsu et al. 2003; Chander et al. 2007). The ;116-nt terminated transcript (T) isindicated, and the asterisk (*) indicates a shorter terminated transcript that is the result of transcription initiating under the control ofthe promoter-distal extended �10 element. (B) Single-round in vitro transcription reactions performed using template #1 (wild-typepromoter-proximal and promoter-distal pause elements) with RNAP holoenzyme (15 nM core reconstituted with either 30 nM s70 wildtype or 30 nM s70 L402F, as indicated). The assay was performed and the gel annotated as in A, except that samples were taken 0.5, 1, 6,18, and 54 min after transcription was initiated. Where indicated, GreB was included in the reactions at 100 nM. (C) As in A, except theassay was performed with RNAP holoenzyme that was reconstituted with 1 mM s70. (D) Graph showing the percentage of s70-dependent pausing detected at the promoter-distal site [distal pause signal/(distal pause signal + terminated signal)] for reactionsperformed in the presence of 30 nM s70 (left graph) as in A, or 1 mM s70 (right graph) as in C. Plotted on the graphs are the mean and SDof three independent experiments. In the right graph, we show that the percent of pausing detected at the distal site on template #2 inthe presence of 1 mM s70 was approximately the same as that detected at the distal site on template #1 in the presence of lowconcentrations (30 nM) of s70 (dotted line).

Deighan et al.

80 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.orgDownloaded from

pause element was mutationally inactivated (template#3) (Fig. 3C). Furthermore, the abundance of the 35- to37-nt transcripts associated with promoter-distal paus-ing was reduced significantly (approximately fivefold)when the PR9 pause element was mutationally inacti-vated (template #2) (Fig. 3C). We conclude that the pres-ence of a s70-dependent pause element in the initialtranscribed region increases the likelihood that s70-de-pendent pausing will occur at a promoter-distal site invivo, presumably by inhibiting loss of s70 during the

earliest phase of elongation, as was revealed by our invitro analysis (see Fig. 2).

Presence of s70-dependent pause element in the initialtranscribed region increases the s70 contentof downstream elongation complexes overdistances of at least 700 bp

Our results obtained in vitro and in vivo suggest thatthe presence of a s70-dependent promoter-proximal pauseelement facilitates recognition of a more downstreampause element because it enhances the s70 content of theearly elongation complex. That is, transcription elonga-tion complexes that arrive at the promoter-distal pauseelement are more likely to contain s70 if they have alreadyengaged a promoter-proximal pause element. We nextused chromatin immunoprecipitation (ChIP) to determinewhether the effect of a promoter-proximal s70-dependentpause element on the s70 content of downstream elonga-tion complexes could be detected throughout a transcrip-tion unit. Using cells containing a single-copy PR9-lacZfusion with either the wild-type pause element or amutationally inactivated pause element (A+2G/T+6G),we assessed the occupancy of both the b subunit andthe s70 subunit of RNAP across each transcription unit.To do this, we determined the fold enrichment of thepromoter/pause region (�92 to +45) and two downstreamregions (centered at positions +339 and +694) in boththe b- and s70-immunoprecipitated material relative toa nontranscribed region of the DNA (within the bgl op-eron). To ensure that promoter-bound transcription com-plexes did not contribute to the immunoprecipitation ofdownstream DNA (due to the difficulty of shearing theDNA to a sufficiently small average fragment length),we separated promoter DNA from downstream DNAusing restriction enzymes (Fig. 4A; see also the Materialsand Methods).

To compare the s70 content of elongation complexes onthe two fusion constructs (PR9-lacZ containing the wild-type or mutated pause element), we calculated fold en-richment ratios (s70/b) by dividing the fold enrichmentof a given DNA region in the s70-immunoprecipitatedmaterial by the fold enrichment of the same region in theb-immunoprecipitated material. Although we observed ageneral decrease in the s70 content of elongation com-plexes with increasing distance from the promoter onboth templates (Fig. 4B), we found that the s70 content ofelongation complexes in both the +339 and +694 regionswas approximately twofold higher when the PR9 pauseelement was intact (see Fig. 4B,D). In contrast, when theChIP assays were done in strains containing chromosom-ally encoded s70 L402F, which is deficient in s70-de-pendent pausing (see Fig. 2B), the fold enrichment (s70/b)ratios were the same within error for each downstreamregion of the DNA regardless of whether the templatecarried a wild-type or mutated pause element (Fig. 4C,D).We conclude, therefore, that the presence of a functionals70-dependent pause element in the initial transcribedregion enhances the wild-type s70 content of downstreamtranscription elongation complexes, and that this effectcan be detected over at least 700 bp.

Figure 3. A promoter �10-like element in the initial tran-scribed region facilitates s70-dependent pausing at a promoter-distal site in vivo. (A) Schematic of templates and LNA probes.Templates #1, #2, and #3 are depicted as in Figure 1. Sequencescomplementary to the +1 to +15 probe (green line) or the +7 to+19 probe (lavender line) are indicated. (B,C) Detection of RNAtranscripts with the +1 to +15 probe (B) or the +7 to +19 probe(C). The in vivo generated RNA samples isolated from a DgreA

strain to facilitate the detection of pause species (Marr andRoberts 2000) were loaded alongside transcripts generated innonradioactive reactions performed in vitro (IVT) using tem-plate #1 in the absence (labeled �) or presence (labeled B) ofGreB; a 10-nt RNA ladder is included (lane M). We note that invivo generated promoter-distal pause products (distal pausecluster) include a faster-migrating species that is likely due tothe action of GreB, as this species is also present when GreB isincluded in the in vitro transcription reaction (see also Fig. 2B).A control experiment excluded the possibility that the apparenteffect of the promoter-proximal pause element on the abun-dance of the promoter-distal pause products is due to an effect ofthe PR9 pause mutations on the efficiency of probe hybridization(Supplemental Fig. S2).

Retention of s70 in elongation complex

GENES & DEVELOPMENT 81

Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.orgDownloaded from

Two control experiments confirmed that the associa-tion of s70 (and b) with downstream DNA was depen-dent on transcription initiating from promoter PR9. First,neither downstream DNA region was enriched abovebackground in either the b- or the s70-immunoprecipi-tated material when PR9 was inactivated (as confirmedby the results of parallel b-galactosidase assays) on thetemplate with the intact pause element (Fig. 4D). Second,the fold enrichment values for the promoter region werehigh, whereas the fold enrichment values for the down-stream DNA regions dropped to background or near back-ground in both the b- and the s70-immunoprecipitatedsamples when the cells were pretreated with rifampicin,which traps RNAP at the promoter (Fig. 4D; Herring et al.2005).

To explore the generality of these observations, wetested the effect of another promoter-proximal s70-de-pendent pause element, which associated with the latepromoter of phage 82. Displaced by 8 bp relative to theposition of the lPR9 pause element, the 82 pause elementinduces pausing at a nascent RNA length of ;25 nt(Goliger and Roberts 1989), as compared with 16–17 ntin the case of the l pause element. Using a pair of 82 PR9-lacZ fusion constructs (bearing the wild-type or a mutated+25 pause element), we again used ChIP to examine theb and s70 occupancies within the promoter/pause regionand two downstream regions (centered at positions +356

and +511) (Fig. 5A). We found that the s70 content ofelongation complexes was between 2.5-fold and threefoldhigher in both the +356 and +511 regions when the 82 PR9

pause element was intact (Fig. 5B,C). As we did in thecase of the l PR9 construct, we inactivated the 82 PR9

promoter and confirmed that the association of s70 (andb) with downstream DNA was dependent on transcrip-tion initiating from this promoter (Fig. 5C). We concludethat a promoter-proximal s70-dependent pause elementcan enhance the s70 content of downstream transcriptionelongation complexes when located in at least two dis-tinct positions within the initial transcribed region.

Discussion

Here we identify s70-dependent pausing as a mechanismby which the sequence of the initial transcribed region

Figure 4. Effect of the lPR9 promoter-proximal s70-dependentpause element on the s70 content of elongation complexes ona lPR9-lacZ fusion. (A) Schematic depiction and relevant se-quence of the single-copy lPR9-lacZ fusions used in the ChIPassay. The �35 and �10 elements of the lPR9 promoter arehighlighted in gray. The �10-like pause-inducing element (po-sitioned between +1 and +6) is highlighted in black. The muta-tions that disrupt the promoter-proximal pause element (on thetemplate designated ‘‘mut pause’’) and the mutations within andupstream of the �35 element that disrupt the lPR9 promoter (onthe template designated ‘‘mut �35’’) are underlined. The blacktwo-headed arrows in the schematic represent the target regions(promoter, +339, +694, and digest control) that were amplifiedby real-time PCR in the ChIP assay. The scissor symbols rep-resent the locations of the restriction enzyme recognition sites(all of which are within the digest control region and outside theother target regions) that were used to separate the b-immuno-precipitated (or s70-immunoprecipitated) promoter region fromthe downstream regions. The efficiency of digestion at thesesites was monitored by quantitative amplification of the digestcontrol region. (B,C) Graphs showing the ChIP fold enrichmentratios (s70/b) from cells containing a lPR9-lacZ fusion witheither the wild-type pause element (WT pause) or an inactivatedpause element (Mut pause), as indicated. The s70/b ratios wereobtained by dividing the fold enrichment over background (bgl)of a given DNA region (+339 or +694) in the s70-immunopre-cipitated material by the fold enrichment of the same regionin the b-immunoprecipitated material (see the Materials andMethods). We note that these fold enrichment ratios do notrepresent the absolute molar ratios of s70 and b in the elonga-tion complex (see the Discussion). In B, the cells contained wild-type s70, whereas in C, the cells contained chromosomallyencoded s70 L402F. The means and SDs of three independentexperiments are graphed. (D) Fold enrichments over bgl for boththe b- and s70-immunoprecipitated DNA from cells containingthe templates shown in A and either wild-type s70 or chromo-somally encoded s70 L402F, as indicated. The fold enrichmentsover bgl were determined as described in the Materials andMethods and represent the means of three independent exper-iments, with SDs in parentheses. These fold enrichment valueswere used to calculate the fold enrichment ratios plotted in B

and C. The cell cultures that were analyzed by ChIP were alsoassayed for b-galactosidase (before the addition of formaldehydeor rifampicin), and the results are expressed in Miller units (MU)with the SDs in parentheses.

Deighan et al.

82 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.orgDownloaded from

can influence both the composition and functional prop-erties of downstream elongation complexes. In particular,our results demonstrate that the presence of a s70-de-pendent pause element in the initial transcribed regioninhibits s70 loss during the earliest stage of elongation,increasing the s70 content of elongation complexes atleast 700 bp into the transcription unit. Moreover, theanalysis of actively transcribing complexes in vitro andin vivo indicates that s70 is retained in a functionallycompetent state that permits it to engage downstreampause-inducing elements.

A model for how the presence of a promoter-proximals70-dependent pause element influences thes70 content of downstream elongation complexes

Before providing a mechanistic explanation for the effectof a promoter-proximal s70-dependent pause element onthe s70 content of downstream elongation complexes, weconsider how the paused early elongation complex atlPR9 is formed. The position of the lPR9 �10-like pauseelement relative to the promoter �10 element (displaceddownstream by 12 bp) suggests that s70 might initiallyengage this element at a nascent RNA length of ;12 nt.Thus, it was proposed that formation of the pausedelongation complex at lPR9 involves the establishmentof sequence-specific interactions between s70 (region 2in particular) and the �10-like element at a nascentRNA length of ;12 nt, followed by several nucleotideaddition steps during which s70 maintains contact withthe pause element, until the transcript reaches a criticallength (16 or 17 nt) at which pausing is detected (Marrand Roberts 2000). Consistent with the suggestion thats70 can maintain contact with the pause element through

several successive nucleotide addition steps, the resultsof ensemble FRET analysis indicate that a s70-dependentpause element positioned from +1 to +6 can stabilize theassociation of s70 with elongation complexes halted afterthe synthesis of RNA transcripts ranging in length from11 nt to at least 15 nt (Nickels et al. 2004). According tothe model, the nucleotide addition steps that occur whiles70 maintains contact with the pause element requireexpansion of the transcription bubble and concomitantDNA scrunching (see Fig. 6A; Marr and Roberts 2000),as has been shown to occur in the context of promoter-bound initial transcribing complexes during the processof promoter escape/abortive initiation (Kapanidis et al.2006; Revyakin et al. 2006). In the case of initial tran-scription, DNA scrunching generates a ‘‘stressed inter-mediate’’ that can be relieved in two ways: Either s70

relinquishes its contacts with the promoter DNA andRNAP escapes the promoter, or the expanded bubble

Figure 5. Effect of the 82 PR9 promoter-proximal s70-dependentpause element on the s70 content of elongation complexes onan 82 PR9-lacZ fusion. (A) Schematic depiction and relevantsequence (see also the legend for Supplemental Fig. S3) of thesingle-copy 82 PR9-lacZ fusions used in the ChIP assay. The �35and �10 elements of the 82 PR9 promoter are highlighted in gray.The +25 pause element consists of a �10-like hexamer (high-lighted in black and positioned from +9 to +14) as well as anupstream TG dinucleotide, and thus is an extended �10-likeelement. The mutations that disrupt the +25 pause element (onthe template designated ‘‘mut pause’’) and the mutations withinthe �10 element that disrupt the 82 PR9 promoter (on the tem-plate designated ‘‘mut �10’’) are underlined. An in vitro tran-scription assay confirmed the elimination of s70-dependentpausing on the mut pause template (Supplemental Fig. S3).The black two-headed arrows in the schematic represent thetarget regions (promoter, +356, +511, and digest control) thatwere amplified by real-time PCR in the ChIP assay. The scissorsymbols represent the locations of the restriction enzyme recog-nition sites (all of which are within the digest control regionand outside the other target regions) that were used to separatethe b-immunoprecipitated (or s70-immunoprecipitated) pro-moter region from the downstream regions. The efficiency ofdigestion at these sites was monitored by quantitative amplifi-cation of the digest control region. (B) Graph showing the ChIPfold enrichment ratios (s70/b) from cells containing a single-copy 82 PR9-lacZ fusion with either the +25 pause element (WTpause) or a mutationally inactivated pause element (Mut pause),as indicated. The s70/b ratios were obtained by dividing the foldenrichment over background (bgl) of a given DNA region (+356or +511) in the s70-immunoprecipitated material by the foldenrichment of the same region in the b-immunoprecipitatedmaterial (see the Materials and Methods). The means and SDsof three independent experiments are graphed. (C) Fold enrich-ments over bgl for both the b- and s70-immunoprecipitatedDNA from cells containing the templates shown in A. The foldenrichments over bgl were determined as described in theMaterials and Methods, and represent the means of three in-dependent experiments with SDs in parentheses. These foldenrichment values were used to calculate the fold enrichmentratios plotted in B. The cell cultures that were analyzed by ChIPwere also assayed for b-galactosidase (before the addition offormaldehyde), and the results are expressed in Miller units(MU) with the SDs in parentheses.

Retention of s70 in elongation complex

GENES & DEVELOPMENT 83

Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.orgDownloaded from

collapses and an abortive transcript is released (Kapanidiset al. 2006; Revyakin et al. 2006). In the case of earlyelongation pausing, the strain generated by DNA scrunch-ing can similarly be resolved in two ways: Either s70

relinquishes its contacts with the pause element andRNAP escapes the pause, or the expanded bubble col-lapses, in this case without release of the nascent RNA,generating a backtracked complex in which the catalyticcenter of the enzyme is no longer in register with the39 end of the nascent transcript (Fig. 6A; Marr and Roberts2000; Perdue and Roberts 2010).

To explain how a promoter-proximal s70-dependentpause element increases the s70 content of downstreamelongation complexes, we propose that the sequence-specific interactions between s70 and the pause elementstabilize the association of s70 with the early elongationcomplex during a ‘‘critical window’’ of nucleotide addi-tion steps when the probability of s70 release is relativelyhigh. Support for the notion of such a critical windowcomes from the results of both ensemble and single-molecule FRET measurements of the s70 content of haltedelongation complexes (Nickels et al. 2004; Kapanidiset al. 2005), which suggest that release of s70 is biphasic,with an initial ‘‘fast’’ phase that occurs after synthesis ofan RNA transcript ;12 nt in length and a subsequent‘‘slow’’ phase. Thus, according to our model, the interac-tion between s70 and an early elongation pause elementstabilizes the association of s70 with the RNAP coreenzyme during critical nucleotide addition steps whens70 release is ‘‘fast’’ (due, perhaps, to clashes between thenascent RNA and specific portions of s70) (for review, see

Mooney et al. 2005) and a significant fraction of thetranscription complexes would otherwise release s70 (Fig.6B). Our finding that the effect of an early elongationpause element on the s70 content of downstream elonga-tion complexes was manifest when the pause elementwas positioned from +1 to +6 (l PR9) or from +9 to +14(82 PR9) suggests that the critical window of fast s70 releasemight occur at different template positions, depending onthe sequence context. Subsequently, as the elongationcomplexes leave the critical region, they appear to entera phase where release of s70 is ‘‘slow,’’ based on the grad-ual decrease in the s70 content of mature elongationcomplexes with increasing distance from the promoterobserved in both prior studies (Raffaelle et al. 2005;Mooney et al. 2009a) and the results presented here(Figs. 4, 5).

Prevalence of promoter-proximal s70-dependentpause elements in vivo

Previous bioinformatics analysis of ;650 promoters sug-gested that s70-dependent promoter-proximal pause ele-ments are associated with ;20% of all promoters in E.coli (Nickels et al. 2004). We took advantage of a morecomprehensive set of 4132 transcription start sites de-fined from high-throughput sequencing of the 59 ends ofprimary RNA transcripts isolated from E. coli (Cho et al.2009) to re-examine this estimate (Supplemental TableS1). The l pause element consists of a three of six matchto the �10 element consensus sequence (TATAAT) inregister +1 (where register is defined by the first base of

Figure 6. Model to explain how a promoter-proximal s70-dependent pause element in-creases the s70 content of downstream elon-gation complexes. (A) Formation of pausedearly elongation complex at lPR9 (see alsoFig. 8 in Perdue and Roberts 2010). (Top left)Domain 2 of s70 (s2) is shown engaging thepromoter-proximal �10-like pause element(black rectangle) at a nascent RNA length of12 nt. (Top right) The nascent RNA is ex-tended to 17 nt via a series of nucleotideaddition steps that occur while s2 maintainscontact with the pause element, requiringDNA scrunching (depicted as ssDNA loopsin the transcription bubble). The scrunchedcomplex can undergo two alternative fates:Either RNAP escapes the pause (bottom

right), requiring that s2 relinquish contactwith the pause element, or the expandedtranscription bubble collapses (bottom left),generating a backtracked complex in whichthe catalytic center of the enzyme is nolonger in register with the 39 end of theRNA transcript. Endonucleolytic cleavageof the transcript (stimulated by Gre factor)(Marr and Roberts 2000) can regenerate

a catalytically proficient complex with a correctly positioned RNA 39 end, allowing the cycle to repeat. (B) Critical window ofnucleotide addition steps during which s70 release is ‘‘fast.’’ In the absence of a promoter-proximal pause element that allows s70 tomaintain stabilizing interactions with the DNA, the transcription complex releases s70 with high probability during a critical windowof early nucleotide addition steps (here when the nascent RNA is extended from 12 to 17 nt).

Deighan et al.

84 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.orgDownloaded from

the �10-like hexamer), including the highly conserved Aand T nucleotides at positions 2 and 6 of the hexamer(McClure 1985), whereas the 82 pause element consistsof a four of six match to the �10 element consensussequence in register +9, again including the highly con-served A and T nucleotides at positions 2 and 6 of thehexamer. For our analysis, we identified transcriptionunits containing a three of six, four of six, five of six, or sixof six match to the�10 element consensus, with an A anda T at positions 2 and 6 of the hexamer, respectively, inregisters +1 to +9; 40% (1654) of the transcription unitscontained at least a three of six match, 20% (817) con-tained at least a four of six match, ;5% (200) containedat least a five of six match, and ;0.3% (12) contained a sixof six match (Table 1). Although we do not know whatfraction of the 4132 transcription start sites used for thisanalysis are associated with a s70-dependent promoter,it is likely to be the major fraction, because most of thetranscripts were present in RNA samples harvested frommid-exponential-phase cells (Cho et al. 2009).

To determine whether the initial transcribed regionsof transcription units (i.e., registers +1 to +9) are enrichedfor putative s70-dependent pause elements, we computedthe probabilities of finding the target sequence motifsby chance within random sequences (Table 1). Based onthis analysis, we would have expected 31% of the tran-scription units to contain at least a three of six match,12.5% to contain at least a four of six match, 2.7% tocontain at least a five of six match, and 0.2% to containa six of six match. This analysis therefore suggests thatinitial transcribed regions are enriched in s70-dependentpause elements. Consistent with this inference, we foundthat, when we searched for the target sequence motifsat five distinct promoter-distal positions (registers +51 to+59, +101 to +109, +151 to +159, +201 to +209, and +251to +259), the percentages of transcription units contain-ing matches in each of these regions approached thepercentages that were calculated based on the probabil-ities of finding the target sequence motifs by chance(Table 1).

Functional roles for s70 during transcription elongation

The results of prior ChIP analyses reveal unexplaineddifferences in the s70 content of transcription elongationcomplexes on different transcription units (Wade andStruhl 2004; Grainger et al. 2005; Reppas et al. 2006;Mooney et al. 2009a). Our finding that the presence ofa promoter-proximal s70-dependent pause element influ-ences the s70 content of transcription complexes up to;700 bp downstream from the transcription start sitesuggests that the sequence of the initial transcribed re-gion may be a key determinant of at least some of thesedifferences. We note that our ChIP data do not permitan estimate of the actual fractional occupancy of s70 inthe elongation complex at any particular position withina transcription unit because we do not know how thecross-linking efficiencies of s70 and RNAP in a promoter-bound or paused early elongation complex compare withthose in an actively transcribing elongation complex.

Using an artificial construct, we demonstrated thatthe presence of a promoter-proximal s70-dependent pauseelement facilitated s70-dependent pausing at a promoter-distal pause site both in vitro and in vivo. Interestingly,promoter-distal s70-dependent pausing has been proposedto occur at a cluster of sites extending as far as ;100 bpdownstream from the transcription start site of the E. colitnaA promoter, which also carries a s70-dependent pauseelement in its initial transcribed region (Hatoum andRoberts 2008). Our findings suggest that the observedpromoter-distal pausing in the tnaA transcription unitmay be influenced by the presence of the s70-dependentpause element in the initial transcribed region.

s70-dependent pausing has been shown to affect theproduction of full-length transcripts, presumably by mod-ulating RNAP density within the transcription unit (seealso Fig. 4D; Hatoum and Roberts 2008). Such effects onRNAP density within a transcription unit could, inprinciple, affect the overall elongation rate and modulatethe responsiveness of RNAP to termination signals by in-fluencing the extent of RNAP ‘‘cooperation’’ (Epshtein

Table 1. Enrichment of putative s70-dependent pause elements in the initial transcribed regions of E. coli transcription units

Match tos70-dependentpause elementa

Number and percentage of matched s70-dependent pause elements in the initialtranscribed regions (registers +1 to +9 [ITR]) of 4132 identified transcription unitsb

and in five downstream regions DR1–DR5c

Probability ofs70-dependent pause

elements in randomized14-nt sequencesd

ITR DR1 DR2 DR3 DR4 DR5 G/C = A/T G/C > A/T

At least threeof six

1654 (40.0%) 1546 (37.4%) 1362 (32.9%) 1323 (32.0%) 1253 (30.3%) 1248 (30.2%) 32.0% 31.0%

At least fourof six

817 (19.8%) 701 (16.9%) 564 (13.6%) 555 (13.4%) 489 (11.8%) 463 (11.2%) 13.2% 12.5%

At least fiveof six

200 (4.8%) 149 (3.6%) 101 (2.4%) 85 (2.1%) 74 (1.8%) 72 (1.7%) 2.9% 2.7%

Six of six 12 (0.3%) 7 (0.2%) 4 (0.1%) 5 (0.1%) 4 (0.1%) 3 (0.1%) 0.2% 0.2%

anAnnnT and at least an X of six match to TATAAT.bThe 4132 transcription start sites were identified by Cho et al. (2009).cRegisters +51 to +59 (DR1), +101 to +109 (DR2), +151 to +159 (DR3), +201 to +209 (DR4), and +251 to +259 (DR5).dAnalysis was performed on 14-nt randomized sequences, assuming each nucleotide was used with equal efficiency (denoted G/C =

A/T), and also using a G + C parameter of 50.8% (denoted G/C > A/T) to reflect the G + C content of E. coli MG1655.

Retention of s70 in elongation complex

GENES & DEVELOPMENT 85

Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.orgDownloaded from

and Nudler 2003). As a component of the elongationcomplex, s70 could also influence transcription throughmechanisms independent of its ability to induce pausing.For example, s70 could influence the accessibility of thetranscription elongation complex to regulatory factors.In this regard, the b9 coiled-coil, which binds s70 region 2,has also been identified as a binding site for at least twoelongation factors, including the highly conserved gen-eral elongation factor NusG and the NusG paralog RfaH(Belogurov et al. 2007; Mooney et al. 2009b; Nickels2009). Essential in E. coli, NusG influences the rate oftranscription elongation and functions as both a termi-nation and an anti-termination factor (for review, seeRoberts et al. 2008). As NusG is expected to compete withs70 for access to the elongation complex (Sevostyanovaet al. 2008), the presence of a promoter-proximal s70-dependent pause element could potentially influence geneexpression by decreasing the NusG content of down-stream elongation complexes. Finally, s70 might exert adirect effect (either positive or negative) on the processof transcription termination (for example, see Arndt andChamberlin 1988).

Materials and methods

Strains and plasmids

A list of strains and plasmids is provided in SupplementalTable S2.

In vitro transcription

His-tagged wild-type s70 and s70 L402F were purified as de-scribed (Nickels et al. 2004). E. coli RNAP core enzyme waspurchased from Epicentre and GreB was a gift from J. Gelles(Brandeis University). Open complexes were formed by incubat-ing 15 nM RNAP (preincubated with either 30 nM wild-types70, 30 nM s70 L402F, or 1 mM wild-type s70) with 20 nM linearDNA template for 5 min at 37°C in transcription buffer (20 mMTris-HCl at pH 8.0, 0.1 mM EDTA, 50 mM KCl, 100 mg/mL BSA,10 mM DTT) plus 200 mM GTP, ATP, and CTP, and 50 mM UTP(supplemented with 1 mCi/mL [a-32P]-UTP). A single round oftranscription was initiated by adding 4 mM MgCl2 and 10 mg/mLrifampicin. Reactions were stopped and processed at the indi-cated times (procedure as described in Nickels et al. 2005), andRNA transcripts were electrophoresed on 12% polyacrylamidesequencing gels. Bands were visualized by PhosphorImager, andthe data were analyzed using ImageQuant software.

Detection of RNA transcripts by hybridization

ML176 cells containing plasmid pFW11tet-PR9_+19, pFW11tet-mut PR9_+19, or pFW11tet-PR9_mut+19 were grown in LB supple-mented with tetracycline (10 mg/mL) to an OD600 of 0.5. Cellsuspensions (5 mL) were mixed with RNAlater (15 mL; Ambion),collected by centrifugation (20 min at 17,000g), and resuspendedin TRI Reagent (1 mL; Molecular Research Center), and RNAspecies <200 nt in size were isolated using mirVana miRNAIsolation kit (Ambion). Transcripts were detected as describedin Goldman et al. (2009).

b-Galactosidase assays

Reporter strain cells (in triplicate) were harvested from culturesgrown at 37°C to mid-exponential phase (OD600 = 0.5) in LB

broth supplemented with tetracycline (10 mg/mL). b-Galactosi-dase assays were performed as described (Nickels 2009) usingmicrotiter plates and a microtiter plate reader. Miller units werecalculated as described (Nickels 2009).

ChIP assay

Cultures were grown as described for the b-galactosidase assays.Formaldehyde-induced cross-linking, cell lysis, and DNA shear-ing were preformed as described (Deighan and Hochschild 2007).When present, rifampicin was added to 150 mg/mL and cells wereincubated for 20 min prior to addition of formaldehyde. Protein/DNA complexes were immunoprecipitated in immunoprecipi-tation buffer (50 mM HEPES at pH 7, 150 mM NaCl, 1% TritonX-100, 0.1% sodium deoxycholate, 1 mM PMSF) using a mono-clonal antibody reactive against the b subunit of RNAP (cloneNT63, Neoclone) or a monoclonal antibody reactive against s70

(clone 2G10, Neoclone) and 20 mL of protein-A/G magneticbeads (New England Biolabs). Immunoprecipitated complexeswere washed once in restriction enzyme buffer 2 (New EnglandBiolabs), and then incubated in the same buffer containing arestriction enzyme cocktail for 1 h at 37°C. The restrictionenzymes cleave within the ‘‘digest control region’’ (a regionnested between the promoter region and the first downstreamregion) (Figs. 4A, 5A) and serve to increase the resolution ofthe assay by separating promoter-associated b (or s70)/DNAcomplexes from downstream regions. For ChIP experimentswith the lPR9-lacZ fusion, the enzymes used were AciI, AhdI,HindIII, XmnI, and NdeI. For ChIP experiments with the 82PR9-lacZ fusion, the enzymes used were BglI, HindIII, NdeI, PstI,and PvuII. Complexes were then washed four times with 13

immunoprecipitation buffer and once with TE buffer (10 mMTris at pH 8, 1 mM EDTA), and were resuspended in elutionbuffer (TE buffer containing 1% SDS). The cross-links werereversed for 7 h at 65°C, and the b- and s70-immunoprecipitatedDNAs were purified using Zymo Research spin columns.

ChIP assay quantitation by real-time PCR

For each ChIP experiment, there were five target regions ofinterest (Figs. 4A, 5A): the promoter region, two downstreamregions within the fused lacZ gene, a digest control region, and,as a background control, a region of the bglF gene within the bgl

operon. The chromosome-encoded bgl operon is transcription-ally silent in wild-type E. coli; therefore, the analysis of bgl

product amplification provided an internal standard for DNAthat was nonspecifically immunoprecipitated by the b and s70

antibodies or by the protein-A/G magnetic beads. All primerpairs (purchased from IDT) (Supplemental Table S3) permittedthe amplification of a single product of the predicted size with;1.93-fold amplification per cycle using 10-mL reactions con-taining iTaq SYBR Green Supermix with ROX (Bio-Rad) and aStepOnePlus Real-Time PCR System (Applied Biosystems). Quan-titative PCRs were run in triplicate for each immunoprecipitatedDNA sample, and the averaged threshold cycle (Ct) of the am-plification products was determined at a specific DRn [(fluores-cence signal of reporter dye/fluorescence signal of passive ROXdye) � baseline]. For each b- and s70-immunoprecipitated sample,the fold enrichment of target DNA regions over background (bgl)was calculated using the formula 1.93Ct(bgl) � Ct(target region). Allfold enrichment values represent the average of three biologicalreplicates, and standard deviations are presented in parentheses(Figs. 4D, 5C). For the digest control regions, the fold enrich-ments over bgl were in the range of 0.03–0.38, indicating thatpromoter-bound transcription complexes did not contribute tothe immunoprecipitation of downstream DNA.

Deighan et al.

86 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.orgDownloaded from

Acknowledgments

We thank J. Gelles and J.W. Roberts for helpful discussion,J. Gelles for purified GreB, and S. Dove for comments on themanuscript. We thank Pinal Kanabar (Waksman Genomics CoreFacility) for analysis of initial transcribed region sequences, PeterHochschild for computing the probabilities that random se-quences contain the target sequence motifs, and Jonathan Livnyfor assistance with generating Supplemental Table S1. Work wassupported by NIH GM44025 to A.H., and a Pew Scholars Awardto B.E.N.

References

Arndt KM, Chamberlin MJ. 1988. Transcription terminationin Escherichia coli. Measurement of the rate of enzymerelease from Rho-independent terminators. J Mol Biol 202:271–285.

Bar-Nahum G, Nudler E. 2001. Isolation and characterization ofs70-retaining transcription elongation complexes fromEscherichia coli. Cell 106: 443–451.

Belogurov GA, Vassylyeva MN, Svetlov V, Klyuyev S, GrishinNV, Vassylyev DG, Artsimovitch I. 2007. Structural basisfor converting a general transcription factor into an operon-specific virulence regulator. Mol Cell 26: 117–129.

Brodolin K, Zenkin N, Mustaev A, Mamaeva D, Heumann H.2004. The s70 subunit of RNA polymerase induces lacUV5

promoter-proximal pausing of transcription. Nat Struct Mol

Biol 11: 551–557.Chander M, Austin KM, Aye-Han N-N, Sircar P, Hsu LM. 2007.

An alternate mechanism of abortive release marked by theformation of very long abortive transcripts. Biochemistry 46:12687–12699.

Cho B-K, Zengler K, Qiu Y, Park YS, Knight EM, Barrett CL, GaoY, Palsson BO. 2009. The transcription unit architecture ofthe Escherichia coli genome. Nat Biotechnol 11: 1043–1049.

Deighan P, Hochschild A. 2007. The bacteriophage lQ anti-terminator protein regulates late gene expression as a stablecomponent of the transcription elongation complex. Mol

Microbiol 63: 911–920.Epshtein V, Nudler E. 2003. Cooperation between RNA poly-

merase molecules in transcription elongation. Science 300:801–805.

Goldman SR, Ebright RH, Nickels BE. 2009. Direct detection ofabortive RNA transcripts in vivo. Science 324: 927–928.

Goliger JA, Roberts JW. 1989. Sequences required for antitermi-nation by phage 82 Q protein. J Mol Biol 210: 461–471.

Grainger DC, Hurd D, Harrson M, Holdstock J, Busby SJ. 2005.Studies of the distribution of Escherichia coli cAMP-receptorprotein and RNA polymerase along the E. coli chromosome.Proc Natl Acad Sci 102: 17693–17698.

Gross CA, Chan C, Dombroski A, Gruber T, Sharp M, Tupy J,Young B. 1998. The functional and regulatory roles of s

factors in transcription. Cold Spring Harb Symp Quant Biol

63: 141–155.Gruber TM, Gross CA. 2003. Multiple s subunits and the

partitioning of bacterial transcription space. Annu Rev Micro-

biol 57: 441–466.Hatoum A, Roberts JW. 2008. Prevalence of RNA polymerase

stalling at Escherichia coli promoters after open complexformation. Mol Microbiol 68: 17–28.

Herring CD, Raffaelle M, Allen TE, Kanin EI, Landick R, AnsariAZ, Palsson BO. 2005. Immobilization of Escherichia coli

RNA polymerase and location of binding sites by use ofchromatin immunoprecipitation and microarrays. J Bacteriol

187: 6166–6174.

Hsu LM, Vo NV, Kane CM, Chamberlin MJ. 2003. In vitro stud-ies of transcript initiation by Escherichia coli RNA Poly-merase. 1. RNA chain initiation, abortive initiation, andpromoter escape at three bacteriophage promoters. Biochem-

istry 42: 3777–3786.Kapanidis A, Margeat E, Laurence TA, Doose S, Ho SO,

Mukhopadhyay J, Kortkhonjia E, Mekler V, Ebright RH,Weiss S. 2005. Retention of transcription initiation factors70 in transcription elongation: Single-molecule analysis. MolCell 20: 347–356.

Kapanidis A, Margeat E, Ho SO, Kortkhonjia E, Weiss S, EbrightRH. 2006. Initial transcription by RNA polymerase proceedsthrough a DNA-scrunching mechanism. Science 314: 1144–1147.

Ko DC, Marr MT, Guo TS, Roberts JW. 1998. A surface ofEscherichia coli s70 required for promoter function and anti-termination by phage l Q protein. Genes Dev 12: 3276–3285.

Marr MT, Roberts JW. 2000. Function of transcription cleavagefactors GreA and GreB at a regulatory pause site. Mol Cell 6:1275–1285.

McClure WR. 1985. Mechanism and control of transcriptioninitiation in prokaryotes. Annu Rev Biochem 54: 171–204.

Mekler V, Kortkhonjia E, Mukhopadhyay J, Knight J, RevyakinA, Kapanidis AN, Niu W, Ebright YW, Levy R, Ebright RH.2002. Structural organization of bacterial RNA polymeraseholoenzyme and the RNA polymerase-promoter open com-plex. Cell 108: 599–614.

Mooney RA, Landick R. 2003. Tethering a s70 to RNA poly-merase reveals high in vivo activity of s factors and s70-dependent pausing at promoter-distal locations. Genes Dev

17: 2839–2851.Mooney RA, Darst SA, Landick R. 2005. s and RNA polymerase:

An on-again, off-again relationship? Mol Cell 20: 335–345.Mooney RA, Davis SE, Peters JM, Rowland JL, Ansari AZ,

Landick R. 2009a. Regulator trafficking on bacterial tran-scription units in vivo. Mol Cell 33: 97–108.

Mooney RA, Schweimer K, Rosch P, Gottesman M, Landick R.2009b. Two structurally independent domains of E. coli NusGcreate a regulatory plasticity via distinct interactions withRNA polymerase and regulators. J Mol Biol 391: 341–358.

Mukhopadhyay J, Kapanidis AN, Mekler V, Kortkhonjia E,Ebright YW, Ebright RH. 2001. Translocation of s70 withRNA polymerase during transcription: Fluorescence reso-nance energy transfer assay for movement relative to DNA.Cell 106: 453–463.

Murakami KS, Masuda S, Darst SA. 2002. Structural basis oftranscription initiation: T. aquaticus RNA polymerase holo-enzyme at 4 A resolution. Science 296: 1280–1284.

Nickels BE. 2009. Genetic assays to define and characterizeprotein–protein interactions involved in gene regulation.Methods 47: 53–62.

Nickels BE, Mukhopadhyay J, Garrity SJ, Ebright RH, HochschildA. 2004. The s70 subunit of RNA polymerase mediates apromoter-proximal pause at the lac promoter. Nat Struct Mol

Biol 11: 544–550.Nickels BE, Garrity SJ, Mekler V, Minakhin L, Severinov K,

Ebright RH, Hochschild A. 2005. The interaction betweens70 and the b-flap of Escherichia coli RNA polymeraseinhibits extension of nascent RNA during early elongation.Proc Natl Acad Sci 102: 4488–4493.

Perdue SA, Roberts JW. 2010. A backtrack-inducing sequence isan essential component of E. coli s70-dependent promoter-proximal pausing. Mol Microbiol 78: 636–650.

Raffaelle M, Kanin EI, Vogt J, Burgess RR, Ansari AZ. 2005.Holoenzyme switching and stochastic release of s factorsfrom RNA polymerase in vivo. Mol Cell 20: 357–366.

Retention of s70 in elongation complex

GENES & DEVELOPMENT 87

Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.orgDownloaded from

Reppas NB, Wade JT, Church GM, Struhl K. 2006. The transi-tion between transcriptional initiation and elongation inE. coli is highly variable and often rate limiting. Mol Cell

24: 747–757.Revyakin A, Liu C, Ebright RH, Strick TR. 2006. Abortive ini-

tiation and productive initiation by RNA polymerase involveDNA scrunching. Science 314: 1139–1143.

Ring BZ, Roberts JW. 1994. Function of a nontranscribed DNAstrand site in transcription elongation. Cell 78: 317–324.

Ring BZ, Yarnell WS, Roberts JW. 1996. Function of E. coli RNApolymerase s factor s70 in promoter-proximal pausing. Cell

86: 485–493.Roberts JW, Yarnell W, Bartlett E, Guo J, Marr M, Ko DC, Sun H,

Roberts CW. 1998. Antitermination by bacteriophage l Qprotein. Cold Spring Harb Symp Quant Biol 63: 319–325.

Roberts JW, Shankar S, Filter JJ. 2008. RNA polymerase elonga-tion factors. Annu Rev Microbiol 62: 211–233.

Sevostyanova A, Svetlov V, Vassylyev DG, Artsimovitch I. 2008.The elongation factor RfaH and the initiation factor s bindto the same site on the transcription elongation complex.Proc Natl Acad Sci 105: 865–870.

Valoczi A, Hornyik C, Varga N, Burgyan J, Kauppinen S, HaveldaZ. 2004. Sensitive and specific detection of microRNAs bynorthern blot analysis using LNA-modified oligonucleotideprobes. Nucleic Acids Res 32: e175. doi: 10.1093/nar/gnh171.

Vassylyev DG, Sekine S, Laptenko O, Lee J, Vassylyeva MN,Borukhov S, Yokoyama S. 2002. Crystal structure of a bacte-rial RNA polymerase holoenzyme at 2.6 A resolution. Nature417: 712–719.

Wade JT, Struhl K. 2004. Association of RNA polymerase withtranscribed regions in Escherichia coli. Proc Natl Acad Sci101: 17777–17782.

Deighan et al.

88 GENES & DEVELOPMENT

Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.orgDownloaded from

10.1101/gad.1991811Access the most recent version at doi: 25:2011, Genes Dev. 

  Padraig Deighan, Chirangini Pukhrambam, Bryce E. Nickels, et al.   functional properties of the bacterial elongation complexInitial transcribed region sequences influence the composition and

  Material

Supplemental 

http://genesdev.cshlp.org/content/suppl/2010/12/28/25.1.77.DC1

  References

  http://genesdev.cshlp.org/content/25/1/77.full.html#ref-list-1

This article cites 43 articles, 14 of which can be accessed free at:

  License

ServiceEmail Alerting

  click here.right corner of the article or

Receive free email alerts when new articles cite this article - sign up in the box at the top

Copyright © 2011 by Cold Spring Harbor Laboratory Press

Cold Spring Harbor Laboratory Press on June 27, 2019 - Published by genesdev.cshlp.orgDownloaded from