You are on page 1of 10

Glycobiology vol. 13 no. 5 pp.

377±386, 2003
DOI: 10.1093/glycob/cwg042

Molecular modeling of glycosyltransferases involved in the biosynthesis of


blood group A, blood group B, Forssman, and iGb3 antigens and their
interaction with substrates

a2,3, Christelle Breton2, Jitka


Helena Heissigerov blood transfusion and organ allo- and xenotransplantation.
Moravcova , and Anne Imberty1,2
3
A and B blood group oligosaccharides are located on cell
2 surfaces (erythrocytes and/or vascular endothelium) of var-
Centre de Recherches sur les Macromolecules Vegetales, CNRS
(affiliated with Universite Joseph Fourier), 601 rue de la Chimie, ious mammals (Oriol, 1987) whereas the xeno-antigen, the
BP 53, 38041 Grenoble cedex 9, France; and 3Institute of Chemical Forssman antigen, and the isogloboside 3 (iGb3) glycolipids
Technology, Technicka 5, 166 28 Prague, Czech Republic are not normally produced by human cells. The terminal
Received on November 18, 2002; revised on December 9, 2002; accepted structures of these oligosaccharides are represented in
on December 17, 2002 Scheme 1.

Downloaded from glycob.oxfordjournals.org by guest on November 6, 2010


The enzymes that are involved in the biosynthesis of
A terminal a1-3 linked Gal or GalNAc sugar residue is the those Gal(NAc)a1-3Gal(NAc)-containing antigens display
common structure found in several oligosaccharide antigens, strong amino acid sequence similarities, suggesting a com-
such as blood groups A and B, the xeno-antigen, the Forssman mon fold, and all belong to the same family, GT6, accord-
antigen, and the isogloboside 3 (iGb3) glycolipid. The enzymes ing to the classification in the CAZY database (http://
involved in the addition of this residue display strong amino afmb.cnrs-mrs.fr/~cazy/CAZY/index.html). The a3-galacto-
acid sequence similarities, suggesting a common fold. From a syltransferase (a3GalT), responsible for the biosynthesis of
recently solved crystal structure of the bovine a3-galactosyl- a-Gal epitope (or xeno-antigen), is found in many mamma-
transferase complexed with UDP, homology modeling meth- lian species, including most primates and New World mon-
ods were used to build the four other enzymes of this family in keys, but not in humans and their closest relatives because
their locked conformation. Nucleotide-sugars, the Mn2 ‡ ion, of the mutational inactivation of the gene (Galili and Swan-
and oligosaccharide acceptors were docked in the models. son, 1991). Species lacking the a3GalT do not have the a-
Nine different amino acid regions are involved in the substrate Gal epitope on glycoconjugates, and about 1% of their
binding sites. After geometry optimization of the complexes circulating antibodies are directed against this antigen,
and analysis of the predicted structures, the basis of the causing the hyperacute rejection in xenotransplantation.
specificities can be rationalized. In the nucleotide-sugar bind- Because several crystal structures of a3GalT have been
ing site, the specificity between Gal or GalNAc transferase solved recently (Boix et al., 2001, 2002; Gastinel et al.,
activity is due to the relative size of two clue amino acids. In 2001), structurally and mechanistically this enzyme is a
the acceptor site, the presence of up to three tryptophan model for the several other related retaining glycosyltrans-
residues define the complexity of the oligosaccharide that ferases of varying donor and acceptor substrate specificity,
can be specifically recognized. The modeling study helps in such as Forssman glycolipid synthase (Forss-S) (Haslam
rationalizing the crystallographic data obtained in this family and Baenziger, 1996), isogloboside 3 synthase (iGb3-S)
and provides insights on the basis of substrate and donor
recognition.

Key words: blood group antigens/galactosyltransferase/


glycosyltransferase/molecular modeling

Introduction
Carbohydrates in the form of glycoproteins and glycolipids
play crucial roles in various signaling and molecular recog-
nition processes, affect the stability and structure of pro-
teins, and are epitopes recognized by the immune system
(Varki, 1993). A core a-galactosyl(1-3)galactose moiety is
the common structure of several oligosaccharide antigens
present on mammalian cell surfaces. These antigens have
been well characterized because of their importance for

1 Scheme 1. Terminal structures of the AB blood group/antigens, the


To whom correspondence should be addressed; e-mail:
imberty@cermav.cnrs.fr xeno-antigen, the Forssman antigen, and the iGb3 antigen.

Glycobiology vol. 13 no. 5 # Oxford University Press 2003; all rights reserved. 377
H. Heissigerova et al.

Table I. Specificity and reaction products of the glycosyltransferases related to blood group A and B synthases

Enzyme Donor Acceptora Product

a3GalT UDP-Gal bGal1,4bGlcNAc-R aGal1,3bGal1,4bGlcNAc-R


GTB UDP-Gal aFuc1,2bGal1,4bGlcNAc-R aGal1,3[aFuc1,2]bGal1,4bGlcNAc-R
iGb3-S UDP-Gal bGal1,4bGlcb-ceramide aGal1,3bGal1,4bGlc1-ceramide
GTA UDP-GalNAc aFuc1,2bGal1,4bGlcNAc-R aGalNAc1,3[aFuc1,2]bGal1,4bGlcNAc-R
Forss-S UDP-GalNAc bGalNAc1,3aGal-R aGalNAc1,3bGalNAc1,3aGal-R

a
The monosaccharides that carry the acceptor hydroxyl are indicated in boldface type.

(Keusch et al., 2000), and the histo-blood group A and B in substrate binding. First evidenced in inverting glycosyl-
glycosyltransferases (GTA and GTB) (Yamamoto et al., transferases for b2-GlcNAc transferase (U È nligil et al., 2000)
1990). and then b4-Gal transferase (Ramakrishnan and Qasba,
Enzymes belonging to this a3-Gal(NAc)T family share 2001), this conformational change has also been demon-
common features. They all use a UDP-nucleotide sugar as strated to occur in retaining glycosyltransferases (Boix et al.,

Downloaded from glycob.oxfordjournals.org by guest on November 6, 2010


donor, retain the configuration of the Gal (or GalNAc) 2001). In a3GalT, opening of the acceptor site is dependent
transferred, and their activity is strictly dependent upon on a donor substrate-induced conformational change (Boix
the presence of a divalent cation (generally Mn2 ‡ ). Never- et al., 2002). Among retaining glycosyltransferases, only
theless, they differ by their fine specificity: a3GalT, blood a3GalT (Boix et al., 2001, 2002) and LgtC (Persson et al.,
group B transferase, and iGb3-S only use UDP-Gal as 2001) have been crystallized in this substrate buried state,
donor, whereas blood group A transferase and Forss-S also called Form II. From the several crystal structures that
use UDP-GalNAc. In addition, the mouse AB glycosyl- have presently been obtained in Form I (open) and Form II
transferase derived from a cis-AB gene can transfer sugar (closed), it can now be inferred that not only the presence of
from both donors (Yamamoto et al., 2001). The amino acid UDP or UDP-sugar is necessary for obtaining the ``locked''
basis for Gal versus GalNAc specificity has been well defined conformation but also that this substrate should be added
for the blood group A and B transferases, which differ by to the crystallization medium. Crystals obtained in the
only four amino acids (Yamamoto and McNeill, 1996). On absence of substrate and then soaked with UDP do bind
the other hand, very little is known about the basis for the the substrate in the active site, but the ordering of loops
acceptor specificity: N-acetyllactosamine and lactose are does not occur in the solid state. Owing to the difficulties in
the acceptor for a3GalT and iGb3-S, respectively. Blood obtaining cocrystals of glycosyltransferases with their sub-
group enzymes require substitution by a L-fucose on posi- strates, molecular modeling is an alternative to approaching
tion 2 of the acceptor galactose, whereas Forss-S requires a the role of flexible loops in substrate binding.
N-acetyl group at the same location. Details for donor and In the present article, we propose to use the recent high-
acceptor specificities are listed in Table I. resolution structure of a3GalT (Boix et al., 2001) as a tem-
Like the majority of glycosyltransferases (Paulson and plate to model the other related enzymes with a3Gal(NAc)
Colley, 1989), a3Gal(NAc)T enzymes are type II membrane transferase activity listed in Table I. Docking of nucleotide-
proteins with a short N-terminal cytoplasmic tail, single- sugars on one hand and of oligosaccharide acceptor on the
transmembrane domain, stem, and C-terminal catalytic other will lead to the comparison of the architecture of the
region. Despite their importance, structural information binding sites. Comparison with the recently solved blood
about glycosyltransferases is rare, and only 12 different gly- group A and B synthases (Patenaude et al., 2002) validates
cosyltransferases have been crystallized until now (see our modeling method and allows for comparing the two
review; U È nligil and Rini, 2000; Breton et al., 2002). Although conformational forms of the enzymes. In addition, the dif-
they share little or no sequence similarity, these 12 glycosyl- ferent sequence motifs involved in substrate binding have
transferases appear to adopt only two different folds, which been defined, and their precise role has been investigated.
have been named BGT fold and SpsA fold for reference to the
first structure solved in each case. Among the known struc-
tures, only five are retaining glycosyltransferases, namely, Results
bovine a3GalT (Boix et al., 2001, 2002; Gastinel et al., 2001),
human blood group A and B synthase (Patenaude et al., Description of the overall structure of the models
2002), a bacterial a4-galactosyltransferase (LgtC) (Persson Figure 1 displays the sequence alignments of the enzymes of
et al., 2001), and rabbit glycogenin that initiates the biosynth- interest for the study. This alignment is the basis of the
esis of glycogen (Gibbons et al., 2002). All of these retaining homology building, and only the catalytic regions are
glycosyltransferases adopt the SpsA fold. shown. This glycosyltransferase family displays a high level
Analysis and comparison of glycosyltransferase crystal of sequence identity. The percentage of identity varies from
structures in the goal of elucidating the basis of substrate 44% to 55% (with the exception of GTA and GTB, which
recognition and catalysis has been complicated by the display 98% identity). There are very few insertions or dele-
occurrence of large movement of one or two loops involved tions in the regions corresponding to secondary structures.
378
Molecular modeling of a3Gal(NAc)transferases

Downloaded from glycob.oxfordjournals.org by guest on November 6, 2010


Fig. 1. Amino acid sequence alignment of bovine a3GalT and related enzymes of the a3Gal(NAc)T family. Secondary structure elements of a3GalT are
indicated above the sequences and numbered as in Boix et al. (2001). Conserved amino acids have a black background and preserved ones a gray
background. Regions involved in ligands binding are boxed. The stars indicate the four amino acids that are different between GTA and GTB sequences.

Loop regions display more variability, but their sizes are phosphate atoms through Lys359 (LBR-H) and Arg365
comparable, thus facilitating the modeling study. (LBR-I), both positions absolutely conserved among the
Because all glycosyltransferases belong to the SpsA fold enzymes studied here.
superfamily, the enzymes modeled here adopt a globular The Gal/GalNAc moiety interacts with different regions:
shape. Figure 2A displays the overall structure of the GTA LBR-B, -C, -E, and -F. Details of the hydrogen bonds
model. One face is responsible for binding the nucleotide- involving the sugar moiety have been listed in Table II.
sugar and the acceptor. Because the crystal structure of Depending on the model, five to seven hydrogen bonds
a3GalT complexed with UDP was taken as the reference are established between the sugar moiety and the proteins
molecule (Boix et al., 2001), all models are in their locked (see Figure 4 for GTA/GalNAc). Four of them are con-
Form II conformation, with the nucleotide-sugar almost served in the whole family: the first aspartate amino acid of
completely buried under the C-terminal region. The accep- the DXD motif (LBR-C) receives a hydrogen bond from O-
tor binding site appears as a deep crevice adjacent to the 3 hydroxyl group. An acidic amino acid from region LBR-F
nucleotide-binding site. These two binding sites are made (Asp316 in a3GalT) receives hydrogen bonds from both
up with residues belonging to nine peptide regions, identi- O-4 and O-6, this latest hydroxyl receiving a hydrogen
fied as ligand binding regions (LBRs) and labeled from A bond from the conserved Ser amino acid of LBR-B. The
to I in Figures 1 and 2. Because there is no significant Gal/GalNAc specificity is ensured by LBR-E, which con-
differences in loop size, the overall structure of the four tains the 277 FYYX(GA)(GA)282 motif. From our modeling,
proteins modeled here does not present large variation, it appears that the possibility to accept an N-acetyl group in
and only one has been displayed. the binding site is controlled by a pair of amino acids
facing each other (His280 and Ala282 in a3GalT). The
The nucleotide-sugar binding site: predicted interactions steric hindrance created by Met266/Ala268 in GTB and
between enzymes and UDP-Gal/GalNAc by His280/Ala282 in a3GalT (His253 in iGb3-S) is clearly
The UDP-sugar binding domain is located between the displayed in Figure 3A. The enzymes that can use GalNAc
central b-sheet (b2±b5) and two long a-helices (a3 and a4) have a glycine at one of these two positions (Leu266/Gly268
and is capped by the C-terminal region. Figure 3A gives in GTA and Gly261/Ala263 in Forss-S), therefore allowing
insights into the binding site pockets of four models. The for an opening of this subsite. These particular two residues
model for binding UDP-Gal by iGb3-S is very similar to the correspond to two of the four amino acids differentiating the
prediction for a3GalT and is not displayed. The Mn2 ‡ human blood group A and B glycosyltransferases.
binding site is made up by the 225 DVD227 (numbering rela-
tive to a3GalT), the very conserved LBR-C region. The The acceptor binding site: interactions between
uridine moiety is bound through the conserved enzyme and oligosaccharide acceptor
134
FA(IV)(GK)(KR)Y139 motif in LBR-A, whereas the Acceptors of various size (disaccharides and trisaccharides)
ribose ring interacts with amino acids of LBR-A, -B, and have been docked into protein models. The molecules that
-C. The C-terminal region makes direct contacts with the have been used in the modeling studies are those listed in
379
H. Heissigerova et al.

Downloaded from glycob.oxfordjournals.org by guest on November 6, 2010


Fig. 2. (A) Graphical representation of the model of human GTA in interaction with Mn2 ‡ , UDP-GalNAc, and trisaccharide acceptor. The Connolly
protein surface is represented with different colors for the different peptide regions involved in substrates binding. These loops are labeled according to
Figure 1. (B) Same representation for the crystal structure of GTA in interaction with Mn2 ‡ , UDP, and H-disaccharide analog (Patenaude et al., 2002).
(C) Stereoscopic representation of the binding site of the GTA model. (D) Same representation for GTA crystal structure. (E) Ribbon representation of
the peptide chain in the GTA model, together with space fill representation of the amino acids involved in capping the nucleotide binding site.

Table I, although the noncarbohydrate part (ceramide) have been color coded in Figure 3B where the proteins are
has not been taken into account in the modeling. When represented by their accessible surfaces. Minor contacts are
compared to the donor nucleotide sugar, it can be stated also established with residues of LBR-E (Tyr278 in a3GT).
that the acceptor oligosaccharides are more exposed to the Details of hydrogen bonds and hydrophobic contacts are
solvent and establish a limited number of contacts with the listed in Tables III and IV.
protein surface. Nevertheless, four regions seem to play an In all enzymes of this family, the primary acceptor is a
important role in binding the acceptor and defining the Gal or GalNAc residue. This can be correlated to a con-
specificity, namely, LBR-D, -F, -G, and -H. These regions served structural feature: LBR-F is almost invariant and

380
Molecular modeling of a3Gal(NAc)transferases

brings a Trp residue in contact with the galactose acceptor


A (Trp314 in a3GalT). As previously observed in several
S185 S185
carbohydrate-binding proteins (Vyas, 1991; Rini, 1995),
the aromatic rings stack against the CH of the galactose
D302 D302

rings and ensure specificity of binding for a galactose at this


E303
E303

G268
A268 position.
L266 M266
For two of the enzymes, a3GalT and iGb3-S, a terminal
GTA GTB
galactose is required for acceptor, and no substitution is
tolerated at the C-2 position of this residue. Examination of
E297
S180 D316
S199 the models indicated that region LBR-H, and parti-
E298
E317 cularly the presence of an additional bulky Trp (Trp356
G261
in a3GalT), is responsible for the fine specificity. The
A263 H280 A282 Trp356 in a3GalT makes a barrier for any substitution on
Forss-S α3GalT
the Gal ring, whereas in the other enzymes the smaller Ala
or Thr residue together with basic residues 323 RK (Forss-S)
or 328 QL (GTA and GTB) form a pocket accommodating
either Fuc or NHAc group on C2 of the Gal ring. The size
B of the first amino acid of region LBR-H (Trp356 in
a3GalT) seems to distinguish whether the first monosac-

Downloaded from glycob.oxfordjournals.org by guest on November 6, 2010


W300 W287
charide of acceptor could be branched or not.
Tryptophan residues are also involved in defining the
H220
F236 H233 H223
K346 K332

acceptor specificity at longer range, that is, with the


W329
G235
A343
L329

reducing sugar of the di- or trisaccharide. Again, a3GalT,


W222 E315
Q328

I316

GTA iGb3-S

W295 W314

H228 Q247
Y231
K340 K359
W250
W356
T337
G230
K324
R323 W249
H342

I343
Forss-S α3GalT

Fig. 3. (A) Models of interaction between different glycosyltransferases


and the nucleotide-sugars. The protein is represented by its
Connolly surface that has been sliced to display the interior of the
binding site. Color coding represents the electrostatic potential (from
red for positively charged area to blue for negatively charged area).
The Mn2 ‡ cation is represented by a magenta sphere and the nucleotide
sugar by sticks. (B) Models of interaction between different
glycosyltransferases and the acceptor oligosaccharide. The Connolly Fig. 4. Stereoscopic representation of the hydrogen bond network between
surface of the protein that has been color coded according GalNAc and surrounding amino acids in the model of the interaction
to Figure 2A. between human GTA and UDP-GalNAc.

Table II. Analysis of the models: hydrogen bonds between the Gal or GalNAc of the nucleotide sugars and the amino acids of the different
glycosyltransferases

Ligand atom a3GalT GTB iGb3-S GTA Forss-S

O (N-acetyl) Gly267(N) Ala263(N)


O (N-acetyl) Gly268(N)
O-2 (Gal) His280(NE2) His253(NE2)
O-3 Asp225(OD1) Asp211(OD1) Asp198(OD1) Asp211(OD1)
O-3 Asp211(OD2) Asp211(OD2) Asp206(OD2)
O-4 Asp316(OD2) Asp302(OD2) Asp289(OD2) Asp302(OD2) Glu297(OE2)
O-4 O6(Acceptor) Gln296NE1)
O-6 Asp316(OD1) Asp302(OD1) Asp289(OD2) Asp302(OD1) Glu297(OE1)
O-6 Asp289(OD1) Trp295(O)
O-6 Ser199(OG) Ser185(OG) Ser172(OG) Ser185(OG) Ser180(OG)

381
H. Heissigerova et al.

which uses lactose or N-acetyllactosamine as acceptor, LBR-E, which contains a variable sequence responsible
displays a Trp residue stacking with the Glc (or GlcNAc) for the specificity toward Gal and GalNAc.
ring at the reducing end, thus limiting the possibilities of  The well-conserved region LBR-F mostly ensures specifi-
substitution or branching for this residue. This selection of city for galacto configuration of the acceptor terminal
the flat, ribbonlike conformationÐwhich can be adopted
residue.
by equatorial±equatorial linked carbohydrate (such as cel-
lulose and chitin, but also lactose and N-acetlyllactosamine)  The variable LBR-H region defines the substitution that
by pavement of Trp residuesÐhas previously been observed can be accepted on this terminal sugar of the acceptor.
in polysaccharide-binding protein module. Blood group A This loop, together with the C-terminal peptide LBR-I,
and B transferases, which use either aFuc1-2bGal1- undergoes the conformational change that locks the
3GlcNAc (type 1) or aFuc1-2bGal1-4GlcNAc (type 2) nucleotide sugar in the binding site. Those two regions
acceptors, present a PG/S motif in the LBR-D region also make contacts with the pyrophosphate group.
instead of the AW motif characteristic of a3GalT.  The variable LBR-D region plays a role in the specificity
for the reducing part of the acceptor.
Role of the nine ligand binding regions  Finally, LBR-G only interacts with Fuc or NHAc
The present dissection of the acceptor binding site moiety of the acceptor in GTA, GTB, and Forss-S.
allows researchers to clarify the role of each binding
regions in cation, donor, and acceptor binding. The amino
acids that are predicted to directly interact with the substrate Discussion

Downloaded from glycob.oxfordjournals.org by guest on November 6, 2010


have been labeled in Figure 5. The specific role of each of
the nine sequence motifs can be summarized as follows. Comparison with the crystal structure of bovine a3GalT
The modeling study started from the crystal structure of the
 The most highly conserved regions, LBR-A and LBR-C, complex a3GalT/UDP (Boix et al., 2001). Further attempts
interact with the uracil and ribose rings and ensure part to crystallize a3GalT in the presence of UDP-Gal yielded to
of the coordination of the Mn2 ‡ cation. the cleavage of the substrate in the active site (Boix et al.,
 Recognition of the sugar moiety of the donor involves 2002). In one structure the cleaved galactose residue is still
loops LBR-B, -C, -E, and -F. A special role is devoted to present in the site. After cleavage, the sugar undergoes

Table III. Analysis of the models: hydrogen bonds between the acceptor sugar and the amino acids of the different glycosyltransferases

Ligand atom a3GalT GTB iGb3-S GTA Forss-S

O40 Gln247(NE2) His233(NE2) His220(NE2) His233(NE2) His228(NE2)


0 00 00 00 00
O5 O3 (GlcNAc) O3 (GlcNAc) O3 (Glc) O3 (GlcNAc)
O60 Tyr278(OH) Tyr264(OH) Tyr251(OH) Tyr264(OH) Tyr259(OH)
O60 Glu317(OE1) Glu303(OE1) Glu290(OE1) Glu303(OE1) Glu298(OE1)
O200 Trp250(NE1) His223(NE2)

Table IV. Analysis of the models: amino acids of the different glycosyltransferases that are involved in van der Waals interaction with the acceptor sugar

a3GalT GTB iGb3-S GTA Forss-S

Gln247 His233 His220 His233 His228


Trp249 Ser235 Trp222
Trp250 Phe236 His223 Phe236 Tyr231
Tyr278 Tyr264 Tyr251 Tyr264 Tyr259
Trp314 Trp300 Trp287 Trp300 Trp295
Glu317 Glu303 Glu290 Glu303 Glu298
Arg323
Leu329 Leu329 Lys324
Trp356 Trp329
Lys359 Lys346 Lys332 Lys346 Lys340
Tyr361 His348 Tyr334 His348

382
Molecular modeling of a3Gal(NAc)transferases

Fig. 5. Alignments of the nine sequence motifs that are predicted to form the binding site. Residues labeling are as follows: plus sign, interacting with Mn2 ‡ ;
open squares, with uridine and ribose; filled diamonds, with pyrophosphate; open circles, with Gal or GalNAc of donor; and filled squares, with
oligosaccharidic acceptor. The amino acids that play a crucial role in specificity have been boxed.

rotation and translation movements of small amplitude C-terminal region interacts with the His78±Ile79 cluster.
that allow it to maximize the number of hydrogen bonds When ordering these two regions in our model of GTA,
in the site. additional contacts are established between the protein and
Our docking of N-acetyllactosamine in the acceptor site the nucleotide-sugar: Lys346 (LBR-H) and Arg252 (LBR-I)
was also done from the a3GalT/UDP complex. Because the of the C-terminal domain make salt bridges to the phos-
crystal structure of the enzyme in complex with LacNAc has phate groups, whereas Val184 (LBR-B) interacts with the
been recently published (Boix et al., 2002), this gives a direct uracil ring. The GalNAc moiety of the nucleotide-sugar
comparison between the model and the crystal structure also presents additional contact with the protein. The orien-

Downloaded from glycob.oxfordjournals.org by guest on November 6, 2010


and therefore a validation of our modeling approach. tation of Gal/GalNAc proposed from the present modeling
Indeed, the location of the acceptor disaccharides is cor- study varies slightly from the one that has been deduced (also
rectly predicted, especially all hydrophobic interaction, with from modeling) but starting from the Form I crystals struc-
a particular role of the tryptophan residues. Small varia- ture, where very few contacts were predicted to occur. In our
tions exist between the predicted and observed hydrogen model, GalNAc is stabilized by a larger number of hydrogen
bond networks, mainly due to the orientation of the hydro- bonds (Figure 4), including Ser185, which was disordered in
xyl group at O-60 . the crystal. Also, the N-acetyl moiety interacted with
Gly268, one of the two crucial amino acids in term of AB
Comparison with the crystal structures of blood specificity (see later discussion), therefore explaining why
group A and B transferases GalNAc is favored over Gal in the larger site of GTA.
In the acceptor site, the differences between Forms I and
Crystal structures of both blood group A and B transferases II are less drastic (Figure 2C and D). Nevertheless, ordering
have been very recently elucidated in the native state and as of bulky Lys346 reduces the space available for the fucose
complexes with UDP and acceptor analog (Patenaude et al., residue. In the present model, this results not only in a
2002). Superimposition of the protein backbone (Asp83± different orientation of the fucose but also for the Gal
Arg176 and Cys196±Pro345) of the model structure on the moiety. This latter residue, presents a slightly different
crystal structure yielded a rms of 0.94 A Ê and 0.87 AÊ for
orientation than in the GTA or GTB crystals, resulting in
GTA and GTB enzymes, respectively, thus confirming the a better stacking with Trp300, as previously observed in the
quality of the homology modeling. In the crystal structures crystal structure of a3GalT/lactose complex (Boix et al.,
complexed with UDP and H-type substrate, two peptide 2002).
regions adjacent to the active site are disordered: one loop
(177±195) and the C-terminus region (346±354) therefore
resulting in the open conformation (Form I) of the binding Comparison with biochemical data on GTA and GTB
site cleft. On the opposite, the modeled structures corre- The differences in amino acids between GTA and GTB are
spond to the Form II conformation, which has been pro- limited to four residues: Arg176Gly, Gly235Ser, Leu266-
posed to be induced by the binding of the nucleotide sugar Met, and Gly268Ala (Yamamoto et al., 1990). Among these
(Persson et al., 2001; Boix et al., 2002; Ramakrishnan et al., four differences, three are located in regions that we identify
2002). The roles of regions LBR-B, LBR-H, and LBR-I in as LBRs. As discussed, Leu266Met and Gly268Ala substi-
binding the ligands could then be inferred from the model. tutions (region LBR-E) have a crucial effect on the shape of
Figure 2 displays a comparison of Form I (crystal) and the nucleotide binding pocket and directly affect the speci-
Form II (model) of GTA. ficity for the nucleotide sugar. Indeed, mutagenesis
The differences are maximum for the nucleotide-sugar studies demonstrated that when only one of the two posi-
binding site (Figures 2C and 2D): this substrate is comple- tions is substituted by its analog counterpart in the other
tely buried in Form II. Side chains of amino acids from the enzyme, the resulting enzyme displays both A and B activity
C-terminal region (Lys346, His348, and Arg252) stack (Yamamoto et al., 1996; Seto et al., 1999). The mouse
together and join amino acids from the basis of the long glycosyltransferase that naturally displays this dual specifi-
a4 helix (Trp181 and Gln182) to form a lid over the pyr- city also has only one difference (Met to Gly) with human
ophosphate moiety of the nucleotide sugar (Figure 2E). BGT (Yamamoto et al., 2001). Our modeling study ratio-
These five amino acids show a high degree of conservation nalizes the concerted action of the two amino acids
among the family of glycosyltransferases studied. Such clos- involved. As shown in Figure 3A, they form together a
ing of the lid by hydrophobic contacts is also observed in bottleneck that size controls the possibility to accept an N-
LgtC structure (Persson et al., 2001) where Pro248 in the acetyl group in a subsite.
383
H. Heissigerova et al.

The substitution Gly235Ser is located in the acceptor site crystal structures that have been determined recently, all
(region LBR-D) and may be involved in small differences in of the enzymes for which Mn2 ‡ is required for activity
acceptor recognition that have been defined by chemical share the same fold, despite a lack of sequence similarities.
mapping of the acceptors (see later discussion). Arg176Gly In such cases, fold recognition methods are a powerful tool
mutation does not affect the A-specificity but gives an 11- for identifying family of enzymes that can share the same
fold increase in kcat (Seto et al., 1997). This amino acid is fold (Breton et al., 2002). Combination of fold recognition
not involved in the substrate binding but is located at the study and molecular modeling may help in predicting the
surface of the enzyme. Analysis of the surface indicates that acceptor specificities of putative glycosyltransferase
it is located about 8 AÊ from LBR-A, which closes the site sequences from newly determined genomes. This approach
above the nucleotide sugar. Depending on the orientation may be helpful in the emerging area of glycogenomics.
of the Arg176 side chain, it can participate to a basic cluster
with K123 and K124 of LBR-A, two basic amino acids of A
and B transferases at the surface of the binding site. Mod- Materials and methods
ifying this basic cluster could affect the turnover of the
nucleotide-sugar and the catalysis products. Progress in Multiple alignment of amino acid sequences was performed
the understanding of the catalytic mechanism is needed to with the ClustalX program (Thompson et al., 1994) using
explain fully the role of these residues. the following sequences: bovine a3GalT (GenBank acces-
Chemical mapping studies with modified synthetic accep- sion number J04989), canine Forss-S (U66140), human
tors demonstrated a special role for O-4 of the galactose GTA (J05175) and GTB (AF134414), and rat iGb3-S

Downloaded from glycob.oxfordjournals.org by guest on November 6, 2010


residue (Lowary and Hindsgaul, 1993, 1994). Indeed, in the (AF248543). The homology modeling COMPOSER
present models, this hydroxyl group is involved in a strong program (Blundell et al., 1988) of the Sybyl software
hydrogen bond with His233 for both GTA and GTB (SYBYL, St. Louis, MO) was used to build the different
enzymes. Mapping studies on the fucose residue indicated enzyme models. Conserved a-helices and b-strands (struc-
some differences in specificity because only the B enzyme turally conserved regions in COMPOSER) were built from
requires the methyl group at C6-Fuc (Mukherjee et al., the highest-resolution structure of a3GalT (Boix et al.,
2000). In our model, this methyl group interacts with 2001; code 1K4V in the Protein Data Bank; Berman et al.,
Leu329 in a region (LBR-G) where there is no difference 2000). Loops were then modeled by using the most similar
between A and B transferases. Nevertheless, in our predic- fragments in a library containing all crystal structures of
tion, the methyl group of the fucose is also spatially close to glycosyltransferases available. Resulting models were then
the region LBR-D, where GTA and GTB differs by a screened using the PROCHECK program (Laskowski et al.,
Gly235Ser substitution. 1993), and backbone conformations lying outside the
allowed regions of Ramachandran map were further opti-
mized. Hydrogen was added on all atoms, and partial
Conclusion
atomic charges were derived using the Pullman procedure.
The elucidation of the catalytic mechanism of glycosyltrans- UDP-Gal/UDP-GalNAc was docked into the binding site
ferases remains one of the most challenging problems in in a conformation and location similar to what has been
structural glycobiology (Ly et al., 2002). Particularly, the observed for UDP-Gal complexed with a3GalT (Boix et al.,
catalytic event that allows transfer of a monosaccharide 2001) and for UDP-2F-Gal complexed with LgtC (Persson
with retention of configuration has not yet been elucidated: et al., 2001). A Mn2 ‡ ion was located between the pyro-
the double displacement reaction via formation of a glycosyl- phosphate group and aspartate groups of the DXD motif of
enzyme intermediate that has been proposed by analogy protein. Atom types and energy parameters available for
with glycosylhydrolases (Gastinel et al., 2001) has been carbohydrates (Imberty et al., 1999) were used together with
revised recently and is now almost abandoned (Boix et al., parameters developed for the sugar±pyrophosphate linkage
2002; Ly et al., 2002), although no clear alternative mechan- (Petrova et al., 1999). For disaccharide acceptors, the con-
ism could be proposed. In this context, the aim of the formation at the glycosidic linkage was selected according
present study was to clarify the role of conformational to crystal structures, when available (PeÂrez et al., 2000), and
changes as well as the basis of substrate and acceptor bind- to previously calculated energy maps (Imberty et al., 1995).
ing in one family of glycosyltransferases and to rationalize In all cases, the monosaccharides on the nonreducing side
their specificities. The sequence motifs playing a role in have been located in the binding site as observed for lactose
ligand binding have been identified: the sugar donor speci- and LacNAc acceptor in complex with a3GalT (Boix
ficity (UDP-Gal versus UDP-GalNAc) is due to the relative et al., 2002).
size of two crucial amino acids, whereas in the acceptor Four models were therefore generated: GTA/UDP-Gal-
site, tryptophan residues play a key role in defining the NAc/aFuc1-2bGal1-4bGlcNAc/Mn2 ‡ , GTB/UDP-Gal/
fine specificity. Because glycosyltransferases are widely aFuc1-2bGal1-4bGlcNAc/Mn2 ‡ , Forss-S/UDP-GalNAc/
used for oligosaccharide synthesis in biotechnological bGalNAc1-3aGal/Mn2 ‡ , and iGb3-S/UDP-Gal/bGal1-
approaches, such a modeling study could provide some 4bGlc/Mn2 ‡ . The complex a3GalT/UDP-Gal/bGal1-
structural basis for rational engineering of specificities and 4bGlcNAc/Mn2 ‡ was also generated for comparison. In
design of new transferase activities. all complexes, several cycles of energy minimization were
The present article has been limited to enzymes belonging performed to optimize the geometry of all ligands and also
to one family of homologous enzymes; it may nevertheless of protein side chains in the binding site and its vicinity.
be extended to other glycosyltransferases. Among the Energy calculations were performed using the TRIPOS

384
Molecular modeling of a3Gal(NAc)transferases

force field (Clark et al., 1989) in the Sybyl package with Imberty, A., Bettler, E., Karababa, M., Mazeau, K., Petrova, P., and
addition of energy parameters developed for carbohydrates Perez, S. (1999) Building sugars: The sweet part of structural biology.
In Vijayan, M., Yathindra, N., and Kolaskar, A.S. (eds.), Perspectives
(Imberty et al., 1999). in structural biology. Indian Academy of Sciences and Universities
Validation of the models was performed by superimposi- Press, Hyderabad, pp. 392±409.
tion with the crystal structure of GTA and GTB enzymes Keusch, J.J., Manzella, S.M., Nyame, K.A., Cummings, R.D., and
complexed with UDP and disaccharidic acceptor analog Baenziger, J.U. (2000) Expression cloning of a new member of the
(code 1LZI and 1LZJ; Patenaude et al., 2002). ABO blood group glycosyltransferases, iGb3 synthase, that directs
the synthesis of isoglobo-glycosphingolipids. J. Biol. Chem., 275,
25308±25314.
Laskowski, R.A., MacArthur, M.W., Moss, D.S., and Thornton, J.M.
Acknowledgments (1993) PROCHECK: a program to check the stereochemical quality of
protein structures. J. Appl. Crystallogr., 26, 283±291.
H.H. is supported by a grant from the French Ministere des Lowary, T.L. and Hindsgaul, O. (1993) Recognition of synthetic deoxy
Affaires Etrangeres. This research was partially supported and deoxyfluoro analogs of the acceptor alpha-L-Fuc p-(1 ! 2)-beta-
by the Assocation pour la Recherche contre le Cancer. D-Gal p-OR by the blood-group A and B gene-specified glycosyl-
transferases. Carbohydr. Res., 249, 163±195.
Lowary, T.L. and Hindsgaul, O. (1994) Recognition of synthetic
O-methyl, epimeric, and amino analogues of the acceptor alpha-L-
Abbreviations Fuc p-(1 ! 2)-beta-D-Gal p-OR by the blood-group A and B gene-
GT, glycosyltransferase; GTA, blood group A transferase; specified glycosyltransferases. Carbohydr. Res., 251, 33±67.
GTB, blood group B transferase; Forss-S, Forssman Ly, H.D., Lougheed, B., Wakarchuk, W.W., and Withers, S.G. (2002)

Downloaded from glycob.oxfordjournals.org by guest on November 6, 2010


Mechanistic studies of a retaining alpha-galactosyltransferase from
synthase; iGb3, isogloboside 3; iGb3-S, isogloboside 3 Neisseria meningitidis. Biochemistry, 41, 5075±5085.
synthase; LBR, ligand binding region; LgtC, a bacterial Mukherjee, A., Palcic, M.M., and Hindsgaul, O. (2000) Synthesis and
a4-galactosyltransferase. enzymatic evaluation of modified acceptors of recombinant blood
group A and B. Carbohydr. Res., 326, 1±21.
Oriol, R. (1987) Tissular expression of ABH and Lewis antigens in
humans and animals: expected value of different animal models in the
References study of ABO-incompatible organ transplants. Transplant. Proc., 19,
Berman, H.M., Westbrook, J., Feng, Z., Gilliland, G., Bhat, T.N., 4416±4420.
Weissig, H., Shindyalov, I.N., and Bourne, P.E. (2000) The protein Patenaude, S.I., Seto, N.O., Borisova, S.N., Szpacenko, A., Marcus, S.L.,
data bank. Nucleic Acids Res., 28, 235±242. Palcic, M.M., and Evans, S.V. (2002) The structural basis for specificity
Blundell, T., Carney, D., Gardner, S., Hayes, F., Howlin, B., Hubbard, T., in human ABO(H) blood group biosynthesis. Nature Struct. Biol., 9,
Overington, J., Singh, D.A., Sibanda, B.L., and Sutcliffe, M. (1988) 685±690.
18th Sir Hans Krebs lecture. Knowledge-based protein modelling and Paulson, J.C. and Colley, K.J. (1989) Glycosyltransferases. Structure,
design. Eur. J. Biochem., 172, 513±520. localization, and control of cell type-specific glycosylation. J. Biol.
Boix, E., Swaminathan, G.J., Zhang, Y., Natesh, R., Brew, K., and Chem., 264, 17615±17618.
Acharya, K.R. (2001) Structure of UDP complex of UDP-galactose: Perez, S., Gautier, C., and Imberty, A. (2000) Oligosaccharide conforma-
b-galactoside-a-1,3-galactosyltransferase at 1.53A resolution reveals a tions by diffraction methods. In Ernst, B., Hart, G., and Sinay, P.
conformational change in the catalytically important C-terminus. (eds.), Oligosaccharides in chemistry and biology: a comprehensive
J. Biol. Chem., 276, 48608±48614. handbook. Wiley/VCH, Weinheim, pp. 969±1001.
Boix, E., Zhang, Y., Swaminathan, G.J., Brew, K., and Acharya, K.R. Persson, K., Ly, H.D., Dieckelmann, M., Wakarchuk, W.W.,
(2002) Structural basis of ordered binding of donor and acceptor Withers, S.G., and Strynadka, N.C.J. (2001) Crystal structure of
substrates to the retaining glycosyltransferase: alpha-1,3 galactosyl- the retaining galactosyltransferase LgtC from Neisseria meningitidis
transferase. J. Biol. Chem., 277, 28310±28318. in complex with donor and acceptor sugar analogs. Nature Struct.
Breton, C., Heissigerova, H., Jeanneau, C., Moravcova, J., and Imberty, A. Biol., 8, 166±175.
(2002) Comparative aspects of glycosyltransferases. In Drickamer, K. Petrova, P., Koca, J., and Imberty, A. (1999) Potential energy
and Dell, A. (eds.), Glycogenomics: the impact of genomics and hypersurfaces of nucleotide-sugars: Ab initio calculations, force-field
informatics in glycobiology. Portland Press, London, pp. 23±32. parametrization, and exploration of the flexibility. J. Am. Chem. Soc.,
Clark, M., Cramer, R.D.I., and van den Opdenbosch, N. (1989) 121, 5535±5547.
Validation of the general purpose Tripos 5.2 force field. J. Comput. Ramakrishnan, B. and Qasba, P.K. (2001) Crystal structure of lactose
Chem., 10, 982±1012. synthase reveals a large conformational change in its catalytic
Galili, U. and Swanson, K. (1991) Gene sequences suggest inactivation of component, the beta1,4-galactosyltransferase-I. J. Mol. Biol., 310,
alpha-1,3-galactosyltransferase in catarrhines after the divergence of 205±218.
apes from monkeys. Proc. Natl. Acad. Sci. USA, 88, 7401±7404. Ramakrishnan, B., Balaji, P.V., and Qasba, P.K. (2002) Crystal structure
Gastinel, L.N., Bignon, C., Misra, A.K., Hindsgaul, O., Shaper, J.H., and of beta-1,4-galactosyltransferase complex with UDP-Gal reveals an
Joziasse, D.H. (2001) Bovine a1,3-galactosyltransferase catalytic oligosaccharide binding site. J. Mol. Biol., 318, 491±502.
domain structure and its relationship with ABO histo-blood group Rini, J.M. (1995) Lectin structure. Annu. Rev. Biophys. Biomol. Struct., 24,
and glycosphingolipid glycosyltransferases. EMBO J., 20, 638±649. 551±577.
Gibbons, B.J., Roach, P.J., and Hurley, T.D. (2002) Crystal structure of Seto, N.O., Palcic, M.M., Compston, C.A., Li, H., Bundle, D.R., and
the autocatalytic initiator of glycogen biosynthesis, glycogenin. J. Mol. Narang, S.A. (1997) Sequential interchange of four amino acids from
Biol., 319, 463±477. blood group B to blood group A glycosyltransferase boosts catalytic
Haslam, D.B. and Baenziger, J.U. (1996) Expression cloning of Forssman activity and progressively modifies substrate recognition in human
glycolipid synthetase: a novel member of the histo-blood group ABO recombinant enzymes. J. Biol. Chem., 272, 14133±14138.
gene family. Proc. Natl. Acad. Sci. USA, 93, 10697±106702. Seto, N.O., Compston, C.A., Evans, S.V., Bundle, D.R., Narang, S.A.,
Imberty, A., Mikros, E., Koca, J., Mollicone, R., Oriol, R., and Perez, S. and Palcic, M.M. (1999) Donor substrate specificity of recombinant
(1995) Computer simulation of histo-blood group oligosaccharides. human blood group A, B and hybrid A/B glycosyltransferases
Energy maps of all constituting disaccharides and potential energy expressed in Escherichia coli. Eur. J. Biochem., 259, 770±775.
surfaces of 14 ABH and Lewis carbohydrate antigens. Glycoconj. J., Thompson, J.D., Gibson, T.J., Plewniak, F., Jeanmougin, F., and
12, 331±349. Higgins, D.G. (1994) The CLUSTAL_X windows interface: flexible

385
H. Heissigerova et al.

strategies for multiple sequence alignment aided by quality analysis Yamamoto, F. and McNeill, P.D. (1996) Amino acid residue at codon 268
tools. Nucleic Acids Res., 22, 4876±4882. determines both activity and nucleotide-sugar donor substrate
È nligil, U.M. and Rini, J.M. (2000) Glycosyltransferase structure and
U specificity of human histo-blood group A and B transferases. In vitro
mechanism. Curr. Opin. Struct. Biol., 10, 510±517. mutagenesis study. J. Biol. Chem., 271, 10515±10520.
È nligil, U.M., Zhou, S., Yuwaraj, S., Sarkar, M., Schachter, H., and
U Yamamoto, F., Clausen, H., White, T., Marken, J., and Hakomori, S.
Rini, J.M. (2000) X-ray crystal structure of rabbit N-acetylglucosami- (1990) Molecular genetic basis of the histo-blood group ABO system.
nyltransferase I: catalytic mechanism and a new protein superfamily. Nature, 345, 229±233.
EMBO J., 19, 5269±5280. Yamamoto, M., Lin, X.H., Kominato, Y., Hata, Y., Noda, R., Saitou, N.,
Varki, A. (1993) Biological roles of oligosaccharides: all of the theories are and Yamamoto, F. (2001) Murine equivalent of the human histo-
correct. Glycobiology, 3, 97±130. blood group ABO gene is a cis-AB gene and encodes a glycosyl-
Vyas, N. (1991) Atomic features of protein±carbohydrate interactions. transferase with both A and B transferase activity. J. Biol. Chem., 276,
Curr. Opin. Struct. Biol., 1, 732±740. 13701±13708.

Downloaded from glycob.oxfordjournals.org by guest on November 6, 2010

386

You might also like