The evolutionarily conserved BTB/POZ domain from the promyelocytic leukemia zinc finger (PLZF) oncoprotein mediates transcriptional repression through the recruitment of corepressor proteins containing histone deacetylases in acute promyelocytic leukemia. We have determined the 2.0 Å crystal structure of the BTB/POZ domain from PLZF (PLZF-BTB/POZ), and have carried out biochemical analysis of PLZF-BTB/POZ harboring site-directed mutations to probe structure-function relationships. The structure reveals a novel α/β homodimeric fold in which dimer interactions occur along two surfaces of the protein subunits. The conservation of BTB/POZ domain residues at the core of the protomers and at the dimer interface implies an analogous fold and dimerization mode for BTB/POZ domains from otherwise functionally unrelated proteins. Unexpectedly, the BTB/POZ domain forms dimer-dimer interactions in the crystals, suggesting a mode for higher-order protein oligomerization for BTB/POZ-mediated transcriptional repression. Biochemical characterization of PLZF-BTB/POZ harboring mutations in conserved residues involved in protein dimerization reveals that the integrity of the dimer interface is exquisitely sensitive to mutation and that dimer formation is required for wild-type levels of transcriptional repression. Interestingly, similar mutational analysis of residues within a pronounced protein cleft along the dimer interface, which had been implicated previously for interaction with corepressors, has negligible effects on dimerization or transcriptional repression. Together, these studies form a structure-function framework for understanding BTB/POZ-mediated oligomerization and transcriptional repression properties.

The PLZF4 gene was first identified as part of a t(11;17) chromosomal translocation with the RARα gene forming the PLZF-RARα fusion protein in APL (1). Unlike the more common APL t(15;17) translocation forming the PML-RARα fusion protein (2, 3), patients harboring the t(11;17) chromosomal translocation are resistant to treatment with pharmacological doses of RAs (4).

The PLZF moiety of the PLZF-RARα fusion protein contains a 120 residue BTB/POZ domain, named for its presence in the Drosophila proteins BTB (5), and its homology with several POZ (6). This domain has been found in an increasing number of proteins in poxvirus, Caenorhabditis elegans, Drosophila, and humans and is generally found at the NH2 terminus of either actin-binding or, more commonly, nuclear transcriptional regulatory proteins (7). Functional studies have shown that the BTB/POZ domain mediates homodimerization (6, 8, 9), heteromultimerization between different BTB/POZ-harboring proteins (6, 10), and transcriptional repression in the case of several DNA regulators harboring BTB/POZ domains (10, 11, 12, 13, 14, 15, 16).

Much of the mechanistic detail for the function of BTB/POZ domains has come from the study of the PLZF and LAZ3/BCL6 (lymphoma-associated zinc finger 3/B cell lymphoma 6) oncoproteins (10, 11). In these cases, the BTB/POZ domain has been shown to promote transcriptional repression through the recruitment of corepressor proteins such as N-CoR and SMRT (17, 18). More recently, the BTB/POZ domain of PLZF has been shown to interact with a protein complex containing N-CoR/SMRT, mSin3A, and the histone deacetylase, HDAC1, to mediate transcriptional repression (19, 20, 21). Moreover, this recruitment has been found to play a major role in the pathogenic effect of the PLZF-RARα fusion protein and for its resistance to treatment with RA. Specifically, a model has been proposed whereby the PLZF-RARα fusion protein acts as a potent transcriptional repressor through the ability of both the RARα (22) and BTB/POZ moieties to recruit the SMRT/N-CoR deacetylase transcriptional repression complex (23, 24). Because RA induces the release of this corepressor complex from RARα (22) but not from PLZF, this model is consistent with the RA resistance of APL patients harboring the PLZF-RARα translocation (25, 26).

Here we present the high resolution crystal structure of the BTB/POZ domain from PLZF and characterize the biochemical properties of PLZF proteins harboring site-directed mutations. The structure provides general insights into the architecture and mode of multimerization for the evolutionarily conserved BTB/POZ domain. Moreover, a correlation of the BTB/POZ domain structure with the dimerization and transcriptional repression properties of PLZF proteins harboring site-directed mutations establishes a structure-function paradigm for understanding the dimerization and transcriptional repression properties of proteins harboring BTB/POZ domains. Finally, the insights provided here provide a framework from which to design PLZF-specific inhibitory molecules that may be used to treat APL patients harboring the PLZF-RARα translocation.

Protein Expression and Purification for Crystallization.

Residues 6–123 of PLZF harboring the BTB/POZ domain and containing the NH2-terminal 6xHis tag sequence MRGSHHHHHHGS (herein called PLZF-BTB/POZ) was overexpressed using a pQE30 T5-polymerase based expression vector in Escherichia coli S9 cells and purified using a combination of anion-exchange (Q-Sepharose) and gel filtration (Superdex-75) chromatography as described elsewhere (8).

SeMet-derivatized PLZF-BTB/POZ protein was prepared by growing pQE30/PLZF-BTB/POZ-transformed E. coli strain B834 (DE3; Novagen) in 4-morpholinepropanesulfonic acid-based minimal media (27) supplemented with 50 mg/l l-SeMet and other amino acids at the suggested concentrations. Cells were grown at 28°C to an A595 of 0.4 and induced with 1 mm isoprpyl-1-thio-β-d-galactopyranoside to an A595 of ∼1.0. The PLZF-BTB/POZ protein was isolated essentially as described for the underivatized protein. Quantitative amino acid analysis of SeMet-derivatized PLZF-BTB/POZ protein confirmed that >90% of the methionine residues had been replaced. After purification, SeMet-derivatized PLZF-BTB/POZ was concentrated to ∼50 mg/ml by centrifugation using a Centricon-10 microconcentrator (Amicon) in a buffer containing 40 mm Tris (pH 8.5), 100 mm NaCl, and 1 mm β-mercaptoethanol and frozen as 50-μl aliquots at −70°C before crystallization. Frozen protein aliquots were thawed for use in crystallization as needed.

Crystallization and Data Collection.

Crystals of underivatized and SeMet-derivatized PLZF-BTB/POZ were prepared using 2-μl hanging drops containing 10 mg/ml PLZF-BTB/POZ, 8% isopropanol, 600 mm MgCl2, 50 mm Tris (pH 8.5), and 50 mm HEPES (pH 6.5) equilibrated over a reservoir containing two times the concentration of salts, buffer, and precipitating agent. Crystals were transiently transferred (for ∼5 min) to a harvest solution composed of salts, buffer, and precipitating agent at the same concentrations as the reservoir solution with the addition of 25% glycerol to facilitate X-ray data collection at cryogenic temperature (−170°C).

MAD data were collected from cryoprotected SeMet-derivatized PLZF-BTB/POZ crystals that were flash frozen in liquid propane and stored in a Dewar-containing liquid nitrogen prior to data collection at 110 K. MAD data were collected at NSLS using beamline X4A equipped with an R-AXIS IV image plate detector. The inverse-beam method was used to record Bijvoet differences from each of four different wavelengths to optimize dispersive differences: upstream remote (λ1 = 0.9878 Å), the downstream remote (λ4 = 0.9667 Å), the inflection (λ2 = 0.9796 Å), and the maximum of X-ray absorption (λ3 = 0.9795 Å). The MAD data were processed with DENZO and SCALEPACK (Ref. 28; Table 1).

Structure Determination and Refinement.

The structure was determined to 2.3 Å resolution using the MAD data collected from SeMet-derivatized PLZF-BTB/POZ crystals. SeMet positions were identified and refined using difference Patterson and difference Fourier synthesis with the PHASES package (Ref. 29; Table 1). Two SeMet sites were unambiguously identified using difference Patterson maps produced with two sets of dispersive signals and three sets of Bijvoet signals, and two additional SeMet sites were confirmed using cross-difference Fourier maps using the first two sites. Phase refinement, using four SeMet sites using all Bijvoet and dispersive differences, and solvent flattening procedures were carried out with the program PHASES (29). The resulting electron density map was of very high quality (Fig. 1 C) and allowed for straightforward tracing of the polypeptide chain with the program O (30) using the SeMet positions as landmarks.

Model refinement was carried out with the program X-PLOR (31) using data collected from SeMet-derivatized PLZF-BTB/POZ crystals at the upstream remote wavelength (0.9878 Å). Before refinement, a randomly selected set of data (10%) was omitted from the refinement and used as a “free data set” to monitor subsequent calculations (32), and the model was refined against the remaining data (90%, working data set). Conventional position refinement was initially carried out at 3.0 Å with X-PLOR. After this procedure, iterative cycles of positional refinement with X-PLOR (33) and manual model building with O (30) using Sigmaa-weighted 2Fo-Fc and Fo-Fc difference maps were extended in steps to resolution limits of 2.7, 2.5, 2.3, and 2.0 Å. The later stages of refinement using both simulated annealing (34) and torsion angle dynamics (35) as implemented in X-PLOR. Also at the later stages of refinement, a bulk solvent correction was applied (36), tightly constrained atomic B-factors were adjusted, and water molecules were built into regions that showed strong Fo-Fc peaks and made stereochemically feasible hydrogen bonds. The correctness of the model was checked against simulated annealing omit maps (37) over the entire structure by omitting 15 residues at a time, and the model was adjusted appropriately. A last round of refinement resulted in a model with good geometry (RMSbond length = 0.005 Å, RMSbond angle = 0.862°) and a working R factor of 25.2% with a free R factor of 27.3% using all reflections between 20 and 2 Å (Table 1). The final model includes residues 7–122 of PLZF-BTB/POZ and 61 water molecules. Residue 6, the NH2-terminal 6xHis-tag and the COOH-terminal residue are not visible in the electron density map. A Ramachandran plot showed no residues of the protein in disallowed regions (38).

Site-directed Mutagenesis.

The plasmids containing the BTB/POZ domain of PLZF (residues 6–123) were constructed by PCR using the plasmid pQE30-PLZF as a template (8). A 5′ oligonucleotide (5′-GGA TCC ACC ATG GGC ATG ATC CAG CTG CAG-3′) with a BamHI site immediately 5′ to a consensus Kozak sequence (ACC) at methionine 6 and a 3′ oligonucleotide (5′-GAT GGA TCC CTA CTC CAG CAT CTT CAG GCA CTG-3′) with a stop codon (TAG) and BamHI site after amino acid 123 were used to amplify the desired sequence. Single amino acid point mutations within the BTB/POZ domain of PLZF6–123 were created using standard PCR-mediated mutagenesis. The mutagenic primers contained the following codons: L21A, CTG to GCG; D35N, GAT to AAT; H64A, CAC to GCC; N66A, AAT to GCT; and Q68A, CAA to GCA. BamHI-digested PCR products were ligated into BamHI-digested pSP73 for in vitro translation and pM2 for in vivo expression. All PCR-derived plasmids were subjected to automated DNA sequencing of both strands to confirm the incorporation of appropriate mutations and integrity of surrounding sequences.

Gel Filtration Analysis of Wild-Type and Mutant PLZF Proteins.

Fifty μl of [35S]methionine-labeled, in vitro-translated PLZF6–123 proteins (SP6 TnT; Promega) were analyzed by gel filtration with a Superdex 200 HR 10/30 column (Pharmacia Biotech, Inc.) equilibrated in PBS (10 mm Na2HPO4, 1.4 mm KH2PO4, 137 mm NaCl, and 2.7 mm KCl, pH 7.0). The column was run at 4°C at a flow rate of 0.3 ml/min, and 1-ml fractions were collected. The protein from each fraction was concentrated by deoxycholate-trichloroacetic acid precipitation (39). The precipitated protein was resuspended in 100 μl of 0.1 m NaOH. Thirty μl of the resuspended protein sample were resolved on a 12% Laemmli SDS-PAGE gel, and PLZF6–123 proteins were visualized by fluorography.

Transient Transfection/Luciferase Assays.

Stable expression of heterologous GAL4-PLZF6–123 fusion proteins was confirmed by transfection in COS-1 cells. One μg of a rabbit anti-GAL4 DBD polyclonal IgG (Santa Cruz Biotech) was used to detect expression of GAL4 fusion proteins by immunoprecipitation of [35S]methionine-labeled cell extracts (40). All transcription/luciferase assays were done in NIH/3T3 cells as described elsewhere (40).

The Overall Structure of the PLZF-BTB/POZ Dimer Is Unique.

The BTB/POZ domain of PLZF (PLZF-BTB/POZ) crystallizes as adimer in space group I222 with one protomer per asymmetric unit with a crystallographic 2-fold passing through the functional dimer. The final model contains residues 7–122 of PLZF, which includes the complete evolutionarily conserved BTB/POZ domain (Fig. 1, A and B). Each subunit is comprised of 5 β-strands and 6 α-helices that associate to form a globular dimer (Figs. 1,D and 2). The dimer has approximate dimensions of 20 Å × 30 Å × 60Å and has the appearance of a “butterfly,” with each subunit forming most of each of the wings (Fig. 2, A and B). The β strands lie on the top and bottom of each of the subunits, and the helices fill the interior and flank the sides of the dimer. There are two pronounced clefts at the top and bottom of the dimer, where the two subunits intersect (Fig. 2 C). The smaller upper cleft has a relatively shallow groove, whereas the larger lower cleft is 20 Å long with a width and depth of 5 and 6 Å, respectively. A search through the structural database with either the monomer or dimer shows no significant homology with other protein structures, suggesting that the PLZF-BTB/POZ domain contains a novel protein fold.

The Core of the PLZF-BTB/POZ Domain Is Conserved within the BTB/POZ Domain Family.

Most of the secondary structural elements of the PLZF-BTB/POZ subunit contribute to stabilization of the hydrophobic core (Fig. 3,A). The only exceptions are strands β1 and β5 and the NH2 terminus of helix α1, which contribute largely to dimerization (Fig. 3, B and C). Val36, Val37, Ile38, Met39, and Val40 from β2; Phe45 and Ala47 from β3; and Tyr70 and Leu72 from β4 form the floor of the hydrophobic core (Fig. 1,C). The sides of the hydrophobic core are stabilized by a large number of residues in the helical segments. In particular, residues from helix α2 (Val52, Leu53, and Ala54), from helix α3 (Phe59, Leu62, and Phe63), from helix α4 (Pro77, Thr79, Phe80, Gln82, Ile83, Leu84, Tyr86, Ala87, and Tyr88), and from helix α5 (Leu99, Leu102, Leu103, Ala105, Ala106, and Leu109) make substantial contributions to the core (Figs. 1,A and 3).

The majority of residues that stabilize the core of the PLZF-BTB/POZ domain are highly conserved within the family of BTB/POZ proteins (Figs. 1,B and 3). This conservation appears to extend beyond the subclass of BTB/POZ domain proteins that function as transcriptional regulators (7). In particular, His48 in the loop between β3 and α2, Leu52 from α2, Phe59 from α3, Tyr88 from α4, and Leu109 from α5 are conserved in virtually all BTB/POZ domains. Other residues that stabilize the core are also highly conserved among BTB/POZ proteins (Fig. 1 B). Taken together, the conservation of residues that stabilize the core of the PLZF-BTB/POZ subunit strongly suggests that other BTB/POZ domains have a homologous subunit tertiary fold.

Dimerization by PLZF-BTB/POZ Implicates a Conserved Mode of Dimerization by Other BTB/POZ Domain Proteins.

Previous studies in our laboratory have used a variety of biophysical techniques to show that the BTB/POZ domain of PLZF forms a dimer with an apparent Kd < 200 nm(8). The structure of the PLZF-BTB/POZ dimer is consistent with the high degree of dimer stability. Overall, there are 23 residues from each subunit that contribute to dimer formation, forming a solvent excluded surface of 2400 Å2 for the dimer (Figs. 1,B and 2 C).

The principle dimer contacts between the PLZF-BTB/POZ subunits are mediated by the β1 and β5 strands and the α1 helix, which are the only secondary structural elements that do not contribute significantly to the subunit core (Fig. 3, B and C). The β1 strand and the NH2 terminus of the α1 helix intrude into the opposing subunit by flanking the core above (β1 and β5 strands) and in the central portion of the butterfly (α1 helix). For simplicity of discussion, we will refer to the symmetry-related subunit of the dimer with a primed (′) designation. The β1 strand is wedged between the β5′ strand and the α6′ helix of the opposing subunit, making sheet interactions with the β5′ strand and van der Waals and hydrogen-bond interactions with other regions of the primed subunit (Fig. 4 B). Specifically, Ile9 makes van der Waals interactions with Leu99 of α5′, Ala96′ in the loop between β5′ and α6′ and Met121 of α6′; Ile11 makes van der Waals interactions with Tyr113, Leu114, Glu117, and Cys118 of helix α6′; and the Nδ2 of Asn13 hydrogen bonds to the side-chain hydroxyl of Tyr86 in helix α4′.

The α1 helix and the proceeding loop is situated between the α1′ and α2′ helices and makes mostly van der Waals interactions with residues within these helices as well as with the α3′ and α4′ helices (Fig. 3 C). Specifically, α1-helix residues His16, Pro17, Leu20, Leu21, and Met27 make a series of van der Waals interactions with residues from helices α1′, α2′, α3′, and α4′. In particular, Leu21 from α1′, Cys54 from α2′, and Ala90 from the loop between α4′ and β5′ play important roles in stabilizing the dimer. Thr32, Leu33, and Asp35 in the loop proceeding helix α1 also contribute to dimer stability. His64 from α3′ plays a particularly important role in this regard.

Nearly 80% of the residues that stabilize the PLZF-BTB/POZ dimer show conservation within the BTB/POZ domain family (Fig. 1 A). In particular, His16 and Leu21 in helix α1 and Asp35 in the loop proceeding the α1 helix are highly conserved and play important roles in dimer stability. All but the aspartic acid play a hydrophobic role in dimer stability. Asp35, in contrast, makes a direct hydrogen bond to Arg49 (moderately conserved within the BTB/POZ family) and a water-mediated hydrogen bond to Asp35′. Taken together, the degree of conservation within residues that stabilize dimer formation suggests that BTB/POZ domains from otherwise unrelated proteins will form dimers with similar quaternary arrangements. Correlating well with our findings, there are several BTB/POZ domains that have been shown to form homodimers. Among them are PLZF (8), ZID (6), Ttk (6), bab (9), and BAZF (11). Indeed, the BTB/POZ domain appears to be an ideally suited dimerization module.

Dimer-Dimer Interactions in the Crystals Implicates a Propensity for Higher Order Oligomerization by BTB/POZ Domains in Vivo.

Comparison of the PLZF-BTB/POZ structure derived here with that of the recently published PLZF-BTB/POZ structure determined by Ahmad et al.(42) shows a high degree of structural similarity between the protein dimers with an RMS deviation between all atoms of 1.1 Å2. Strikingly, this comparison also shows that although the two structures were obtained in different crystal lattice environments, both show structurally homologous dimer-dimer interactions in the crystal lattice (Fig. 4,A). These dimer-dimer contacts bury a total of 1200 Å2 of solvent excluded surface and is largely mediated by 2 4-stranded antiparallel β-sheet involving β1 and β5′ from one dimer with the corresponding segments of the symmetry related dimer. In addition, Ala96 in the loop between β5 and α5 makes a van der Waals contact with Met121 at the COOH-terminus of α6 in the symmetry related dimer. The α6-mediated interactions at the dimer-dimer interface are somewhat more extensive in the PLZF-BTB/POZ structure described by Ahmad et al.(42) because their α6 helix contains an additional turn of secondary structure. It is interesting to note that the mode of dimer-dimer interaction observed in the crystals does not prohibit the formation of extended dimer-dimer interactions, which would result in the formation of even higher order multimers (Fig. 4, B and C). Taken together, these observations suggest that the BTB/POZ domain may mediate the formation of higher-order multimers in vivo.

Correlating well with our findings, two recent studies have shown that the BTB/POZ domain of the GAGA transcription factor directly mediates the formation of higher-order oligomers to bind multiple GAGA sites that are found in natural target promoters in vivo(41, 43). Moreover, the formation of these higher-order oligomers have been shown to be correlated with the cooperative nature of GAGA transcription factor binding to multiple DNA-binding sites and correlated with the finding that this cooperativity is strictly dependent on the presence of the GAGA BTB/POZ domain. Interestingly, natural GAGA promoters display a large degree of variability between GAGA sites, also correlating well with the relatively flexible dimer-dimer interactions seen in the crystals.

Our findings of higher-order BTB/POZ oligomers may also explain other studies that find that the BTB/POZ domains from some proteins mediate specific hetero-oligomers. For example, Ttk can form oligomers with itself and with the GAGA protein but not with the BTB/POZ region of ZID (6). In addition, BAZF has been shown to form oligomers with itself and with BCL6 (11). Taken together, the BTB/POZ domain appears to be an ideally suited module for both homo and hetero protein multerimerization.

Biochemical Analysis of PLZF Proteins Harboring Site-directed Mutations Shows That Dimerization Is Required for Transcriptional Repression.

The recent study by Ahmad et al.(42) has suggested that a pronounced cleft along the bottom of the PLZF dimer (Fig. 2,C) may be a site of corepressor interaction. To directly test this hypothesis and to directly probe the functional significance of dimer formation by PLZF, we carried out site-directed mutagenesis coupled with biochemical analysis of these mutant proteins. We prepared five different site-directed mutations that fell into two subgroups (Fig. 2,C). The first two (L21A and D35N) involved residues that play critical roles in dimerization; and the second group (H64A, N66A, and Q68A) involved residues in the pronounced cleft at the base of the PLZF-BTB/POZ domain dimer. Each of the mutants were tested for both dimerization and transcriptional repression properties. To evaluate the effects of these mutations on the dimerization of the BTB/POZ domain, the wild-type PLZF6–123 and each mutant described herein were in vitro translated and then subjected to gel filtration (Fig. 5). Consistent with expectations, the wild-type PLZF6–123 protein, as well as each of the proteins harboring mutations in the cleft region, eluted from the sizing column at a molecular weight consistent with a dimeric PLZF-BTB/POZ species. In addition, proteins harboring mutations in the dimerization interface of PLZF-BTB/POZ eluted in two peaks, one near the void volume (Mr ≤ 670,000), indicative of protein aggregates and another at a position consistent with a monomeric PLZF-BTB/POZ domain (Mr 17,000). These results indicated that the L21A and D35N mutant proteins were defective in dimer formation.

To evaluate any effects these mutations may have on the transcriptional repression function of the PLZF BTB/POZ domain, heterologous fusions between the DNA binding domain of GAL4 (amino acids 1–147) and each of the BTB/POZ domain mutants described herein were created (Fig. 6). Each fusion protein demonstrated stable and comparable nuclear-localized expression in COS-1 cells (Fig. 6,B), suggesting comparable DNA-binding properties for each of our protein fusions. The wild-type GAL4-PLZF6–123 BTB/POZ domain and each mutant version thereof were cotransfected with 2 TK-luciferase reporter downstream of four copies of the GAL4 consensus UAS in NIH/3T3 cells (Fig. 6,A). The wild-type GAL4-PLZF6–123 protein demonstrated significant repression of the luciferase reporter gene activity (Fig. 6,C). Expression of the of the D35N mutant protein, which fails to dimerize properly (Fig. 5,B), demonstrated a significantly impaired repression function (Fig. 6,C). To a lesser extent, the repression function of the L21A mutant protein was also impaired (Fig. 6,C). All other mutants (H64A, N66A, and Q68A) demonstrated negligible effects on the repression function of the BTB/POZ domain of PLZF (Fig. 6 C). Significantly, these data suggest that the repression function of the PLZF-BTB/POZ domain is strictly dependent upon homodimerization. Moreover, these data suggest that the cleft region of the PLZF-BTB/POZ domain may not play a significant role in the transcriptonal repression property of PLZF.

Recently, Huynh and Bardwell (10) have carried out a site-directed mutagenesis study of the BTB/POZ domain of BCL6 with the goal of disrupting corepressor function. In this study, the authors targeted NH2-terminal residues that are highly conserved in proteins that showed interaction with the corepressor proteins N-CoR and SMRT and that mediated transcriptional repression (BCL6, PLZF, ZID, GAGA, and vaccinia virus SalF17R). Mutations were made to the corresponding residues of the BTB/POZ harboring protein, Protein A from the mod(mdg4) gene, which failed to show binding to N-CoR and SMRT and which failed to mediate transcriptional repression. A correlation of the phenotype of these mutations with the PLZF-BTB/POZ structure shows that the two mutations that disrupt corepressor binding and function (Leu to Ser at position 21, and Met-Arg to Ser-Leu at positions 27 and 28 using PLZF numbering) would also disrupt dimerization of the PLZF-BTB/POZ region (Figs. 2,C and 3,C). An asparagine to histidine change at position 25 (using PLZF numbering) did not have a detectable effect on N-CoR and SMRT interaction or transcriptional repression. The structure of the PLZF-BTB/POZ domain shows that Asp25 is packed on the interior of the protein, thus playing an important role in protein folding (Fig. 3 A). Modeling of a histidine in position 25 shows that it could carry out a comparable role in BTB/POZ domain stability. Taken together, our results and the results of Huynh and Bardwell (10) underscore the importance of BTB/POZ domain dimerization for N-CoR and SMRT-mediated transcriptional repression.

The precise mode of corepressor recognition by PLZF must await the structure of an appropriate PLZF-BTB/POZ/corepressor complex. Nonetheless, the structural and functional information provided here provides a framework from which to use structure/function analysis to better understand the N-CoR and SMRT binding and repression properties of the BTB/POZ domain of PLZF. Moreover, the information provided here provides a conceptual and structural scaffold from which to design PLZF-specific inhibitory molecules that may target the dimerization and/or interaction of PLZF with corepressor proteins to be used to treat APL patients harboring the PLZF-RARα translocation.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

      
1

Supported by a grant from the Leukemia Research Foundation (to R. M.) and NIH Basic Cancer Research Training Grant CA09171 (to D. C. S). F. J. R. was supported in part by NIH Grant CA52009, Core Grant CA10815, Core Grant DK50306, and Grants DK49210, GM54220, DAMD17-96-1-6141, and ACS NP-954; the Irving A. Hansen Memorial Foundation; the Mary A. Rumsey Memorial Foundation; and the Pew Scholars Program in the Biomedical Sciences.

                  
4

The abbreviations used are: PLZF, promyelocytic leukemia zinc finger; RAR, retinoic acid receptor; APL, acute promyelocytic leukemia; RA, retinoic acid; BTB, Broad complex, Trametrack, and Bric a brac; POZ, poxvirus and zinc finger proteins; SMRT, silencing mediator of retinoid and thyroid receptor; SeMet, selenomethionine; MAD, multiwavelength anomalous dispersion.

Fig. 1.

The BTB/POZ domain family and its relationship to PLZF-BTB/POZ. A, schematic bar diagram of PLZF and related proteins. The BTB/POZ domain and zinc fingers (Zn) are indicated for PLZF, BCL-6, and BAZF. B, sequence alignment of BTB/POZ domains. Among the 50+ BTB/POZ domains known to date, a panel of 16 representative sequences that are most homologous with PLZF and that have been shown to function as transcriptional regulators are aligned (CLUSTAL program) and displayed (BOXSHADE program). Black and gray backgrounds are used to indicate identical and/or conserved residues found in at least 50% of the proteins at a given position, respectively. Secondary structural elements within the BTB/POZ domain of PLZF are shown above the sequence alignment. •, residues that are buried within the core of the monomer; ▵, residues that are buried upon dimer formation. C, experimental MAD electron density map after solvent flattening at a resolution of 2.0 Å showing β-strands 2, 3, and 4 that stabilize the hydrophobic core of the PLZF-BTB/POZ domain. The map is contoured at 1.3 sigma. D, topology diagram of the BTB/POZ domain of PLZF. The β5′ strand (from the opposing subunit) is illustrated because its presence induces the β1 strand.

Fig. 1.

The BTB/POZ domain family and its relationship to PLZF-BTB/POZ. A, schematic bar diagram of PLZF and related proteins. The BTB/POZ domain and zinc fingers (Zn) are indicated for PLZF, BCL-6, and BAZF. B, sequence alignment of BTB/POZ domains. Among the 50+ BTB/POZ domains known to date, a panel of 16 representative sequences that are most homologous with PLZF and that have been shown to function as transcriptional regulators are aligned (CLUSTAL program) and displayed (BOXSHADE program). Black and gray backgrounds are used to indicate identical and/or conserved residues found in at least 50% of the proteins at a given position, respectively. Secondary structural elements within the BTB/POZ domain of PLZF are shown above the sequence alignment. •, residues that are buried within the core of the monomer; ▵, residues that are buried upon dimer formation. C, experimental MAD electron density map after solvent flattening at a resolution of 2.0 Å showing β-strands 2, 3, and 4 that stabilize the hydrophobic core of the PLZF-BTB/POZ domain. The map is contoured at 1.3 sigma. D, topology diagram of the BTB/POZ domain of PLZF. The β5′ strand (from the opposing subunit) is illustrated because its presence induces the β1 strand.

Close modal
Fig. 2.

Structure of the PLZF-BTB/POZ dimer. A, a schematic of PLZF-BTB/POZ viewed perpendicular to the 2-fold axis of the dimer. The primary subunit is shown in green, and the primed subunit is shown in blue. B, a schematic of PLZF-BTB/POZ viewed parallel to the 2-fold axis of the dimer. C, a space-filling model of the PLZF-BTB/POZ dimer. The position of single amino acid substitutions that were biochemically characterized in this study (Leu21, Asp35, His64, Asn66, and Gln68) are shown in yellow.

Fig. 2.

Structure of the PLZF-BTB/POZ dimer. A, a schematic of PLZF-BTB/POZ viewed perpendicular to the 2-fold axis of the dimer. The primary subunit is shown in green, and the primed subunit is shown in blue. B, a schematic of PLZF-BTB/POZ viewed parallel to the 2-fold axis of the dimer. C, a space-filling model of the PLZF-BTB/POZ dimer. The position of single amino acid substitutions that were biochemically characterized in this study (Leu21, Asp35, His64, Asn66, and Gln68) are shown in yellow.

Close modal
Fig. 3.

Details of the BTB/POZ domain dimer. A, BTB/POZ conserved residues mapped onto the PLZF-BTB/POZ structure. Highly conserved residues (shaded black in Fig. 1 B) are mapped onto a schematic of the PLZF-BTB/POZ dimer, illustrating that the majority of conserved residues function to stabilize the core of the monomer and the protein dimer. B, β1 strand-mediated interactions that stabilize the dimer. Residues that are associated with dimer stability are indicated as light shaded colored side chains superimposed onto a schematic of the protein dimer. C, α1 loop-mediated interactions that stabilize the dimer.

Fig. 3.

Details of the BTB/POZ domain dimer. A, BTB/POZ conserved residues mapped onto the PLZF-BTB/POZ structure. Highly conserved residues (shaded black in Fig. 1 B) are mapped onto a schematic of the PLZF-BTB/POZ dimer, illustrating that the majority of conserved residues function to stabilize the core of the monomer and the protein dimer. B, β1 strand-mediated interactions that stabilize the dimer. Residues that are associated with dimer stability are indicated as light shaded colored side chains superimposed onto a schematic of the protein dimer. C, α1 loop-mediated interactions that stabilize the dimer.

Close modal
Fig. 4.

Dimer-dimer interactions in the PLZF-BTB/POZ crystals. A, a backbone trace of the protein regions that mediate dimer-dimer interactions in the crystals are shown. The primary PLZF-BTB/POZ dimer is shown in blue, and the symmetry-related dimer is shown in green (brackets, secondary structural elements). Backbone hydrogen bonds within the same dimer are shown in a light shade of the respective color, and hydrogen bonds between dimers are shown in red. Ala96 and Met121, which make van der Waals interactions at the dimer interface, are also indicated as red side chains. A superimposition of the dimer-dimer interaction mediated in the PLZF-BTB/POZ crystals (RMS deviation for all atoms is 1.3 Å2) described by Ahmad et al.(42) is shown in gray. B and C, schematic representations showing the organization of the proposed higher order BTB/POZ domain oligomers.

Fig. 4.

Dimer-dimer interactions in the PLZF-BTB/POZ crystals. A, a backbone trace of the protein regions that mediate dimer-dimer interactions in the crystals are shown. The primary PLZF-BTB/POZ dimer is shown in blue, and the symmetry-related dimer is shown in green (brackets, secondary structural elements). Backbone hydrogen bonds within the same dimer are shown in a light shade of the respective color, and hydrogen bonds between dimers are shown in red. Ala96 and Met121, which make van der Waals interactions at the dimer interface, are also indicated as red side chains. A superimposition of the dimer-dimer interaction mediated in the PLZF-BTB/POZ crystals (RMS deviation for all atoms is 1.3 Å2) described by Ahmad et al.(42) is shown in gray. B and C, schematic representations showing the organization of the proposed higher order BTB/POZ domain oligomers.

Close modal
Fig. 5.

Effects of single amino acid substitutions on the dimerization properties of the PLZF BTB/POZ domain. A, in vitro translation of wild-type and PLZF6–123-BTB/POZ proteins containing single amino acid substitutions (L21A, D35N, H64A, N66A, and Q68A). Molecular weight standards and a negative control in which a translation reaction was done in the absence of RNA polymerase are indicated. B, Superdex 200 HR 10/30 gel filtration fractions of in vitro-translated, 35S-labeled wild-type and mutant PLZF6–123-BTB/POZ proteins were resolved in SDS-PAGE gels and visualized by fluorography. Elution of molecular weight standards was determined in a parallel gel filtration experiment under exactly the same conditions. Fractions with peak levels of molecular weight standards are indicated at the top of the figure.

Fig. 5.

Effects of single amino acid substitutions on the dimerization properties of the PLZF BTB/POZ domain. A, in vitro translation of wild-type and PLZF6–123-BTB/POZ proteins containing single amino acid substitutions (L21A, D35N, H64A, N66A, and Q68A). Molecular weight standards and a negative control in which a translation reaction was done in the absence of RNA polymerase are indicated. B, Superdex 200 HR 10/30 gel filtration fractions of in vitro-translated, 35S-labeled wild-type and mutant PLZF6–123-BTB/POZ proteins were resolved in SDS-PAGE gels and visualized by fluorography. Elution of molecular weight standards was determined in a parallel gel filtration experiment under exactly the same conditions. Fractions with peak levels of molecular weight standards are indicated at the top of the figure.

Close modal
Fig. 6.

Effects of single amino acid substitutions on the transcriptional repression properties of the PLZF BTB/POZ domain. A, schematic diagram illustrating the plasmids used in transfection of NIH/3T3 cells. GAL4 fusion proteins of the wild-type and PLZF6–123 proteins containing single amino acid substitutions are shown. B, similar expression levels of GAL4-PLZF6–123 fusion proteins was confirmed by transient transfection in COS-1 cells, followed by immunoprecipitation of [35S]methionine-labeled cell extracts with anti-GAL4 DBD polyclonal IgG. Closed arrowhead, GAL4 protein; open arrowhead, GAL4-KRAB protein as a positive control. Arrow, wild-type and mutant GAL4- PLZF6–123 fusion proteins. Molecular weight standards are indicated. C, the relative levels of transcriptional repression of the GAL4-PLZF6–123 fusion proteins are indicated in terms of fold-repression. Values were calculated by comparing normalized luciferase activities with cells transfected in the absence of a GAL4 effector protein. Cotransfection with a pcDNA3-β-galactosidase expression plasmid was used to normalize all luciferase values for transfection efficiency. Bars, SEs observed for three independent transfections (each performed in duplicate).

Fig. 6.

Effects of single amino acid substitutions on the transcriptional repression properties of the PLZF BTB/POZ domain. A, schematic diagram illustrating the plasmids used in transfection of NIH/3T3 cells. GAL4 fusion proteins of the wild-type and PLZF6–123 proteins containing single amino acid substitutions are shown. B, similar expression levels of GAL4-PLZF6–123 fusion proteins was confirmed by transient transfection in COS-1 cells, followed by immunoprecipitation of [35S]methionine-labeled cell extracts with anti-GAL4 DBD polyclonal IgG. Closed arrowhead, GAL4 protein; open arrowhead, GAL4-KRAB protein as a positive control. Arrow, wild-type and mutant GAL4- PLZF6–123 fusion proteins. Molecular weight standards are indicated. C, the relative levels of transcriptional repression of the GAL4-PLZF6–123 fusion proteins are indicated in terms of fold-repression. Values were calculated by comparing normalized luciferase activities with cells transfected in the absence of a GAL4 effector protein. Cotransfection with a pcDNA3-β-galactosidase expression plasmid was used to normalize all luciferase values for transfection efficiency. Bars, SEs observed for three independent transfections (each performed in duplicate).

Close modal
Table 1

Crystallographic data

Unit cell parameters      
PLZF-BTB/POZ (residue 7-122; four ordered SeMet)      
Space group I222 a = 38.8 Å  b = 78.2 Å c = 85.2 Å  
Unit cell parameters      
PLZF-BTB/POZ (residue 7-122; four ordered SeMet)      
Space group I222 a = 38.8 Å  b = 78.2 Å c = 85.2 Å  
Data collection (between 20 and 2 Å)
Wavelength (Å)Unique reflectionsTotal reflectionsCompletenessRmergeaAve I/ς
λ10.9879 8,790 115110 96.7% 7.0% 35.5 
λ20.9795 16,465 115154 97.3% 7.2% 27.0 
λ30.9791 16,460 116531 97.3% 7.8% 26.8 
λ40.9667 15,963 109193 94.3% 8.0% 25.5 
Data collection (between 20 and 2 Å)
Wavelength (Å)Unique reflectionsTotal reflectionsCompletenessRmergeaAve I/ς
λ10.9879 8,790 115110 96.7% 7.0% 35.5 
λ20.9795 16,465 115154 97.3% 7.2% 27.0 
λ30.9791 16,460 116531 97.3% 7.8% 26.8 
λ40.9667 15,963 109193 94.3% 8.0% 25.5 
Phasing statisticsb (between 20 and 2 Å)Refinement statisticsc
Difference (w.r.t λ1)Phasing powerdFOMResolution range Number of refl. Rworking/Rfreee (%)Between 20 and 2 Å 7889 (I/ς>2)F 25.2/27.3
CentricAccentric
λ2disp 2.43 0.541 0.368 RMS deviations Bonds 0.005 Å 
λ2anom 2.63 0.335   Angles 0.862° 
λ3disp 1.67 0.445 0.284 Baverage (no. of atoms) Protein 29.5 (931) 
λ3anom 3.35 0.384   Water 35.6 (61) 
λ4anom 2.16 0.292   Mg+2 26.0 (1) 
Combined FOM  0.694     
Phasing statisticsb (between 20 and 2 Å)Refinement statisticsc
Difference (w.r.t λ1)Phasing powerdFOMResolution range Number of refl. Rworking/Rfreee (%)Between 20 and 2 Å 7889 (I/ς>2)F 25.2/27.3
CentricAccentric
λ2disp 2.43 0.541 0.368 RMS deviations Bonds 0.005 Å 
λ2anom 2.63 0.335   Angles 0.862° 
λ3disp 1.67 0.445 0.284 Baverage (no. of atoms) Protein 29.5 (931) 
λ3anom 3.35 0.384   Water 35.6 (61) 
λ4anom 2.16 0.292   Mg+2 26.0 (1) 
Combined FOM  0.694     
a

Rmerge = ∑ | I − < I > |/∑ I

b

PHASES output

c

Refinement is against λ1 data.

d

Phasing power = [∑|FH|2/∑ (FPH.obsFPH.calc)2]1/2.

e

Rfree is calculated for 10% of the data.

f

Around 600 reflections that are close to two outermost ice rings are excluded from refinement.

We thank Craig Ogata and his staff for access to and help on beamline X4A at NSLS; David Speicher (Wistar Institute microchemistry core facility) for mass spectrometry and NH2-terminal sequencing analysis of SeMet-derivatized PLZF-BTB/POZ protein; and Greg Van Duyne, Mitch Lewis, Dan King, Ravi Venkataramani, Yi Mo, Stacey Benson, and Kunchithapadam Swaminathan for useful discussions. Coordinates for the PLZF-BTB/POZ structure can be obtained from the Research Collaboratory for Structural Bioinformatics (RCSB009519).

1
Chen Z., Brand N. J., Chen A., Chen S-J., Tong J-H., Wang Z. Y., Waxman S., Zelent A. Fusion between a novel Kruppel-like zinc finger gene and the retinoic acid receptor-α locus due to a variant t(11;17) translocation associated with acute promyelocytic leukemia.
EMBO J.
,
12
:
1161
-1167,  
1993
.
2
Rowley J. D., Golomb H. M., Dougherty C. 15/17 translocation: a consistent chromosomal change in acute promyelocytic leukaemia [letter].
Lancet
,
1
:
549
-550,  
1977
.
3
Zelent A. Translocation of the RARα locus to the PML of PLZF gene in acute promyelocytic leukemia.
Br. J. Haematol.
,
86
:
451
-460,  
1994
.
4
Licht J. D., Chomienne C., Goy A., Chen A., Scott A. A., Head D. R., Michaux J. L., Wu Y., DeBlasio A., Miller W. H., Jr, Zelenetz A. D., Willman C. L., Chen Z., Chen S-J., Zelent A., Macintyre E., Veil A., Cortes J., Kantarjian H., Waxman S. Clinical and molecular characterization of a rare syndrome of acute promyelocytic leukemia associated with translocation (11;17).
Blood
,
85
:
1083
-1094,  
1995
.
5
Zollman S., Godt D., Prive G. G., Couderc J-L., Laski F. The BTB domain, found primarily in zinc finger proteins, defines an evolutionarily conserved family that includes several developmentally regulated genes in Drosophila.
Proc. Natl. Acad. Sci. USA
,
91
:
10717
-10721,  
1994
.
6
Bardwell V. J., Treisman R. The POZ domain: a conserved protein-protein interaction motif.
Genes Dev.
,
8
:
1664
-1677,  
1994
.
7
Albagli O., Dhordain P., Deweindt C., Lecocq G., Leprince D. The BTB/POZ domain: a new protein-protein interaction motif common to DNA- and actin-binding proteins.
Cell Growth Differ.
,
6
:
1193
-1198,  
1995
.
8
Li X., Lopez-Guisa J. M., Ninan N., Weiner E. J., Rauscher F. J., III, Marmorstein R. Overexpression, purification, characterization, and crystallization of the BTB/POZ domain from the PLZF oncoprotein.
J. Biol. Chem.
,
272
:
27324
-27329,  
1997
.
9
Chen W., Zollman S., Couderc J. L., Laski F. A. The BTB domain of bric a brac mediates dimerization in vitro.
Mol. Cell. Biol.
,
15
:
3424
-3429,  
1995
.
10
Huynh K. D., Bardwell V. J. The BCL-6 POZ domain and other POZ domains interact with the co-repressors N-CoR and SMART.
Oncogene
,
17
:
2473
-2484,  
1998
.
11
Okabe S., Fukuda T., Ishibashi K., Kojima S., Okada S., Hatano M., Ebara M., Saisho H., Tokuhisa T. BAZF, a novel BCL6 homolog, functions as a transcriptional repressor.
Mol. Cell. Biol.
,
18
:
4235
-4244,  
1998
.
12
Numoto M., Niwa O., Kaplan J., Wong K. K., Merrell K., Kamiya K., Yanagihara K., Calame K. Transcriptional repressor ZF5 identifies a new conserved domain in zinc finger proteins.
Nucleic Acids Res.
,
21
:
3767
-3775,  
1993
.
13
Albagli O., Dhordain P., Bernardin F., Quief S., Kerckaert J-P., Leprince D. Multiple domains participate in distance-independent LAZ3/BCL6-mediated transcriptional repression.
Biochem. Biophys. Res. Commun.
,
220
:
911
-915,  
1996
.
14
Chang C-C., Ye B. H., Chaganti R. S. K., Dalla-Favera R. BCL-6, a POZ/zinc-finger protein, is a sequence-specific transcriptional repressor.
Proc. Natl. Acad. Sci. USA
,
93
:
6947
-6952,  
1996
.
15
Seyfert V., Allman D., He Y., Staudt L. M. Transcriptional repression by the proto-oncogene BCL-6.
Oncogene
,
12
:
2331
-2342,  
1996
.
16
Deweindt C., Albagli O., Bernardin F., Dhordain P., Quief S., Lantoine D., Kerckaert J-P., Leprince D. The LAZ3/BCL6 oncogene encodes a sequence-specific transcriptional inhibitor: a novel function for the BTB/POZ domain as an autonomous repressing domain.
Cell Growth Differ.
,
6
:
1495
-1503,  
1995
.
17
Dhordain P., Albagli O., Lin R. J., Ansieau S., Quief S., Leutz A., Kerckaert J-P., Evans R. M., Leprince D. L. Corepressor SMART binds the BTB/POZ repressing domain of the LAZ3/BCL6 oncoprotein.
Proc. Natl. Acad. Sci. USA
,
94
:
10762
-10767,  
1997
.
18
Hong S-H., David G., Wong C-W., Dejean A., Privalsky M. L. SMART corepressor interacts with PLZF and with the PML-retinoic acid receptor α (RARα) and PLZF-RARα oncoproteins associated with acute promyelocytic leukemia.
Proc. Natl. Acad. Sci. USA
,
94
:
9028
-9033,  
1997
.
19
David G., Alland L., Hong S-H., Wong C-W., Depinho R., Dejean A. Histone deacetylase associated with mSin3A mediates repression by the acute promyelocytic leukemia-associated PLZF protein.
Oncogene
,
16
:
2549
-2556,  
1998
.
20
Lin R. J., Nagy L., Inoue S., Shao W. L., Miller W. H., Evans R. M. Role of the histone deacetylase complex in acute promyelocytic leukaemia.
Nature (Lond.)
,
391
:
811
-814,  
1998
.
21
Grignani F., De Matteis S., Nervi C., Tomassoni L., Gelmetti V., Cioce M., Fanelli M., Ruthardt M., Ferrara F. F., Zamir I., Seiser C., Lazar M. A., Minucci S., Pelicci P. G. Fusion proteins of the retinoic acid receptor-α recruit histone deacetylase in promyelocytic leukaemia.
Nature (Lond.)
,
391
:
815
-818,  
1998
.
22
Chen J. D., Evans R. M. A transcriptional co-repressor that interacts with nuclear hormone receptors.
Nature (Lond.)
,
377
:
454
-457,  
1995
.
23
Heinzel T., Lavinsky R. M., Mullen T-M., Soderstrom M., Laherty C. D., Torchia J., Yang W-M., Brard G., Ngo S. D., Davie J. R., Seto E., Eisenman R. N., Rose D. W., Glass C. K., Rosenfeld M. G. A complex containing N-CoR, mSin3 and histone deacetylase mediates transcriptional repression.
Nature (Lond.)
,
387
:
43
-48,  
1997
.
24
Alland L., Muhle R., Hou H., Potes J., Chin L., Schreiber-Agus N. Role for N-CoR and histone deacetylase in Sin3-mediated transcriptional repression.
Nature (Lond.)
,
387
:
49
-55,  
1997
.
25
He L. Z., Guidez F., Tribioli C., Peruzzi D., Ruthardt M., Zelent A., Pandolfi P. P. Distinct interactions of PML-RARα and PLZF-RARα with co-repressors determine differential responses to RA in APL.
Nat. Genet.
,
18
:
126
-135,  
1998
.
26
Guidez F., Huag W., Tong J-H., Dubois C., Balitrand N., Waxman S., Michaux J. L., Martial P., Degos L., Chen Z., Chomienne C. Poor response to all-trans retinoic acid therapy in a t(11;17) PLZF/RARα patient.
Leukemia (Baltimore)
,
8
:
312
-317,  
1994
.
27
Neidhardt F. C., Bloch P. L., Smith D. F. Culture medium for enterobacteria.
J. Bacteriol.
,
119
:
736
-747,  
1974
.
28
Otwinowski Z. Oscillation data reduction program Sawyer L. Isaacs N. Bailey S. eds. .
Proceedings of the CCP4 Study Weekend: Data Collection and Processing
,
:
56
-62, SERC Daresbury Laboratory Warrington, United Kingdom  
1993
.
29
Furey W., Swaminathan S. PHASES-95: a program package for the processing and analysis of diffraction data from macromolecules Carter C. W. Sweet R. M. eds. .
Methods in Enzymology
,
277
:
590
-620, Academic Press Orlando, FL  
1997
.
30
Jones T. A., Zou J. Y., Cowen S. W. Improved methods for building protein models in electron density maps and the location of errors in these models.
Acta Crystallogr. A
,
47
:
110
-119,  
1991
.
31
Brunger A. T. X-PLOR 3.1, a system for X-ray crystallography and NMR Yale University Press Haven, CT  
1992
.
32
Brunger A. T. The free R value: a novel statistical quantity assessing the accuracy of crystal structures.
Nature (Lond.)
,
335
:
472
-474,  
1992
.
33
Brunger A. T., Kuriyan J., Karplus M. Crystallographic R factor refinement by molecular dynamics.
Science (Washington DC)
,
235
:
458
-460,  
1987
.
34
Brunger A. T., Krukowski A. Slow-cooling protocols for crystallographic refinement by simulated annealing.
Acta Crystallogr. A
,
46
:
585
-593,  
1990
.
35
Rice L. M., Brunger A. T. Torsion angle dynamics: reduced variable conformational sampling enhances crystallographic structure refinement.
Proteins
,
19
:
277
-290,  
1994
.
36
Jiang J. S., Brunger A. T. Protein hydration observed by X-ray diffraction: solvation properties of penicillopepsin and neuraminidase crystal structures.
J. Mol. Biol.
,
243
:
100
-115,  
1994
.
37
Hodel A., Kim S-H., Brunger A. T. Model bias in macromolecular crystal structures.
Acta Crystallogr. A
,
48
:
851
-858,  
1992
.
38
Kleywegt G. J., Jones T. A. Phi/Psi-chology: Ramachandran revisted.
Curr. Biol.
,
4
:
1996b
1395
-1400,
39
Peterson G. L. A simplification of the protein assay method of Lowry et al. which is more generally applicable.
Anal. Biochem.
,
83
:
346
-356,  
1977
.
40
Ryan R. F., Schultz D. C., Ayyanathan K., Singh P. B., Friedman J. R., Fredericks W. J., Rauscher F. J. KAP-1 corepressor protein interacts and colocalizes with heterochromatic and euchromatic HP1 proteins: a potential role for Kruppel-associated Box-zinc finger proteins in heterochromatin-mediated gene silencing.
Mol. Cell. Biol.
,
19
:
4366
-4378,  
1999
.
41
Espinas M. L., Jimenez-Garcia E., Vaquero A., Canudas S., Bernues J., Azorin F. The N-terminal POZ domain of GAGA mediates the formation of oligomers that bind DNA with high affinity and specificity.
J. Biol. Chem.
,
274
:
16461
-16469,  
1999
.
42
Ahmad K. F., Engel C. K., Prive G. G. Crystal structure of the BTB domain from PLZF.
Proc. Natl. Acad. Sci. USA
,
95
:
12123
-12128,  
1998
.
43
Katsani K. R., Hajibagheri M. A. N., Verrijzer C. P. Co-operative DNA binding by GAGA transcription factor requires the conserved BTB/POZ domain and reorganizes promoter topology.
EMBO J.
,
18
:
698
-708,  
1999
.