11
Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc. Annotated Classic The Insulator Activity of CTCF Victor G. Corces 1, * 1 Department of Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322, USA *Correspondence: [email protected] We are pleased to present a series of Annotated Classics celebrating 40 years of exciting biology in the pages of Cell . This install- ment revisits “The Protein CTCF Is Required for the Enhancer Blocking Activity of Vertebrate Insulators” by Dr. Gary Felsenfeld and his colleagues. Here, Dr. Victor G. Corces comments on the landmark discovery of the insulator activity of CTCF from Felsenfeld, who has pioneered the work decoding the function of CTCF in chromatin organization and transcriptional regulation. Each Annotated Classic offers a personal perspective on a groundbreaking Cell paper from a leader in the field with notes on what stood out at the time of first reading and retrospective comments regarding the longer term influence of the work. To see Victor G. Corces’ thoughts on different parts of the manuscript, just download the PDF and then hover over or double-click the highlighted text and comment boxes on the following pages. You can also view Corces’ annotation by opening the Comments navigation panel in Acrobat.

The Insulator Activity of CTCF - Cell · Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc. Annotated Classic The Insulator Activity of CTCF Victor G. Corces1,* 1Department of Biology,

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: The Insulator Activity of CTCF - Cell · Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc. Annotated Classic The Insulator Activity of CTCF Victor G. Corces1,* 1Department of Biology,

Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc.

Annotated Classic

The Insulator Activity of CTCFVictor G. Corces1,*1Department of Biology, Emory University, 1510 Clifton Road NE, Atlanta, GA 30322, USA*Correspondence: [email protected]

We are pleased to present a series of Annotated Classics celebrating 40 years of exciting biology in the pages of Cell. This install-ment revisits “The Protein CTCF Is Required for the Enhancer Blocking Activity of Vertebrate Insulators” by Dr. Gary Felsenfeld and his colleagues. Here, Dr. Victor G. Corces comments on the landmark discovery of the insulator activity of CTCF from Felsenfeld, who has pioneered the work decoding the function of CTCF in chromatin organization and transcriptional regulation.

Each Annotated Classic offers a personal perspective on a groundbreaking Cell paper from a leader in the field with notes on what stood out at the time of first reading and retrospective comments regarding the longer term influence of the work.

To see Victor G. Corces’ thoughts on different parts of the manuscript, just download the PDF and then hover over or double-click the highlighted text and comment boxes on the following pages. You can also view Corces’ annotation by opening the Comments navigation panel in Acrobat.

Page 2: The Insulator Activity of CTCF - Cell · Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc. Annotated Classic The Insulator Activity of CTCF Victor G. Corces1,* 1Department of Biology,

Cell, Vol. 98, 387–396, August 6, 1999, Copyright 1999 by Cell Press

The Protein CTCF Is Requiredfor the Enhancer Blocking Activityof Vertebrate Insulators

al., 1985). When scs elements are placed on either sideof a gene for eye color and introduced into Drosophila,the resulting flies all have similar eye color independentof the transgene’s site of integration, an indication thatscs has protected the reporter gene from both negative

Adam C. Bell, Adam G. West, and Gary Felsenfeld*Laboratory of Molecular BiologyNational Institute of Diabetes and

Digestive and Kidney DiseasesNational Institutes of HealthBethesda, Maryland 20892-0540 and positive endogenous influences, or “position ef-

fects” (Kellum and Schedl, 1991, 1992). Another Dro-sophila insulator element, gypsy, was first identified be-

Summary cause of its ability to block the action of an enhancer ona promoter when it lies between them, but not otherwise

An insulator is a DNA sequence that can act as a barrier (Holdridge and Dorsett, 1991; Geyer and Corces, 1992;to the influences of neighboring cis-acting elements, Dorsett, 1993). Studies of these elements have led topreventing gene activation, for example, when located a working definition of insulators: they protect againstbetween an enhancer and a promoter. We have identi- position effects and/or they block enhancer action in afied a 42 bp fragment of the chicken b-globin insulator directional manner. In the cases of both scs9 and gypsy,that is both necessary and sufficient for enhancer proteins have been identified that bind specifically toblocking activity in human cells. We show that this the DNA elements and are, at least in part, responsiblesequence is the binding site for CTCF, a previously for mediating insulator activity (Geyer and Corces, 1992;identified eleven–zinc finger DNA-binding protein that Zhao et al., 1995).is highly conserved in vertebrates. CTCF sites are Insulator elements have also been identified in verte-present in all of the vertebrate enhancer-blocking ele- brates (Chung et al., 1993, 1997; Robinett et al., 1997;ments we have examined. We suggest that directional Zhong and Krangel, 1997), but the protein(s) respons-enhancer blocking by CTCF is a conserved component ible for their activity has been elusive. In earlier workof gene regulation in vertebrates. from our laboratory, we described a 1.2 kb DNA element

with strong enhancer blocking activity, which was de-rived from the 59 end of the chicken b-globin locus

Introduction(Chung et al., 1993, 1997). This region contains a DNaseI–hypersensitive site, present in all tissues (Reitman and

Enhancer-mediated activation is a fundamental mecha-Felsenfeld, 1990), and its position coincides almost ex-nism of gene regulation in eukaryotes. Enhancers canactly with the region of transition between an activeact over large distances to activate transcription, inde-chromatin conformation, marked both by histone hyper-pendent of their orientation and position relative to theacetylation and heightened sensitivity to DNase I, andpromoter. In many cases, if given access, enhancersan inactive domain extending farther 59 that is insensitivecan act promiscuously to activate transcription of heter-to nuclease and less highly acetylated (Hebbes et al.,ologous promoters. Genome sequencing has revealed1994).many cases where differentially regulated genes neigh-

Recently we reported that we were able to reduce thisbor each other at distances over which enhancers mightelement to a 250 bp “core” fragment that accounted foract, yet the genes are independently regulated. Takena significant portion of the globin insulator enhancertogether, these facts suggest the need for mechanismsblocking activity (Chung et al., 1997). We describe herethat prevent the inappropriate action of an enhancer onthe identification of a single DNA-binding site withina neighboring locus (see for example Hagstrom et al.,the core that is necessary and sufficient for enhancer1996 and Zhou et al., 1996). This restriction must beblocking. We have purified a protein that binds to thisachieved, at least in some cases, without impeding thesite and show that the affinity of various mutant bindingaction of the enhancer within its native locus. A DNAsites for this protein is directly proportional to their ca-element able to function in this way would in effectpacity to act as insulators in vivo. The DNA-bindingconstitute a boundary to the action of an enhancer,protein responsible for this activity is CTCF (CCCTC-preventing it from acting across the boundary, whilebinding factor), a highly conserved and ubiquitous DNA-otherwise leaving the enhancer unimpeded. This prop-binding protein implicated in both transcriptional silenc-erty is one of the defining characteristics of a kind ofing and activation (Baniahmad et al., 1990; Lobanenkovregulatory element only recently recognized: the insula-et al., 1990; Klenova et al., 1993; Filippova et al., 1996;tor (Kellum and Elgin, 1998; Bell and Felsenfeld, 1999;Burcin et al., 1997; Vostrov and Quitschke, 1997).Udvardy, 1999).

We find that functional CTCF-binding sites are alsoThe first DNA sequences to be described as havingthe properties of an “insulator” were the scs and scs9 present in other insulators from diverse vertebrate spe-elements of Drosophila, initially identified as marking the cies. Although these elements derive from a variety ofchromatin boundaries of a heat shock locus (Udvardy et genetic loci, they are all located between independently

regulated genes. We suggest that directional enhancerblocking by CTCF is a conserved functional component* To whom correspondence should be addressed (e-mail: gxf@

vger.niddk.nih.gov). of vertebrate domain boundaries.

Victor Corces
Highlight
Victor Corces
Highlight
The reader may notice the use of the terms "insulator", "barrier," and "boundary" throughout the paper. These terms were employed indistinguishably at the time. Later on, Felsenfeld and colleagues proposed to use "insulator" to denote sequences capable of inhibiting enhancer-promoter interactions, whereas the use of "barrier" was restricted to sequences that could inhibit the spreading of heterochromatic regions.
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
The identification of the scs and scs' elements by Schedl and colleagues took a lot of courage, and I don't think a grant application proposing these experiments would be funded today. The working hypothesis underlying this work was that puffs of Drosophila polytene chromosomes are regions of "open" chromatin, and there must be a boundary that delimits the spread of a puff. Given the resolution of polytene chromosomes, the location of this boundary can only be guessed to be within a 100 kb region, approximately. The identification of the scs and scs' sequences within this large regions was certainly a tour de force at the time.
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
CTCF is not only conserved in vertebrates but also present in Drosophila and most bilaterians. This explains an important observation in the original 1993 manuscript by Felsenfeld and colleagues describing the beta-globin insulator: the DNA sequence from chickens also acted as an insulator in Drosophila. This makes sense now when one considers that the binding sites for CTCF in Drosophila and vertebrates are almost identical.
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
The identification of an insulator element in the gypsy retrotransposon has a long and interesting history. Ed Lewis at Caltech had found a recessive mutation that he called "suppressor of Hairy-wing" because it was able to revert the phenotype of a second mutation named Hairy-wing. He then tested the ability of su(Hw) to suppress the phenotypes of other mutations he had in the lab, and he found that indeed the phenotypes of mutations in other genes such as yellow, forked, and Ultrabithorax could also be reverted to wild-type when in the presence of su(Hw) mutations. Like much of his work, these findings were never published. Luckily Matt Meselson did a sabbatical in Lewis' lab in 1978. An important insight to explain these observations came from his realization that not all alleles of a gene were responsive to the effect of su(Hw), and that only spontaneous, but not chemically or X-ray induced, alleles would be affected. He then hypothesized that all spontaneous alleles must have been caused by the insertion of the same transposable element. When Bender and Hogness isolated the Ultrabithorax gene, they realized that all Ubx alleles responsive to mutations in su(Hw) were caused by insertion of a transposon they called gypsy. Years later, it was found that a specific sequence of gypsy that interacted with the Su(Hw) protein could function as an insulator and was able to inhibit promoter-enhancer interactions. This could then explain Lewis' original observation: mutations of su(Hw) would inactivate the insulator and reverse the effect of gypsy on the enhancers of the gene surrounding the insertion site.
Victor Corces
Highlight
Drosophila insulators appeared to pose both barrier and enhancer-blocking activity whereas the 42 bp CTCF insulator described here did not. In retrospect, this is probably a consequence of the way in which barrier function was measured in Drosophila, using transgenes displaying a variegated phenotype and generated by P-element insertion. We now know that P-element-containing transgenes tend to insert into regulatory regions of genes and, thus, the "barrier" assay was probably also measuring interactions between enhancers and promoters.
Victor Corces
Highlight
Victor Corces
Highlight
Page 3: The Insulator Activity of CTCF - Cell · Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc. Annotated Classic The Insulator Activity of CTCF Victor G. Corces1,* 1Department of Biology,

Cell388

Figure 1. Fine Mapping of the Insulator Core

(A) The position of HS4 was measured by comparing the migration of the fragment generated by limited digestion of chicken erythrocytechromatin with DNase I with the migration of genomic DNAs of known length. The position of HS4 relative to previously defined DNase Ifootprints (Chung et al., 1997) is indicated. (B-D) Results of enhancer blocking assays in which the elements indicated were placed betweenenhancer and promoter as shown in (E). A schematic of each inserted element is shown, as well as the relative numbers of colonies observed,and the numerical value of the insulation effect (fold insulation) relative to the uninsulated controls (No Insert [pNI] and l DNA [pJC3-4]). Thedata presented in this figure represent the average of at least 4 independent assays. (B) Enhancer blocking activity of fragments of the core.(C) Effect on enhancer blocking of deletion of footprinted regions from the core. (D) Increasing enhancer blocking observed when insulatingelements were multimerized. (E) A schematic diagram of the construction used to test various DNA fragments for enhancer blocking activity.

Results responsible for the majority of the enhancer blockingactivity of the core. While deletion of FI slightly increasedthe enhancer blocking effect, deletion of FII and FIIIWe showed in earlier studies that a 1.2 kb DNA element

at the 59 end of the chicken b-globin locus, correspond- significantly reduced enhancer blocking activity (Figure1C). Deletions of FIV and FV were essentially neutral.ing to a constitutive DNase I–hypersensitive site (59HS4),

functions as an insulator in an enhancer blocking assay. Insertion of an increasing number of copies of the FII/III fragment between the enhancer and the promoterThis assay tests the ability of a sequence to prevent

activation of a gene for neomycin resistance by a strong resulted in a stepwise increase in blocking activity; anincrease was also observed for the 1.2 kb insulator andenhancer when the construct is stably transformed into

an erythroleukemia cell line (Chung et al., 1993). The the core (Figure 1D). Taken together these results showthat the FII/III fragment is the functional enhancer-insulator effect is manifested by a marked reduction in

the number of colonies resistant to G418 only when blocking region of the core.Further analysis of the FII/FIII fragment showed thatthe globin insulator is placed between enhancer and

promoter. By this same assay, we showed that a large removal of the “spacer” sequence between FII and FIIIresulted in even stronger blocking activity (Figure 2A).part of the insulator activity is contained in a 250 bp

GC-rich “core” fragment at the 59 end of the 1.2 kb In fact, by removing sequences adjacent to FII, we ob-tained a 42 bp sequence spanning FII that alone pos-element (Chung et al., 1997). HS4 maps precisely within

this core region, consistent with its significance in vivo sessed a blocking activity nearly equal to that of the full1.2 kb insulator. Consistent with its functional impor-(Figure 1A).tance, we note that the position of FII is coincident withthat of HS4 in nuclei (Figure 1A). Importantly, enhancerFine Mapping of Directional

Enhancer-Blocking Sequences blocking by FII displays the same position-dependenceas that observed for the full 1.2 kb insulator (Figure 2B;DNase I footprinting of the core revealed five protected

regions (FI to FV, illustrated in Figure 1A; Chung et al., Chung et al., 1997). When placed either upstream ofthe enhancer or downstream of the promoter, FII has1997). We chose to focus on this fragment to identify

an insulator protein–binding site. We divided the core essentially no effect on colony number in our assay.Thus, in order to affect expression, FII must be locatedinto separate segments and carried out enhancer blocking

assays with each fragment. Splitting the core between between the enhancer and the promoter.FII and FIII generated two fragments (FI/FII and FIII/IV/V) each of which had some enhancer blocking activity. Identification of a Candidate

Enhancer-Blocking ProteinHowever, a fragment containing only FII and FIII hadgreater activity than the entire core (FII/III, Figure 1B). Since the FII fragment had the strongest activity, we

focused our attention on identifying proteins that boundDeletion analyses confirmed that FII and FIII were

Victor Corces
Highlight
We now know that CTCF binds in the spacer region between nucleosomes, and that CTCF binding sites are hypersensitive to DNase I. Therefore, the overlap between FII and the HS4 hypersensitive site was an important insight suggesting that Felsenfeld and colleagues were on the right track to find the protein responsible for insulator function.
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
The slight increase in insulator activity by deletion of FI and the slight decrease by deletion of FIII is not considered further in the manuscript and it is not clear whether it is due to the binding of additional proteins or to alterations in the binding of CTCF. In vitro, high affinity interaction of CTCF with DNA requires a sequence of around 50 bp. Experiments by Pugh and colleagues using ChIP-exo have recently defined the CTCF binding site and the nucleotides required for interaction in exquisite detail. However, we still lack a crystal structure of CTCF bound to DNA and, therefore, we do not understand the details of how this protein interacts with its target. In principle, it is possible that FI and FIII are involved in proper interaction of CTCF with its DNA target but it is also possible that other proteins that interact with CTCF make contacts with these sequences.
Victor Corces
Highlight
Victor Corces
Highlight
Felsenfeld and colleagues would later find that the 1.2 kb region contains two separate functional elements, one responsible for the enhancer blocking activity analyzed here and the other responsible for the separation of the two domains containing active and silenced chromatin found at the edge of the beta-globin locus. The two activities require different proteins.
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
The authors emphasize at different points in the manuscript that the DNase hypersensitive site is both specially strong and constitutive across cell types. Recent work comparing the strength of DNase hypersensitivity and protein DNA occupancy suggests a direct correlation between the two. The strength of the 5'HS4 site may then reflect occupancy by other proteins in addition to CTCF, including, as we now know, cohesin. We also know now that 60%–70% of CTCF sites in the genome are conserved across different cell types, and that these conserved sites are more likely to function as enhancer blockers, rather than enhancer facilitators, and are more likely to be present at TAD borders (see below).
Victor Corces
Highlight
Page 4: The Insulator Activity of CTCF - Cell · Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc. Annotated Classic The Insulator Activity of CTCF Victor G. Corces1,* 1Department of Biology,

A Vertebrate Enhancer-Blocking Protein389

each of these proteins for this sequence. A deletion of4 base pairs within the region that overlaps both thea2- and the Su[Hw]-binding sites had no effect on theblocking activity of the FII/III fragment (Figure 2D). Fur-thermore, a 100 bp fragment, derived from the Drosoph-ila gypsy element, that contains three canonical Su[Hw]-binding sites, had no activity in our assay. We concludethat neither the Su[Hw] nor the a2 site can account forthe activity of FII.

Likewise, in the context of FII, mutation of the Sp1consensus had no effect on blocking activity; in fact,mutation of each of the three potential Sp1-binding sitesin FII/III resulted in substantially increased activity (Fig-ure 2C). Sp1 may act as an inhibitor of enhancer blockingin our assay. This may also explain the inhibitory effectof the “spacer” sequence between FII and FIII notedabove.

To determine which sequences within FII are respon-sible for its activity we made multiple transversions(C→A and G→T) across the 59, middle (M), and 39 regionsof the fragment (Figure 3A). All of these reduced theactivity of FII, but changes at the 39 end of the fragment(x39) caused a complete loss of enhancer blocking activ-ity. Likewise, deletion of 10 bp from both ends (DF)or a reversal of the sequence 59→39 (rev) resulted insubstantial losses in activity.

Armed with this information, we searched for a proteinthat bound to FII with a competition profile that matchedthe sequence specificity observed in the enhancerblocking assay. To do this, we made nuclear extractsfrom the human erythroleukemic cell line K562 (the cellline in which the enhancer blocking assay is performed)and from adult chicken red blood cells (since this insula-tor is a chicken element). Identical patterns were ob-tained with these two extracts in a gel mobility-shiftassay (Figure 3B). In each case two major complexeswere observed when the extract was incubated with aFigure 2. Identification of a Minimal Enhancer-Blocking Site

60 bp probe spanning footprint II. The upper complex(A) A 90 bp fragment spanning FII and FIII was subjected to furtherdeletion. Deleted regions are indicated by a dashed line. The effects could be supershifted with an antibody against Sp1 andof these deletions on enhancer blocking are shown. (B) Examination was competed by a 100-fold excess of an unlabeled Sp1of the effect of the relative positions of enhancer and promoter on consensus–binding site. We conclude that this complexthe enhancer blocking effect of FII in the colony assay. (C) Enhancer

contains Sp1. In contrast, the lower complex was neitherblocking activities of mutants of FII or FII/III aimed at assessing thesupershifted by anti-Sp1, nor was its binding influencedcontribution of binding sites for Sp1 or (D) a2 and Su(Hw) bindingby an excess of Sp1 consensus–binding site. More im-site homologies. (E) Alignments of Sp1, a2, and Su(Hw) binding sites

with FII. The grayed base pairs TAAT were deleted to examine the portantly, the affinity of the lower complex for variouscontribution of the a2 and Su(Hw) homologies to enhancer blocking mutants of FII paralleled, with striking accuracy, theand DNA binding; the grayed CC→AA mutation was used to reduce enhancer blocking activities of those same fragmentsSp1 binding to FII and FII/III (Anderson and Freytag, 1991). The

(Figure 3C). The slight deviations we observed couldenhancer blocking data presented in this figure represent the aver-arise from differential effects of the mutations on theage of 2–5 independent assays for each construction.binding of the insulator protein and Sp1 which, as weshowed earlier, complicate interpretation of the in vivoresults.

to it. A comparison of the sequence of FII with that ofknown transcription factor–binding sites revealed sev-

A Single Protein Is Responsible for the Sequenceeral potentially significant homologies. An Sp1 consen-Specificity of Enhancer Blockingsus sequence lies in the middle of the fragment and aProbing a blot of nuclear proteins with labeled FII re-sequence homologous to a yeast a2-binding site (Sauervealed a single FII-specific DNA-binding protein with anet al., 1988) overlaps a partial match to the binding siteapparent size of z140 kDa (Figure 4A). The affinity ofof the Drosophila protein suppressor of Hairy-wingthis protein for FII variants was identical to that observed(Su[Hw]) (Figure 2E; Geyer and Corces, 1992). To testin the gel-shift assay and was also in good agreementwhether any of these homologies could account for thewith the enhancer blocking data (Figure 3C and 4A). Weblocking activity of FII, we introduced mutations that

were predicted to reduce dramatically the affinity of purified this protein from chicken red blood cell nuclear

Victor Corces
Highlight
The gypsy insulator contains 12 binding sites for Su(Hw). Subsequent experiments showed that three of them are not sufficient for insulator function in Drosophila either.
Victor Corces
Highlight
Page 5: The Insulator Activity of CTCF - Cell · Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc. Annotated Classic The Insulator Activity of CTCF Victor G. Corces1,* 1Department of Biology,

Cell390

Figure 3. Sequence Specificity of Enhancer Blocking and Nuclear Factor Binding by FII

(A) The enhancer blocking capacity of the indicated FII variants was measured in the colony assay.(B) Gel mobility shift assays of FII and nuclear extracts from human K562 and chicken red blood cells (RBC). Cold competitors as indicatedwere added at a 100-fold molar excess in these experiments (sequences are in [A]; N is a nonspecific oligo, a(Sp1) indicates addition of Sp1antiserum).(C) Comparison of the capacity of the indicated FII mutants to (i) act as insulators in the colony assay (black), (ii) compete with FII for bindingto a candidate insulator protein in gel shift (striped), and (iii) compete with FII for binding to CTCF in a Southwestern assay (stippled). Datawere normalized with FII activity considered as 100% in each assay.

extract by conventional chromatography. Throughout Conservation of Sequence amongVertebrate Insulatorsthe purification, the elution profiles of the FII binding

activities were identical in gel-shift and Southwestern Because CTCF is highly conserved among vertebrates,we examined whether CTCF sites are present in otherassays (data not shown). This protein bound tightly to

S, CM, and hydroxyapatite columns, and eluted with a vertebrate insulators. Two such elements have recentlybeen described. A 1.4 kb fragment found in the in-peak at z330 kDa on gel filtration (Figure 4B). Coomas-

sie staining of gels from the final hydroxyapatite frac- tergenic spacer region of the ribosomal RNA genes ofXenopus laevis, termed the repeat organizer (RO), hastions revealed a single protein with an apparent molecu-

lar weight of z140 kDa corresponding to the position been shown to prevent enhancer action in a directionalmanner (Robinett et al., 1997). The 39 half of this se-of the FII Southwestern activity. The sequences of four

internal peptides (Figure 4C) from the 140 kDa DNA- quence is composed of seven tandem repeats of anz100 bp GC-rich sequence (Labhart and Reeder, 1987).binding component of our final purified fraction all per-

fectly matched the predicted sequence of a previously This sequence bears significant homology with CTCFsites including FII (Figure 6B). Moreover, a DNA fragmentcloned 11–zinc finger DNA-binding protein, CTCF (Klen-

ova et al., 1993; Filippova et al., 1996). spanning one repeat unit of RO binds specifically, albeitweakly, to CTCF in vitro (Figure 7A). In our in vivo assay,Consistent with this identification, in vitro–translated

CTCF binds to FII with a sequence specificity identical the full-length RO element conferred considerable en-hancer blocking activity, and a single copy of the 100to that observed in our gel-mobility shift and enhancer

blocking assays (Figure 5). As is expected, this protein bp repeat from this element had weak enhancer blockingactivity on its own (Figure 7B).also binds to other previously characterized CTCF sites

(Figure 5, lanes 9–11), and these sites also act as en- Another vertebrate insulator, BEAD-1, is a 1.6 kb en-hancer-blocking element derived from the human T cellhancer blockers in our assay (Figure 5). Alignment of

these CTCF sites with FII reveals a conserved region receptor a/d locus (Zhong and Krangel, 1997). Best-fit alignment of this element with various CTCF sitesthat has been shown to be critical for binding of CTCF

to these other sites (Filippova et al., 1996; Burcin et al., revealed a good match between FII and a sequenceroughly at the center of this element (BEAD-A in Figure1997; Vostrov and Quitschke, 1997). We note that it was

mutation of this conserved 39 sequence that completely 6C). In fact, a DNA fragment containing this region alsobound specifically to purified chicken CTCF (Figure 7A).abrogated CTCF binding and enhancer blocking in our

assays (see x39 in Figure 3 and alignments in 6A). Consistent with these observations, both full-length

Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
The finding that CTCF, an evolutionarily conserved protein, was responsible for the insulator function found in the chicken beta-globin locus was a landmark finding. Until then, most hard-core scientists working on transcription considered insulators as curious oddities found in Drosophila but of little importance to the issue of gene expression, since insulator-binding proteins of Drosophila appeared not to be conserved in vertebrates. Here, a conserved protein previously shown to be important in transcription turned out to be crucial for insulator function. From this time on, talks on insulators at transcription meetings were no longer scheduled on the last day, after the bus to the airport had already left.
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
The binding site of CTCF found here, and those now known at many genomic locations, contains a CpG site that can be methylated. This modification interferes with the binding of CTCF to its target. This may be an important mechanism to regulate binding of CTCF at specific sites in the genome, and may underlie cell-type-specific roles of this protein during cell lineage commitment.
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
At the time, several different sequences with insulator function had been described in both Drosophila and vertebrates. In Drosophila, these various insulator sequences bound different proteins, but none of these proteins had been found in vertebrates. The finding that all previously characterized insulator sequences in vertebrates could interact with CTCF was at once satisfying and perplexing. To date, CTCF is the main DNA-binding insulator protein found in vertebrates, and it is still not clear why Drosophila, with a 20-fold smaller genome, would require a more complex set of insulators to control the expression of its genes.
Victor Corces
Highlight
Victor Corces
Highlight
Page 6: The Insulator Activity of CTCF - Cell · Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc. Annotated Classic The Insulator Activity of CTCF Victor G. Corces1,* 1Department of Biology,

A Vertebrate Enhancer-Blocking Protein391

Figure 4. Purification of an FII-Binding Factor

(A) Southwestern assay reveals sequence-specific FII binding at z140 kDa apparentmolecular weight in protein fractions ob-tained during different stages of purification.Identical blots were probed simultaneouslywith labeled FII in the presence of a 50-foldexcess of the indicated unlabeled cold com-petitor.(B) An outline of the scheme used to purifythe FII-binding factor.(C) A representative example of a Coomassie-stained gel of the purified fractions elutedfrom the hydroxyapatite column is shown onthe left with the internal peptide sequencesobtained from the indicated band shown. Theresult of a Southwestern assay of FII bindingto this fraction is shown in the right panel.

BEAD-1 and the CTCF-binding BEAD-A element defined However, mutations of the 39 end of this site did abolishenhancer blocking.here were effective enhancer-blocking elements in our

assay (Figure 7B). Furthermore, deletion of the BEAD-A When FII was used as a probe in gel retardation ex-periments with crude extracts, two major complexessequence from BEAD-1 largely eliminated the activity of

the larger element. were observed. One of these was attributable to Sp1.The other had properties implicating it in insulating activ-ity: it was competed by any DNA that was also activeDiscussionin the enhancer blocking assay, but not by any of theinactive mutated sequences. Similar gel shift patternsA major defining property of insulators is their abilitywere obtained with extracts from chicken erythrocyteto interfere with enhancer–promoter interaction whennuclei and the human erythroleukemia line K562, inplaced between them. Our laboratory previously haswhich the enhancer blocking assays were carried out.shown that a 1.2 kb element at the 59 end of the chickenWe used this result to purify the protein responsible forb-globin locus has such enhancer blocking activity. Herethis complex and found that it was identical to a knownwe identify a small DNA sequence motif within this insu-protein, CTCF. Consistent with this, in vitro–translatedlator that is sufficient to account for most of its abilityCTCF bound FII with sequence specificity identical toto block enhancers. A 42 bp fragment containing thethe original complex. Furthermore, the original complexmotif was able to suppress enhancer activity in a direc-was supershifted with an antibody against CTCF.tional manner about as well as the full 1.2 kb element

CTCF is an 82 kDa protein with 11 zinc fingers (Filip-from which it was derived. Although the fragment con-pova et al., 1996) and is characterized by an unusuallytains binding sites for Sp1 and the yeast a2 repressor,

mutating these sites had no effect on blocking activity. extensive DNase I footprint (51 bp) when bound to its

Victor Corces
Highlight
Later work by Felsenfeld and colleagues would show that four Zn fingers are sufficient for proper CTCF binding. It is still unclear what the role of the other seven fingers is. One possibility is that these other fingers interact with adjacent nucleotides in a site-specific manner, distorting the structure of CTCF at specific sites in the genome and allowing it to interact with different proteins. Alternatively, these fingers may directly interact with other proteins. We now know that CTCF is present together with cohesin at many of its genomic locations and that cohesin is essential for insulator function, but Felsenfeld and colleagues have shown that the C terminus of CTCF, and not the Zn fingers, are involved in interactions with the SA2 subunit of the cohesin complex. Other proteins such as Kaiso, YY1, CHD8, PARP1, MAZ, JUND, ZNF143, PRDM5, and nucleophosmin have been reported to directly or indirectly interact with CTCF, and it is possible that the Zn fingers not involved in DNA binding are involved in interactions with some of these proteins.
Victor Corces
Highlight
Victor Corces
Highlight
Page 7: The Insulator Activity of CTCF - Cell · Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc. Annotated Classic The Insulator Activity of CTCF Victor G. Corces1,* 1Department of Biology,

Cell392

Figure 5. FII Binding and Enhancer Blockingby CTCF

Purified FII-binding factor (lane 1) and invitro–translated CTCF (lanes 2–11) have iden-tical specificity for FII (compare to Figure 3)and identical migration in a gel shift assaywhen bound to FII (lanes 2–8) or previouslycharacterized CTCF sites from the chickenc-myc promoter (lane 9), the chicken lyso-zyme promoter (lane 10) or the human amy-loid b protein promoter (lane 11). The righttable summarizes the capacity of these CTCFsites to act as enhancer blockers in the col-ony assay. Data are the average of two inde-pendent measurements.

site on DNA, consistent with an involvement of several these elements requires recognition of particular insula-tor sequences by specific DNA-binding proteins. In Dro-fingers in typical binding sites. It migrates anomalously

on acrylamide/SDS gels, which accounts for the dis- sophila, three seemingly unrelated DNA-binding pro-teins are required for the action of distinct insulatorscrepancy in apparent molecular weight (Klenova et al.,

1997). Studies of CTCF in other systems suggest that it (Spana et al., 1988; Holdridge and Dorsett, 1991; Geyerand Corces, 1992; Zhao et al., 1995; Ohtsuki and Levine,can play a variety of regulatory roles. It binds to the

promoter of the amyloid b protein precursor and causes 1998). These proteins act, presumably in concert withother factors, to somehow restrict the action of an en-transcriptional activation (Vostrov and Quitschke, 1997),

but when it interacts with sites in the c-myc oncogene hancer in a directional manner (Kellum and Elgin, 1998;Bell and Felsenfeld, 1999; Udvardy, 1999). While theit causes repression (Filippova et al., 1996). It is also

capable of acting in synergy with certain thyroid hor- mechanism through which insulator action is achievedremains unknown, sequence-specific binding by thesemone receptor binding sites both in repression and in

T3 induction (Baniahmad et al., 1990). Not all of the 11 proteins is a necessary step in defining this process. Ourdata strongly support the notion that CTCF is capablezinc fingers of the protein are involved in binding to

the sites that have been examined so far. Furthermore, of directing enhancer blocking in vertebrates. First, bywhittling down the chicken b-globin insulator to a mini-different sites employ partially different subsets of fin-

gers to contact the DNA (Filippova et al., 1996). One can mal active element, we identified a CTCF site that playsan essential role in that element’s activity. Accurate ap-imagine that the characteristics of the binding site would

have a large influence on the conformation of the protein, praisal of the full enhancer blocking capacity of thiselement was complicated by the presence of Sp1 sites,the nature of its interactions with cofactors, and its ulti-

mate biological effect. whose activity apparently masked that of CTCF in ourassay. By mutating the Sp1 sites in this region (FigureThe best characterized insulators are found in Dro-

sophila. The directional enhancer blocking activity of 2C, FII/III-DSp1*), however, the “full” 9-fold enhancer

Figure 6. Sequence Homologies among CTCFSites and Vertebrate Insulators

(A) Alignment of FII with other known CTCFsites reveals a conserved 39 region which cor-responds to the sequence altered in the x39

mutant (see Figure 3).(B) Alignment of the 100 bp repeats of theXenopus RO element and FII.(C) Alignment of FII with a homologous site(BEAD-A) in the BEAD-1 element from the hu-man T cell receptor a/d locus.

Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
From a mechanistic perspective, it is still unclear whether the original roles assigned to CTCF as an activator or a repressor can be explained by its function as an insulator. Although it seems unlikely, it is still possible that at very specific sites CTCF associates with proteins that mediate a mechanistically different role for CTCF to activate transcription. Perhaps the original role as a transcription factor can be explained by its ability to facilitate enhancer-promoter interactions at some sites in the genome (see below). However, the evidence of a role for CTCF as a classical repressor is scant.
Victor Corces
Highlight
Victor Corces
Highlight
It is now well established that the function of CTCF as an insulator depends on its ability to mediate interactions between distant sites in the genome. Interestingly, at some genomic sites these interactions are involved in recruiting enhancers to the appropriate promoter, whereas at other sites CTCF seems to block interactions between sequences located at either side. But from a mechanistic perspective, we still don't understand how insulators block enhancer-promoter interactions.
Victor Corces
Highlight
Victor Corces
Highlight
Page 8: The Insulator Activity of CTCF - Cell · Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc. Annotated Classic The Insulator Activity of CTCF Victor G. Corces1,* 1Department of Biology,

A Vertebrate Enhancer-Blocking Protein393

of a stably integrated reporter gene, the reporter is pro-tected both against variation in expression from one lineto another, and also against extinction of expressionover a period of at least 40–80 days in culture (Pikaartet al., 1998). Preliminary results (F. Recillas-Targa et al.,unpublished) indicate that this activity depends uponsequences other than FII within the insulator. The com-plete activity of the b-globin 59 insulator element is thuslikely to involve multiple components.

We find that the enhancer blocking activity of the 59b-globin insulator is dependent upon CTCF, and thatsimilar sequences are present in the two other verte-brate insulators. The first of these is the BEAD-1 elementfound in the human T cell receptor (TCR) a/d locus(Zhong and Krangel, 1997). BEAD-1, which has strongdirectional enhancer blocking properties, is located be-tween TCRd gene segments and TCRa joining gene seg-ments. It has been proposed that BEAD-1 prevents ad-specific enhancer from acting on the a genes early inT cell development. We have shown here that BEAD-1contains a CTCF-binding site and that this site is respon-sible for a large portion of the observed enhancerblocking activity. We have also examined the enhancerblocking activity of an element that derives from the59-boundary of the chicken lysozyme gene (Stief et al.,1989). Consistent with the findings of Stief et al., wefound that this element conferred considerable en-hancer blocking activity in our assay and that this ele-ment contains (at least) two bona fide binding sites forCTCF, both of which are independently capable of en-hancer blocking in our assays (data not shown).

Figure 7. Conservation of Sequence-Specific Enhancer Blocking A specialized insulator element has been describedActivity among Vertebrate Insulators

within the Xenopus ribosomal RNA repeats (Robinett(A) Gel mobility shift assays with FII, RO542–90, and BEAD-A70 as

et al., 1997). A 100 bp repeating sequence within thisprobes reveal sequence-specific binding to partially purified CTCF.element has significant homology with FII, binds CTCF,An antibody raised against a C-terminal peptide of CTCF specificallyand confers enhancer blocking in our assay. In the sys-supershifts all three complexes. PI indicates the addition of preim-

mune IgY and I indicates the addition of anti-CTCF (C-terminal) IgY. tem described by Robinett et al., enhancer blocking by(B) Enhancer blocking activities of vertebrate insulators. These data the full RO element was only revealed in constructionsare the average of at least two independent experiments with the where the relative position of enhancer, promoter, andexception that the data for the RO elements is from a single determi- insulator elements mimicked the in vivo arrangement ofnation.

these elements. In our assay, this element was effectivein enhancer blocking, whereas analogous constructions

blocking capacity of this element was revealed. This failed to reveal the enhancer blocking capacity of thisactivity is equal to that observed with the entire 1.2 kb element in Xenopus oocytes. These discrepancies mayinsulator (Chung et al., 1993). Our conclusions are further be explained by differences in the enhancer and pro-supported by the observation that, in vivo, the enhancer moter functions in these systems or they may reflectblocking activity of various mutated FII fragments accu- species or cell type–specific differences in the contribu-rately parallels the capacity of these fragments to bind tion of CTCF, or other factors, to the enhancer blockingCTCF in vitro. Finally, in every case we examined, bind- capacity of RO. To date, a Xenopus homolog of CTCFing sites for CTCF were capable of directional enhancer has not been identified. However, given the fact thatblocking in vivo. Taken together, our results show that this protein is highly conserved in other vertebrates (Filip-CTCF binding sites are necessary and sufficient for en- pova et al., 1996), it is reasonable to suppose that suchhancer blocking activity in our assay. The fact that a homolog exists.several DNA-binding proteins have been implicated in There seems little doubt that CTCF plays a major roleinsulator function in Drosophila may indicate that en- in the enhancer blocking activity of all of these elements.hancer-blocking proteins other than CTCF will ultimately Our focus on FII in the b-globin insulator led us to thisbe found in vertebrates. However, the presence of CTCF conserved attribute of vertebrate insulators. A minorsites in each of the vertebrate loci shown thus far to contribution to enhancer blocking is also made by FIII.have this activity is strong evidence for a conserved role This may arise from a weak CTCF consensus in this site.of CTCF in insulator function in vivo. Taken together, our results suggest a conserved and

As we have pointed out in the Introduction, the b-globin perhaps widely used function of insulators, involving59 insulator element shares with the Drosophila insu- CTCF, in the maintenance of distinct regulatory regions.lators the additional ability to protect against position Indeed, recent results in our laboratory (N. Saitoh eteffects. When two copies of the entire 1.2 kb fragment al., unpublished) show that the 39 end of the chicken

b-globin locus is marked by a hypersensitive site withcontaining the b-globin 59 HS4 are placed on either side

Victor Corces
Highlight
It is interesting that, as mentioned above, other DNA-binding insulator proteins have not been yet found in vertebrates. The best candidate for a novel insulator protein in vertebrates is TFIIIC, which is a transcription factor required for RNA Polymerase III function. First, this protein has been shown to posses barrier function in yeast. Second, tRNA genes and SINE elements, both of which contain binding sites for TFIIIC, can function as insulators in reporter assays similar to those used in this paper. TFIIIC binds in the genome of mammals at many sites independent of the presence of RNA polymerase III and, therefore, these sites could represent a second type of insulator in vertebrates. Interestingly, many sites of TFIIIC in the genome are located in close proximity to CTCF sites. This distribution could explain the apparent dual role of CTCF as an enhancer blocker and an enhancer facilitator if, for example, sites containing both proteins act as the former. More recently it has been found that CTCF can associate with RNA, including both noncoding and mRNAs. It is then also possible that the presence of specific RNAs can alter the functional activity of CTCF at certain sites in the genome.
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Page 9: The Insulator Activity of CTCF - Cell · Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc. Annotated Classic The Insulator Activity of CTCF Victor G. Corces1,* 1Department of Biology,

Cell394

the AscI site of this plasmid are located between the enhancersimilar properties to 59HS4 and that it also contains a(mouse HS2) and the reporter (“g-neo”). The following primers wereCTCF-binding site. This element is located between theused in PCR amplifications to generate fragments for cloning intoglobin genes and a nearby gene encoding an odorantthe AscI site of pNI: AC1F, AGGCGCGCCTGGGAGCTCACGGGGAC

receptor (Bulger et al., 1999) further substantiating the AGCCCCC; AC1R, AGGCGCGCCTGGGAGCGCCGGACCGGAGCGnature and likely function of these boundary elements. It GAG; AC2F, AGGCGCGCCGGCTCCGCTCCGGTCCGGCGCTCC;

AC2R, AGGCGCGCCTGTCATTCTAAATCTCTCTTTCAGC; DIACF,may be that in some situations, where enhancer blockingAGGCGCGCCGCCCCCAGGGATGTAATTACGTCC; DIIacF, AGCCCactivity is all that is required, a CTCF-binding site alone isCCCCCCAAA GCCCCCAGGGATGGGGGCAGCAGCGAGCCGC; DI-sufficient, whereas in the case of a permanent chromatinIacR, GGCGGCTCGCTGC TGCCCCCATCCCTGGGGGCTTTGGGGdomain boundary, such as that found at the 59 end ofGGGGGC; DIIIACF, CCGAGCCGGCAGCGTGCGGGGACAG; DIIIACR,

the chicken b-globin locus, additional components are CCCGCACGCTGCCGGCTCGGCGGACCGGAGCGGAGCCCCG;involved. We surmise that, even in those cases where DIVACF, CCTCTGAACGCTTCTCGCTGCTCTT; DIVACR, CAGCGAG

AAGCGTTCAGAGGCCTTCCCCGTGCCCGGGCTG; DVACR, AGGConly CTCF sites are present, the activity of CTCF re-GCGCCGCCCAGGTGTCTGCAGGCTCAAAGAGC; BEADascF, AGGquires the participation of other proteins, just as theCGCGCCGAATTCCAGAAATCTTTGATTTCAGATGCT; BEADascR,directional enhancer blocking activity of the suppres-AGGCGCGCCGGATCCCACTCTTAGCCATTATACTGCATTG; BEAD-sor of Hairy-wing protein involves interaction with theDAF, TGAGCATCTTCAGGGCCCCTGGATTCCATTTCAGAGCTTCC

Mod(mdg4) protein (Gerasimova et al., 1995; Gdula et al., GGTTCTC; BEADDAR, ATCCAGGGGCCCTGAAGATGCTCA. The1996; Georgiev and Kozycina, 1996; Gdula and Corces, core, FI/FII, FIII/IV/V, DFI, and DFV were generated by PCR using

the plasmid p501 (Reitman and Felsenfeld, 1990) as a template1997; Gerasimova and Corces, 1998). We are currentlyand the primer pairs AC1F/AC2R, AC1F/AC1R, AC2F/AC2R, DIACF/exploring the possibility that proteins that copurifiedAC2R, and AC1F/DVACR, respectively. Deletions of FII, FIII, and FIVwith CTCF in our preparations may participate in insula-from the core were accomplished by two-step, overlapping PCR.tor function.For each deletion, a pair of intermediate fragments was generated

We have demonstrated that in our system a CTCF site by PCR in separate reactions using p501 as the template and themust be located between enhancer and promoter to primer pairs AC1F/DIIacR, DIIacF/AC2R, AC1F/DIIIACR, DIIIACF/

AC2R, AC1F/DIVACR, and DIVACF/AC2R. The products of each ofinfluence expression. This result depends only on thethese reactions were gel purified, mixed pairwise to generate theposition of the insulator; it can be inserted in eitherappropriate templates, and the final products were amplified withorientation with equal effect (data not shown). TheseAC1F/AC2R. The full-length BEAD-1 fragment was generated byproperties must be reconciled with our partial under-PCR from K562 genomic DNA with primers BEADascF and BEAD-

standing of how enhancers function. Various models ascR. The fragment BEADDA was generated in a two-step, overlap-have been proposed to account for enhancer blocking. ping PCR reaction, first using the BEAD-1 fragment as a template

and the primers BEADascF/BEADDAR and BEADDAF/ BEADascRThey fall into two broad categories: steric models andin separate reactions, then mixing the gel-purified products of thesetracking models (Kellum and Elgin, 1998; Bell andreactions with primers BEADascF and BEADascR to generate theFelsenfeld, 1999; Udvardy, 1999). Steric mechanismsfinal product by PCR. A 1.6 kb fragment containing the full-lengthpostulate that insulators partition enhancer and pro-RO element was subcloned into Ecl136II cut pJC5-4 after liberation

moter into two separate domains that are inaccessible of this fragment from p0,1 (Robinett et al., 1997) by digestion withto each other. Tracking models suppose that some acti- Ecl136II and PvuII. All other enhancer-blocking fragments were gen-

erated by direct synthesis of the appropriate complementary oligo-vating signal must travel along the DNA from enhancernucleotides on an ABI 394 DNA synthesizer. The top strands ofto promoter, and that the insulator blocks this transmis-these were: FII/III, AGGCGCGCCGGGATGTAATTACGTCCCTCCCCsion. Our inability to distinguish among these modelsCGCTAGGGGGCAGCAGCGAGCCGCCCGGGGCTCCGCTCCGGTresults, in part, from an incomplete understanding ofCCGGCGCTCCCCCCGCATCCCCGAGGGCGCGCCT; FII/III-DSp1*,

how enhancers work. AGGCGCGCCGGGATGTAATTACGTCCCTAACCCGCTAGGGGGCWe conclude that CTCF is likely to play a role in the AGCAGCGAGCCGAACGGGGCTCCGCTCCGGTCCGGCGCTAACC

CCGCATCCCCGAGGGCGCGCCT; FII/III-Da2, AGGCGCGCCGGGAenhancer blocking function of many insulator elements.TGTACGTCCCTCCCCCGCTAGGGGGCAGCAGCGAGCCGCCCGGIn the case of the 59-globin insulator, an independentlyGGCTCCGCTCCGGTCCGGCGCTCCCCCCGCATCCCCGAGGGCregulated gene encoding a folate receptor has recentlyGCGCCT; D spacer, AGGCGCGCCCCCAGGGATGTAATTACGTCC

been identified 59 of the globin locus (Prioleau et al., CTCCCCCGCTAGGGGGCAGCACCGGTCCGGCGCTCCCCCCGC1999). These genes are close enough to each other that ATCCCCGAGCCGGGGCGCGCCT; DIIN, AGGCGCGCCGGGGGCAthe regulatory elements of the two loci might influence GCAGCGAGCCGCCCGGGGCTCCGCTCCGGTCCGGCGCTCCCC

CCGCATCCCCGAGGGCGCGCCT; DIIIN, AGGCGCGCCCCAAAGCeach other inappropriately in the absence of an insula-CCCCAGGGATGTAATTACGTCCCTCCCCCGCTAGGGGGCAGCAtor. A rather similar situation exists in the case of the TGCGAGCCGCCCGGGGCTCCGCGGCGCGCCT; FII, AGGCGCGCCcell receptor locus (Zhong and Krangel, 1997) where theCCCAGGGATGTAATTACGTCCCTCCCCCGCTAGGGGGCAGCAG

BEAD insulator may prevent inappropriate activity of an GCGCGCCT; FIII, AGGCGCGCCCCGGTCCGGCGCTCCCCCCGCenhancer. The presence of CTCF sites in these quite ATCCCCGAGCCGGGGCGCGCCT; gypsy-3, AGGCGCGCCAAAAT

ACATTGCATACCCTCTTTTAATAAAAAATATTGCATACGTTGACGAdifferent genetic loci implies that the role of such sitesAACAAATTTTCGTTGCATACCCAATAAAAGGCGCGCCT; mycFV,in the establishment of enhancer boundaries is likelyAGGCGCGCCGGGGGGGGGCACGGAGCCCCTCGGCCGCCCCCTto be a conserved and important component of geneCGCGGCGCGCCCTCCCCGCTCACGGAGCCCGCGCGGAGCCGG

regulation. GGGCGAGGCGCGCC; lys, AGGCGCGCCTTTAGCTGCATTTGACATGAAGAAATTGAGACCTCTACTGGATAGCTATGGTATTTACATGT

Experimental Procedures CTTTTTGCTTAGTTACTAGGCGCGCC; Apb, AGGCGCGCCCCCTCCCGGCGCGAGCGGGCGCAGTTCCCCGGCGGCGCCGCTAGGGG

Plasmid Constructions and Oligonucleotides TCTCTCTCGGGTGCCGAGCGGGGTGGGCCGGATAGGCGCGCC;The plasmid pNI was the base plasmid for enhancer blocking RO100, AGGCGCGCCGGGGACCCGATTCGGGGTCGGGGCCCCGassays. pNI was generated by replacing the SacI copy of the 1.2 GGGGTGCCCGCTAAGGGGCCCCGGGGGGCCCTCCCGGCGAAGkb insulator in pJC5-4 (Chung et al., 1993) with an AscI linker after AGGGGCCCATTGGCGCGCCT; BEAD-A, AGGCGCGCCGTGGAAG

AGGGATGTTGAGGGCCCAGGGGCTGCCTTGCCGGTGCATTGGCdigestion of this plasmid with Ecl136II. Fragments subcloned into

Victor Corces
Highlight
Victor Corces
Highlight
This statement proved to be extremely insightful. Recent studies using HiC to map all possible interactions in the nucleus suggest that the genome is organized into domains called TADs. TADs are regions with a high frequency of intra-TAD interactions and a low frequency of inter-TAD interactions. The low frequency of inter-TAD interactions creates boundaries between TADs. CTCF is present both inside TADs (85% of all CTCF sites) and at TAD boundaries (15% of all CTCF sites). Inside TADs, CTCF appears to promote interactions, perhaps between enhancers and promoters, whereas at TAD boundaries CTCF appears to interfere with interactions between flanking adjacent sequences present inside TADs. These two apparently opposite roles may be related to the nature of other proteins associated with CTCF at different sites. TAD boundaries are enriched for SINE elements and tRNA genes, suggesting that perhaps association of CTCF with TFIII is responsible for its function in the establishment of TAD boundaries. As suggested above, this would also explain the apparent contradictory roles of CTCF as an enhancer blocker and an enhancer facilitator.
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
These studies eventually resulted in the identification of nucleophosmin as a CTCF-associated protein required for its localization to nucleolar sites. In retrospect, it is interesting that cohesin, which has a much more widespread role in CTCF function, was not identified during this work.
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
This question has not been satisfactorily answered to date. Evidence accumulated since the publication of this manuscript strongly support the idea that enhancers work by forming loops that allow interactions between the transcription complex and enhancer-bound transcription factors. However, mechanistically, it is unclear why the partition of such a loop in two separate loops would interfere with enhancer-promoter interactions. On the other hand, given what we have learned about how enhancers control transcription, the existence of a signal that must travel along the DNA from the enhancer to the promoter seems unrealistic.
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Victor Corces
Highlight
Page 10: The Insulator Activity of CTCF - Cell · Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc. Annotated Classic The Insulator Activity of CTCF Victor G. Corces1,* 1Department of Biology,

A Vertebrate Enhancer-Blocking Protein395

TGCCCAGGCCTGCACTGCCGCCTGCCGGCAGGGGTCCAGTCC supplemented with 0.25% nonfat dry milk, 5 mg/ml poly-dI/dC, and3 pmol of labeled probe in a final volume of 20 ml, washed threeACGAGACCCAGCTCCCTGCTGGCGGAAGGGCGCGCCT. All FII

mutants were identical to FII except for those bases indicated in times for 10 min in the same buffer without probe and exposed tofilm. In the example shown in Figure 4A, samples from an early pilotlowercase in Figure 3C. For use in the enhancer blocking assay,

complementary single-stranded oligonucleotides were purified by purification were loaded as follows: fractions of a large S sepharose,second small S sepharose, and subsequent Q sepharose columndenaturing PAGE, annealed, and subcloned into pNI. The FII site

was also generated with NdeI sites at its ends for cloning upstream were loaded from left to right in each of the panels shown andprobed as indicated in the figure legend.of the enhancer in pNI to generate FII-UP. To generate FII-DOWN,

FII was digested out of pNI-FII, the ends were flushed with Klenow,and XbaI linkers (New England Biolabs) were added for cloning into Protein Purification and Translationthe XbaI site of pNI. Nuclear extracts from K562 cells and whole chicken blood were

prepared essentially as previously described (Evans et al., 1988).For purification of the FII-binding protein, nuclei were prepared fromEnhancer Blocking Assay6 liters of whole chicken blood (Pelfreez Biologicals) and extractedEnhancer blocking assays were performed as previously describedin buffer C: 20 mM HEPES (pH 7.9), 420 mM NaCl, 5 mM MgCl2, 0.2(Chung et al., 1993, 1997). Briefly, 20 mg of each construct wasmM EDTA, and 1 mM DTT. The resulting extract was diluted to 150linearized by SalI digestion, phenol-chloroform extracted, ethanolmM NaCl/20% glycerol in the same buffer and fractionated on aprecipitated, and quantified by UV absorption. Twenty nanograms500 ml SP sepharose column (Pharmacia) using a 0.15–1 M NaClof each DNA was then electroporated into 1 3 107 K562 cells and,linear gradient. Active fractions were pooled, diluted to z150 mMafter 24 hr of recovery, cells were plated in soft agar with geneticinNaCl, and fractionated on a 25 ml CM sepharose column with aat 750 mg/ml. Colonies were counted after 3 weeks of selection and0.15–1 M NaCl gradient. These fractions were pooled and loadedthe colony number was normalized to that obtained with pNI oronto a 2.6/60 cm Sephacryl S-300 gel filtration column (Pharmacia)pJC3-4 (Chung et al., 1993).preequilibrated with 20 mM HEPES (pH 7.9), 150 mM NaCl, 5 mMMgCl2, 0.2 mM EDTA, and 1 mM DTT. Active fractions were pooled,DNase I–Hypersensitive Site Analysisdialyzed into 10 mM potassium phosphate (pH 8.0), 150 mM NaCl,Nuclei were isolated from adult chicken red blood cells essentially5 mM MgCl2, 1 mM DTT, 20% glycerol, and loaded onto a 25 mlas described previously (Bresnick and Felsenfeld, 1994) except thatMacro-Prep ceramic hydroxyapatite column (Bio-Rad). This column0.2 mM EGTA was included in all buffers. After incubation of thewas eluted with a 10–800 mM phosphate gradient at pH 8.0.nuclei with varying concentrations of DNase I for 5 min at roomThroughout the isolation all buffers were supplemented with 1 mMtemperature, the reaction was terminated by the addition of SDSPMSF, 0.7 mg/ml pepstatin, and 0.5 mg/ml leupeptin and maintainedand genomic DNA was purified. To map precisely the position ofat 48C. Fractions pooled from the gel filtration, and all subsequentHS4, DNase I digested and undigested genomic DNAs (10 mg) werebuffers, were supplemented with 40 mg/ml bestatin and 200 mg/further digested with StyI to generate an z1 kb parent fragmentml AEBSF. For peptide sequencing, 1 ml of a final active fractionthat spanned the insulator core. This DNA was then digested with(representing z1/10th of our final yield and z5 mg of purified z140the enzymes indicated in Figure 1A. Digested DNA was resolved onkDa protein) was TCA precipitated, resolved on a 7% Tris-acetatea 1.3% agarose gel and subjected to Southern blotting by standardSDS-PAGE (Novex), transferred to PVDF, stained with amido black,techniques using a 503 bp StyI-SacI fragment of p501 (Reitman andand internal protein sequence was obtained at the Rockefeller Uni-Felsenfeld, 1990) as a probe.versity Protein/DNA Technology Center by in situ digestion withendoproteinase Lys-C followed by HPLC purification of individual

DNA Binding Assayspeptides and Edman sequence determination of each peptide. In

All DNA binding assays were carried out in a binding buffer com-vitro–translated human CTCF was obtained using the plasmid

posed of 20 mM HEPES (pH 7.9), 150 mM KCl, 5 mM MgCl2, 1 mMp4B7.1 (obtained from V. Lobanenkov) as a template for in vitro

DTT. For gel mobility shift assays, DNA binding was carried out attranscription by T7 polymerase according to the manufacturer’s

room temperature for 30 min in binding buffer plus 5% glycerol,instructions (Ambion, “Message Machine”), followed by in vitro

20–40 fmol of end-labeled probe, poly-dI/dC at 50–100 mg/ml, 0.5%translation of the resulting RNA in a nuclease-treated rabbit reticulo-

Triton X-100, and 1–5 ml of protein in 20 ml. Probes were oligonucleo-cyte system (Promega).

tide duplexes identical to those used for sub-cloning into the en-hancer-blocking vector. The RO and BEAD sites, trimmed down for

Acknowledgmentsgel shifts, were RO542–90, 59-GGCAGGGGACCCGATTCGGGGTCGGGGCCCCCGGGGGTGCCCGCTAAGGGGCCCCGGGGGGCCCTC

We thank V. Lobanenkov for providing CTCF expression constructsCCGGCGAAGAGGGGCCCATTGGGA and BEADA-70, 59-TGCATTGand C. Robinett for providing the RO constructs as well as helpfulGCTGCCCAGGCCTGCACTGCCGCCTGCCGGCAGGGGTCCAGTCinformation regarding the repeat sequences. We are grateful toCACGAGACCCAGCTCCCTGC. Cold competitor duplexes weremembers of the laboratory for useful discussions and support.added simultaneously with labeled probes at 50-fold (Figures 3C,

5, and 7A) or 100-fold (Figure 3B) molar excess. Supershifts wereReceived May 17, 1999; revised June 23, 1999.carried out by preincubating the appropriate proteins in binding

buffer for 2 hr at 08C with an antibody raised against a C-terminalpeptide (APNGDLTPEMILSMMD) of CTCF or against Sp1 (Santa ReferencesCruz Biotechnology), followed by a 30 min 238C incubation withDNA. In the RO gel shifts in Figure 7A, 100 mg/ml poly-dG/dC (Roche Anderson, G.M., and Freytag, S.O. (1991). Synergistic activation ofMolecular Biochemicals) was substituted for poly-dI/dC. For South- a human promoter in vivo by transcription factor Sp1. Mol. Cell.western assays, proteins were TCA precipitated, resolved by SDS- Biol. 11, 1935–1943.PAGE, transferred to PVDF, and then denatured and renatured by Baniahmad, A., Steiner, C., Kohne, A.C., and Renkawitz, R. (1990).successive 10 min incubations in binding buffer supplemented with Modular structure of a chicken lysozyme silencer: involvement ofguanidine hydrochloride at 4.8, 3, 1.5, and 0.75 molar. After a further an unusual thyroid hormone receptor binding site. Cell 61, 505–514.10 min wash in binding buffer, the blots were blocked in binding

Bell, A.C., and Felsenfeld, G. (1999). Stopped at the border: bound-buffer plus 5% nonfat dry milk for 16 hr at 48C. An FII probe wasaries and insulators. Curr. Opin. Genet. Dev. 9, 191–198.generated for Southwestern assays by annealing a full-length topBresnick, E.H., and Felsenfeld, G. (1994). Dual promoter activationstrand (CCCAGGGATGTAATTACGTCCCTCCCCCGCTAGGGGGCAby the human beta-globin locus control region. Proc. Natl. Acad.GCAGGCGCGCCT) to a short 39 complementary primer (AGGCGCGSci. USA 91, 1314–1317.CCTGCTGC). This partial duplex was extended with Klenow in the

presence of [a-32P]dCTP resulting in a probe identical to that used Bulger, M., von Doorninck, J.H., Saitoh, N., Telling, A., Farrell, C.,Bender, M.A., Felsenfeld, G., Axel, R., and Groudine, M. (1999).in gel-shift assays, but with 10 labeled phosphates per molecule.

Blots were probed for 3 hr at room temperature in binding buffer Conservation of sequence and structure flanking the mouse and

Page 11: The Insulator Activity of CTCF - Cell · Cell 158, August 14, 2014, 2014 ©2014 Elsevier Inc. Annotated Classic The Insulator Activity of CTCF Victor G. Corces1,* 1Department of Biology,

Cell396

human beta-globin loci: the beta-globin genes are embedded within differentially expressed in multiple forms. Mol. Cell. Biol. 13, 7612–an array of odorant receptor genes. Proc. Natl. Acad. Sci. USA 96, 7624.5129–5134. Klenova, E.M., Nicolas, R.H., U, S., Carne, A.F., Lee, R.E., Lobanen-Burcin, M., Arnold, R., Lutz, M., Kaiser, B., Runge, D., Lottspeich, kov, V.V., and Goodwin, G.H. (1997). Molecular weight abnormalitiesF., Filippova, G.N., Lobanenkov, V.V., and Renkawitz, R. (1997). Neg- of the CTCF transcription factor: CTCF migrates aberrantly in SDS-ative protein 1, which is required for function of the chicken lysozyme PAGE and the size of the expressed protein is affected by the UTRsgene silencer in conjunction with hormone receptors, is identical and sequences within the coding region of the CTCF gene. Nucleicto the multivalent zinc finger repressor CTCF. Mol. Cell Biol. 17, Acids Res. 25, 466–474.1281–1288. Labhart, P., and Reeder, R.H. (1987). DNA sequences for typicalChung, J.H., Whiteley, M., and Felsenfeld, G. (1993). A 59 element ribosomal gene spacers from Xenopus laevis and Xenopus borealis.of the chicken b-globin domain serves as an insulator in human Nucleic Acids Res. 15, 3623–3624.erythroid cells and protects against position effect in Drosophila.

Lobanenkov, V.V., Nicolas, R.H., Adler, V.V., Paterson, H., Klenova,Cell 74, 505–514.

E.M., Polotskaja, A.V., and Goodwin, G.H. (1990). A novel sequence-Chung, J.H., Bell, A.C., and Felsenfeld, G. (1997). Characterization specific DNA binding protein which interacts with three regularlyof the chicken beta-globin insulator. Proc. Natl. Acad. Sci. USA 94, spaced direct repeats of the CCCTC-motif in the 59-flanking se-575–580. quence of the chicken c-myc gene. Oncogene 5, 1743–1753.Dorsett, D. (1993). Distance-independent inactivation of an enhancer Ohtsuki, S., and Levine, M. (1998). GAGA mediates the enhancerby the suppressor of Hairy-wing DNA-binding protein of Drosophila. blocking activity of the eve promoter in the Drosophila embryo.Genetics 134, 1135–1144. Genes Dev. 12, 3325–3330.Evans, T., Reitman, M., and Felsenfeld, G. (1988). An erythrocyte- Pikaart, M.J., Recillas-Targa, F., and Felsenfeld, G. (1998). Lossspecific DNA-binding factor recognizes a regulatory sequence com- of transcriptional activity of a transgene is accompanied by DNAmon to all chicken globin genes. Proc. Natl. Acad. Sci. USA 85,

methylation and histone deacetylation and is prevented by insula-5976–5980.

tors. Genes Dev. 12, 2852–2862.Filippova, G.N., Fagerlie, S., Klenova, E.M., Myers, C., Dehner, Y.,

Prioleau, M.-N., Nony, P., Simpson, M., and Felsenfeld, G. (1999).Goodwin, G., Neiman, P.E., Collins, S.J., and Lobanenkov, V.V.

An insulator element and condensed region separate the chicken(1996). An exceptionally conserved transcriptional repressor, CTCF,b-globin locus from an independently regulated erythroid-specificemploys different combinations of zinc fingers to bind divergedfolate receptor gene. EMBO J., 18, 4035–4048.promoter sequences of avian and mammalian c-myc oncogenes.Reitman, M., and Felsenfeld, G. (1990). Developmental regulationMol. Cell Biol. 16, 2802–2813.of topoisomerase II sites and DNase I–hypersensitive sites in theGdula, D.A., and Corces, V.G. (1997). Characterization of functionalchicken beta-globin locus. Mol. Cell. Biol. 10, 2774–2786.domains of the su(Hw) protein that mediate the silencing effect ofRobinett, C.C., O’Connor, A., and Dunaway, M. (1997). The repeatmod(mdg4) mutations. Genetics 145, 153–161.organizer, a specialized insulator element within the intergenicGdula, D.A., Gerasimova, T.I., and Corces, V.G. (1996). Genetic andspacer of the Xenopus rRNA genes. Mol. Cell. Biol. 17, 2866–2875.molecular analysis of the gypsy chromatin insulator of Drosophila.Sauer, R.T., Smith, D.L., and Johnson, A.D. (1988). Flexibility of theProc. Natl. Acad. Sci. USA 93, 9378–9383.yeast alpha 2 repressor enables it to occupy the ends of its operator,Georgiev, P., and Kozycina, M. (1996). Interaction between muta-leaving the center free. Genes Dev. 2, 807–816.tions in the suppressor of Hairy wing and modifier of mdg4 genes of

Drosophila melanogaster affecting the phenotype of gypsy-induced Spana, C., Harrison, D.A., and Corces, V.G. (1988). The Drosophilamutations. Genetics 142, 425–436. melanogaster suppressor of Hairy-wing protein binds to specific

sequences of the gypsy retrotransposon. Genes Dev. 2, 1414–1423.Gerasimova, T.I., and Corces, V.G. (1998). Polycomb and trithoraxgroup proteins mediate the function of a chromatin insulator. Cell Stief, A., Winter, D.M., Stratling, W.H., and Sippel, A.E. (1989). A92, 511–521. nuclear DNA attachment element mediates elevated and position-

independent gene activity. Nature 341, 343–345.Gerasimova, T.I., Gdula, D.A., Gerasimov, D.V., Simonova, O., andCorces, V.G. (1995). A Drosophila protein that imparts directionality Udvardy, A. (1999). Dividing the empire: boundary chromatin ele-on a chromatin insulator is an enhancer of position-effect variega- ments delimit the territory of enhancers. EMBO J. 18, 1–8.tion. Cell 82, 587–597. Udvardy, A., Maine, E., and Schedl, P. (1985). The 87A7 chromomere.Geyer, P.K., and Corces, V.G. (1992). DNA position-specific repres- Identification of novel chromatin structures flanking the heat shocksion of transcription by a Drosophila zinc finger protein. Genes Dev. locus that may define the boundaries of higher order domains. J.6, 1865–1873. Mol. Biol. 185, 341–358.Hagstrom, K., Muller, M., and Schedl, P. (1996). Fab27 functions Vostrov, A.A., and Quitschke, W.W. (1997). The zinc finger proteinas a chromatin domain boundary to ensure proper segment specifi- CTCF binds to the APBbeta domain of the amyloid beta-proteincation by the Drosophila bithorax complex. Genes Dev. 10, 3202– precursor promoter. Evidence for a role in transcriptional activation.3215. J. Biol. Chem. 272, 33353–33359.Hebbes, T.R., Clayton, A.L., Thorne, A.W., and Crane-Robinson, Zhao, K., Hart, C.M., and Laemmli, U.K. (1995). Visualization of chro-C. (1994). Core histone hyperacetylation co-maps with generalized mosomal domains with boundary element-associated factor BEAF-DNase I sensitivity in the chicken beta-globin chromosomal domain. 32. Cell 81, 879–889.EMBO J. 13, 1823–1830.

Zhong, X.P., and Krangel, M.S. (1997). An enhancer-blocking ele-Holdridge, C., and Dorsett, D. (1991). Repression of hsp70 heat

ment between alpha and delta gene segments within the humanshock gene transcription by the suppressor of hairy-wing protein

T cell receptor alpha/delta locus. Proc. Natl. Acad. Sci. USA 94,of Drosophila melanogaster. Mol. Cell Biol. 11, 1894–1900.

5219–5224.Kellum, R., and Elgin, S.C. (1998). Chromatin boundaries: punctuat-

Zhou, J., Barolo, S., Szymanski, P., Levine, M. (1996). The Fab–7ing the genome. Curr. Biol. 8, R521–R524.element of the bithorax complex attenuates enhancer2promoter

Kellum, R., and Schedl, P. (1991). A position–effect assay for bound- interactions in the Drosophila embryo. Genes Dev. 10, 3195–3201.aries of higher order chromosomal domains. Cell 64, 941–950.

Kellum, R., and Schedl, P. (1992). A group of scs elements functionas domain boundaries in an enhancer-blocking assay. Mol. Cell.Biol. 12, 2424–2431.

Klenova, E.M., Nicolas, R.H., Paterson, H.F., Carne, A.F., Heath,C.M., Goodwin, G.H., Neiman, P.E., and Lobanenkov, V.V. (1993).CTCF, a conserved nuclear factor required for optimal transcrip-tional activity of the chicken c-myc gene, is an 11-Zn-finger protein