You are on page 1of 9

PROTEIN STRUCTURE AND FOLDING:

Crystal Structure of Alkaline Phosphatase


from Human Placenta at 1.8 Å Resolution:
IMPLICATION FOR A SUBSTRATE
SPECIFICITY

Marie Hélène Le Du, Torgny Stigbrand,


Michael J. Taussig, André Ménez and Enrico
A. Stura

Downloaded from http://www.jbc.org/ at UNIV OF UTAH on November 22, 2014


J. Biol. Chem. 2001, 276:9158-9165.
doi: 10.1074/jbc.M009250200 originally published online December 20, 2000

Access the most updated version of this article at doi: 10.1074/jbc.M009250200

Find articles, minireviews, Reflections and Classics on similar topics on the JBC Affinity Sites.

Alerts:
• When this article is cited
• When a correction for this article is posted

Click here to choose from all of JBC's e-mail alerts

This article cites 33 references, 8 of which can be accessed free at


http://www.jbc.org/content/276/12/9158.full.html#ref-list-1
THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 276, No. 12, Issue of March 23, pp. 9158 –9165, 2001
© 2001 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

Crystal Structure of Alkaline Phosphatase from Human Placenta at


1.8 Å Resolution
IMPLICATION FOR A SUBSTRATE SPECIFICITY*

Received for publication, October 10, 2000, and in revised form, December 15, 2000
Published, JBC Papers in Press, December 20, 2000, DOI 10.1074/jbc.M009250200

Marie Hélène Le Du‡§, Torgny Stigbrand¶, Michael J. Taussig储, André Ménez‡,


and Enrico A. Stura‡§
From the ‡Département d’Ingénierie et d’Etudes des Protéines (DIEP), Commissariat à l’Energie Atomique,
C. E. Saclay, 91191 Gif-sur-Yvette Cedex, France, ¶Department of Immunology, Umeå University Sweden, and
储Laboratory of Molecular Recognition, The Babraham Institute, Babraham, Cambridge CB2 4AT, United Kingdom

Human placental alkaline phosphatase (PLAP) is one clustered on the same chromosome. The fourth (NSAP) is non-
of three tissue-specific human APs extensively studied specific and can be found in bone, liver, and kidney. It is about
because of its ectopic expression in tumors. The crystal 50% identical with the other three, and its gene is located on

Downloaded from http://www.jbc.org/ at UNIV OF UTAH on November 22, 2014


structure, determined at 1.8-Å resolution, reveals that another chromosome (1–3). Like the other mammalian phos-
during evolution, only the overall features of the en- phatases, PLAP is post-translationally modified. This includes
zyme have been conserved with respect to Escherichia two N-glycosylation sites and a glycosylphosphatidylinositol
coli. The surface is deeply mutated with 8% residues in anchor with which these APs are tethered to the membrane.
common, and in the active site, only residues strictly The release of the enzyme may occur through the action of
necessary to perform the catalysis have been preserved.
phosphatidylinositol-specific phospholipase C.
Additional structural elements aid an understanding of
Irrespective of whether they are of mammalian or bacterial
the allosteric property that is specific for the mamma-
origin, APs catalyze the hydrolysis of phosphomonoesters (4)
lian enzyme (Hoylaerts, M. F., Manes, T., and Millán, J. L.
(1997) J. Biol. Chem. 272, 22781–22787). Allostery is prob- with release of inorganic phosphate and alcohol. PLAP has only
ably favored by the quality of the dimer interface, by a low sequence identity with the Escherichia coli enzyme (30%),
long N-terminal ␣-helix from one monomer that em- but residues involved in the active site of the enzyme (Ser-92,
braces the other one, and similarly by the exchange of a Asp-91, Arg-166) and the ligands coordinating the two zinc
residue from one monomer in the active site of the other. atoms and the magnesium ion are conserved. Therefore, the
In the neighborhood of the catalytic serine, the orienta- catalytic mechanism, which was deduced from the structure of
tion of Glu-429, a residue unique to PLAP, and the pres- the E. coli AP (5), was proposed to be similar in eukaryotic APs.
ence of a hydrophobic pocket close to the phosphate In E. coli, the mechanism involves the activation of the cata-
product, account for the specific uncompetitive inhibi- lytic serine by a zinc atom, the formation of a covalent phos-
tion of PLAP by L-amino acids, consistent with the ac- phoseryl intermediate, the hydrolysis of the phosphoseryl by a
quisition of substrate specificity. The location of the water molecule activated by a second zinc atom, and the release
active site at the bottom of a large valley flanked by an of the phosphate product or its transfer to a phosphate acceptor
interfacial crown-shaped domain and a domain contain- (6). However, as judged from various studies using mammalian
ing an extra metal ion on the other side suggest that the
APs, PLAP presents novel catalytic characteristics. It is an
substrate of PLAP could be a specific phosphorylated
allosteric enzyme (7), and it is uncompetitively inhibited by
protein.
some L-amino acids, namely L-Phe, L-Trp, and L-Leu (8 –10).
The tissue specificity of the human enzymes has been corre-
lated to an additional function (3). In NSAP, this specificity
Alkaline phosphatases (E.C.3.1.3.1) (APs)1 form a large fam-
involves binding to collagen (11, 12). In PLAP, evidence has
ily of dimeric enzymes common to all organisms. In man, three
accumulated over the past few years suggesting a role in cell
out of four AP isozymes are tissue-specific, one is placental
division in both normal and transformed cells (13–16). This role
(PLAP), the second is from germ cell (GCAP), and the third is
probably occurs through its phosphatase activity against phos-
intestinal. They are 90 –98% homologous, and their genes are
phorylated proteins (14, 17). In addition, it has been suggested
that PLAP may be involved in the transfer of maternal IgG to
* Work at the Babraham Institute was supported by a grant from the the fetus (18 –20).
Biotechnology and Biological Sciences Research Council, Swindon, PLAP is one of the first proteins found to be ectopically
United Kingdom (to M. J. T.), and work at University of Umeå was
supported by Cancerfonden. The costs of publication of this article were
expressed by cancer cells, leading to the concept that dysregu-
defrayed in part by the payment of page charges. This article must lation of embryonic genes plays a significant role in the cancer
therefore be hereby marked “advertisement” in accordance with 18 process (21). During the last 30 years, many clinical reports
U.S.C. Section 1734 solely to indicate this fact. have been published concerning PLAP and its use as a tumor
The atomic coordinates and structure factors (code 1EW2) have been
deposited in the Protein Data Bank, Research Collaboratory for Struc-
marker. However, the results are often controversial, mainly
tural Bioinformatics, Rutgers University, New Brunswick, NJ because the assays cannot distinguish between the closely re-
(http://www.rcsb.org/). lated tumor markers GCAP and PLAP (3). The problem of
§ To whom correspondence should be addressed. E-mail: mhledu@ cross-reactivity in the assays reveals that there is a need to
cea.fr (for M. H. L.-D.) or estura@cea.fr (for E. A. S.).
1
The abbreviations used are: AP, alkaline phosphatase; PLAP, pla-
elucidate the three-dimensional structure of at least one hu-
cental AP; GCAP, germ cell AP; NSAP, non-specific AP; ECAP, E. coli man AP.
AP. PLAP is a good representative of all human APs because of

9158 This paper is available on line at http://www.jbc.org


Crystal Structure of AP from Human Placenta at 1.8-Å Resolution 9159
TABLE I
Data collection, phasing, and refinement statistics
Data collection Diffraction limit (Å) 1.9 Å 1.7 Å

Resolution used (Å) (last shell) 50.0-2.1 Å (2.22-2.13 Å) 22.0-1.82 Å (1.87-1.82 Å)


Number of reflections 30158 41585
I/␴(I) (last shell) 8.6 (2.8) 8.5 (1.6)
Completeness (last shell) 90.0% (87.6%) 79.6% (67.6%)
Rsym (last shell) 14.4% (59.1%) 8.2% (48.8%)
Molecular replacement Resolution (Å) 8.0-3.5 Å
R-factor (%) (2nd solution) 50.2% (51.3%)
Correlation factor (2nd solution) 18.5% (14.5%)
Refinement Resolution (Å) 22.0-1.82 Å
R-factor (%) 18.5%
R-free (%) 24.2%
Metal peak height (B-factor after Zn1 33 ␴ 32 ␴ (20.1 Å2)
refinement)
Zn2 34 ␴ 30 ␴ (19.2 Å2)
Mg3 19 ␴ 28 ␴ (14.5 Å2)
X4 18 ␴ 24 ␴ (15.7 Å2)

the high sequence homology between these enzymes. The crystals or difficulty in crystallization, suggesting that the presence of
knowledge of its three-dimensional structure is crucial to elu- longer sugar chains hinders crystallization. PLAP (0.17 mM) and p-

Downloaded from http://www.jbc.org/ at UNIV OF UTAH on November 22, 2014


nitrophenyl phosphate (45 mM) were mixed in the volumetric ratio 4:1
cidating the distinctive properties of eukaryotic APs, which
before setting up the drops. The reservoir solution consisted of 14%
cannot be predicted with confidence by analyzing the bacterial polyethylene glycol 4,000 to 20% polyethylene glycol 3,350, 100 mM
AP structure. These properties include the uncompetitive sodium cacodylate, 2 mM zinc acetate, pH 6.5. The crystals grew as long
mechanism of inhibition, the allosteric property related to cat- needles with well defined pyramidal ends, 0.05 ⫻ 0.05 mm in cross-
alytic activity, and the elements related to tissue specificity. In section, often as long as the whole length of the drop.
addition, the location of the residues specific to PLAP may help Cryogenic and Heavy Atom Soaking—Screening for cryo-protection
conditions was carried out for crystals to be analyzed with synchrotron
in the design of new diagnostic tools to eliminate the cross-
radiation. To make up the cryo-protectant, glycerol was added to the
reactivity observed in the current ones. We present here the working solution with 0 –15% xylitol. A few crystals were added to this
crystal structure of PLAP at 1.8-Å resolution and a number of solution. If the crystals dissolved, extra precipitant was added until the
structural elements that may explain the specific properties of crystals were stable for a prolonged period of time (at least 20 min).
PLAP and of other mammalian APs. Crystals were manipulated in the cryo-solution using loops. CryoLoops,
mounting pins, and crystal caps were purchased from Hampton Re-
MATERIALS AND METHODS search (Laguna Hills, Ca). Heavy atom soaking was carried out using
Purification—PLAP was purified according to the method described 75–300 ␮l of soaking solutions in depression plates.
by Holmgren et al. (22). The frozen placenta, SS homozygote phenotype, X-ray Data Collection—A first crystal was shock-cooled at 80 K and
was slowly thawed and cleaned from blood and membranes and cut into used to collect a native data set at 1.9-Å resolution. After a short
pieces. Phosphate-buffered saline in the same amount as the weight of soaking (20 min) in a solution containing 3 mM p-chlormercuri benzene
the pieces was added, and the mixture was homogenized. An equal sulphonate (PCMBS) in cryo-solution, a second crystal was shock-cooled
volume of n-butanol was added, and PLAP was extracted by stirring for at 80 K. One set of data was collected at 1.7-Å resolution. Both data sets
30 min. After centrifugation at 7000 ⫻ g for 30 min, the upper butanol were collected at Laboratoire pour l’Utilisation du Rayonnement Elec-
phase was removed, and the aqueous phase was heat-inactivated for 30 rromagnetique, station D41, on a Mar Research image-plate area de-
min at 56 °C. After a similar centrifugation and filtration, acetone was tector. Data processing and internal scaling were carried out with HKL
added (the same volume as the aqueous phase), and the mixture was (24) (Table I). The space-group determination was performed using the
stored at ⫺40 °C for 1 h. After centrifugation at 11000 ⫻ g for 40 min, autoindexation procedure as implemented in Denzo, and the enanti-
the pellet was dissolved in 10 mM Tris, 2 mM MgCl2, pH 7.0, by stirring omer determination was performed by observation of the reflections
and by dialyzing extensively against the same solution. The solution extinction in the output file. The crystal belongs to the monoclinic space
was then applied to a DEAE-Sephadex column and washed with Tris- group C2221 with a ⫽ 88.8 Å, b ⫽ 114.5 Å, c ⫽ 106.9 Å. There is one
buffered saline, pH 7.0. PLAP was eluted with a gradient from Tris- monomer per asymmetric unit with a Vm of 2.74, corresponding to 50%
buffered saline to Tris-buffered saline ⫹ 0.2 M NaCl; the A280 and the solvent in the unit cell.
catalytic phosphatase activity were determined. The pooled active frac- Structure Resolution—The structure was solved by molecular re-
tions were dialyzed against 0.1 M glycine and concentrated by DIAFLO placement, using the program AMoRe (25), and the coordinates of one
ultrafiltration to 3 ml. This fraction was applied to a preparative iso- monomer of the wild type E. coli alkaline phosphatase (ECAP) (entry
electric focusing column (Ampholytes pH 3–10, Amersham Pharmacia code 1alk) (5) and the native data set. The best solution obtained was in
Biotech), eluted 48 h later and assayed for catalytic activity. The active the resolution shell of 8.0 to 3.5 Å, with a weak correlation coefficient
fractions were pooled and gel-filtered on a Sephadex G-200 column in (18.1) and a poor contrast between the first and second solution (Table
Tris-buffered saline. I). However, when applying the rotation matrix and translation to the
Crystallization—The protein was concentrated to 10 –16 mg/ml in 10 coordinates file, we observed that the dimer was recreated along a
mM Tris, 2 mM MgCl2, 0.02% NaN3, pH 7.0, using an Amicon Centricon crystallographic 2-fold axis, which validated this solution as the correct
30. Crystallization screening and growth of crystals were carried out one.
using sitting drop vapor diffusion plates (23). Crystals were obtained Model Building and Refinement—The re-building of the model was
from 12% polyethylene glycol 4,000, 2 mM zinc acetate (essential), 200 limited by the low phasing power of the ECAP model due to the low
mM imidazole malate, pH 5.5– 6.5. Other buffers such as acetate, cit- (30%) sequence identity between the two APs. Standard sequence align-
rate, and cacodylate gave similar results. Magnesium is not needed in ment programs gave misleading results and uninterpretable maps. The
the reservoir solution because 2 mM MgCl2 is present in the storage method of hydrophobic cluster analysis as coded in the program HCA
buffer. Large crystals were first grown in the presence of 10 mM p- (26) was applied to both ECAP and PLAP sequences. The subsequent
nitrophenyl phosphate, which is a substrate used in colorimetric reac- hydrophobic patch alignment resulted in a corrected sequence align-
tions that is hydrolyzed by PLAP, the solution turning yellow on mixing ment between the two. The starting model contained most of the ECAP
with the protein solution. The solubility of the PLAP substrate/product main chain without loops or insertions, leaving only the side chains for
complex increases with increasing concentrations of p-nitrophenyl the conserved residues. The program WARP version 5.0 plus the rou-
phosphate, whereas the addition of the product, phosphate, augments tine Side Dock, kindly provided by A. Perrakis (27), was crucial in the
precipitation. Solubility variations are also observed from preparation rebuilding of the model as well as the data set collected at 1.7-Å
to preparation, possibly reflecting variations in the amount and type of resolution. The final PLAP model could then be re-built rapidly. In the
glycosylation. Higher solubility preparations correlate with poor quality active site, the peak height in the Fo ⫺ Fc map calculated from the
9160 Crystal Structure of AP from Human Placenta at 1.8-Å Resolution
native date set allowed us to unambiguously model a zinc at M1 and M2 of each monomer are controlled by the conformation of the
and a magnesium at the M3 position, as in the E. coli structure. The second AP subunit (7). The principle of allostery implies that
peak height in the Fo ⫺ Fc map calculated from the PCMBS data was
high at M3, but the geometry was in agreement with a magnesium. In
the binding of the substrate in one active site will affect the
addition, after refinement with a magnesium at M3, the B-factor of this second binding site. The crystal form studied here requires the
atom was equivalent to those of its ligands, and no residual density was presence of the substrate p-nitrophenyl phosphate to grow.
observed in the final Fo ⫺ Fc map. The statistics of the final model as Therefore, the protein structure described corresponds to a
given by XPLOR (36) are summarized in Table I. PROCHECK (28) was conformation of the enzyme activated by substrate.
used to analyze the structure geometry. The three-dimensional struc-
A second item to consider is the hydrophobic character of the
tures of PLAP and ECAP were superimposed with the iterative pro-
gram ALIGN (29). The coordinates have been deposited to the PDB at dimer interface of PLAP. Less than 30% of the residues are
Brookhaven (entry code 1EW2). involved in hydrogen-bonding interactions, which confer flexi-
bility on the interface. The surface buried in the dimer inter-
RESULTS AND DISCUSSION
face is slightly larger in PLAP (4150 Å2) than in ECAP (3900
Overall Structure—The overall structure of PLAP is a dimer; Å2). However, the residues involved in dimerization are mainly
each monomer contains 479 residues, 4 metal atoms, 1 phos- different, as is the nature of the interface. In ECAP, 36 out of
phate ion, and 603 water molecules. The two monomers are 82 total residues at the dimer interface are involved in hydro-
related by a 2-fold crystallographic axis. The surface of PLAP is gen bonds, whereas in PLAP only 24 out of 83 residues are
poorly conserved with that of the E. coli enzyme, with only 8% involved in hydrogen bonding interactions, and only 9 of these
residues in common, although the core is preserved. PLAP are conserved between the two. It could be speculated that in
possesses additional secondary structure elements comprising PLAP many hydrogen bond interactions have been replaced by
an N-terminal ␣-helix (residues 9 –25), forming an arm that less specific van der Waals contacts, which are more likely to

Downloaded from http://www.jbc.org/ at UNIV OF UTAH on November 22, 2014


embraces the other monomer; an ␣-helix and a ␤-strand in a allow rearrangement of the two monomers.
highly divergent region (residues 208 –280) and a different Biochemical studies performed on PLAP showed that resi-
organization of the small ␤-sheet in domain 365– 430. PLAP
dues Asn-84, Tyr-367, and Glu-429 are involved in the allostery
has two glycosylation sites at Asn-122 and Asn-249 that vary in
(7). Our structure brings additional information concerning the
their degree of glycosylation depending of the placental sample
precise role of these residues. First, Asn-84 is located at the
(Fig. 1). Two disulfide bridges are located at different positions
dimer interface with its ␦ oxygen within hydrogen-bonding
from those in the E. coli enzyme. One is close to the first
distance of Asp91-N (3.2 Å) and probably intervenes through
glycosylation site and may rigidify the loop that carries the
this residue in the stabilization of the catalytic Ser-92. On the
carbohydrate chain. The other is located near to Asp-481,
same loop, Val-85 and Asp-86 interact with Ala-1 and Ile-2
where the glycosylphosphatidylinositol anchor is attached, and
from the other monomer, and Lys-87 interacts with Leu-369
may stabilize the orientation of PLAP relative to the mem-
and Tyr-367, also from the other monomer. Therefore, Asn-84
brane. In the active site, only the residues that are essential to
may affect the enzyme activity through its interaction with
the catalysis are preserved, i.e. the catalytic serine, the three
Asp-91 and may intervene in the allostery process because of
metal ion sites (Zn1, Zn2, and Mg3) and their ligands, whereas
its involvement in the dimerization (Fig. 4). Second, Tyr-367 is
most of the surrounding residues are different (Fig. 2). The
located at the entrance of the active site of the second mono-
upper part of the active site contains a large number of basic
mer. Its hydroxyl is located 6.1 Å from the phosphate and 3.1 Å
residues, and the lower part contains mainly aliphatic or hy-
drophobic residues. Among these hydrophobic residues, we ob- from His-432, which in turn chelates the zinc atom Zn1. The
serve Tyr-367, which belongs to the second monomer. The network consisting of Tyr-367 (monomer1)-His-432 (mono-
consequences of such a pattern of residues are typical for mam- mer2)-Zn1 (monomer2)-PO4 (monomer2) might affect the step
malian APs and will be discussed below. of phosphoseryl hydrolysis by modulating the reactivity of Zn1
Half of the enzyme surface corresponds to three clearly iden- (Fig. 4). The presence in the active site of a residue from the
tifiable regions whose sequences largely vary among human second monomer and its probable involvement in the enzyme
APs and are lacking in nonmammalian enzymes (Fig. 3). The reactivity is a new structural feature that could not have been
first is the long N-terminal ␣-helix described above. The sec- postulated from the ECAP structure, as the loop containing
ond, an interfacial “crown domain,” is formed by the insertion residues 366 to 375 in PLAP is an insertion with respect to
of a 60-residue segment (366 – 430) from each monomer. It ECAP.
consists of two small interacting ␤-sheets, each composed of In their study, Hoylaerts et al. (7) report that the mutation
three parallel strands and surrounded by six large and flexible E429G affects the affinity of Zn1 for PLAP (7). Glu-429 is
loops containing a short ␣-helix. Third, a “metal binding do- solvent-accessible and located at the entrance of the cleft,
main,” comprised of 76 residues (209 –285) and folded into two which leads to the active site where the O⑀1 oxygen interacts
␤-strands flanked by two ␣-helices. It includes the glycosyla- with the N␦ nitrogen of His-320 (distance ⫽ 3.4Å). In turn,
tion site at Asn-249, stabilized by a stacking interaction with His-320-N⑀ interacts with Zn1 (distance ⫽ 2.4 Å). Therefore,
Trp-248, and an additional metal ion. In the Fo ⫺ Fc map, the network consisting of Glu-429-His-320-Zn1-PO4 might also
calculated from the native data set, the peak for this metal affect the hydrolysis of the phosphoseryl moiety through the
corresponds to about 10 electrons. Its height and the crystalli- destabilization of Zn1 (Fig. 4). Therefore, Glu-429 may not be
zation conditions suggest the presence of a magnesium, but its directly involved in the allostery process but probably affects
coordination by carboxylates from Glu-216, Glu-270, and Asp- the activity and the metals in the enzyme.
285 by Phe-269-CO and a water molecule is also in agreement Finally, the N-terminal ␣-helix described above is removed
with a calcium ion (30) (Fig. 1c). The crown domain and the from the rest of its own monomer, but it interacts with the
metal binding domain correspond to novel folds, since a search second monomer with a buried surface area of 555 Å2, suggest-
for other structurally related domains using the program DALI ing an involvement in enzyme dimerization. Residues of the
(31) yielded no significant matches. N-terminal extremity interact with Arg-370 from loop 366 –375
Allosteric Process—One important property that differenti- and with Asn-106 and Arg-117 from the second monomer.
ates mammalian APs from their bacterial counterpart is the Therefore, the interaction of the N-terminal ␣-helix with the
allostery observed when the enzyme is fully metalized. Mu- second monomer may help stabilize Tyr-367 and, therefore,
tagenesis studies reveal that stability and catalytic properties affect the enzymatic activity (Fig. 4).
Crystal Structure of AP from Human Placenta at 1.8-Å Resolution 9161

Downloaded from http://www.jbc.org/ at UNIV OF UTAH on November 22, 2014


FIG. 1. Electron density for carbohydrates and fourth metal ion. a and b, glycosylation site connected to Asn-122 corresponding,
respectively, to the reported 1.8-Å structure (a) and to a 2.1-Å data set still under refinement (b) (Table I). c, carbohydrate connected to Asn-249
in stacking with Trp-248 and coordination of the fourth metal ion with Glu-216, Phe-269-CO, Glu-270-O⑀2, Asp-285, and one water molecule. In
each case, the 2 Fo ⫺ Fc map is shown in blue and is contoured at the 1.2␴ level. The Fo ⫺ Fc map is shown in green and contoured at 10␴ level.
This figure was made with TURBO (34).

Specific Uncompetitive Inhibition—A second kinetic property Glu-429 is of particular importance as it is the only active site
of human APs, not shared by their bacterial ancestors, is that residue specific to PLAP. The corresponding residue is a serine
they are stereospecifically inhibited by L-amino acids through in intestinal AP, a glycine in GCAP, and a histidine in NSAP
an uncompetitive mechanism. This type of rare inhibition im- (8 –10). The structure confirms the involvement of Arg-166 and
plies that the inhibitor can bind to the enzyme-substrate com- Glu-429 and also allows precise description of the hydrophobic
plex but not to the enzyme alone. In the case of a two-step pocket, which probably stabilizes the hydrophobic moiety of the
mechanism like that of PLAP, the complex can be the initial inhibitor. Located at the entrance of the cleft, which leads to
enzyme-substrate or any of the intermediate complexes. Previ- the active site, Glu-429 borders a pocket that extends from the
ous studies on PLAP have shown that the molecular mecha- catalytic Ser-92 to the phosphate product. Arg-166 is found at
nism of this inhibition involves Arg-166 and Glu-429 and sug- the edge of a second pocket around the active site, also bordered
gests the presence of a hydrophobic pocket in the active site. by Phe-107, Gln-108, and Tyr-367 from the second monomer
9162 Crystal Structure of AP from Human Placenta at 1.8-Å Resolution

FIG. 2. Active site of PLAP. A Corey-


Pauling-Koltun representation of the ac-
tive site pocket of PLAP is shown. The
residues conserved with ECAP are repre-
sented in white. The substituted residues
are colored according to the residue type,
Phe, Trp, and Tyr are violet; Asn, Cys,
Gln, Met, Ser, and Thr are green; Arg,
His, and Lys are blue; Asp and Glu are
red; Ala, Gly, Ile, Leu, Pro, and Val are
yellow. Residues from the second mono-
mer are pink. This figure was made with
TURBO.

Downloaded from http://www.jbc.org/ at UNIV OF UTAH on November 22, 2014


FIG. 3. Overall structure. The overall
structure of PLAP is shown in ribbon rep-
resentation with the residue side chains
of the three extra domains in ball and
stick representation. The monomer I is
shown in pale green, monomer II in blue,
N-terminal ␣-helix in red, crown domain
in orange, and metal binding domain in
yellow. This figure was made with MOL-
MOL (35).

(Fig. 2). In Fig. 5, we have modeled the inhibition of PLAP by complexes of PLAP and L-amino acids should further clarify the
a L-Phe amino acid. This model shows that the geometry of role of these two pockets and the mechanism of uncompetitive
Glu-429, Arg-166, and the hydrophobic pocket is ideal to fit an inhibition.
L-amino acid such as L-Phe. The hydrophobic moiety of the Elements Determining Enzyme Specificity—Although the
ligand would be stabilized in the hydrophobic pocket, its car- target of each human AP remains unknown, the tissue speci-
boxyl group by Arg-166 and its amide group by Glu-429. This ficity strongly suggests an adaptation for these enzymes to-
topology matches both the stereo-specificity and uncompetitive ward specific proteins. For example, loop 400 – 430 of NSAP is
character of the inhibition. directly involved in the binding of collagen (12). Although the
The knowledge of the structural organization of these pock- case of PLAP is more controversial, our structural data allow
ets offers a molecular basis for the design of more specific us to speculate that the enzyme may have a specific target,
inhibitors, which may be useful in drug therapy when involve- whereas the bacterial enzyme has none. First of all, in the
ment of PLAP in tumor growth is confirmed. Our study of the active site itself, there are two pockets that may accommodate
Crystal Structure of AP from Human Placenta at 1.8-Å Resolution 9163

FIG. 4. Residues involved in the al-


lostery. Ribbon representation of the ac-
tive site is shown, with monomer 1 in
green, and monomer 2 in magenta. The
network Glu-429-His-320-Zn1 is circled
in blue, the network Tyr-367(#2)-His-
432(#1)-Zn1 in yellow, and the residues
involved in the stabilization of the N-ter-
minal from the second monomer are cir-
cled in brown.

Downloaded from http://www.jbc.org/ at UNIV OF UTAH on November 22, 2014


FIG. 5. Uncompetitive inhibition.
Shown is modeling of L-Phe uncompeti-
tive inhibitor in the active site of PLAP.
The L-Phe amino acid is in stick represen-
tation colored in yellow. PLAP is in Corey-
Pauling-Koltun representation with one
monomer colored in white and the second
colored in pink. Residues of PLAP inter-
acting with L-Phe are colored in residue
type: acidic in red, basic in blue, neutral
in green, aromatic in violet. The metal
ions are colored in orange. This figure was
made with TURBO.

residues from the target protein. Should such a protein bind at from the metal binding domain. This loop interacts with and
this position, Glu-429 would be ideally positioned to stabilize may be stabilized by the putative calcium ion. It could therefore
the ester moiety of the substrate in the initial step of the be responsible for the synergism observed between calcium and
catalytic process. This residue is probably a key determinant PLAP activity by intervening in substrate binding (15). In
for substrate selection. In addition, the hydrophobic pocket summary, the organization of the extended active site would be
extends on one side of the active site into a large surface of consistent with PLAP interacting with a large substrate. Such
about 1700 Å2, which with the exception of Asp-173, comprises a substrate is likely to be a phosphorylated protein.
only neutral or hydrophobic residues from both monomers. The Comparison with the Other Human Alkaline Phospha-
size and character of this region is similar to that typically tases—An important issue in the use of PLAP as a cancer
observed in protein-protein interactions (32), and it would be marker is the assessment of the potential to differentiate it
an ideal location to stabilize a protein substrate. As we move from the other human alkaline phosphatases present in serum.
away from the core of the active site, we notice a large valley All mammalian APs have closely related sequences, conserving
with an accessible surface of 4000 Å2, flanked on each side by all the three specific elements: the four cysteines involved in
the crown and metal binding domains (Fig. 3). The bottom of the disulfide bridges, the glycosylation sites, and the residues
this valley is also lined by the loop containing residues 270 –285 that coordinate the fourth metal ion. Hence, it is likely that the
9164 Crystal Structure of AP from Human Placenta at 1.8-Å Resolution
same ion and glycosylation sites are present in all APs. The
core is thus largely preserved, and the divergence between the
four proteins is concentrated in the elements of specificity.
PLAP and GCAP are the closest 2, with only 10 substitutions
between them (33). Eight of these positions have sufficient
solvent exposure to differentiate between the surfaces of these
two enzymes (Fig. 6). There are 58 substitutions between PLAP
and intestinal AP, corresponding to about 12% of all residues
(Fig. 6). Half of these are situated on the specific elements that
encompass less than one-third of the protein residues. The
greatest diversity exists between PLAP and NSAP, with 209
substitutions (Fig. 6). 56% of the 161 residues in the specific
elements are substituted in NSAP, consistent with their in-
volvement in the mediation of specificity and modulation of
activity. Finally, three epitopic surfaces distinguish PLAP from
the other APs. One corresponds to the active site pocket in
proximity to Glu-429, the second corresponds to the metal-
binding site, and the third corresponds to the region comprising
Glu-15 and Ile-67 (Fig. 6).
Conclusion—The structure of human PLAP, the first struc-

Downloaded from http://www.jbc.org/ at UNIV OF UTAH on November 22, 2014


ture of a eukaryotic example of this family of enzymes, gives
some hints regarding the evolution of this enzyme. Compared
with the bacterial homologues, the catalytic requirement has
helped the preservation of the overall core of the enzyme, the
main secondary structure, and the residues necessary for the
catalysis. However, the development of a placenta in mammals
has been accompanied by the emergence of tissue specificity
and the coadaptation of the enzyme to a specific protein target.
The acquisition of allostery and the uncompetitive inhibition
imply that this enzyme can be regulated in vivo. These new
properties have led to extensive structural modification both in
the active site and at the enzyme surface, in agreement with
the hypothesis that the target of PLAP may be a phosphoryl-
ated protein.

Acknowledgments—We are very grateful to Dr. J. C. Boulain and


Dr. J. B. Charbonnier for fruitful discussion during the course of this
study. We also thank Dr. A. Poupon for help in the use of the HCA
program and A. Bentley for help in the data collections at LURE station
W41.

REFERENCES
1. Millán, J. L. (1988) Anticancer Res. 8, 995–1004
2. Millán, J. L. (1992) Clin. Chim. Acta 209, 123–129
3. Millán, J. L., and Fishman, W. H. (1995) Crit. Rev. Clin. Lab. Sci. 32, 1–39
4. Schwartz, J. H., and Lipmann, F. (1961) Proc. Natl. Acad. Sci. U. S. A. 47,
1996 –2005
5. Kim, E. E., and Wyckoff, H. W. (1991) J. Mol. Biol. 218, 449 – 464
6. Coleman, J. E. (1992) Annu. Rev. Biophys. Biomol. Struct. 21, 441– 483
7. Hoylaerts, M. F., Manes, T., and Millán, J. L. (1997) J. Biol. Chem. 272,
22781–22787
8. Hummer, C., and Millán, J. L. (1991) Biochem. J. 274, 91–95
9. Watanabe, T., Wada, N., Kim, E. E., Wyckoff, H. W., and Yang Chou, J. (1991)
J. Biol. Chem. 266, 21174 –21178
10. Hoylaerts, M. F., Manes, T., and Millán, J. L. (1992) Biochem. J. 286, 23–30
11. Wu, L. N. Y., Genge, B. R., Lloys, G. C., and Wuthier, R. E. (1991) J. Biol.
Chem. 266, 1195–1203
12. Bossi, M., Hoylaerts, M. F., and Millán, J. L. (1993) J. Biol. Chem. 268,
25409 –25416
13. Chan, J. R., and Stinson, R. A. (1986) J. Biol. Chem. 261, 7635–7639
14. Telfer, J. F., and Green, C. D. (1993) FEBS Lett. 329, 238 –244
15. She, Q., Mukherjee, J. J., Huang, J., Crilly, K. S., and Kiss, Z. (2000) FEBS
Lett. 469, 163–167
16. Dabare, A. A., Nouri, A. M., Cannell, H., Moss, T., Nigam, A. K., and Oliver,
R. T. (2000) Urol. Int. 63, 168 –174
17. Butikofer, P., Vassella, E., Ruepp, S., Boschung, M., Civenni, G., Seebeck, T.,
FIG. 6. Comparison of human alkaline phosphatases. a, surface Hemphill, A., Mookherjee, N., Pearson, T. W., and Roditi, I. (1999) J. Cell
representation of the overall structure of the dimer of PLAP colored in Sci. 112, 1785–1795
ivory with the residues differing from the sequences of GCAP in color. 18. Makiya, R., and Stigbrand, T. (1992) Eur. J. Biochem. 205, 341–345
Residues unique to PLAP are blue, and residues conserved with at least 19. Makiya, R., and Stigbrand, T. (1992) Biochem. Biophys. Res. Commun. 182,
one other human AP are orange. b, sequence alignment of PLAP, GCAP, 624 – 630
20. Stefaner, I., Stefanescu, A., Hunziker, W., and Fuchs, R. (1997) Biochem. J.
intestinal AP, and NSAP. The first line of the alignment corresponds to
327, 585–592
the consensus sequence for human APs. Also included are residues 21. Fishman, W. H., Inglis, N. R., Stolbach, L. L., and Krant, M. J. (1968) Cancer
unique to PLAP, shown as stars highlighted in blue. Residues that are Res. 36, 935–938
not conserved with PLAP are highlighted in green, and residues homol- 22. Holmgren, P. A., Stigbrand, T., Damber, M. G., and von Schoultz, B. (1979)
ogous with PLAP are highlighted in yellow. Obstet. Gynecol. 54, 631– 634
23. Stura, E. A (1999) Seeding Techniques in Crystallization of Nucleic Acids and
Crystal Structure of AP from Human Placenta at 1.8-Å Resolution 9165
Proteins (Ducruix, A., and Giegé, R., eds) pp. 177–206, Oxford University 30. Mac Phalen, C. A., Strynadka, N. C. J., and James, M. N. G. (1991) Adv.
Press, Oxford Protein Chem. 42, 77–144
24. Otwinowski, Z., and Minor, W. (1997) Methods Enzymol. 276, 307–326 31. Holm, L., and Sander, C. (1998) Nucleic Acids Res. 26, 316 –319
25. Navaza, J., and Saludjian, P. (1997) Methods Enzymol. 276, 581–594 32. Lo Conte, L., Chothia, C., and Janin, J. (1999) J. Mol. Biol. 285, 2177–2198
26. Callebaut, I., Labesse, G., Durand, P., Poupon, A., Canard, L., Chomilier, J., 33. Hoylaerts, M. F., and Millán, J. L. (1991) Eur. J. Biochem. 202, 605– 616
Henrissat, B., and Mornon, J. P. (1997) Cell. Mol. Life. Sci. 53, 621– 645 34. Roussel, A., and Cambillau, C. (1989) Silicon Graphics Geometry Partner
27. Perrakis, A., Morris, R., and Lamzin, V. S. (1999) Nat. Struct. Biol. 6, 458 – 463 Directory, pp. 77–78, Silicon Graphics, Mountain View, CA
28. Laskowski, R. A., MacArthur, M. W., Moss, D. S., and Thornton, J. M. (1993) 35. Koradi, R., Billeter, M., and Wüthrich, K. (1996) J. Mol. Graph. 14, 29 –32,
J. Appl. Crystallogr. 26, 283–291 51–55
29. Satow, Y., Cohen, G. H., Padlan, E. A., and Davies, D. R. (1986) J. Mol. Biol. 36. Brünger, A. T. (1992) X-PLOR, A system for X-ray Crystallography and NMR.
190, 593– 604 Yale University Press, New Haven, CT, USA

Downloaded from http://www.jbc.org/ at UNIV OF UTAH on November 22, 2014

You might also like