Download as pdf or txt
Download as pdf or txt
You are on page 1of 24

doi:10.1006/jmbi.2001.4780 available online at http://www.idealibrary.com on J. Mol. Biol.

(2001) 310, 635±658

The Dimerization Domain of HNF-1a a: Structure and


Plasticity of an Intertwined Four-helix Bundle with
Application to Diabetes Mellitus
Narendra Narayana, Qing-xin Hua and Michael A. Weiss*

Department of Biochemistry Maturity-onset diabetes mellitus of the young (MODY) is a human gen-
Case Western Reserve etic syndrome most commonly due to mutations in hepatocyte nuclear
University, 10900 Euclid factor-1a (HNF-1a). Here, we describe the crystal structure of the HNF-
Avenue, Cleveland 1a dimerization domain at 1.7 A Ê resolution and assess its structural plas-
OH 44106-4935, USA ticity. The crystal's low solvent content (23 %, v/v) leads to tight packing
of peptides in the lattice. Two independent dimers, similar in structure,
are formed in the unit cell by a 2-fold crystallographic symmetry axis.
The dimers de®ne a novel intertwined four-helix bundle (4HB). Each pro-
tomer contains two a-helices separated by a sharp non-canonical turn.
Dimer-related a-helices form anti-parallel coiled-coils, including an
N-terminal ``mini-zipper'' complementary in structure, symmetry and
surface characteristics to transcriptional coactivator dimerization cofactor
of HNF-1 (DCoH). A con¯uence of ten leucine side-chains (®ve per pro-
tomer) forms a hydrophobic core. Isotope-assisted NMR studies demon-
strate that a similar intertwined dimer exists in solution. Comparison of
structures obtained in multiple independent crystal forms indicates that
the mini-zipper is a stable structural element, whereas the C-terminal
a-helix can adopt a broad range of orientations. Segmental alignment of
the mini-zipper (mean pairwise root-mean-square difference (rmsd) in Ca
coordinates of 0.29 A Ê ) is associated with a 2.1 A
Ê mean Ca rmsd displace-
ment of the C-terminal coiled-coil. The greatest C-terminal structural vari-
ation (4.1 AÊ Ca rmsd displacement) is observed in the DCoH-bound
peptide. Diabetes-associated mutations perturb distinct structural features
of the HNF-1a domain. One mutation (L12H) destabilizes the domain
but preserves structural speci®city. Adjoining H12 side-chains in a
native-like dimer are predicted to alter the functional surface of the mini-
zipper involved in DCoH recognition. The other mutation (G20R), by
contrast, leads to a dimeric molten globule, as indicated by its 1H-NMR
features and ¯uorescent binding of 1-anilino-8-naphthalene sulfonate. We
propose that a glycine-speci®c turn con®guration enables speci®c inter-
actions between the mini-zipper and the C-terminal coiled-coil.
# 2001 Academic Press
Keywords: diabetes mellitus; gene regulation; isotope-edited NMR; leucine
*Corresponding author zipper; protein structure

Abbreviations used: ANS, 1-anilino-8-naphthalene sulfonate; DCoH, dimerization cofactor of HNF-1; DG, distance
geometry; DM, diabetes mellitus; 4HB, four-helix bundle; HNF-1a, hepatocyte nuclear factor-1a; HNF-p1,
dimerization domain of hepatocyte nuclear factor-1a (residues 1-32); LZ, leucine zipper; MODY, maturity-onset
diabetes mellitus of the young; MPD, methyl-2,4-pentanediol; MSe and Nle designate selenomethionine and
norleucine, respectively (amino acids are otherwise designated by standard single-letter code); NOE, nuclear
Overhauser enhancement; PDB, RCSB Protein Data Bank; SA, simulated annealing.
E-mail address of the corresponding author: weiss@biochemistry.cwru.edu

0022-2836/01/030635±24 $35.00/0 # 2001 Academic Press


636 An Intertwined Four-helix Bundle in HNF-1

Introduction four-helix bundles (4HB) have been proposed.33,34


We have recently suggested that residues 8-19 and
Maturity-onset diabetes mellitus of the young 190 -80 comprise an anti-parallel coiled-coil (``mini-
(MODY) is a monogenic human syndrome zipper'') along one face of the 4HB and that this
arising from impaired glucose-stimulated face mediates binding of HNF-1a to the saddle-
insulin secretion by the pancreatic b cell in the shaped coactivator DCoH.34 Recruitment of DCoH
islet of Langerhans.1 Six MODY genes have (not itself a DNA-binding protein) into a speci®c
been identi®ed, ®ve of which encode preinitiation complex, although not necessary for
speci®c transcription factors. The most common HNF-1a/DNA recognition, enhances by HNF-1a-
form (MODY3) is caused by heterozygous directed transcriptional activation.30 Symmetry-
mutations in hepatic nuclear factor HNF-1a.2 ± 8
based modeling suggested that the HNF-1a mini-
Although HNF-1a is also an important regulator
of hepatic and renal gene expression, the zipper sits atop the saddle to form an intermolecu-
MODY3 phenotype is restricted to the pancreatic lar four-helix bundle{.34 Essential features of this
b-cell{. Homozygous or compound heterozygous model have been veri®ed recently by X-ray crystal-
mutations in human HNF-1a have not been lography (Figure 1(a)).35
observed and are presumably incompatible with We describe here the crystal structure of the
post-natal life. Homozygous disruption of HNF- dimerization domain of HNF-1a at 1.7 A Ê resol-
1a in the mouse leads to massive liver failure, ution. Experimental phases were obtained by
Fanconi's renal dysfunction, and perinatal death.9 multi-wavelength anomalous dispersion using
In the b-cell, HNF-1a functions as a weak activa- selenomethionine (MSe) as scatterer. Because the
tor of the insulin gene and a putative cassette of native amino-terminal methionine residue was
genes required for regulated insulin secretion.10,11 not suf®ciently ordered, MSe was substituted for
This HNF-1a-regulated cassette is not well
one of the leucine side-chains. The structure
characterized.
HNF-1a is composed of four functional reveals an intertwined a-helical dimer anti-paral-
regions: an amino-terminal dimerization domain lel in orientation. ``Eisenberg''-like helix
(residues 1-32); a bipartite DNA-binding motif swapping36,37 between protomers leads to a
with variant homeodomain (residues 98-280); novel intertwined topology. Because such swap-
a carboxy-terminal transactivation domain ping was unanticipated by previous NMR mod-
(281-631); and a ¯exible linker (residues 33-97) els,33,34 we have reinvestigated the structure in
that connects the dimerization and DNA-binding solution. Dimer-related nuclear Overhauser
domains.12 ± 15 Excepting this linker, MODY effects (NOEs), de®ned by asymmetric 13C-isoto-
mutations occur in each region.16,17 The present pic labeling,38 are consistent with the overall
study focuses on the N-terminal dimerization orientation of protomers as observed in the crys-
domain,18,19 an autonomous module12,14,20,21 tal structure. Flexibility among N and C-terminal
required for the protein's gene-regulatory
segments is observed in solution and rationalizes
activity.8 This module also functions as a target
of transcriptional coactivator dimerization patterns of thermal (B) factors. Recent indepen-
cofactor of HNF-1 (DCoH).22 ± 30 An homologous dent crystallographic analyses of the HNF-1a
leucine-rich motif occurs in HNF-1b (also a domain by Alber and co-workers39 enable com-
MODY gene)31 and mediates combinatorial parison of the peptide structure in different crys-
homo- and heterodimerization.32 tal forms and lattice environments. Such
The structure of the dimerization domain has comparison suggests that the 4HB contains a
been investigated by circular dichroism (CD) and rigid element (the mini-zipper) and an adjustable
1
H-NMR spectroscopy. Although initially proposed element (variably oriented C-terminal a-helices).
to form a leucine zipper (LZ; parallel coiled-coil), These elements are connected by a tight turn
NMR studies have demonstrated the presence of a unrelated to classical b- or g-turn families.40,41
helix-turn-helix secondary structure.21 Possible sol- Diabetes-associated mutations in this turn and
ution structures based on parallel or anti-parallel
adjoining N-terminal a-helix2,5,42 destabilize the
4HB by distinct mechanisms. Whereas a destabi-
{ Designation of HNF-1a as a hepatic factor re¯ects lizing mutation in the mini-zipper (L12H) pre-
the history of its discovery and role in liver-speci®c serves the domain's architectural speci®city, a
gene regulation.15 Heterozygous mutations in the coding
region or promoter of the human HNF-1a gene have no
mutation in the turn (G20R) leads to a molten
known effect on hepatic function in contrast to globule. We propose that a glycine-speci®c turn
impairment of b-cell function.10 architecture is required for speci®c packing of
{ In crystallographic studies of the HNF-p1/DCoH side-chains of the C-terminal coiled-coil against
complex,35 a-helical orientations in the intermolecular side-chains of the N-terminal mini-zipper. Studies
4HB were described as opposite to those predicted.34
This is incorrect: predicted and observed helical
of the diabetes-associated variants suggest that a
orientations are congruent (see the Supplementary frustrated turn alters the global topography of
Material). the free-energy landscape.
An Intertwined Four-helix Bundle in HNF-1 637

Figure 1. Structure of the dimerization domain of HNF-1a. (a) Ribbon representation of co-crystal structure of
DCoH dimer (blue) complexed with HNF-p1 (red and green) (accession number 1F93).35 The arrow indicates 2-fold
rotation axis. (b) Crystal structure of isolated HNF-p1w dimer determined in this study. Red and green cylinders rep-
resent a-helices 1 and 2 in the two protomers. Each protomer comprises a helix-turn-helix secondary structure with
MODY mutation site G20 (represented as a yellow sphere) within a non-canonical turn. a-Helix 1 (residues 4-19) is
longer than a-helix 2 (residues 23-32). (c) Stereo view of a portion of 2Fo ÿ Fc electron density map calculated using
1.7 AÊ resolution data. The map is contoured at the 1.5s level and superimposed on the re®ned model. MSe denotes
selenomethionine residue.
638 An Intertwined Four-helix Bundle in HNF-1

Results (residues 3-30) yields a root-mean-square deviation


(rmsd) of 0.24 A Ê.
Because molecule 1 is better ordered than mol-
The crystal structure (HNF-1a residues 1-32 with ecule 2, analysis focuses on molecule 1 unless
W33 extension{ and MSe13 substitution{; desig- otherwise stated. Residues 1-3 form an extended
nated HNF-p1w) was determined by multiple strand followed by a-helix 1 (residues 4-19), a
anomalous dispersion (MAD) phasing and re®ned sharp turn (20-22), a-helix 2 (residues 23-32) and
to 1.7 AÊ resolution (Rcrystˆ0.231 and Rfreeˆ0.245; W33. No interhelical interaction (<3.5 A Ê ) is
see Table 1). Electron density (2FoÿFc) correspond- observed in the protomer. Within the asymmetric
ing to residues 10-16 is shown in Figure 1(c). The unit, molecules 1 and 2 are connected loosely
asymmetric unit contains two independent poly- through a-helices 2 and 20 (interface depicted in
peptides and 55 ordered water molecules}. The Supplementary Material). The surface area buried
polypeptide molecules are not related by the phys- at this interface is only 846 AÊ 2. By contrast, mol-
iological dimer axis (see below). Molecule 1 is bet- ecules 1 and 2 each form extensive interactions
ter ordered than molecule 2; respective mean with another polypeptide in the unit cell related
thermal B-factors are 15.9 AÊ and 23.0 AÊ 2. The con- by a crystallographic 2-fold symmetry axis
®gurations of non-glycine residues are in the most (Figure 1(b)). The surface area buried at this inter-
preferred region of the Ramachandran plot. Resi- face is 1741 A Ê 2. The putative symmetry-related
due G20 in each polypeptide is in the generously dimer is consistent with prior analyses of NOEs in
allowed L-region adjoining the aL island. Mol- solution33,34 and in accord with the co-crystal struc-
ecules 1 and 2 each contain two a-helices con- ture of a HNF-p1/DCoH complex (Figure 1(a)).35
nected by a sharp reversal segment, rendering a V We therefore regard this symmetry-related packing
shape to the protomer's overall architecture as the biological dimer. The orientation between
(Figure 1(b)). The angle between a-helices is 37 in protomers within dimers 1 and 2 in the unit cell
each case. Alignment of the two polypeptides by are essentially identical.
least-squares superposition of main-chain atoms
Dimer topology and interactions
{ HNF-p1w differs from the peptide studied by Alber between protomers
and colleagues (HNF-p1)35 by the presence of an
additional C-terminal tryptophan-amide (W33; in place a-Helices 1 and 2 each form anti-parallel coiled-
of P33 in native HNF-1a sequence), N-terminal coils with dimer-related mates to de®ne opposite
substitution of methionine by norleucine, and surfaces of the 4HB. Each coiled-coil remains con-
substitution of L13 by MSe for MAD phasing. W33 tiguous throughout its length. Helix 1 contacts
appears to stabilize the structure,34 presumably via side- helix 20 , whereas helix 10 contacts helix 2. Despite
chain contacts observed in solution and not re¯ected in this novel topology, individual coiled-coils are sep-
the crystal structure (see the Supplementary Material). arately superimposable on those of a conventional
Norleucine was employed to avoid methionine 4HB (see Supplementary Material). As
oxidation. Se-Met at position 13 was also employed by
expected,43,44 the helices in HNF-p1w contain 3.6
Rose et al. 39 for MAD phasing.
{ Substitution of L13 by MSe has no effect on the residues per turn. Distances of closest approach
between helical axes are 10.9 A Ê (N-terminal coiled-
a-helix content of the HNF-1a dimer, as inferred by CD.
However, the variant dimer is less stable; Gu, as coil) and 9.3 A Ê (C-terminal coiled-coil). In accord
estimated by CD-detected guanidine titration, is with classical expectations,45 the angle between
2.4(0.2) kcal/mol (see the Supplementary Material). helices 1 and 10 is 30  and that between helices 2
The relative orientation of N and C-terminal a-helices in and 20 is 23  . The crossing angle between N and
the crystal structure of HNF-p1w differs from that seen C-terminal coiled-coils is 26  . Although the overall
in the DCoH co-crystal structure. 35 These differences structure resembles that observed in a peptide-
are retained in a preliminary electron-density map of DCoH complex35 and peptide dimers in different
native (L13) HNF-p1w peptide crystals (N.N. and
crystallographic environments,39 the precise orien-
M.A.W., unpublished results) and so are unrelated to
the MSe13 substitution. Similar results have been tation of coiled-coils differs (see Discussion and
obtained by Rose et al.,39 who provide ®ve sets of Supplementary Material).
dimer-speci®c coordinates: (i) and (ii) two independent The dimer interface is stabilized by extensive
dimers in the asymmetric unit in wild-type HNF-p1; (iii) hydrophobic and van der Waal's interactions
and (iv) two independent dimers of an MSe12 analogue between protomers and by symmetry-related
and (v) one MSe-13 analogue. Of these ®ve dimers, only hydrogen bonds between the side-chains Q9 and
the MSe-13 structure exhibits exact 2-fold symmetry in S190 (and likewise between Q90 and S19). Unlike
the dimer. Quantitative comparisons of these and the coiled-coils,44,46 there is no salt-bridges between
present structure are described in Discussion (Figures 7 protomers. The dimer's hydrophobic core contains
and 8, and Table 4).
a cluster of leucine side-chains. A prominent fea-
} In deposited coordinates, residues in molecules 1
and 2 are labeled 1-33 and 36-63, respectively. Residues ture is the packing of L12 and MSe13 (L13 in the
34, 35, and 64-66 are disordered and hence absent from native sequence) against L120 , MSe130 , L160 , L210 ,
the coordinate set. The 55 ordered water molecules are and L260 (Figure 2). Eight leucine and two MSe
numbered 67-121. In the text and Figures, primed labels side-chains thus contribute to the intertwined inter-
denote dimer-related residues. face. The side-chains of L12 and L120 (a site of
An Intertwined Four-helix Bundle in HNF-1 639

Table 1. Statistics for data collection, phasing, and re®nement


A. Data collection and phasing
Space group P21212
Ê)
Unit cell (A aˆ28.42, bˆ42.19, cˆ42.43
Vm (AÊ 3/Da) 1.6 (two protomers/asymmetric unit; 23% solvent content)
Resolution (A Ê) 1.7 (MAD)
Data set (Se scatterer) Inflexion (li) Peak (lp) Remote (lr)
Wavelength (A Ê) 0.9800 0.9794 1.0030
Observed reflections 47,253 46,501 29,713
Unique reflections 5693 5669 5778
Completeness (%) 95.2 94.8 96.6
Average I/sa 26.8 (4.3) 26.1 (4.1) 23.9 (3.7)
Rmerge (%)a,b 3.8 (2.6) 4.0 (2.6) 3.7 (2.4)
Mean figure of merit (FOM)c 0.90

B. Refinement statistics
Resolution range (A Ê) 40-1.7
Rcryst (%) (F > 3sF)d 23.1
Rfree (%) (F > 3sF) 24.5
Reflections (F > 3sF; completeness)
Working set 4949 (83 %)
Test set 296 (5%)
Number of non-H atoms
Protein 458
Solvent 55
Average B-values (A Ê 2)
Protein 19.1
Solvent 28.1
Ramachandran plot (%)
Most favored 98.1
Additionally allowed 1.9
a
Values in parentheses indicate the highest resolution shell.
b
Rmerge ˆ hklijIi(hkl) ÿ hI(hkl)ij/hkliIi(hkl). Friedel-mates were collected at wavelengths li and lp.
c
MAD phases were computed using data between 20 and 2.5 A Ê resolution. FOM ˆ hcos (a)i, in which a is the error in phase
angle of a re¯ection.
d
Rcryst ˆ hkljFobs(hkl) ÿ Fcalc (hkl)j/hklFobs(hkl). The structure was re®ned using X-ray data collected at a remote wavelength.

MODY mutation; see Discussion) are contiguous The intertwined 4HB contains a non-
and contribute to the non-polar surface of the canonical turn
mini-zipper. The N-terminal portion of helix 1 con-
tacts the symmetry-related turn as the side-chain of
Q9 contacts S190 , G200 , and L210 . The side-chains of a-Helix 1 is connected to a-helix 2 by a sharp
K23, L26, I27, A29, L30, E32, and W33 (and their turn whose structure differs from that of classical b
symmetry-related mates) mediate interactions or g-turn families.40,41,47 The turn (comprising resi-
between helix 2 and helix 20 . dues 19-22) exhibits successive backbone (f,c)
At the interface between helices 1 and 20 , key angles of (78.7  , ÿ0.5  ), (97.0  , 23.7  ), (ÿ52.6  ,
contacts occur between the side-chains of MSe13 136.4  ), and (ÿ85.5  , 156.3  ). Similar torsion
and L17 (helix 1) and L260 (helix 20 ). L17 also con- angles occur in diverse crystallographic
tacts A290 and E320 . Preliminary studies of iso- environments39 and in the peptide-DCoH com-
morphic crystals containing the native L13 peptide plex.35 Whereas the angle between helices 1 and 2
reveal an essentially identical structure at 1.7 A Ê in the present structure is 37  , however, that in the
resolution with ten leucine side-chains in the core. bound peptide is 65  .35 The conformation of the
Of the two molecules in the asymmetric unit, elec- turn's second residue (G20) lies in the right side of
the Ramachandran plot (i.e. positive f angle near
tron density for W33 is seen only in molecule 1.
Ê ) by the aL island). Although G20 is invariant among
The indole side-chain is surrounded (<4.5 A
mammalian HNF-1a and HNF-1b sequences
residues 29 through 32 and L170 , L210 , S220 , K230 , (Figure 3), L-amino acids would not be excluded
and L260 (see the Supplementary Material) The on the basis of a Ramachandran criterion. Molecu-
main-chain amide nitrogen atom of W33 forms a lar modelling nonetheless suggests that the b car-
hydrogen bond with the carbonyl oxygen atom of bon atom of an L-amino acid would clash with the
G31 and so provides a C-cap to a-helix 2. Lattice carbonyl oxygen atom of the preceding residue
contacts to W33 in molecule 1 are observed from (S19). The mutation G20R has been observed in a
symmetry-related (in addition to dimer-related) MODY pedigree42 and causes a profound decre-
polypeptides; these lattice contacts involve residues ment in the dimer's thermodynamic stability
1-5. (Gu > 4 kcal/mol (1 cal ˆ 4.184 J); Table 2).34
640 An Intertwined Four-helix Bundle in HNF-1

Figure 2. Leucine-rich cluster in


HNF-p1w dimer. (a) and (b) Rib-
bon representations (related by
180o about the horizontal axis pas-
sing between L12 and L26, and
L120 and L260 ) of the HNF-p1w
dimer. CPK models of leucine side-
chains that form the leucine-rich
cluster in core. In one protomer,
the ribbon is shown in black and
the leucine residues in red (L12,
MSe13, L16, L21, L26). In the other
protomer, the ribbon is colored
gray and leucine side-chains green
(L120 , MSe130 , L160 , L210 , L260 ).
Each protomer contributes four leu-
cine and one selenomethionine resi-
due. (c) A stereo view of the
leucine-rich cluster shown in (a).

Flexible regions and disorder dimer's secondary structure in solution as de®ned


by NOEs comprises a-helices 8-19 and 22-29 (see
Electron density is well de®ned for all residues the Supplementary Material), the N-terminal
in molecule 1, whereas in molecule 2 the N-term- a-helices in the crystal extend from residues 4-19 in
inal two and C-terminal three residues are disor- both molecules in the asymmetric unit. This differ-
dered. Mean main-chain B-factors for a-helix 1 in ence in helical length is unlikely to be due to the
molecules 1 and 2 are 12.6 and 16.5 A Ê 2, respect- presence of an organic cosolvent (25 % methyl-2,4-
ively, and for a-helix 2, 14.1 and 29.2 AÊ 2, respect- pentane-diol; MPD) under conditions of crystalliza-
ively. Corresponding side-chain B-values for tion, as the circular dichroism (CD) spectrum of
a-helix 1 in molecules 1 and 2 are 18.0 and 19.5 A Ê 2, the dimer is unaffected by the cosolvent (see the
respectively, and for a-helix 2, 21.8 and 30.5 A2, Ê Supplementary Material).
respectively. (Residue-speci®c B-factors are pro- Previous NMR analyses employed NOE patterns
vided as Supplementary Material.) The apparent (such as strong dNN and nested (i,i ‡ 3) daN con-
¯exibility of residues 1-2 and 31-33, as inferred tacts) to delimit a-helices.21,33,34 It is possible that
from higher thermal values in molecule 1 and such analyses, although assigning stable helical
absence of density in molecule 2, is corroborated elements correctly, failed to discern a signi®cant
by 1H-NMR motional narrowing.34 Whereas the helical bias among an ensemble of populated
An Intertwined Four-helix Bundle in HNF-1 641

Table 2. Mutant domains: guanidine denaturation studies and NMR chemical shifts
m(kcal molÿ1
Analogue Conc. (mM) Gu Gu Cmid Mÿ1) gc gu
A. Thermodynamic parametersa
wt 5 11.9  0.1 - 4.1  0.07 1.27  0.02 ÿ6.69 5.26  0.1
wt-KEK 5 11.8  0.1 ÿ0.1  0.2 3.9  0.06 1.32  0.02 ÿ6.69 5.10  0.1
L12H 5 8.2  0.2 ÿ3.7  0.3 6.1  0.20 0.25  0.01 ÿ6.69 1.52  0.2
G20R-KEK 5 6.9  0.4 ÿ5.0  0.5 0.4  0.05 0.67  0.08 ÿ6.69 0.24  0.4
G20R-KEK 60 6.8  0.6 ÿ5.1  0.7 2.0  0.30 0.74  0.10 ÿ5.33 1.45  0.6

B. 1H-NMR differences in chemical shift (L12H)b


Residue HN CaH CbH Other
Leu5 0.1
Gln9 0.23
Glu11 0.1 CgH2 0.1
Ala15 ÿ0.17, ÿ0.72
Leu16 0.4 CdH3 0.2
a
Guanidine denaturation data were ®tted according to a two-state model (see Materials and Methods). Rows 1-4 were published
in part by Hua et al.34 Gu indicates the change in free energy on unfolding as corrected to standard conditions (1 M peptide
concentration and 4  C). Gu ˆ gc ‡ gu, where gu is obtained by curve ®tting and gc is a correction factor that depends on dimer
concentration and temperature. Cmid is de®ned as the denaturant concentration at which 50 % of the protein is unfolded. The m
value is the slope d(Gu)/d(M). KEK designates N-terminal extension Lys-Glu-Lys.
b
Chemical-shifts are given relative to 3,3,3-trimethylsilylpropionate (0 ppm) at pH 7.0 and 25  C. Only differences > 0.1 ppm in
magnitude are shown. Positive value present resonances that are up®eld in the native spectrum relative to the spectrum of the L12H
analogue.

N-terminal structures. To investigate this possi- de®ned by contacts between side-chains, L12-L160 ,
bility, nascent helical propensity was probed by MSe13-MSe130 , and L13-L160 . Contiguity between
two additional and independent NMR parameters: the C-terminal helices is established by contacts
(i) 1Ha and 13Ca chemical shifts;48 and (ii) confor- between K23-L300 , K23-W330 , L26-L260 , L26-L300
mationally sensitive 3JaN coupling constants.49,50 and I27-W330 . The intertwined helix 1/20 relation-
Whereas residues in the 8-18 and 23-30 segments ship is veri®ed by contacts between side-chains
exhibit 3JaN values less than 5 Hz in accord with MSe13-L260 and L17-W330 . Representative NOE
their helical NOE patterns, 3JaN values of residues data and corresponding structural crystal models
4-6 are 6.7, 6.8 and 6.0 Hz, respectively (3JaN of are provided in Figure 4. Within interacting side-
residue 7 could not be obtained due to line-broad- chains, the details of which protons are in closest
ening). Further, whereas the 1Ha and 13Ca chemical proximity across the dimer interface can differ
shift index for residues 7-18 and 23-30 is consistent between crystal and solution structure (unpub-
with a-helix, index values for residues 4-6 are lished results). Dimer-related NOEs inconsistent
inconsistent with helix. These results suggest that with the crystal structure are observed involving
residues 4-6 do not exhibit a strong a-helical bias the side-chains of L5, L8, and W33, presumably
in solution. Accordingly, we imagine that the due to ¯exibility in solution (see the Supplemen-
environment of residues 1-7 in the crystal lattice tary Material). Similarly, W33-speci®c ring-current
favors N-terminal extension of a-helix 1. In the shifts predicted on the basis of the crystal structure
crystal structure of the DCoH-bound peptide35 of molecule 1 are not observed in solution (see the
a-helix 1 begins at residue 6, intermediate between Supplementary Material) in accord with this resi-
the NOE-de®ned a-helix in solution and that due's apparent disorder in molecule 2.
observed in the present structure.

Contrasting effects of clinical mutations


The solution structure resembles the
crystal structure Previous studies have established that mutations
L12H and G20R permit native or near-native helix
To investigate the relationship between the top- content (as inferred from CD) but perturb the stab-
ology of the dimer in the crystal and in solution, a ility of the dimer (Table 2).34 Thermodynamic par-
uniformly 13C-labeled peptide was prepared in ameters derived from guanidine denaturation
Escherichia coli and mixed with the unlabeled pep- studies by the two-state formalism suggest that the
tide to obtain an equilibrium between isotopic G20R mutation causes a larger decrement in stab-
homodimers and heterodimers. NOEs were then ility (Gu ÿ 5.0 kcal/mol) than does L12H
identi®ed between 13C-attached protons and (Gu ÿ3.7 kcal/mol). The structural basis of this
12
C-attached protons at the dimer interface. Dimer- difference is enigmatic, as the L12H substitution in
related NOEs diagnostic of an intertwined top- the dimer's non-polar interface would seem to be
ology are in overall accord with the crystal struc- less conservative. Because this conclusion depends
ture (Table 3). The phase of the mini-zipper is on the appropriateness of the two-state model, CD
642 An Intertwined Four-helix Bundle in HNF-1

Figure 3. (a) Sequence alignment of HNF-1a and HNF-1b and (b) models showing proposed volume compensation.
(a) Peptide sequences of HNF-1a dimerization domain (1-32) available at the SWISS-PROT annotated protein
sequence data base release 39.9 of 22-Nov-2000 (http://www.expasy.ch/sprot/sprot-top.html)84 are aligned. Invariant
and diabetes-related mutational sites L12 and G20 are shown in red. I27L polymorphism63 is indicated by letter L (in
green). In the lower part of (a), peptide sequences of HNF-1b dimerization domain (1-32) are aligned. Invariant resi-
dues L12 and G20 are in red. An asterisk (*) at position 18 indicates a difference in the type of amino acid relative to
HNF-1a. Valine residues are found at positions 21 and 25 in HNF-1b compared to L21 and A25 in HNF-1a (T25 in
chicken). These valine residues, proposed volume-®lling substitutions in HNF-1b, are bracketed. (b) The left
®gure represents a CPK model of a putative G21-G25 crevice obtained by modifying residues at position 21 and 25
in HNF-p1w protomer. The stereo view of the same protomer without modi®cation displays the native volume-®lling
side-chains of L21 (red) and A25 (pink) in HNF-p1w protomer.

spectra were obtained as a function of peptide con- conducted at a peptide concentration of 5 mM, at
centratrion (in the range 5-75 mM) as an explicit which the G20R analogue (unlike native and L12H
probe of the stability of the dimer. The spectrum of dimers) is substantially dissociated. To address the
the native dimer is invariant in this concentration possibility that the unfolding curve and apparent
range in accord with its low dissociation constant Gu value (which presumably re¯ected only the
(of the order of 1 nM; a precise estimate requires fraction of dimer) were unreliable at this concen-
the assumption of no residual structure in the tration, the guanidine titration was repeated at a
monomer). By contrast, the inferred values of Gu peptide concentration of 60 mM to ensure a predo-
predict that the spectrum of the G20R analogue minance of dimers. Although the unfolding curve
would exhibit marked concentration-dependent is shifted to more negative (native-like) values of
changes in this range, whereas the L12H analogue ellipticity (Figure 5), derived thermodynamic par-
would exhibit slight but discernible changes. This ameters are essentially unchanged (Table 2). The
is indeed observed (Figure 5). These data thus cor- statistical r2-value is 0.9996.
roborate the order-of-magnitude estimates of G Inspection of the ®t between the two-state theor-
provided by the two-state model. Nevertheless, the etical curve and the G20R titration data points at
original CD titration studies by Hua et al.34 were 60 mM reveals that systemic deviations occur
An Intertwined Four-helix Bundle in HNF-1 643

Table 3. Dimer-related NOEs and distances diagnostic of intertwined topology


Proton(s) Proton(s) Ê)
Distance across dimer (A Ê)
Distance in protomer (A
A. Phase of helix 1/helix 10 mini-zipper
L12d2-CH3 L160 d2- CH3 3.4 7.4
L13d1-CH3 L130 d1-CH3 6.2 -
L13d1-CH3 L160 d1-CH3 5.3 5.4

B. Intertwined helix 1/helix 20 relationshipsa


L13d1-CH3 L260 d2-CH3 5.3 4.6
L17d2-CH3 W330 C4Hb 5.5 14.9

C. Phase of helix 2/20 contact


L26d2-CH3 L260 d2-CH3 3.92 -
Distances shown represent carbon-carbon distances in crystal structure. Corresponding hydrogen-hydrogen distances were
estimated using the Biopolymer facility of InsightII (Molecular Simulations, Inc.).
a
Dimer-related NOEs are observed between W330 C4H and a set of protons in helix 1 (T10 gCH3, L13 dCH3 and A14 bH3) that
are inconsistent with the present structure (see the Supplementary Material) but would be consistent with the bound conformation35
if extended to W33 by model building. A dimer-related NOE is also observed between L5 dCH3 and L210 dCH3. This contact,
involving the ¯exible N-terminal segment, is inconsistent with the present structure and that described by Rose et al.35 The methyl
resonances of L5 exhibit anomalous broadening rather than motional narrowing (as exhibited by residues 1-3), presumably due to
millisecond exchange among multiple conformations.
b
Dimer-related NOEs are observed between L17 d2-CH3 and W330 C4H, which were classi®ed correctly in the side-by-side model
and are in accord with the present structure.

despite the reassuring r2 value (i.e. close to unity). domain's near-native helix content as probed by
Points fall beneath the line at guanidine concen- CD.
trations 0-1 M, above the line at guanidine concen- The anomalous NMR features of the G20R
trations 1-3.5 M, beneath the line from 3.5-5 M, domain suggest that the variant dimer is an a-heli-
and above the line from 5-7 M (see the Supplemen- cal molten globule. Because G20 adopts a positive
tary Material). Such systematic deviations, not f angle in the crystal structures, we imagine that
observed in ®tting the native and L12H transitions, substitution by Arg could lead to frustration of the
are distinct from random scatter and suggest that turn and could, in turn, disallow the native interdi-
the transition is not two-state. To investigate poss- gitation of side-chains between N and C-terminal
ible origins of the G20R analogue's anomalous coiled-coils. Interconversion of the G20R structure
titration behavior, 1H-NMR spectra were obtained among a range of non-optimal helix-packing
(Figure 5(b)). Whereas native and L12H samples schemes would rationalize both the analogue's
give rise to a spectrum typical of a globular non-two-state unfolding transition and anomalous
domain (with sharp and well-resolved resonances; NMR features. To test this model, we employed
spectra (b)a and (b)b, respectively), the G20R 1-anilino-8-naphthalene sulfonate (ANS) as a probe
dimer gives rise to an anomalous spectrum charac- of the molten-globule state{. The ANS ¯uorescence
terized by limited chemical-shift dispersion and emission spectra of the native and L12H domains
differential line broadening (spectra (b)d and (b)e). are similar in magnitude to that of a control globu-
lar protein (hen egg-white lysozyme). By contrast,
Sharp 1H-NMR resonances from the ¯exible N ter-
enhanced ANS ¯uorescence emission is observed
minus region are retained. The extent of line broad-
in the presence of the G20R domain at a peptide
ening is not signi®cantly affected by peptide
concentration of 100 mM. This enhancement is
concentration in the range 0.2-1.5 mM, suggesting
dimer-speci®c, as its extent diminishes as the pep-
that broadening is due to conformational exchange tide concentration is reduced, indicating, as
that is intermediate on the millisecond time-scale expected, little or no binding of the dye to the
of NMR chemical shifts. Gel-®ltration chromatog- monomer.
raphy at a peptide concentration of 0.5 mM under The quality of the NMR spectrum of the L12H
NMR conditions demonstrates that the G20R pep- dimer, by contrast to that of the G20R dimer, per-
tide is predominantly dimeric; no higher-order mits conventional sequential assignment (see the
aggregation is observed. Line-broadening and lim- Supplementary Material). The pattern of chemical
ited dispersion preclude conventional 2D-NMR shifts is similar to that of the native spectrum.
analysis (see the Supplementary Material). The Only ®ve residues (5, 9, 11, 15 and 16) exhibit
extensive perturbations manifest in the NMR spec- changes in chemical shift greater than 0.1 ppm in
trum of the G20R analogue contrast with the magnitude (Table 2(b)), The largest change occurs
at the Ha position of A15 (0.72 ppm) and may
{ ANS, a hydrophobic organic dye,86 exhibits little re¯ect the net ring currents of H12 and H120 . Small
binding to native globular domains but can enter the perturbations are observed at neighboring protons
¯uid non-polar interior of a molten globule.87 Such at positions 11 (0.1 ppm change in a g-methylene
binding shields the dye from solvent quenching and so resonance) and 16 (0.2 ppm change in a methyl res-
gives rise to enhanced ANS ¯uorescence. onance related to L12 by (i,i ‡ 4) and dimer-related
644 An Intertwined Four-helix Bundle in HNF-1

Figure 4. The dimer's intertwined topology is retained in solution. (a) and (b) Molecular models and (c) and (d)
corresponding NOE spectra. (a) Anti-parallel contacts in mini-zipper. Stereo view of stick (gray) representation of
N-terminal helices (residues 5-19 and 50 -190 ) in the HNF-p1w dimer. Hydrogen bonding interactions between Q9 and
S190 , and Q90 and S19 are shown by broken lines. The side-chains of Q9 and Q90 are colored in red, S19 and S190 in
green. The asterisk (*) represents the crystallographic 2-fold axis perpendicular to the plane of paper. The methyl
groups of MSe13 and MSe130 are shown as spheres. Residues L12 and L120 are near the 2-fold axis. Corresponding
dimer-related NOEs are shown in (c). (b) Dimer-related contacts re¯ecting intertwined topology. A signature of the
intertwined dimer is provided by key contacts between MSe13-L260 and L17-W330 . Stereo view of ribbon (gray) rep-
resentation of HNF-p1w protomer. The side-chains of L21, L26 and W33 are shown in gray, cyan and gray colors,
respectively. Residues 60 through 190 are shown as green sticks. The methyl groups in L21, L26, W33, MSe130 , L170
and amino group of Q9 are shown as spheres. (c) The 2D NOESY spectrum in H2O contains the following cross-
peaks: (a) Q9-He1-L210 -Ha, (b) Q9-He1-S190 -Hb, (c) Q9-He1-L210 -Hg, (d) Q9-He1-L210 -Hb1, (e) Q9-He1-L210 -Hb2, (f) W33-
H5-T100 -Ha, (g) W33-H6-T100 -Ha, (h) W33-H6-A140 -Hb, (i) W33-H7-A140 -Hb, (j) W33-H5-K230 -Hg and/or T100 -Hg, (k)
W33-H6-K230 -Hg and/or T100 -Hg, (l) W33-H7-K230 -Hg and/or T100 -Hg; (a0 ) Q9-He2-S190 -Ha, (b0 ) Q9-He2-S190 -Hb1, (c0 )
Q9-He2-L210 -Hg, (e0 ) Q9-He2-L210 -Hb2. Arrow indicates NOE between L21-NH and L17-Ha. (d) The 2D projection of
13
C intermolecular NOE 3D spectrum of 13C-HNF-p1w and unlabeled peptide dimer in 2H2O. The spectrum contains
the following inter-subunit NOEs: (m) L13-Hd2-L260 -Hd2, (n) L13-Hd2-L160 -Hd2, (o) L17-Hd2-L300 -Hd2, (p) L30-Hd2-L170 -
Hd2, (q) L30-Hd2-L130 -Hd2, (r) L16-Hd1-L130 -Hd2, (s) L26-Hd2-L130 -Hd2, (t) ambiguous, I27-H1-I270 -H1 and/or L12-Hd1-
L120 -Hd1, (u) L26-Hd-L260 -Hd2 and possibly I27-Hd-I270 -Hd at up®eld edge, (m0 ) L13-Hd1-L260 -Hd2, (n0 ) L13-Hd1-L160 -
Hd2, (o0 ) L17-Hd1-L300 -Hd2, (r0 ) L16-Hd1-L130 -Hd1 and (s0 ) L26-Hd-L130 -Hd1.

contacts). A similar correspondence is observed in NOEs suggests strongly that the variant side-chain
comparison of NOESY spectra: diagonal plots of is accommodated within a native-like fold. Surpris-
inter-residue contacts are essentially identical ingly, the pK51
a of the H12 imidazole ring (7.2) is
(Figure 6). Although isotope-directed NMR studies not shifted to low values, indicating that the
of the L12H dimer have not been performed, the variant structure is compatible with protonation of
overall similarities between chemical shifts and the imidazole side-chains.
An Intertwined Four-helix Bundle in HNF-1 645

Figure 5. Diabetes-associated G20R mutation induces a molten-globule state. (a) CD and ¯uorescence studies of
peptide analogues. ((a)a and (a)b) CD studies of G20R and L12H domains at successive peptide concentrations:
75 mM (®lled circles), 50 mM (open circles), 25 mM (®lled triangles), 15 mM (®lled squares), and 5 mM (open triangles).
Explicit dissociation of the G20R dimer at this concentration range corroborates the estimate of its thermodynamic
destabilization (Gu) by two-state modelling of denaturant-induced unfolding. (a)c Denaturant-induced unfolding
studies of the G20R domain at high and low peptide concentrations (60 and 5 mM) in each case demonstrate non-sig-
moidal transitions and yield similar estimates of Gu. The unfolding curve of the L12H analogue by contrast retains
a sigmoidal character and exhibits a less marked gu, in accord with the concentration-dependence of its CD spec-
trum. (a)d ANS ¯uorescence studies of HNF-1 peptide analogues. Binding of the hydrophobic dye to the G20R ana-
logue is indicated by enhanced ANS ¯ourescence quantum yield, whereas residual ANS ¯uorescence in the presence
of the native and L12H domains is similar to that observed in the presence of a control globular protein (hen egg-
white lysozyme). (b) 1D 1H-NMR spectra of the HNF-1 peptide and analogues: a, native HNF-p1W; b, L12H ana-
logue of HNF-p1W; c, KEK-extended HNF-p1W; d, G20R analogue of KEK-extended HNF-p1W at 1 mM peptide
concentration; and e, G20R analogue of KEK-extended HNF-p1W at 0.2 mM peptide concentration. The attenuation
of chemical-shift dispersion and differential resonance broadening in the G20R dimer provides evidence of a molten-
globule state.
646 An Intertwined Four-helix Bundle in HNF-1

Discussion
4HBs are ubiquitous structural motifs exhibiting
a rich diversity of tertiary structures.52,53 Such
motifs provide favorable models for studies of pro-
tein folding and design.54,55 Parallel dimeric 4HBs
are of particular importance in transcription factors
containing a helix-loop-helix DNA-binding
domain.56 ± 58 To our knowledge, the anti-parallel
HNF-1a domain represents the smallest 4HB motif,
a novel intertwined dimer. Mutations in this
domain are associated with diabetes mellitus in
humans.16,42,59,60

Comparison of HNF-p1 peptides in different


crystal forms
Crystal structures of related HNF-p1 peptides
(see footnote to page 638) were described recently
in independent studies by Alber and co-workers.39
While the manuscript was in review, multiple coor-
dinate sets were deposited in the RCSB Protein
Data Bank, permitting detailed comparison of
structures. The coordinates were of the native
HNF-p1 peptide and two Se-Met analogues at pos-
itions 12 and 13. Crystallization conditions and
crystal forms in each case differ from those
described here (a summary of crystallographic
information is given in the Supplementary
Material). The availability of multiple independent
structures provides an opportunity to evaluate the
conformational repertoire of the peptide structure
and effects of crystal packing.
Comparison of the present structure with those
described by Rose et al.39 was performed by least-
squares superposition of equivalent atoms. Two
alignment schemes were investigated. The ®rst
Figure 6. Diagonal plots of (a) inter-residue NOEs in employed an overall alignment of all main-chain
native dimer and (b) L12H variant dimer demonstrate atoms in the dimer. Residue-speci®c mean rmsd
structrural similarity. Contacts between side-chains are values for main-chain and side-chain atoms are
shown in lower right-hand triangles; contacts between shown in Figure 7(a) and (b). Whereas low main-
main-chain atoms or between main-chain and side-chain chain rmsd values are observed for two structures
atoms are shown in upper left-hand triangles. Red boxes
(wild-type dimer bd and MSe-13 dimer; superim-
in (a) indicate dimer-related NOEs, including those
given in Table 2B; green boxes in (a) indicate contacts posed ®lled green and blue triangles, respectively),
likely to include both intraprotomeric and dimer-related large values are observed throughout the sequence
contributions. Blue boxes in (b) highlight contacts by for one MSe12 dimer (designated ac; open red dia-
mutant H12 side-chain. No distinction is made in (b) monds). For the other wild-type dimer (designated
between intraprotomeric and dimer-related contacts as ac; open green triangles) and the other MSe12
isotope-edited spectra were not obtained. Contacts a-e dimer (designated bd; ®lled red diamonds), low
in (a) are present in native spectrum but not in that of rmsd values are observed in the mini-zipper and
L12H variant, whereas contact a in (b) is present in the larger values in the C-terminal a-helices. In each
variant spectrum but not in the native spectrum. Assign- case, side-chain rmsd values are lower in the
ments are: a, 30-Ha/12-CdH3; b, 14- Ha/23-Hb2; c, 23-
interior than among solvent-exposed residues.
Ha/16- CdH3; d, 17- HN/24- Cb; e, 23- Ha/17- Cd1, a, 3-
Hb/29- Cb. (Corresponding distances in crystal struc- Structural variability among C-terminal segments
tures are given in the Supplementary Material.) Open can give rise to extremely large side-chain rmsd
squares indicate possible contacts rendered ambiguous values (3-8 A Ê ). Of the ®ve independent dimers
by resonance overlap. An asterisk in (a) indicates described by Rose et al.,39 the two most similar to
dimer-related NOEs inconsistant with crystal structure that obtained here are the wild-type bd dimer and
(molecule 1) MSe-13 dimer (mean main-chain rmsd of 0.25 A Ê
and 0.24 AÊ , respectively, for residues 5-28 and 50 -
280 ).
The overall main-chain alignment described
above suggests an asymmetry in degree of struc-
An Intertwined Four-helix Bundle in HNF-1 647

Figure 7. Comparison of HNF-p1 peptide structures by rmsd in two alignment schemes. (a) and (b) Alignment
according to the main-chain atoms including all four helices in the dimer (residues 5-28 and symmetry-related mates)
suggests the extent of overall structural variation. (c) and (d) Alignment according to the main-chain atoms of mini-
zipper only (residues 5-19 and symmetry-related mates) highlights the segmental mobility of the C-terminal coiled-
coil and invariance of N-terminal mini-zipper. Five peptide dimers determined by Rose et al.39 are compared to mol-
ecule 1 of the present study. Abbreviations used for peptide dimers are the same as in Table 4, Wt-ac (open green tri-
angles), wt-bd (®lled green triangles), MSe12-ac (open red diamonds), MSe12-bd (®lled red diamonds), and MSe-13
(®lled blue triangles). Main-chain rmsd values are shown in (a) and (c), whereas side-chain rmsd values are shown in
(b) and (d). In each panel, positions of the four helices are indicated by ®lled or open bars at top. Because HNF-p1
dimers except MSe-13 lack exact 2-fold symmetry39 (unlike the present crystal form), rmsd values for residues 1-32
and 10 -320 are depicted separately. Values for residues whose positions were not de®ned in the electron density in the
corresponding structure are omitted.

tural similarity or difference: larger rmsd values Converse protomer-speci®c alignment on helix 2
tend to occur in the C-terminal a-helix than in the (not shown) likewise results in skew positions of
N-terminal a-helix. To explore this apparent asym- helix 1. Dimer-speci®c alignment on the mini-zip-
metry further, we aligned the structures according per (Figures 7(c) and 8(b)) reveals invariance of the
to Ca atoms in (a) the N-terminal a-helix of an iso- mini-zipper and variable positioning of the C-term-
lated protomer (residues 9-19) and (b) the mini-zip- inal coiled-coil. The converse alignment on the
per (residues 9-19 and 90 -190 ). These alignments are C-terminal coiled-coil (residues 23-28 and 230 -280 )
illutrated in Figure 8. The protomer-speci®c align- by contrast fails to yield a consistent view of the
ment (Figure 8(a)) demonstrates close superposi- aligned helices (not shown). We therefore regard
tion of helix 1 with variable orientation of helix 2. the mini-zipper alignment as providing physical
648 An Intertwined Four-helix Bundle in HNF-1

Figure 8. Least-squares superpo-


sition of HNF-p1 peptides. (a) Indi-
vidual protomers of HNF-p1
dimers35 were aligned with equiv-
alent Ca atoms of residues 9-19 of
the present HNF-p1w protomer
shown in red. The wild-type proto-
mers are in cyan (PDB ID: 1g39),
MSe12 protomers are in green
(PDB ID: 1g2y), the MSe-13 proto-
mer is in blue (PDB ID: 1g2z). Only
Ca atoms are displayed in (a)-(c).
The rmsd of superposition ranges
between 0.1 and 0.25 A Ê . (b) The
HNF-p1 dimers from Rose et al.35,39
were aligned with equivalent Ca
atoms of the mini-zipper segment
(residues 9-19 and 90 -190 ). The col-
oring scheme is the same as in (a).
The HNF-p1 dimer in DCoH-HNF-
p1 complex (PDB ID 1f93) is shown
in black. There is an excellent
agreement in each case with rmsd
of superposition less than 0.4 A Ê
indicating rigid mini-zipper.
(c) Same superposition as in (b) but
a different view from that seen in
(b) showing the variable helices 2.
The protomers are colored as in
(b). For clarity, only the C-terminal
helices 2 of different dimers are
shown except for the reference
HNF-p1 dimer (red), where all its
Ca atoms are shown.

insight into the modular composition of the precision of packing of the mini-zipper is, by con-
domain. Residue-speci®c rmsd values in this align- trast, characteristic of a native state.
ment are shown in Figure 7(c) and (d). An asym- The HNF-1a dimerization domain provides a
metry in rmsd values between N and C-terminal recognition surface for binding transcriptional
helices is consistent and striking. An overview is coactivator DCoH.30 Differences between structures
provided by mean rmsd values for Ca atoms by of free and bound peptides suggest that the HNF-
segment (Table 4A); corresponding interhelical 1a dimer undergoes a limited but detectable
angles in HNF-p1 peptides are given in Table 4B. change in conformation upon binding to DCoH.
Whereas the angle between mini-zipper helices is In the absence of additional co-crystal structures,
nearly invariant, large variations are observed in it is not known whether the distinctive structural
all other inter-helical angles: the angle between N features of the bound HNF-p1 dimer are a general
and C-terminal helices in a protomer, angle consequence of DCoH binding or simply one of
between C-terminal helices, and crossing-angle several possible bound conformations. The extent
between mini-zipper and C-terminal coiled-coil. of induced ®t is thus an unresolved issue.
The variable tertiary structure of the C-terminal Together, observations of higher thermal factors
helices is reminiscent of a molten globule, a col- for helix-2 and different relative orientations
lapsed state containing ordered elements of sec- between helix 2 and 20 in isolated and bound struc-
ondary structure of variable orientation.61 The tures suggest that helix 2 is adjustable by rigid-
An Intertwined Four-helix Bundle in HNF-1 649

Table 4. Least-squares superposition and interhelical angles in HNF-p1 peptides


Residues MSe13* Wt-ac Wt-bd MSe12-ac MSe12-bd MSe-13 DCoH-HNF
A. rmsd (AÊ) of least-squares C superposition of mini-zipper
a a

9-19 90 -190 0.20 0.21 0.27 0.30 0.35 0.21 0.37


23-28 230 -280 0.27 2.40 0.18 3.30 2.50 0.29 4.10
B. Interhelical angles in HNF-p1 peptidesb
Angles (deg.) MSe13 Wt-ac Wt-bd MSe12-ac MSe12-bd MSe-13 DCoH-HNF
y1 37 39 37 42 43 37 65
y2 30 28 29 28 26 30 22
y3 23 12 16 3 20 18 9
y4 ÿ26 ÿ42 ÿ29 ÿ41 ÿ38 ÿ28 ÿ63
a
rmsd is the root-mean-square standard deviation calculated with reference to the better-ordered molecule (MSe13) in the present
structure. MSe13* is the less-ordered dimer in the present study. Wt-ac and Wt-bd represent wild-type dimers of subunits A and C,
and B and D, respectively (PDB ID: 1g39). MSe12-ac (subunits A and C) and MSe12-bd (subunits B and D) are HNF-p1 dimers with
Se-Met at position 12 (PDB ID: 1g2y). MSe-13 (HNF-p1 with Se-Met at position 13) dimer consists of subunits A and its 2-fold
related molecule (PDB ID: 1g2z). DCoH-HNF (subunits E and F of HNF-p1 peptide; PDB ID: 1f93).
b
y1 is the angle between helix 1 and helix 2 in the protomer. y2 is the interhelical angle between the mini-zipper helices. y3 is the
interhelical angle between interprotomer C-terminal helices 2 and 20 . y4 is the crossing angle between the C-terminal and N-terminal
helices.

body movement. We propose that helix 2 can of the helix 1/10 and 2/20 contacts. A detailed
adopt different relative orientations without per- description of the dimer0 s solution structure and
turbing helix 1 and 10 (Figures 7 and 8), and is in dynamics is in preparation. It is important to note
agreement with the recent crystal structures of that the confounding features of W33 in the orig-
HNF-p1 peptides in different crystal forms.39 inal NMR analysis did not arise due to inclusion of
a non-native C-terminal residue with aberrant
Relationship of structure in crystal and properties. Rather, the anomalous NMR behavior
in solution of W33 provides a faithful re¯ection of the anoma-
lous molten character of the C-terminal coiled-coil
The crystal structure of HNF-p1w differs from as revealed by comparison of multiple crystal
either parallel or anti-parallel 4HB models pro- structures (see Figures 7 and 8, and above discus-
posed on the basis of NMR studies.33,34 The anti- sion). It will be of future interest to investigate
parallel model successfully predicted the existence which crystal form or subset of crystal forms best
of an N-terminal coiled-coil (mini-zipper) in accord represents the ensemble of structures in solution.
with dimer-related NOEs (9-190 and 9-210 ; That 33 unique NMR spin systems are observed
Figure 4(a) and (c)). Nevertheless, the putative 4HB indicates that interconversion among structural
was envisaged as consisting of side-by-side proto- substates is fast on the time-scale of chemical shifts
mers rather than intertwined a-helices.34 A major (<1 ms).
role in the NMR analysis was played by prominent
long-range NOEs involving the indole resonances Symmetry-based modelling retrodicts
of W33 (Supplementary Material in Hua et al.).34 peptide-DCoH complex
Although contacts between W33 and L170 were
correctly interpreted in the original analyses, a Rigid-body docking of the HNF-p1w structure
side-by-side model was suggested by multiple and transcriptional coactivator DCoH provides an
additional NOEs from the indole ring to the side- opportunity to evaluate the strengths and limi-
chains of residues 10, 13, and 14. These are incon- tations of symmetry-based modelling.34 The recent
sistent with the crystal structure (see the Sup- availability of a co-crystal structure35 enables
plementary Material) but presumably arise due to evaluation of previous predictions and retrodic-
¯exibility in solution leading to multiple confor- tions based on the present structure. We ®rst dis-
mations. The criterion of consistency employed in cuss overall features of the HNF-1a/DCoH
the original distance-geometry (DG) modelling complex and then molecular details of the inter-
procedure34 did not consider possible confounding face. As predicted,34 HNF-1a/DCoH recognition is
effects of multiple W33 conformations. Analysis of mediated by formation of an intermolecular 4HB
inferred interproton distances in the crystallo- employing the HNF-1a mini-zipper and convex
graphic dimer emphasizes the importance of NOEs (``top'') surface of the DCoH saddle (Figure 1(a))35
between degenerate (symmetry-related) 1H-NMR (see also the Supplementary Material). Such bind-
resonances, unobservable in the absence of isotopic ing utilizes corresponding symmetry axes. The
labeling. Asymmetric 13C labeling allows such con- HNF-1a dimer has exact 2-fold symmetry, whereas
tacts to be de®ned. Key examples are provided by the DCoH tetramer is a dimer of saddle-shaped
strong NOEs between methyl resonances of dimers with a 4HB at its interface; each protomer
MSe13-MSe130 and L26-L260 . These contacts, incon- contributes one helix to the 4HB with approximate
sistent with a side-by-side model, de®ne the phase dihedral (222) symmetry. The interface superim-
650 An Intertwined Four-helix Bundle in HNF-1

Figure 9. Modeling of HNF-p1/DCoH complex. (a) Anti-parallel N-terminal helices 1 and 10 in HNF-p1 dimer are
shown as red ribbons. The rest of the HNF-p1 molecule is omitted for clarity. DCoH dimer (subunits A and B from
PDB code 1DCO) is shown as green ribbon. The 2-fold symmetry axes of DCoH dimer and the HNF-p1 dimer are
aligned. This results in the formation of a 4HB at the interface of DCoH and HNF-p1. The crossing angle between the
HNF-p1 helices and the DCoH helices is ÿ40  compared to ÿ27  in the co-crystal structure.35 (b) A diagram showing
relative orientaions of helices in the 4HB formed in the DCoH tetramer and DCoH/HNF-p1 complex. The left dia-
gram shows the dihedral symmetry of helical orientations in the DCoH tetramer. The right diagram shows the pre-
dicted 2-fold symmetry of the orientations of helices at the interface of the DCoH dimer and the HNF-p1 dimer,34
veri®ed in the DCoH/HNF-p1 crystal structure.35 Although the helical directions seen in the DCoH tetramer can
occur in the DCoH/HNF-p1 complex, it is predicted not to be favorable due to steric clashes and smaller buried sur-
face area upon dimerization (see the text). (c) The modeled interactions between the L12 residue (red) in HNF-p1 and
residues F47 (green) and T51 (violet) in DCoH are displayed by a dotted van der Waal's surface. Thick and thin rib-
bons correspond to DCoH and HNF-p1 helices, respectively. The stereo diagram depicts the ®t of L12 and L120 into
the hole formed by F47, T51 and their symmetry-related residues (F470 and T510 ).

poses these symmetry axes. Our previous docking Alignment of symmetry axes
exercise, although employing a side-by-side model
of the anti-parallel HNF-1a 4HB, nonetheless cap- The importance of symmetry in HNF-1a/DCoH
tured essential features of the intermolecular 4HB recognition has long been recognized. A previous
interface (see the Supplementary Material). In light model of the HNF-1a dimer as a parallel 4HB33
of the present crystal structure, we revisit the thus posed a structural dilemma: the mismatch of
docking problem to consider how reliably would symmetries appeared to preclude any simple
symmetry-based docking predict the actual mechanism of DCoH binding.25 Characterization of
structure of the complex.35 an anti-parallel mini-zipper, although incorrect in
An Intertwined Four-helix Bundle in HNF-1 651

its details, resolved this incongruity and led to cor- F47 and T51 and their 2-fold-related mates
rect positioning of HNF-1a atop the DCoH (Figure 9(c)), as observed in the complex.35 The
saddle.34 In this model, as in the co-crystal struc- model further retrodicts contacts from Q9, A15 and
ture (Figure 1(a)),35 DCoH recognition is mediated S19 in HNF-p1 to L55, T51-R52, and L55 in DCoH,
by HNF-p1w helices 1 and 10 , whereas helices 2 respectively, also in accord with the co-crystal
and 20 are distal to the interface. The model cor- structure. Farther from the symmetry axes,
rectly de®ned the relative orientations of individ- however, several intermolecular interactions are
ual not retrodicted immediately. These include S6-E58,
a-helices in the intermolecular 4HB (Figure 9(a) Q7-R45, L8-E58, E11-R45, E18-R52, and S19-K59.
and (b)), which differ from that in the DCoH This limitation is due to a combination of factors.
homotetramer.23 (i) Rigid-body modelling neglects local induced ®t
The above features are retained on docking of or plasticity of side-chains in either protein. DCoH
the present crystal structure. Signi®cantly, the residues N44, R45, R52, E58, and K59, each
improved resolution of helical phases makes poss- involved in interactions with HNF-p1, assume
ible retrodiction of possible side-chain contacts. different side-chain conformations in the peptide-
Fortuitously, the length of a-helix 1 in HNF-p1w is DCoH complex35 relative to the DCoH homo-
the same as those of contact helices in the DCoH tetramer.23 Relocation of the DCoH side-chain of
4HB (15 residues). Formation of the HNF/DCoH E58, for example, is necessary to relieve a potential
4HB is predicted to bury a total surface area of steric clash with HNF-p1 residue Q7. (ii) Whereas
1820 A Ê 2. These values are similar to those observed differences in interhelical angles across the inter-
in the co-crystal structure, wherein a total surface molecular 4HB yield negligible errors at the center
area of 1960 A Ê 2 is buried.35 The rmsd of super- of symmetry (i.e. near L12 and L120 ), progressively
posed C atoms of helix-1 and helix-10 in HNF-p1w
a
larger errors occur farther from the center.
and equivalent helices in the DCoH tetramer is These comparisons suggest that in the absence of
1.1 AÊ . Symmetry-based docking of the present signi®cant conformational changes, symmetry-
crystal structure thus yields the correct overall fea- based modelling provides a robust method for pre-
tures of the complex: N-terminal helices (1 and 10 ) diction of the overall features of a complex and
are ``sandwiched'' between C-terminal helices (2 some of its key contacts. Rigid-body docking is
and 20 ) of the HNF-p1w dimer and a pair of DCoH insuf®cient, however, to predict all the details of
helices atop the saddle resulting in a six-helix the contact surfaces. Predictions may be improved
bundle. by explicit consideration of side-chain electrostatic
An alternative possible mode of recognition and functionalities. Inspection of electrostatic sur-
employing HNF-1a helices 2 and 20 was evaluated faces in the present case strongly suggests the
as a control. An intermolecular 4HB may be possibility of the observed ionic interactions (such
obtained readily by inversion of the HNF-1a as E11-R45 and E18-R52)35 with modest local
domain atop the saddle. Such an ``upside-down'' changes in side-chain con®guration.
model in fact yields helical orientations whose
symmetry is similar to that of the DCoH homote-
Comparison of HNF-1a
a and HNF-1b
b
tramer. Because a-helices 2 and 20 are shorter than
a-helices 1 and 10 (eight versus 15 residues), how- HNF-1a and HNF-1b belong to a family of
ever, this alternative model exhibits a 32 % homo- and heterodimeric transcription factors reg-
reduction in buried surface area (to 1135 A Ê 2) In ulating expression of many genes.15,22,62 Each binds
addition, the side-chains exposed on the surface of to speci®c DNA sites as a dimer and forms hetero-
helices 2 and 20 are less compatible (with respect to dimers in vitro and in vivo. Alignment of HNF-1a
polarity and charge) with the juxtaposed surface of and HNF-1b sequences is shown in Figure 3(a).
the DCoH saddle than are the side-chains at the Diabetes-related mutational sites L12 and G20 are
surface of the mini-zipper. Helices 1 and 10 in the invariant (highlighted in red). In HNF-1b positions
present crystal structure are longer (by two resi- 21 and 25 contain valine residues in place of L21
dues) than in the peptide-DCoH complex, wherein and A25 in HNF-1a domain (brackets, lower panel
residues 1-6 assume a different conformation. In of Figure 3). The paired valine residues in HNF-1b
the model, residues 1-5 of HNF-p1w exhibit a ster- presumably function as compensating substitutions
ic clash with DCoH. Because these residues are to ®ll the volume occupied in the present structure
¯exible in solution, the potential clash could be by adjoining L21-A25 side-chains. Molecular mod-
avoided readily. elling of L21G (red) and L25G (pink) creates a cre-
vice, as depicted on the left side of Figure 3(b). In
Retrodicted side-chain environments the crystal structure, this crevice is ef®ciently ®lled
by the close-packing of side-chains L21 and A25
The side-chains on the mini-zipper's surface (right side of Figure 3(b)). Given the conservation
nearest to the HNF-1a dimer axis are L12 and L120 , of the remaining residues lining the HNF-1b's
a site of diabetes-related mutation. Alignment of putative DCoH-binding surface, we expect that an
HNF-1a- and DCoH symmetry axes automatically homologous complex would indeed be formed. In
positions the methyl groups of L12 and L120 within HNF-1b, position 18 has a serine residue in place
a hydrophobic pocket formed by DCoH residues of the glutamic acid residue in HNF-1a (asterisk in
652 An Intertwined Four-helix Bundle in HNF-1

Figure 3(a), lower panel). E18 is not involved in similarities in chemical shifts and NOEs suggest
interactions between protomers, suggesting that that a native-like fold is maintained. To explore
this subsitution is neutral with respect to dimeriza- possible structural consequences of the L12H substi-
tion. However, because the E18 side-chain carboxy- tution and their implications for DCoH recognition,
late group forms an ionic interaction with R52 of an homology model was constructed based on the
DCoH,35 it is possible that the presence of S18 in native crystal structure. The modelling procedure
HNF-1b would weaken binding to DCoH. To our employed the distance-geometry protocol of Havel
knowledge, binding of HNF-1b or the HNF-1a/ & Snow,64 wherein NOEs were simulated based on
HNF-1b heterodimer to DCoH has not been the crystal structure (see Materials and Methods).
demonstrated. (Restraint information and statistical parameters are
provided as Supplementary Material.) A consistent
DG/SA model was obtained, suggesting that the
Environment of clinical mutation sites
adjoining H12 side-chains are accommodated
I27L polymorphism readily in the mini-zipper interface with only local
conformational adjustments. The overall fold and
The substitution I27L (highlighted in green in structure of the variant mini-zipper are shown in
Figure 3(a), upper panel) is a common polymorph- Figure 10(c) and (d). Although L12 has been
ism in diabetic and non-diabetic populations unas- described as a core side-chain, the methyl reson-
sociated with MODY.2 The I27 side-chain is ances in fact contribute to the ¯at hydrophobic sur-
exposed on the surface of helix 2; its interactions face of the native dimer (Figure 10(a) and (b)). The
differ in detail between the present structure and L12H model predicts a striking remodelling of this
the peptide-DCoH co-crystal structure (see above). surface in which the contiguous imidazole rings
Surprisingly, this polymorphism has been shown give rise to a weakly polar protuberance
recently to be an independent correlative determi- (Figure 10(e)). Exposure of the titratable Ne edge of
nant of insulin sensitivity due to hepatic or periph- the imidazole ring to solvent is supported by the
eral insulin-resistance.63 It is not known whether robustness of the fold to low pH.
the mutation alters the regulation of glucose
metabolism directly or exists in linkage disequili-
G20R MODY mutation
brium with another polymorphic gene regulating
metabolism. G20 lies in a non-canonical turn (Figure 1(b))
and appears to direct the relative orientations of
L12H MODY mutation helices in the peptide's ``active'' conformation. Its
main-chain atoms are hyperexposed in both an
L12 is involved in van der Waal's interactions extracted protomer and actual dimer with a frac-
with symmetry-related L120 and L160 side-chains tional exposed surface of 0.64. Although the pat-
and projects to the surface of the mini-zipper tern of torsion angles in the turn differs from those
(Figures 2 and 10). The side-chain thus contributes of classical turns, a similar pattern is observed in
to both the stability and functional surface of the helix-turn-helix motif of the arc repressor (PDB
dimer. The side-chain is partially buried upon code 1BDT).65 The structures are otherwise unre-
dimerization; in the extracted monomer, the frac- lated. MODY-associated substitution G20R is
tional exposed surface is 0.55, whereas in the associated with a profound thermodynamic desta-
dimer it is 0.26 (a complete set of residue-speci®c bilization (Table 2; and see Hua et al.34) and loss of
accessibilities is provided in the Supplementry architectural speci®city. Although the variant
Material). L12 is invariant among mammalian dimer retains near-native helix content as inferred
HNF-1a and HNF-1b sequences (Figure 3(a)). Mol- from CD studies, a combination of features (non-
ecular modelling suggests that substitutions L12G two-state unfolding, binding of ANS, loss of
or L12A would result in signi®cant packing defects chemical-shift dispersion with conformational
(cavities of respective volumes 20 and 15 A Ê 3). broadening) indicate that the variant dimer forms
MODY-associated mutation L12H effects a signi®- a molten globule. We speculate that local frustra-
cant reduction in thermodynamic stability (G tion of the sharp turn precludes ef®cient packing
value 3.7(0.3) kcal/mol; Table 2)34 and accord- of the C-terminal coiled-coil against the mini-zip-
ingly weakens dimerization. L12 makes buried per. A possible mechanism of frustration is
contacts with dimer-related residues of HNF-p1 suggested by the crystal structure: given the pre-
and the DCoH dimer in peptide-DCoH complex.35 sent backbone torsion angles at residues 19 and 20,
Consistent with this location, L12H disrupts bind- any L-amino acid substitution at position 20 would
ing of HNF-1a to DNA and DCoH.35 Overexpres- not be sterically favorable. The distance between
sion of the variant intact protein restores activation the carbonyl oxygen atom of residue 19 (ith pos-
of a reporter gene to native levels.8 ition) and the Cb of residue 20 (position i ‡ 1)
Crystals of an L12H analogue of HNF-p1w could would be less than 2.4 A Ê , which predicts a steric
not be obtained; however, the analogue's NMR clash; by contrast D-amino acids should be accom-
spectrum is nonetheless similar to that of the native modated readily. The proposed mechanism pre-
dimer. Although isotope-assisted NMR studies of dicts that the G20R perturbation is not side-chain
the L12H analogue were not performed, overall speci®c; i.e. G20A and other analogues would exhi-
An Intertwined Four-helix Bundle in HNF-1 653

Figure 10. Surface representation of HNF-p1w dimer and L12H homology model. (a) Electrostatic potential of the
DCoH-binding surface of HNF-p1w (stereo pair). The coloring code is red, <ÿ10 kT/e; white, ÿ10kT/e to 10kT/e;
and blue, >10kT/e. Residues E11 and E18 are involved in ion-pair interactions with DCoH residues.35 The exposed
concave surface is complementary in shape to the top of the DCoH saddle as predicted by model building studies
(see the text) and observed in the recent co-crystal structure.35 (b) The DCoH-binding surface of HNF-p1w dimer has
a leucine-rich stripe (leucine residues 5, 8, 12, 13, 16 and 21, and their symmetry-related residues) shown in green.
The orientation of molecular surface (white) of the HNF-p1w dimer is same as in (a). L12 is located in the middle of
the leucine-rich stripe. Residues 1-4 and 10 -40 have been omitted for clarity. (c)-(e) Homology model of the L12H var-
iant dimer. (c) The variant side-chain is compatible with a native overall main-chain fold. One protomer is shown in
cyan and the other in red; terminal residues 1-4 and 10 -40 are disordered in accord with NMR evidence of ¯exibility.
(d) The structure of the mini-zipper is predicted to accommodate a pair of histidine side-chains (yellow) with
exposure of polar Ne ring protons. Neighboring side-chains are in positions similar to those in crystal structure.
(e) The surface of the variant mini-zipper is predicted to contain a weakly polar protuberance (yellow) due to the
edges of the two His side-chains. This change in surface character would be expected to perturb DCoH binding. (a),
(b) and (e) Generated using the program GRASP;85 (c) and (d) (and other Figures) were made using InsightII (MSI,
Inc., San Diego).

bit similar instabilities. It is unclear why the puta- The structure consists of a novel intertwined four-
tive steric clash cannot be relieved by local confor- helix bundle with non-canonical turn. Multiple
mational adjustments to give an altered but structural comparisons suggest strongly that the
architecturally speci®c structure. domain consists of a rigid element, an N-terminal
anti-parallel mini-zipper, and a plastic element, the
C-terminal coiled-coil. Two types of plasticity are
Concluding remarks
observed. (i) The immediate N and C-terminal seg-
The dimerization domain of HNF-1a provides ments can be a-helical or disordered depending on
an opportunity to compare structures at high resol- packing environment. Corresponding ¯exibility in
ution in multiple crystallographic environments. solution has been demonstrated by 1H-NMR
654 An Intertwined Four-helix Bundle in HNF-1

motional narrowing, chemical-shift patterns, of interactions as part of a structural switch


selected main-chain coupling constants and, in the between functional states. An example of such a
case of W33, multiple inter-residue NOEs inconsist- dynamic switch has been characterized recently in
ent with a single unqiue structure. (ii) Plasticity a cytoplasmic signalling pathway.73
exists at the interface between the mini-zipper and
the overlying C-terminal a-helices leading to differ-
ent patterns of side-chain packing. Although the
Materials and Methods
non-canonical turn and intertwined topology are Peptide synthesis
maintained consistently, striking differences occur
in possible inter-helical angles. The collection of The HNF-1a domain with MSe substitution at position
HNF-p1 crystal structures is reminiscent of insulin, 13 was prepared by solid-phase synthesis (Gryphon
whose many high-resolution crystal structures like- Sciences, Inc., South San Fransisco, CA). Sample purity
was greater than 98 % as estimated by analytical reverse-
wise exhibit a combination of invariant and plastic phase HPLC. Fidelity of synthesis was veri®ed by
features.66,67 The most divergent HNF-1a structure ion-spray mass spectrometery. The dimeric status of
is that observed in a speci®c complex with DCoH. the peptide was veri®ed by analytical gel-®ltration
Because recognition is mediated by the invariant chromatography.
mini-zipper and the molten C-terminal coiled-coil,
such induced ®t appears to involve a transmission Crystallization and X-ray data collection
of conformation change. Transmission of confor-
mational change among crystal structures of insu- The peptide was crystallized by the hanging drop/
lin has been highlighted by the late D. C. Hodgkin vapor diffusion method. The peptide was made 3 mM in
and colleagues68 and, as in the HNF-1a/DCoH 50 mM Tris-HCl (pH 7.0), 2 mM DTT. Crystals were
complex, may facilitate receptor binding.69 grown at 4  C by mixing equal volumes of peptide sol-
The present study suggests new directions for ution (3 ml) and reservoir solution (3 ml) containing 20 %
future study. First, given the collection of equival- (v/v) methyl-2,4-pentanediol (MPD), 50 mM Tris-HCl
(pH 7.5), and 1 mM DTT. Crystals mounted on nylon
ent crystal structures, which subset of confor- loops were transferred to stabilization buffer (100 mM
mations best represents the structure in solution? Tris-HCl (pH 7.5), 30 % MPD or 30 % PEG400) and ¯ash-
Although the present isotope-edited studies verify frozen in liquid nitrogen before data collection at 100 K.
the overall intertwined topology and phase of the X-ray data were collected at beamline 19-ID at the
mini-zipper, more detailed heteronuclear NMR stu- Advanced Photon Source (APS), Argonne National Lab-
dies of structure and dynamics will be required to oratory. Multiple anomalous dispersion (MAD) data sets
assess the inter-helical packing scheme(s) and esti- were collected at three wavelengths: remote (lr), peak
mate the range of possible conformational change. (lp), and at the inversion point (li) of selenium atom
Laboratory-frame approaches based on residual (Table 1). The crystals belong to orthorhombic space
group P21212 with a ˆ 28.42 A Ê , b ˆ 42.19 A Ê , and
dipolar couplings are likely to be informative. Ê . There are two molecules in the asymmetric
c ˆ 42.43 A
Second, what is the role of the non-canonical turn unit with a solvent content of 23 % (v/v). Raw data were
in enabling a speci®c architecture to be obtained? processed and scaled using the HKL2000 package.74
The role of turns in general in 4HBs has been the Data collection statistics are presented in Table 1.
subject of debate.70,71 We have suggested a speci®c
structural mechanism for the invariance of glycine
Structure determination and refinement
(G20) and predict that any L-amino acid (but not
D-amino acids) at position 20 would block speci®c The two selenium positions corresponding to two
folding to create a novel molten globule. If so, the independent molecules (molecules 1 and 2) in the asym-
HNF-1a motif would provide an example of an metric unit were determined by computing anomalous
energy landscape72 whose shape and depth and dispersive- difference Patterson maps using program
depend on a particular turn orientation. Finally, CNS.75 Heavy-atom positions were re®ned against MAD
what is the biological signi®cance of the domain's data with remote wavelength as reference using data
between 40 and 3.0 A Ê resolution. Further density modi®-
asymmetric plasticity? A possible clue is provided Ê resolution using
cation and phase extension to 2.5 A
by the unexplained G31D diabetes-associated CNS yielded a high-quality MAD-phased electron den-
mutation. Unlike L12H and G20R, the G31D var- sity map. It was possible to trace the two helices in mol-
iant would seem not to affect the domain's struc- ecule 1 and one helix in molecule 2. A polyalanine
ture, stability or interaction with DCoH.35 An model was constructed for molecule 1 and for one helix
intriguing possibility is that in vivo HNF-1a inter- in molecule 2 using program O76 and re®ned using data
acts with DCoH and with other transcriptional co- collected at remote wavelength (lr). Structure re®nement
activators or co-repressors and that such (to date) was done using CNS. Overall anisotropic scale factors
uncharacterized interactions are disrupted by the and a bulk solvent corrections were applied and a maxi-
G31D mutation. This possibility in turn suggests a mum likelihood amplitude-based target function were
employed.77 Simulated-annealing (SA) torsion-angle
structural model in which the plastic portion of the
re®nement78 and individual B-factor re®nement was per-
HNF-1a domain adopts one orientation on binding formed as implemented in CNS. Several rounds of alter-
to DCoH and another conformation on binding to nate SA re®nement and model rebuilding were
a putative second regulatory cofactor. Plasticity in performed with gradual increments in the data resol-
the packing of the mini-zipper and C-terminal ution. Each residue in molecule 1 was seen in the elec-
a-helices may thus extend the domain's repertoire tron density map, whereas helix 2 in molecule 2 was
An Intertwined Four-helix Bundle in HNF-1 655

poorly de®ned and built residue-by-residue in iterative obtained for the equilibrium mixture: 13C-1H HSQC,
13
re®nement cycles. Water molecules that are at a potential C-1H 2D HSQC-TOCSY (mixing time of 45 ms), and
13
hydrogen bonding distance from the protein or another C-1H intermolecular 2D NOESY.83
water molecule were added in ®nal rounds of re®ne-
ment. SA omit maps were computed by omitting two Circular dichroism
residues at a time for both molecules 1 and 2, and the
model rebuilt. In molecule 2 the ®rst two N-terminal Spectra were obtained using an Aviv spectropolari-
residues and the last three C-terminal residues are not meter equipped with thermister temperature control.
seen in the map and are considered disordered. The ®nal Samples were placed in a 1 mm path-length quartz
model consists of molecule 1 (33 residues), molecule 2 cuvette. Peptide concentrations in the presence of MPD
(28 residues), and 55 water molecules. The structure was were corrected for volume change of solvent miscibility.
re®ned to a ®nal crystallographic R of 23.1 % using F > 3 The buffer solutions corresponded to crystallization
sF between 40 and 1.7 A Ê resolution. The Rfree for the conditions.
re®ned model was 24.5 %. Some of the ®nal re®nement
statistics are given in Table 1. Average B-factors for mol-
ecule 1, molecule 2, and water molecules are 15.9 A Ê 2, L12H homology modelling
23.0 AÊ 2, and 28.1 A Ê 2, respectively. The ®nal structure
A molecular model of the L12H variant dimer was
was validated using PROCHECK.79 constructed using the DG/SA protocol as described:64
1212 homology distance restraints were imposed, sup-
HNF-DCoH model building plemented by one key H12-speci®c NOE (H12-L160 , pre-
sumed to be inter-protomeric). In brief, inter-proton
The crystal structure of HNF-p1w was used to dock distances were inferred from the crystal structure (using
against the crystal structure of the DCoH dimer (sub- the HBUILD facility in InsightII (Molecular Simulations,
units A and B of PDB code 1DCO) using the InsightII Inc.)) for residues 5-33, excepting the side-chain of resi-
molecular graphics package (MSI, Inc.). The 2-fold axis due 12; no restraint was included for residues 1-4 or 10 -40
of HNF-p1w was aligned on the 2-fold axis of DCoH, in light of NMR evidence of ¯exibility. Distance bounds
superposing dimer-related anti-parallel helices of HNF- were assigned to conventional NMR ranges (i.e. strong
p1 and the equivalent helices (without considering the (<2.7 AÊ ), medium (<3.4 A Ê ) or weak (<5 A Ê )) based on
helix direction) in the DCoH tetramer (subunits C and inferred values in the crystal structure. a-Helix-related
D). Superposition employed sheer rotation about the hydrogen bonds were imposed by pairs of distance
aligned axis (Figure 9(a)). This procedure results in a restraints (between the carbonyl oxygen atom of residue
model for the HNF-p1w/DCoH complex with a 4HB i and the amide proton of residue i ‡ 4 (2.3-2.6 A Ê ) and
interface. The model predicts different relative helical between the carbonyl oxygen atom of residue i and the
orientations for this 4HB compared to the DCoH tetra- amide nitrogen atom of residue i ‡ 4 (2.7-3.3 A Ê )) for resi-
mer: the direction of the helices in HNF-p1w dimer are dues 10-19 and 26-30. Dihedral restraints on main-chain
reversed compared to the helices in DCoH tetramer f values were assumed for helical residues (residues 6-
(Figure 9(b)) in accord with modelling studies by Hua 19 and 22-30; f ˆ ÿ 65(40)  ); both f and c values
et al.34 and the co-crystal structure.35 Cavity volumes were imposed for G20 and G200 (f ˆ 70(40)  and
were computed using the program surfnet.80 c ˆ ÿ 150(40)  ); and side-chain w1 and w2 dihedral
angles were imposed for residues 12 and 13 (40  of the
value in the present crystal structure). The main-chain
Expression and labeling of HNF-1a
a peptide rmsd between the present structure and the L12H model
Ê . No signi®cant restraint
(residues 5-28 and 50 -280 ) is 1.1 A
A DNA segment encoding HNF-p1w was prepared
violation is observed in the ®nal homology ensemble
by polymerase chain reaction (PCR) using the human
(see the Supplementary Material, where a summary of
HNF-1a cDNA as template. The PCR product was
restraint information is provided).
cloned into BamHI and EcoRI sites of plasmid pMW1
previously employed to express phage l N peptides.81
The HNF-1a peptide was expressed as a thrombin- Atomic coordinates
cleavable fusion protein with staphylococcal nuclease
as described.82 The expressed protein contained an Atomic coordinates (1JB6) and X-ray data (Y1JB6Sp)
N-terminal hexa-His tag to enable af®nity puri®cation. have been deposited with the RCSB Protein Data Bank..
Biosynthetic 13C labeling was accomplished in an overex-
pressing strain of Escherichia coli by growth in M9 mini-
mal medium containing [13C]glucose as sole carbon
source. The biosynthetic peptide contained 36 residues,
including three non-native N-terminal residues (GSE); Acknowledgments
the C terminus was not amidated.
We thank M. Shoham for allowing us to use his beam
time to perform our MAD experiment at the Advanced
Isotope-assisted NMR studies Photon Source (APS). Use of the Argonne National Lab-
oratory structural biology center beam line 19-ID at the
The following spectra were obtained for the 13C- APS is supported by the US Department of Energy,
labeled peptide homodimer: 13C-1H HSQC, 13C-1H 2D Of®ce of Biological and Environmental Research under
HSQC-TOCSY (mixing time of 45 ms), 13C-1H 3D contract W-31-109-ENG-38. We thank V. Srajer at the
HCCH-TOCSY, 13C-1H 3D NOESY-HSQC (mixing time BIOCARS beam line 14-bm-D at the APS for the initial
of 250 ms). Complete assignments were obtained. To stages of this study; S. Soleiman for help in using O pro-
evaluate intermolecular NOEs, an equimolar mixture of gram on their workstation; N. Phillips and M. Hake for
the synthetic peptide HNF-p1w and the labeled biosyn- gel-®ltration experiments; A. Stern for ring-current shift
thetic peptide was prepared. The following spectra were calculations; S. Korolev and R. Immormino for help in
656 An Intertwined Four-helix Bundle in HNF-1

data collection at APS; W. Jia for CD experiments and caused by an insulin secretion defect. J. Clin. Invest.
related Figures; and S. Nakagawa (The University of 99, 582-591.
Chicago) and J. Wilken (Gryphon Sciences, Inc.) for 12. Frain, M., Swart, G., Monaci, P., Nicosia, A.,
peptide synthesis. M.A.W. thanks the faculty of the Cold Stamp¯i, S., Frank, R. & Cortese, R. (1989). The
Spring Harbor course on macromolecular crystallogra- liver-speci®c transcription factor LFB1 contains a
phy for their encouragement. This work was supported, highly diverged homeobox DNA binding domain.
in part, by the Diabetes Research & Training Center at Cell, 59, 145-157.
The University of Chicago and by a grant from the NIH 13. Mendel, D. B. & Crabtree, G. R. (1991). HNF-1, a
to M.A.W. member of a novel class of dimerizing homeodo-
main proteins. J. Biol Chem. 266, 677-680.
14. Tomei, L., Cortese, R. & De Francesco, R. (1992).
A POU-A related region dictates DNA binding
References speci®city of LFB1/HNF1 by orienting the two
1. Velho, G. & Froguet, P. (1997). Maturity-onset XL-homeodomains in the dimer. EMBO J. 11, 4119-
diabetes of the young (MODY), MODY genes and 4129.
non-insulin-dependent diabetes mellitus. Diabetes 15. Tronche, F. & Yaniv, M. (1992). HNF-1, a homeodo-
Metab. 23, 34-37. main member of the hepatic transcription regulatory
2. Yamagata, K., Oda, K., Kaisaki, P. J., Menzel, S., network. BioEssays, 14, 579-587.
Furuta, H., Vaxillaire, M. et al. (1996). Mutations in 16. Iwasaki, N., Oda, N., Ogata, M., Hara, M., Hinokio,
the hepatocyte nuclear factor-1a gene in MODY-3. Y., Oda, Y. et al. (1997). Mutations in the hepatocyte
Nature, 384, 455-458. nuclear factor- lalpha/MODY3 gene in Japanese
3. Glucksmann, A., Lehto, M., Tayber, O., Scotti, S., subjects with early- and late-onset NIDDM. Diabetes,
Berkemeier, L., Pulido, J. et al. (1997). Novel 46, 1504-1508.
mutations and a mutational hotspot in the MODY3 17. Yamada, S., Nishigori, H., Onda, H., Utsugi, T.,
gene. Diabetes, 46, 1081-1086. Yanagawa, T., Maruyama, T. et al. (1997). Identi®-
4. Hansen, T., Eiberg, H., Rouard, M., Vaxillaire, M., cation of mutations in the hepatocyte nuclear factor
Moller, A. M., Rasmussen, S. K. et al. (1997). Novel (HNF)-la gene in Japanese subjects with IDDM. Dia-
MODY3 mutations in the hepatocyte nuclear factor- betes, 46, 1643-1647.
1alpha gene: evidence for a hyperexcitability of pan- 18. Nicosia, A., Monaci, P., Tomei, L., De Francesco, R.,
creatic beta-cells to intravenous secretagogues in a Nuzzo, M., Stunnenberg, H. & Cortese, R. (1990). A
glucose-tolerant carrier of a P447L mutation. Dia- myosin-like dimerization helix and an extra-large
betes, 46, 726-730. homeodomain are essential elements of the tripartite
5. Vaxillaire, M., Rouard, M., Yamagata, K., Oda, N., DNA binding structure of LFB1. Cell, 61, 1225-1236.
Kaisaki, P. J., Boriraj, V. V. et al. (1997). Identi®- 19. Chouard, T., Blumenfeld, M., Bach, I.,
cation of nine novel mutations in the hepatocyte Vandekerckhove, J., Cereghini, S. & Yaniv, M.
nuclear factor 1-a gene associated with maturity- (1990). A distal dimerization domain is essential for
onset diabetes of the young (MODY3). Hum. Mol. DNA-binding by the atypical HNF1 homeodomain.
Genet. 6, 583-586. Nucl. Acids Res. 18, 5853-5863.
6. Elbein, S. C., Teng, K., Yount, P. & Scroggin, E. 20. De Francesco, R., Pastore, A., Vecchio, G. & Cortese,
(1998). Linkage and molecular scanning analyses of R. (1991). Circular dichroism study on the confor-
MODY3/hepatocyte nuclear factor-1a gene in typi- mational stability of the dimerization domain of
cal familial type 2 diabetes: evidence for novel transcription factor: LFB1. Biochemistry, 30, 143-147.
mutations in exons 8 and 10. J. Clin. Endocrinol. 21. Pastore, A., De Francesco, R., Barbato, G.,
Metab. 83, 2059-2065. Castiglione, Morelli M. A., Motta, A. & Cortese, R.
7. Yamagata, K., Yang, Q., Yamamoto, K., lwahashi, (1991). 1H resonance assignment and secondary
H., Miyagawa, J., Okita, K. et al. (1998). Mutation structure determination of the dimerization domain
P291fsinsC in the transcription factor hepatocyte of transcription factor LFB1. Biochemistry, 30, 148-
nuclear factor-1a is dominant negative. Diabetes, 47, 153.
1231-1235. 22. Hansen, L. P. & Crabtree, G. R. (1993). Regulation of
8. Yamada, S., Tomura, H., Nishigori, H., Sho, K., the HNF-1 homeodomain proteins by DCoH. Curr.
Mabe, H., Iwatani, N. et al. (1999). Identi®cation of Opin. Genet. Dev. 3, 246-253.
mutations in the hepatocyte nuclear factor-1alpha 23. Endrizzi, J. A., Cronk, J. D., Wang, W., Crabtree,
gene in Japanese subjects with early-onset NIDDM G. R. & Alber, T. (1995). Crystal structure of DCOH,
and functional analysis of the mutant proteins. Dia- a bifunctional protein-binding transcriptional coacti-
betes, 48, 645-648. vator. Science, 268, 556-559.
9. Pontoglio, M., Barra, J., Hadchouel, M., Doyen, A., 24. Ficner, R., Sauer, U. H., Stier, G. & Suck, D. (1995).
Kress, C., Bach, J. P. et al. (1996). Hepatocyte nuclear Three-dimensional structure of the bifunctional pro-
factor 1 inactivation results in hepatic dysfunction, tein PCD/DCoH, a cytoplasmic enzyme interacting
phenylketonuria, and renal fanconi syndrome. Cell, with transcription factor HNF1. EMBO J. 14, 2034-
84, 575-585. 2042.
10. Byrne, M. M., Sturis, J., Menzel, S., Yamagata, K., 25. Cronk, J. D., Endrizzi, J. A. & Alber, T. (1996). High-
Fajans, S. S., Drons®eld, M. J. et al. (1996). Altered resolution structures of the bifunctional enzyme and
insulin secretary responses to glucose in diabetic transcriptional coactivator DCOH and its complex
and nondiabetic subjects with mutations in the dia- with a product analogue. Protein Sci. 5, 1963-1972.
betes susceptibility gene MODY3 on chromosome 26. Suck, D. & Ficner, R. (1996). Structure and function
12. Diabetes, 45, 1503-1510. of PCD/DCoH, an enzyme with regulatory proper-
11. Lehto, M., Tuomi, T., Mahtani, M. M., Widen, E., ties. FEBS Letters, 389, 35-39.
Forsblom, C., Sarelin, L. et al. (1997). Characteriz- 27. Rhee, K. H., Stier, G., Becker, P. B., Suck, D. &
ation of the MODY3 phenotype. Early-onset diabetes Sandaltzopoulos, R. (1997). The bifunctional protein
An Intertwined Four-helix Bundle in HNF-1 657

DCoH modulates interactions of the homeodomain 44. O'Shea, E. K., Klemm, J. D., Kim, P. S. & Alber, T.
transcription factor HNF1 with nucleic acids. J. Mol. (1991). X-ray structure of the GCN4 leucine zipper,
Biol. 265, 20-29. a two-stranded, parallel coiled-coil. Science, 254, 539-
28. Sourdive, D. J., Transy, C., Garbay, S. & Yaniv, M. 544.
(1997). The bifunctional DCOH protein binds to 45. Crick, F. H. C. (1953). The packing of a-helices:
HNF1 independently of its 4-alpha-carbinolamine simple coiled-coils. Acta Crystallog. 6, 689-697.
dehydratase activity. Nucl. Acids Res. 25, 1476-1484. 46. Lupas, A. (1996). Coiled coils: new structures and
29. Johnen, G. & Kaufman, S. (1997). Studies on the new functions. Trends Biochem. Sci. 21, 375-382.
enzymatic and transcriptional activity of the dimeri- 47. Wintjens, R. T., Rooman, M. J. & Wodak, S. J.
zation cofactor for hepatocyte nuclear factor 1. Proc. (1996). Automatic classi®cation and analysis of
Natl Acad. Sci. USA, 94, 13469-13474. alpha alpha-turn motifs in proteins. J. Mol. Biol. 255,
30. Mendel, D. B., Khavari, P. A., Conley, P. B., Graves, 235-253.
M. K., Hansen, L. P., Admon, A. & Crabtree, G. R. 48. Wishart, D. S. & Sykes, B. D. (1994). Chemical shifts
(1991). Characterization of a cofactor that regulates as a tool for structure determination. Methods Enzy-
dimerization of a mammalian homeodomain pro- mol. 239, 363-392.
tein. Science, 254, 1762-1767. 49. Vuister, G. W. & Ad, Bax. (1993). Quantitative J
31. Horikawa, Y., Iwasaki, N., Hara, M., Furuta, H., correlation: a new approach for measuring homo-
Hinokio, Y., Cockburn, B. N. et al. (1997). Mutation nuclear three-bond J(HNHa) coupling constants in
in hepatocyte nuclear factor-1 beta gene (TCF2) 15
N-enriched proteins. J. Am. Chem. Soc. 115, 7772-
associated with MODY. Nature Genet. 17, 384-385. 7777.
32. Bach, I. & Yaniv, M. (1993). More potent transcrip- 50. Grzesiek, S., Kuboniwa, H., Hinck, A. P. & Bax, A.
tional activators or a transdominant inhibitor of the (1995). Multiple-quantum line narrowing for
HNF1 homeoprotein family are generated by measurement of Ha-Hb J couplings in isotopically
alternative RNA processing. EMBO J. 12, 4229-4242. enriched proteins. J. Am. Chem. Soc. 117, 5312-5315.
33. Pastore, A., De Francesco, R., Castiglione, Morelli 51. Wuthrich, K. (1976). NMR in Biological Research:
M. A., Nalis, D. & Cortese, R. (1992). The dimeriza- Peptides and Proteins, American Elsevier Publishing
tion domain of LFB1/HNF1 related transcription Company, Inc., New York.
factors: a hidden four helix bundle? Protein Eng. 5, 52. Weber, P. C. & Salemme, F. R. (1980). Structural and
749-757. functional diversity in 4-alpha-helical proteins.
34. Hua, Q. X., Zhao, M., Narayana, N., Nakagawa, Nature, 287, 82-84.
S. H., Jia, W. & Weiss, M. A. (2000). Diabetes- 53. Kamtekar, S. & Hecht, M. H. (1995). Protein motifs.
associated mutations in a b-cell transcription factor
7. The four-helix bundle: what determines a fold?
destabilize an anti-parallel ``mini-zipper'' in a dimer-
FASEB J. 9, 1013-1022.
ization interface. Proc. Natl Acad. Sci. USA, 97, 1999-
54. Osterhout, J. J., Handel, T., Na, G., Toumadje, A.,
2004.
Long, R. C., Connolly, P. J. et al. (1992). Characteriz-
35. Rose, R. B., Bayle, J. H., Endrizzi, J. A., Cronk, J. D.,
ation of the structural properties of a1B, a peptide
Crabtree, G. R. & Alber, T. (2000). Structural basis of
designed to form a four-helix bundle. J. Am. Chem.
dimerization, coactivator recognition and MODY3
Soc. 114, 331-337.
mutations in HNF-1a. Nature Struct. Biol. 7, 744-748.
36. Bennett, M. J., Choe, S. & Eisenberg, D. (1994). 55. DeGrado, W. F., Summa, C. M., Pavone, V., Nastri,
Domain swapping: entangling alliances between F. & Lombardi, A. (1999). De novo design and struc-
proteins. Proc. Natl Acad. Sci. USA, 91, 3127-3131. tural characterization of proteins and metallopro-
37. Schlunegger, M. P., Bennett, M. J. & Eisenberg, D. teins. Annu. Rev. Biochem. 68, 779-819.
(1997). Oligomer formation by 3D domain swap- 56. Ferre-D'Amare, A. R., Prendergast, G. C., Ziff, E. B.
ping: a model for protein assembly and misassem- & Burley, S. K. (1993). Recognition by Max of its
bly. Advan. Protein Chem. 50, 61-122. cognate DNA through a dimeric b/HLH/Z domain.
38. Pachter, R., Arrowsmith, C. H. & Jardetzky, O. Nature, 363, 38-45.
(1992). The effect of selective deuteration on magne- 57. Ellenberger, T., Fass, D., Arnaud, M. & Harrison,
tization transfer in larger proteins. J. Biomol. NMR, S. C. (1994). Crystal structure of transcription factor
2, 183-194. E47: E-box recognition by a basic region helix-loop-
39. Rose, R. B., Endrizzi, J. A., Cronk, J. D., Holton, J. & helix dimer. Genes Dev. 8, 970-980.
Alber, T. (2000). High-resolution structure of the 58. Ma, P. C., Rould, M. A., Weintraub, H. & Pabo,
HNF-1a dimerization domain. Biochemistry, 39, C. O. (1994). Crystal structure of MyoD bHLH
15062-15070. domain-DNA complex: perspectives on DNA recog-
40. Chou, P. Y. & Fasman, G. D. (1977). b-Turns in pro- nition and implications for transcriptional activation.
teins. J. Mol. Biol. 115, 135-175. Cell, 77, 451-459.
41. Rose, G. D., Gierasch, L. M. & Smith, J. A. (1985). 59. Okita, K., Yang, Q., Yamagata, K., Hangenfeldt,
Turns in peptides and proteins. Advan. Protein Chem. K. A., Miyagawa, J., Kajimoto, Y. et al. (1999).
37, 1-109. Human insulin gene is a target gene of hepatocyte
42. Ng, M. C., Cockburn, B. N., Lindner, T. H., Yeung, nuclear factor-1alpha (HNF-1alpha) and HNF-1beta.
V. T., Chow, C. C., So, W. Y. et al. (1999). Molecular Biochem. Biophys. Res. Commun. 263, 566-569.
genetics of diabetes mellitus in Chinese subjects: 60. Ellard, S. (2000). Hepatocyte nuclear factor 1 alpha
identi®cation of mutations in glucokinase and hep- (HNF-1alpha) mutations in maturity-onset diabetes
atocyte nuclear factor-1alpha genes in patients with of the young. Hum. Mutat. 16, 377-385.
early-onset type 2 diabetes mellitus/MODY. Diabet. 61. Ptitsyn, O. (1996). How molten is the molten
Med. 16, 956-963. globule? Nature Struct. Biol. 3, 488-490.
43. Banner, D. W., Kokkinidis, M. & Tsernoglou, D. 62. De Simone, V. & Cortese, R. (1991). Transcriptional
(1987). Structure of the ColE1 rop protein at 1.7 A Ê regulation of liver-speci®c gene expression. Curr.
resolution. J. Mol. Biol. 196, 657-675. Opin. Cell Biol. 3, 960-965.
658 An Intertwined Four-helix Bundle in HNF-1

63. Chiu, K. C., Chuang, L. M., Ryu, J. M., Tsai, G. P. & 78. Rice, L. M. & Brunger, A. T. (1994). Torsion angle
Saad, M. F. (2000). The I27L amino acid polymorph- dynamics: reduced variable conformational
ism of hepatic nuclear factor-1alpha is associated sampling enhances crystallographic structure re®ne-
with insulin resistance. J. Clin. Endocrinol. Metab. 85, ment. Proteins: Struct. Funct. Genet. 19, 277-290.
2178-2183. 79. Laskowski, R. A., Macarthur, M. W., Moss, D. S. &
64. Havel, T. F. & Snow, M. E. (1991). A new method Thornton, J. M. (1993). PROCHECK: a program to
for building protein conformations from sequence check the stereochemical quality of protein struc-
alignments with homologues of known structure. tures. J. Appl. Crystallog. 26, 283-291.
J. Mol. Biol. 217, 1-7. 80. Laskowski, R. A. (1995). SURFNET: a program for
65. Schildbach, J. F., Karzai, A. W., Raumann, B. E. & visualizing molecular surfaces, cavities, and inter-
Sauer, R. T. (1999). Origins of DNA-binding speci- molecular interactions. J. Mol. Graph. 5, 323-330.
®city: role of protein contacts with the DNA back- 81. Su, L., Radek, J. T., Hallenga, K., Hermanto, P.,
bone. Proc. Natl Acad. Sci. USA, 96, 811-817. Chan, G., Labeots, L. A. & Weiss, M. A. (1997).
66. Bentley, G., Dodson, E., Dodson, G., Hodgkin, D. & RNA recognition by a bent alpha-helix regulates
Mercola, D. (1976). Structure of insulin in 4-zinc transcriptional antitermination in phage lambda.
insulin. Nature, 261, 166-168. Biochemistry, 36, 12722-12732.
67. Blundell, T. L., Cut®eld, J. F., Cut®eld, S. M., 82. King, C. Y. & Weiss, M. A. (1993). The SRY
Dodson, E. J., Dodson, G. G., Hodgkin, D. C. et al. high-mobility-group box recognizes DNA by partial
(1971). Atomic positions in rhombohedral 2-zinc intercalation in the minor groove: a topological
insulin crystals. Nature, 231, 506-511. mechanism of sequence speci®city. Proc. Natl Acad.
68. Chothia, C., Lesk, A. M., Dodson, G. G. & Hodgkin, Sci. USA, 90, 11990-11994.
D. C. (1983). Transmission of conformational change 83. Lee, W., Revington, J. M., Arrowsmith, C. & Kay,
in insulin. Nature, 302, 500-505. L. E. (1994). A pulsed ®eld gradient isotope-®ltered
69. Derewenda, U., Derewenda, Z., Dodson, E. J., 3D 13C HMQC-NOESY experiment for extracting
Dodson, G. G., Reynolds, C. D., Smith, G. D. et al. intermolecular NOE contacts in molecular com-
(1989). Phenol stabilizes more helix in a new sym- plexes. FEBS Letters, 350, 87-90.
metrical zinc insulin hexamer. Nature, 338, 594-596. 84. Bairoch, A. & Apweiler, R. (2000). The SWISS-PROT
70. Zhou, H. X., Hoess, R. H. & DeGrado, W. F. (1996). protein sequence database and its supplement
In vitro evolution of thermodynamically stable turns. TrEMBL in 2000. Nucl. Acids Res. 28, 45-48.
Nature Struct. Biol. 3, 446-451. 85. Nicholls, A., Bharadwaj, R. & Honig, B. (1993).
71. Brunet, A. P., Huang, E. S., Huf®ne, M. E., Loeb, GRASP: graphical representation and analysis of
J. E., Weltman, R. J. & Hecht, M. H. (1993). The role surface properties. Biophys. J. 64, 166-170.
of turns in the structure of an alpha-helical protein. 86. Stryer, L. (1965). The interaction of a naphthalene
Nature, 364, 355-358. dye with apomyoglobin and apohemoglobin, a
72. Wang, J., Onuchic, J. & Wolynes, P. (1996). Statistics ¯uorescent probe of non-polar binding sites. J. Mol.
of kinetic pathways on biased rough energy land- Biol. 13, 482-495.
scapes with applications to protein folding. Phys. 87. Goto, Y. & Fink, A. L. (1989). Conformational states
Rev. Letters, 76, 4861-4864. of beta-lactamase: molten-globule states at acidic
73. Kim, A. S., Kakalis, L. T., Abdul-Manan, N., Liu, and alkaline pH with high salt. Biochemistry, 28, 945-
G. A. & Rosen, M. K. (2000). Autoinhibition and 952.
activation mechanisms of the Wiskott-Aldrich syn-
drome protein. Nature, 404, 151-158.
74. Otwinowski, Z. & Minor, W. (1997). Processing of Edited by M. F. Summers
X-ray diffraction data collected in oscillation mode.
Methods Enzymol. 276, 307-326.
75. Brunger, A. T., Adams, P. D., Clore, G. M., DeLano, (Received 18 January 2001; received in revised form 10
W. L., Gros, P., Grosse-Kunstleve, R. W. et al. (1998). May 2001; accepted 10 May 2001)
Crystallography & NMR system: a new software
suite for macromolecular structure determination.
Acta Crystallog. sect. D, 54, 905-921.
76. Jones, T. A., Zou, J.-Y., Cowan, S. W. & Kjeldgaard,
M. (1991). Improved methods for building protein
models in electron density maps and the location of
errors in these models. Acta Crystallog. sect. A, 47,
110-119.
77. Adams, P. D., Pannu, N. S., Read, R. J. & Brunger, http://www.academicpress.com/jmb
A. T. (1997). Cross-validated maximum likelihood
enhances crystallographic simulated annealing Supplementary Material comprising 11 Figures
re®nement. Proc. Natl Acad. Sci. USA, 94, 5018-5023. and six Tables is available on IDEAL

You might also like