You are on page 1of 9

Letters in Drug Design & Discovery, 2006, 3, 675-682 675

Prediction of the Conformation of the Human P2X7 Receptor


Peter P. Mager* , Peter Illes and Horst Walther

Institute of Pharmacology and Toxicology, University of Leipzig, Saxony, Germany


Received June 15, 2006: Revised July 25, 2006: Accepted July 26, 2006
Abstract: There is a hope that inhibitors of the purinoreceptor P2X7, a ligand-gated ion channel, would have a
beneficial role in the therapy of inflammatory processes. Therefore, the P2X7 channel is a putative drug target
but neither X-ray nor NMR analyses have been made. It was attempted to predict the conformations of P2X
receptor subunits using homology-based comparative modelling and threading. The modelling could not be
carried out because of missing template proteins. The paper reviews a new method to predict the conformation
of proteins. The method is exemplified on the human P2X7 (h-P2X7) receptor. The coordinates of the spatial
structure of the h-P2X7 protein were determined by a profile-based neural network prediction. The
conformation was geometry optimised using the quantum chemistry RHF/3-21G minimal basic set and all-
atom molecular mechanics AMBER force field. A dielectric constant of ε = 3.5 was used to simulate the
lipophilic environment of the membrane-bound glycoprotein. The h-P2X7 protein has a number of interacting
peptide modules. The discovery of peptide module raises the possibility of exploiting structure-based
strategies to design h-P2X7 inhibitors.
Keywords: Structural bioinformatics, Proteomics, Drug target, Human P2X7 receptor, Ligand-gated ion channel.

INTRODUCTION State-of-the-art homology modelling techniques and


automatic ab initio prediction methods could also not solve
The membrane-embedded purinoreceptor P2X family
the problem (see Appendix). Therefore, a hybrid technique
belongs to the large class of membrane-embedded
has been proposed which consists of structural
glycoproteins. It has two transmembrane domains and a core
bioinformatics, secondary structure prediction, subsequent
of five extracellularly occurring disulfide bonds. Neither X-
geometry optimisation, and comparative sequence-function
ray nor NMR analyses have been made because of technical
analysis [6].
difficulties. Currently, seven different P2X receptor proteins
(P2X1 to P2X7) have been cloned. This review summarises the method using the h-P2X7
receptor subunit. It is shown that the new technique led to
The P2X7 subunit is an unusually large P2X protein
the discovery of interacting peptide modules. The modules
which can operate as trimeric, membrane-embedded,
may provide new insights for further experimentation and
monovalent-bivalent (Na+ , K+ , Ca2+ ) cation channel. It
serve as an explanatory guide for designing selectively acting
opens on binding of extracellularly occurring adenosine 5'-
h-P2X7 inhibitors.
triphosphate (ATP) to produce postsynaptic depolarisation
and excitation [1,2]. Additionally, agonist stimulation for
more than a few seconds causes a reversible expansion of the METHODOLOGY
P2X7 selectivity filter. The nonselective pore (with
The amino acid sequences of the human P2X7 (h-P2X7)
unknown stoichiometry and function) allows for the passage
receptor subunit have been taken from the Swiss Protein
of large molecules (≤ 900 Da). The human P2X7 (h-P2X7)
Sequence Database (Swiss-Prot, Swiss Institute of
protein is an ubiquitous receptor in many tissue types. It
Bioinformatics, and European Bioinformatics Institute,
differs from the other P2X subtypes in additionally coupling
EMBL). The primary accession number is Q99572. Among
to numerous other signalling cascades (e.g., a rapid release
the large number of approaches of secondary structure
of inflammatory cytokines), and influences actin
prediction, the PROFsec method was applied [7-11]. The
cytoskeleton organisation for many macrophage functions
results were used as starting point for a subsequent analysis.
including cell fusion, phagocytosis and apoptosis. The
To convert the two-dimensional sketch into a reasonable
P2X7 subunit plays a role as global thermistor of immune
conformational structure, the PROFsec results were uploaded
function, e.g., in astrocyte-mediated inflammation [3] of
into HyperChem program (Hypercube Inc., 419 Phillip
neurodegenerative diseases such as Alzheimer’s disease. The
Street, Waterloo, Ontario N2L 3X2, Canada). The side
misfolded Aβ(1-42) amyloid peptide of Alzheimer’s disease
chains of the h-P2X7 protein were added by building the
[4] can modulate the h-P2X7 receptor [5].
model in stereochemically preferred conformers using a
Unfortunately, sufficiently close targets to confidently library of protein bonds, angles, and torsion-angles
model the sequence of P2X proteins were not available. distributions. The tertiary and supersecondary folds are build
and then optimised from an assembly of secondary units by
*Address correspondence to this author at the Research Group of producing a single low-energy conformer.
Pharmacochemistry, Institute of Pharmacology and Toxicology, University
of Leipzig, D-04107 Leipzig, Härtelstr. 16-18, Saxony, Germany; The bonding states of the cysteines of h-P2X7 subunit
Tel: +49(0)041974636-2; Fax: +49(0)041974636-9, were determined by the CYSPRED program [12] and
E-mail: magp@medizin.uni-leipzig.de multiple alignment analysis [13,14] in combination with

1570-1808/06 $50.00+.00 © 2006 Bentham Science Publishers Ltd.


676 Letters in Drug Design & Discovery, 2006, Vol. 3, No. 10 Mager et al.

experimentally obtained results reviewed elsewhere [6]. conformationally flexible fragment (amino acid subsequences
Multiple alignments of P2X sequences were carried out in 398-595) which may be termed "winking-cloud“. It is only
three steps: all sequences were compared to each other observed in P2X7 subunits.
(pairwise alignments), a dendogram was constructed
describing an approximate grouping of sequences by The four putative nucleotide binding domains (NBDs)
similarity, and the final multiple alignment was carried out with positively charged Arg and Lys residues that are
using the dendogram as guide. The approaches used here are completely conserved in all species-dependent P2X subunits
the ClustalV and ClustalW algorithms. The PAM100 (with exception of NDB2 in mouse P2X4) are as follows in
Dayhoff Mutation Data Matrix (ClustalV) and the BLOSUM h-P2X7 [6]: NBD1: 63-67, NBD2: 188-194, NBD3: 292-
series (ClustalW) were applied to weight different pairs of 299, NBD4: 307-314.
aligned amino acids. The results suggested five-disulfide The putative protein kinase C (PKC) phosphorylation
bond pairs of the h-P2X7 receptor: pair 1: C119-C168, pair regulator domains are as follows: PKC1: 15-17 (conserved
2: C129-C152, pair 3: C135-C162, pair 4: 216-226, pair 5: in all species-dependent P2X subunits), PKC2: 28-30,
C260-C269 [6]. Geometry optimisation occurred by the PKC3: 47-49, PKC4: 124-126, PKC5: 131-133, PKC6:
HyperChem program (see above). It was assumed that the 149-151, PKC7: 204-206, PKC8: 274-276, PKC9: 397-
geometry optimised h-P2X7 receptor subunit is monomeric, 399, PKC10: 467-469, PKC11: 490-492, and PKC12: 555-
unoccupied with any ligand, and charged. The intramolecular 557. The h-P2X7 subunit does not include tyrosine protein
hydrogen bonding forces were included in the calculation kinase phosphorylation sites but a number of kinase-II
process. A hybrid QC/MM-model was used. It consists of phosphorylation sites (6-9, 86-89, 390-393, 402-405, 420-
the quantum chemistry (QC) RHF/3-21G minimal basic set 423, 508-511).
at the restricted Hartee-Fock level and all-atom molecular
mechanics (MM) force field. The partial charges were derived The extracellularly occurring N-glycosylation attachment
from restrained electrostatic potential fits to ab initio sites are N-gly1: 187-190 (strictly conserved in all species-
distributions obtained at the RHF/3-21G level (Gaussian94, dependent P2X subunits with exception of mouse P2X4), N-
Gaussian Inc., Carnegie Office Park, Pittsburgh, PA 15106). gly2: 202-205, N-gly3: 213-216, N-gly4: 241-244, and N-
As MM force field, the AMBER96 force field (Assisted gly5: 284-287 (Fig. 1).
Model Building and Energy Refinement 96) was applied.
Some regions in proteins represent classes of typically
The dielectric constant of ε = 3.5 simulated the behaviour of
aligned sequence motifs that identify potentially interesting
the membrane-embedded h-P2X7 subunit, and the interior of
motifs of a protein family. They are used to characterise the
the protein. To avoid interactions between the nonbonded
protein family because they encode protein folds and
atoms by a sharp cutoff which can cause discontinuities in
functionalities more flexibly and powerfully than can single
the potential surface, a smoothing applied from the inner
motifs. The general signature of P2X receptors represents the
radius (10 Angstroms) to the outer radius (14 Angstroms)
identifier code of evolutionarly selected functions and for the
was used. The structure was refined using a conjugate
maintenance of its spatial environment. The fingerprint is
gradient minimiser (Fletcher-Reeves modification of the
commonly found in the whole P2X receptor family and
Polak-Ribiere method). Convergence was obtained when the
observed in the extracellular domain. Related to the h-P2X7
gradient root mean square RMS was RMS ≤ 0.05
protein, the extracellular subsequence 249-275 represents the
kcal/Angstroms x mol. The results of the geometry
ATP-gated P2X receptor signature [6]. Pro271 is a part of
optimisation were checked by the Ramanchandran plot,
the conserved sequence motif. Three neighboured residues of
rotamer analysis, all-atom contact dots, and C(β) deviation
Pro271 are also completely conserved: C-x(1)-P-y(1)-Y-z(1)-
[6].
F where x(1), y(1), and x(1) are unspecified amino acids.
Naturally occurring posttranslational phosphorylation and Pro271 maintains probably an anti-misfolding capacity of
attachment sites were screened by using the knowledge-based the P2X receptor signature.
ScanPROSITE regular expression search via PROSITE's
interface [16]. The one-letter amino acid code is here used. Fig. 1 illustrates the predicted backbone structure of the
The general PROSITE signature SY-x(0,2)-[AL]-K-{P} h-P2X7 protein and the location of important domains.
describes a subpeptide where SY must be present, it is
followed by no amino acid or two unspecified amino acids,
then it follows A or L, the subsequent K must be present, COMPARISON OF STRUCTURAL FOLDS WITH
and P is not allowed. KcsA
The framework for the molecular architecture of ion
channels is the KcsA protein. It has been found that the
DOMAINS prokaryotic two-transmembrane K+ -KcsA channel and the
The categorisation of the domains into two intracellular P2X1, P2X3, and P2X4 receptor subtypes share a similar
cytoplasmic domains (ICDs), two transmembrane domains structural fold [6,17,18] although the primary structures and
(TMDs), and an extracellular domain (ECD) is as follows functions are quite different. The conclusion is also
[6]: the N-terminal ICD1: amino acid positions 1-25; the supported by the result of the h-P2X7 receptor. It appears
TMD1: 26-48; the ECD: 49-330 (a globular glycoprotein that the common folds of these cation channels represent an
domain with an asymmetrical charge distribution and a evolutionarily selected structure, independently of their
twisted loop, four putative nucleotide binding domains, and specific volume and function, that is, it seems that overall
five disulfide bridges); the TMD2: 331-350; and the C- three-dimensional shapes are better conserved during
terminal ICD2: 351-595. The ICD2 includes a large, bulky, evolution than sequences.
Conformation of h-P2X7 Letters in Drug Design & Discovery, 2006, Vol. 3, No. 10 677

Fig. (1). Result of structural bioinformatics applied to the membrane-embedded, monomeric h-P2X7 receptor. The result from
computational tools is a prediction and subject to further experimental verification. The structure is viewed parallel to the plane of
the membrane. The fully functional receptor is a trimeric ion channel. For visual clarity, the backbone structure of the amino acids is
illustrated. There are two intracellular domains, a N-terminal ICD1 (amino acid of the positions 1-25, and a C-terminal ICD2 (351-595,
it includes the "winking-cloud" in positions 398-595 which is a large, bulky, conformationally flexible fragment), two
transmembrane domains called TMD1 (26-48, and TMD2 (331-350), and an extracellular domain ECD (49-330, which is a globular,
twisted glycoprotein loop with an asymmetrical charge distribution and a disulfide core (marked by short yellow lines; pair 1: C119-
C168, pair 2: C129-C152, pair 3: C135-C162, pair 4: 216-226, pair 5: C260-C269). Colors: red, nucleotide binding domains NBD1
(63-67), NBD2 (188-194), NBD3 (292-299), NDB4 (307-314); yellow: PKC phosphorylation sites PKC1 (completely conserved, 15-
17), PKC2 (28-30), PKC3 (47-44), PKC4 (124-126), PKC5 (131-133), PKC6 (149-151), PKC7 (204-206), PKC8 (274-276), PKC9
(397-399), PKC10 (467-469), PKC11 (490-492), PKC12 (555-557); green: N-glycosylation attachment sites N-gly1 (completely
conserved, 187-190, not shown here due to visual clarity), N-gly2 (202-205, not shown here due to visual clarity), N-gly3 (213-216),
N-gly4 (241-244), N-gly5 (284-287); violet (left): signalling peptide (1-44); violet (right): cavity and gate/filter of the channel-
forming motif (332-344, 345-353); white: trafficking motif (383-387); dark blue: fingerprint (249-275). The kinase II
phosphorylation sites are not shown here.
PEPTIDE MODULES such interacting peptide modules of h-P2X7, some should
be mentioned:
The prototype of a protein that requires interacting
peptide modules is the acetylcholinesterase [15]. It was (1) There are many differences between the "orthodox ion
suggested that a functional P2X channel requires also a channels" and the membrane-embedded P2X ion
precise coupling between distinct regulator modules that are channel. The filter and the gate of P2X receptors are
important for signal transduction [6]. Among the number of commonly localised by an area at the end of the
678 Letters in Drug Design & Discovery, 2006, Vol. 3, No. 10 Mager et al.

TMD2 and the beginning of the ICD2. It has been protein includes the amino acid positions YYRKK in
proposed that the door/cavity module of the h-P2X7 positions 383-387. The motif is in neighbourhood to
channel is formed by the amino acids in positions the PKC9 (TLK, 397-399) and kinase-II
332-344, and the gate/filter module is located in the phosphorylation sites (SIVE, 390-393) and probably
positions 345-353 [6]. The channel of the P2X involved in the maintenance of cell-surface
receptors is gated by ATP but the ATP binding center expression. A number of other C-terminal trafficking
is outside the proper channel. The octapeptide module motifs that are not conserved must be envisaged, too.
NFRYAKYY of the NBD3 (292-299, the completely (4) An example of a triad of sterically interacting peptide
conserved residues are marked in bold-face letters) lies modules is the nucleotide binding domain NBD4
directly over the top of the door of the putative (RTLIKAFG, 307-314), the N-glycosylation
channel-forming motif. It was suggested that ATP- attachment site N-gly5 (NVSL, 284-287), and the
complexed NBD3 opens the channel, and an unbound PKC5 phosphorylation site (SDR, 131-133). Fig. 1
NBD3 closes the channel, that is, a periodic illustrates the modules. The interaction of ATP with
conformational switch of the channel forming motif the NBD4 of the h-P2X7 receptor takes place in the
may occur. The residues which put into the cavity depth of the NBD4 pocket. The positive charge of the
and contribute to hydrogen-bonding forces are Arg and Lys residues supply charge shielding of the
probably involved in the control of the transport of negatively charged phosphate groups. Further details
hydrated cations through the channel. Dimeric models of an interaction of ATP with nucleotide binding
for P2X2 and P2X3 channel constructs have been domains see literature [6,22]. As expected, R307Q
proposed in literature [6,19]. polymorphism causes loss of function of the h-P2X7
(2) The second example deals with NBD2 (FTVLIKN, protein [23].
188-194) which is in direct neighbourhood to the
extracellularly occurring regulator domain PKC7
(TTR, 204-206) and two N-glycosylation attachment INTERNALISATION
sites, the conserved N-gly1 (NFTV, 187-190) and N- Subdomains are also involved in internalisation of the
gly2 (NYTT, 202-205). Fig. 1 illustrates the areas. complete protein. Regulation of the protein by ligand-
Experimental results have shown that the priming mediated internalisation consists of the formation of a
effect of nucleotides might be attributable to a complex between the target protein and other proteins (e.g.,
phosphorylation of the PKC sites of the ectodomain GRK-3-, clathrin-, β-arrestin-2-, and dynamin-mediated
[20]. internalisation of h-P2X7 into clathrin domains), formation
It is commonly accepted that only the extracellular of vesicles, transport to endosomes, and release of the target
domain of the P2X receptor has N-glycosylation protein. Then, fully assembled proteins are delivered back to
addition sites. The following rules were here used to their functional location, while misfolded proteins are
predict putative N-glycosylation sites. The peptide N- directed to lysosomes where their degradation occurs.
x(1)-[ST] represents the general signature of N- Unfortunately, the various proposed peptide clusters
glycosylation with the site Asn. The presence of the [2,24,25] for an endocytosis of internalised P2X receptors
imino acid proline between Asn and Ser/Thr will are not conserved.
inhibit N-glycosylation. The hydrophilic The h-P2X7 protein starts probably its endocytosis with
oligosaccharide chains are bound to the Asn residues the N-terminal intracellular ICD1 (Fig. 1). It includes the
of N-glycosylation addition sites (Fig. 1) and modify only completely conserved PKC phosphorylation site
the surface properties, and consequently, the energy of (PKC1, TNK, positions 15-17), the nonconserved PKC2
solvation, transport, and membrane-binding (TIK, 28-30), and a kinase-II phosphorylation site (SCSD,
properties of the protein. They may shield a large 6-9) which are parts of the signalling peptide (1-44). The
selection of the protein surface and are probably signalling peptide starts with the known methionine residue.
important for the expression at the cell surface of P2X The most likely cleavage site of signalling peptides is
channels. It appears that the N-glycosylation addition identified by an artificial neural network and a hidden
sites N-gly1 and N-gly2 of h-P2X7 are required for Marcovian model between the positions 44 and 45 [6]. Such
the proper folding and arrangement of the cleavage sites are one of the targets for designing specific
neighboured peptide modules, that is, they have a and efficient inhibitors. The T18 of P2X5 is homologous
stabilising effect on the structure of the NBD2 and with the T12 of P2X7, and it was shown that the T18A
PKC7. As the N-glycosylation addition sites mutant blocks P2X5 internalisation did not show ATP-
contribute in part to NBD2 and PKC7, the effect of activated currents [26].
single-point mutants can hardly be controlled. The
Asn residue N187 of the N-glycosylation attachment An example that extracellular domains may also
site of h-P2X7 is homologous with N182 of rat contribute to internalisation is probably the N-glycosylation
P2X2. Replacement of the double mutants 182/239 addition site N-gly4 (NFSD, 241-244; Fig. 1). Due to the
and 182/298 of Asn residues of the N-glycosylation twisted nature of the globular coil of the extracellular
addition sites of rat P2X2 by Gln led to a domain, the PKC3 phosphorylation site SDK (47-49) and
nonfunctional P2X2 subunit [21]. the signalling peptide are in steric neighbourhood to N-gly4
(Fig. 1).
(3) The non-functionality of P2X mutants is also due to
trafficking problems. The conserved C-terminal Quite recently, it was reported that ATP downregulates
trafficking motif Y-x(2)-[KRQT]-K of the h-P2X7 the P2X7 protein by ligand-mediated internalisation and a
Conformation of h-P2X7 Letters in Drug Design & Discovery, 2006, Vol. 3, No. 10 679

pathway that does not depend on cation flux [27,28]. In residue) lead to loss-function polymorphisms of the human
homology to the results of other proteins, it may be P2X7 protein [36,37]. The mutant E496A impairs the ATP-
speculated that an agonist-promoted down-regulation of P2X induced IL-1β and IL-18 release from monocytes [38].
receptors is also related to the N-gly4 subdomain. The N- The LPS-binding domain was identified between
glycosylation site (N187) of the β2-adrenergic receptor residues 573 and 590 which controls trafficking of h-P2X7
contributes to the internalisation during a long-term agonist and, there fore, modulates availability and activity during an
exposure, and the mutant N187D failed to display long-term inflammatory action [35]. The mutants R578E and K579E
agonist-promoted down-regulation [29]. N-linked glycosyla- of h-P2X7 led to trafficking defects [39] but it must be
tion sites of the H+ -K+ -ATPase β-subunit are critical for stated that the dibasic amino acids R578 and K579
folding and protect the internalised protein from targeting to (positions related to h-P2X7) are only conserved in human,
the degradation pathways [30]. rat and mouse P2X7, but in frog P2X7 they are missing and
replaced by E544 and T545. However, the number of
THE "WINKING-CLOUD" OF THE P2X7 PROTEIN matches of the "winking-cloud" of the P2X7 family with
proinflammatory cytokines, such as the interleukins IL-1β,
Although the C-terminal portion might to be essential IL-4, IL-6, IL-10, IL-13, IL-18, 1β (which must be activated
for multiple functions, little information is known regarding by caspase-1), is negligible.
the structural motifs that are involved in receptor
localisation, trafficking, and signal transduction. It was
suggested that the fragment 573-590 of the ICD2 of h-P2X7 PROLINE RESIDUES
also contains several protein-protein and protein-lipid
interaction motifs [31], which may interact with areas that The hydrophobic, nonpolar imino acid proline plays a
are involved in ligand-induced gating of the channel [32] and particular role in proteins, it forms a peptidyl-prolyl bond
promote lipid-dependent endocytosis [33]. that is incompatible with the peptide bond geometries. The
following extracellularly occuring proline residues are
The subdomain 398-595 ("winking-cloud", lipid- strictly conserved in all species-dependent P2X subunits (the
interaction motif) includes three PKC phosphorylation sites position is related to h-P2X7): P96, P169, P227 (with
(PKC10: 467-469, PKC11: 490-492, and PKC12: 555-557) exception of mouse-P2X7 where Pro is replaced by Ser), and
as regulator domains, three calcium/cyclic nucleotide- P271. Pro169 of h-P2X7 deserves special attention. It is
independent kinase-II phosphorylation sites (402-405, 420- included in the completely conserved subsequence WCP-
423, 508-511), and two putative N-myristoylation addition x(1)-E and contains the "proline-switch motif" E-x(1)-P that
sites (504-509, 586-591), which are probably responsible for has the potential to mediate conformer-specific ligand
irreversible lipid binding. Once added, the N-myristoyl binding resulting in an allosteric activation of the active site.
group increases the lifetime of the protein. Thus, the
myristoylated and surrounding motifs may contribute to the Like Gly, Asn, Ser and Asp, proline residues generally
targeting to specific intracellular compartments, reversible prefer turns or loops and have α- and β-breaking influence
interactions with transmembrane lipids and proteins, and onto the local conformation [6]. An analysis of the
transport. Some of these peptide modules are able to interact configuration of the modelled P2X3 has shown that the
(Fig. 1). The intracellularly occurring P474 located within peptide bonds nearly always have a trans configuration
the ICD2) s only present in the "winking cloud" of P2X7 which is energetically more favourable than cis [6]. The
(human, rat, mouse, frog). The signature is [SP]-P-x(1)-WC- frequency of a cis and trans configuration of proline residues
[QK]. depends, at least in part, on the neighboured residues. In the
modelled P2X3 protein, the cis configuration of proline was
The localisation of P2X7 to the cell surface is tightly found more frequently [6].
regulated by a C-terminal interaction motif resembling a
binding site for the subcellularly located human The high activation barrier (14-24 kcal/mol) for the
lipopolysaccharide receptor [34]. The lipopolysaccharide slowly occuring cis/trans-isomerisation (time constants of 10
receptor (LPS-R) regulates upon nucleotide stimulation a to 100 sec) through a planar transition state could be
variety of cell signalling mechanisms. Experimental studies involved in the rate-limiting step of slow-folding kinetics of
have shown that h-P2X7 modulates the production of proteins, adds probably to their flexibility, and assists in
numerous LPS-R-stimulated inflammatory mediators, e.g., conformational changes due to changes in the solvent-
proinflammatory cytokines, NF-κB-dependent inflammatory accessible, extracellular surface area [40-43]. While most
cytokines, and macrophage production of TNF-α [34,35]. structures reveal peptidyl-prolyl bonds that adopt either cis
The "winking-cloud" of the P2X7 family (studied species: or trans configuration, other exhibit conformational
human, rat, mice, frog) includes conserved fragments that heterogeneity about one or more peptidyl-prolyl bonds. It
match with important areas of the LPS-R. Related to the should be added here that the possible existence of both
amino acid positions of human P2X7, the subdomains are as isomeric forms of the native conformation may only be
follows: [LF]-[KR]-x(1,2)-F-x(1)-D (398-405), V-x(5)-G- observed in solution and in membrane-embedded protein but
x(2,3,4)-Q-x(3)-G (412-426), [EQ]-L-x(6)-G (496-504), and not in the crystal structures. Furthermore, crystal structures
W-x(8,9)-FA-x(0,7)-[IVG] (556-568; x = 0 is valid for the may not be of sufficient resolution to provide evidence for
P2X7 family, V of [IVG] is present in frog P2X7, x = 7 and occurring cis/trans-isomerisation. An inhibitor of the
G are found in PLS-R). The mutant E496A (replacement of isomerisation is the azetidine-2-carboxylic acid.
the charged, hydrophilic Asp in position 496 by the The peptidyl-prolyl-cis/trans isomerase family isomerises
hydrophobic Ala residue) or the mutant I568N (replacement the [TS]-x(0)-P motif which is a target for a phosphorylation
of the lipophilic Ile in position 568 by the hydrophilic Asn of Thr/Ser by proline-directed protein kinases. The reversible
680 Letters in Drug Design & Discovery, 2006, Vol. 3, No. 10 Mager et al.

phosphorylation is also called proline-directed of the maximum-likelihood criterion (g = 0.29, 5%


phosphorylation, it is a major signalling mechanism in significance level). The standard error of the regressand was
diverse cellular processes such as cell proliferation and 0.67, and the test statistics for examining the null
differentiation [40,41,43]. The P2X7 subunit does not hypothesis of zero regression coefficients were TS1 = 2.68,
include SP, TP, or the reverse Pro-Thr/Ser, in contrast to the TS 2 = 2.93, and TS3 = 2.32 (critical quantile g = 2.07 at
other P2X subunits (S289P290 in P2X4, S86P287 in the 5% significance level). There was no significant
P2X1; T18P19 in P2X1, T30P31 in P2X2, T17P18 in collinearity and multicollinearity, no high-leverage point,
P2X4, T110P11 in P2X5, T109P11 in P2X6; P330T331 in and no influential point. Other parameters, such as the
P2X1, P340T341 in P2X2; the positions are related to logarithm of the distribution coefficient P (octanol/water, pH
human proteins). However, the strictly conserved Pro96 7.4), and an indicator variable XD which parameterises the
(related to h-P2X7) of the complete P2X family is part of bridges X = CH2CH2 (XD = 1) and CH2 (XD = 0) of the
the [TS]-x(1)-P signature of the P2X7 subfamily where x(1) lead structure (Fig. 1), were insignificant. There was a hope
denotes a single apolar amino acid. The function of x(1) is to improve the fit by a generalised-regressionartificial-neural
still an open question (hydrophobic distance spacer?). network (GRNN) using an unconstrained genetic algorithm.
In contrast to the regression analysis, the genetic GRNN
ignored relevant and added noisy descriptors although the
PERSPECTIVES IN MEDICINAL CHEMISTRY goodness-of-fit criterion was strongly improved due to
This review dealt with a hybrid approach consisting of overfitting (R2 = 0.98). Consequently, the genetic GRNN
structural bioinformatics, secondary structure prediction, was considered as supplementary tool but could not replace
subsequent geometry optimisation, comparative sequence- the traditional QSAR methodology.
function analysis, and experimental findings. It was
surprising to see that the predicted structural P2X1, P2X2,
P2X3, and P2X4 structures [6,17-19,22] agreed with the
P2X models proposed from biochemical, neurophysio-
logical, pharmacological, and mutation experiments [1-
NH
2,20,44-46]. The same conclusion can be drawn with respect
to P2X7 [47]. N
X
Lead molecules for h-P2X7 inhibitors are reviewed
elsewhere [50]. Originally developed as inhibitor of the
calcium/calmodulin-dependent protein kinase-II, 1-N,O-
bis(1,5-isoquinolinesulfonyl)-N-methyl-L-tyrosyl)-4-phenyl- R
piperazine derivatives (KN-62) have been recognised as Fig. (2). 4,5-diarylimidazolines as inhibitors of the human
selectively acting antagonists of the h-P2X7 protein. KN-62 P2X7 receptor, a potential target for a therapy of Alzheimer’s
analogues block currents in cells expressing the h-P2X7 disease and rheumatoid arthritis.
receptor but have little effect on rat P2X7 [48-51]. However,
neither of their actions on h-P2X7 appears to be related to Although the so-called computational molecular docking
calmodulin or the calcium/calmodulin-dependent protein problem is far from being solved and there are limitations in
kinase-II. Quantitative structure-activity relationships of KN- each docking strategy, significant progress has been made
62 analogues [6] have shown the role of steric, electronic, [55], and the predicted conformation of the ligand-gated h-
and hydrogen-bonding properties of the substituents while P2X7 protein may be useful for designing experiments
lipophilic parameter seem to play a less important role. directed towards the understanding of blocking mechanisms.
For the large h-P2X7 protein, subdomain docking is
Potent h-P2X7 inhibitors are also 4,5-diarylimidazolines. probably the only rational way. In particular, the interacting
The search for novel 4,5-diarylimidazolines (Fig. 2) occurred peptide modules are potential targets. Their discovery raises
by an extensive use of high throughout screening the possibility of exploiting structure-based strategies to
approaches, utilising diverse compound library sources [52]. develop h-P2X7 inhibitors that are of paramount
As regressand, the unscaled 50% inhibitor concentration was pharmacological importance.
employed (Y = IC50). Because of the presence of flagged
observations, a major problem in developing a QSAR
equation was to select suitable chemical descriptors from a APPENDIX: MODEL LIMITATIONS
pool of variables. The "best way" was to employ a To avoid semantic confusion, it should be noted that
simultaneous one-regression/one-observation leaving-out "prediction" is defined in a probabilistic sense. Matches to
resampling of regression analysis [53,54]. The final generic rules do not mean "this is true" but rather "this
regression equation was equal to might be true". Only biological and chemical knowledge can
IC50 = -0.35 - 1.20(σ+ ) - 1.18(pKH) - 1.42(Es(ortho)) determine whether or not these predictions are meaningful.
(sample size of the training set: n = 26) where σ+ is an Methods of secondary structure prediction are based on
electronic substituent constant, pKH is the negative algorithms that concentrate on predicting the most suitable
logarithm of the equilibrium constant KH of hydrogen conformation on the basis of the properties and interactions
binding between substituted benzenes and phenol as of amino acids. As underlying theories are usually applied:
hydrogen donor, and Es(ortho) is Taft's steric constant for information and statistical theories, homology method, k-
ortho substituents. The squared multiple correlation nearest-neighbours (kNN), hierarchical (HNN) and
coefficient R2 = 0.62 was larger than the critical quantile g bidirectional recurrent (BRNN) methodologies of artificial
Conformation of h-P2X7 Letters in Drug Design & Discovery, 2006, Vol. 3, No. 10 681

neural network (ANN) analyses, deterministic methods of 2006) but there was no success because the input sequence
combinatorial optimisation, empirical rules, and hybrid was too long so that Rosetta simulations and HMMstr-CM
techniques. The approaches vary in their average accuracy but contact maps have been disabled. The limit is less than 150
the majority of the predicted secondary structure cluster do amino acids.
not differ markedly.
The P2X subunits can form oligomers that are viewed as
The following methods were used for comparing the probably trimeric assemblies consisting of homomeric
results of secondary structure prediction: GOR (versions I and/or heteromeric subunits which differ in their biological
and IV of the Garnier approach), SOPMA (self-optimised and chemical properties. With respect to the complete P2X7
prediction with alignments of homologous sequences), subunit, the modelling of trimeric protein is not possible
APSSP and APSSP2 (combination of nearest neighbour due to the large number of amino acids. Furthermore, the
with ANNs and multiple alignment analysis), SSpro2 and sites of contacts are unknown. Therefore, the proposal of a
SSpro8 (BRNN), PSIpred (prediction based on position- referee to imagine the possible trimeric structure cannot be
specific scoring matrices incorporating four feedforward realised at the present state of knowledge.
ANNs), SAM-HMM and SAM-T99-2d (prediction by
combining kNN and multiple sequence alignment), Molecular dynamics (MD) simulation can add explicit
PROFsec (a profile-based ANN prediction from multiple water molecules around a protein, the use of a periodic box
sequence alignments), PSA (probabilistic discrete state-space prevents solvent molecules from diffusing away or
model with optimal filtering and smoothing methods), and evaporating during a MD calculation. Unfortunately, the
DSSP (pattern recognition of hydrogen-bonded and P2X7 molecule is too large (bounding box 103, 92, 274
geometrical features). An example of hybrid methods is a Angstroms) to solvate in a box containing the TIP3P model
multivariate linear regression combination (MLRC) which of water molecules, this is no drawback, however. Compared
consists of the SOPMA, GORIV, and SIMPA96 methods. with the use of continuum electrostatics, MD is often
insufficient to describe solvation effects due to the lack of
The majority of the programs tended to overestimate the description for electronic polarisations. Although the
role of α-helices in transmembrane domains because the constant-dielectric and distance-dependent dielectric
generic algorithms were intentionally constructed from a set functions are not based on rigorous physics, they provide a
of a priori model assumptions. For example, low-resolution relatively fast evaluation of solvation effects and the second
experiments suggested that most helices span over 17-25 derivatives of the electrostatic energy. The native fold in
residues and most loops between two helices are longer than solutions was simulated by varying the dielectric constant ε
15 residues. For high-resolution data it was found that only (1-4 scale factors: electrostatic 0.5, van-der-Waals 0.5). The
50% fall into this interval and that 50% of the loops were protein is dehydrated if ε = 1 is used. A model that captures
shorter than 10 residues. Secondary structure prediction the ionisation properties of surface residues of proteins (ε =
methods use often low-resolution input data, however. 42) is the solvent-accessibility model. In the Poison-
Furthermore, the hypothesis that 80% of proteins have α- Boltzmann model, the dielectric constant is much lower (ε =
helical transmembrane domains have been generalised. Thus, 4). A dielectric constant of ε = 3.5 simulates an apolar
the frequency of helices in TMDs is overestimated. The environment, the behaviour of the membrane-embedded
PROFsec method simulates the influence of α-helical P2X7 subunit, and the interior of the protein. When
breaking amino acids in a more satisfactory degree. conformational relaxation is treated explicitly, ε = 3.5 yields
It was believed at first glance that ATP-binding proteins reasonable results, especially, when the neutral and ionised
with two transmembrane domains can be used as putative states of the groups are treated separately. The buried charged
templates in a homology-based molecular modelling groups make minimal contact with side chain or backbone
process. Many modern tools of homology-based molecular polarised atoms. Thus, ε = 3.5 was thought to include
modelling are Web server based as opposed to installable contributions by relaxation of permanent dipoles in addition
software for local use key. Unfortunately, sufficiently close to electronic polarisability.
targets were not available (e.g., ProModII of the To build dimeric P2X channel constructs of the 22
SwissModel Server, Aug 22, 2001, and Jun 02, 2003; 3D- amino acids that form the proper channel, distance restraint-
JIGSAW, PSI-BLAST, and CPHmodels Servers, Nov 05, based protein-protein docking was applied [6,18]. The
2002; SDSC Server, Nov 13, 2002; Superfamily HMM resulting constructs were again geometry optimised [6]. The
Library and Genome Assignments Server, Jun 02, 2003). proposed dimeric model satisfied the rules of a general
Other programs presented outputs but indicated that the fold architecture (length, diameters) of P2X ion channels but the
recognition results were probably not worthy of attention transport of hydrated cations and counterions in the central
(3D-PSSM Server, Nov 05, 2002; mGenTHREADER, Nov cavity could not be modelled. However, the building of
06, 2002). Modelling techniques such as Modeller or the trimeric channel constructs failed. Furthermore, to simulate a
respective modules of SYBYL or MOE also could not solve cation flux, the dielectric constant of the open P2X ion
the problem. channel (ε = 3.5) must be converted to the local dielectric
A referee has stated that some ab initio methods deal behaviour of intra-channel water (ε = 80) ad bulk solution
successfully with the 3D packing of secondary structure conditions of hydrated cations (ε = 82, ionic strength 0.1M;
elements. The Monte Carlo fragment insertion method for ε = 97, ionic strength 0.01M). As consequence, a channel
tertiary structure prediction (ROSETTA) and its improved construct must be viewed with at least two dielectric
version did not give results, however. This first fully regions. When cations are included in the gate, the pattern of
automatic ab initio prediction method of the I- proton sharing is altered, and the ionisation state of the
sites/HMMstr/ROSETTA server has been used (Jun 12, channel side chains is changed. The solution of these
682 Letters in Drug Design & Discovery, 2006, Vol. 3, No. 10 Mager et al.

problems is computationally difficult and further research is [28] Kochukov, M. Y.; Ritchie, A. K. Am. J. Physiol. Cell Physiol.,
2004, 287, C992-C1002.
needed to develop suitable algorithms. [29] Mialet-Perez, J.; Green, S. A.; Miller, W. E.; Liggett, S. B. J.
In summary: template proteins that confidently model Biol. Chem., 2004, 279, 38603-38607.
[30] Vagin, O.; Turdikulova, S.; Sachs, G. J. Biol. Chem., 2004, 279,
the sequence of P2X7 receptor protein, and novel ab initio 39026-39034.
methods that were able to present predictions of medium- [31] Denlinger, L. C.; Fisette, P. L.; Sommer, J. A.; Watters, J. J.;
sized and larger proteins, are not available up-to-now. Under Prabhu, U.; Dubyak, G. R.; Proctor, R. A.; Bertics, P. J. J.
these circumstances, it is sometimes better to use a simple Immunol., 2001, 167, 1871-1876.
method that works (PROFsec and subsequent geometry [32] Klapperstück, M.; Büttner, C.; Schmalzing, G.; Markwardt, F. J.
Physiol. (Lond.), 2001, 534, 25-35.
optimisation) than state-of-the-art methods that cannot work [33] Markwardt, F. Letter to the author, Sep 11, 2003.
up-to-now for large proteins. [34] Denlinger, L. C.; Angelini, G.; Schell, K.; Green, D. N.;
Guadarrmara, A. G.; Prasbhu, U.; Coursin, D. B.; Bertics, P.;
Hogan, K. J. Immunol., 2005, 174, 4424-4431.
REFERENCES [35] Ferrari, D.; Pizzirani, C.; Adinolfi, E.; Memoli, R.; Curti, A.;
Idzko, M.; Panther, E.; Di Virgilio, F. J. Immunol., 2006, 176,
[1] North, R. A. Physiol. Rev., 2002, 82, 1013-1067. 3877-3883.
[2] Vial, C.; Roberts, J. A.; Evans, R. J. Trends Pharmacol. Sci., 2004, [36] G, B. J.; Zhang, W. Y.; Worthington, R. A.; Sluyter, R.; Dao-
25, 487-493. Ung, L. P.; Petrou, S.; Barden, J. A.; Wiley, J. S. J. Biol. Chem.,
[3] DiVirgilio, F. Immunol. Today, 1995, 16, 524-528. 2001, 276, 11135-11142.
[4] Mager, P. P. Med. Res. Rev., 1998, 18, 403-430. [37] Wiley, J. S.; Dao-Ung, L. P.; Li, C.; Shemon, A. N.; Gu, B. J.;
[5] Rampe, D.; Wang, L.; Ringheim, G. E. J. Neuroimmunol., 2004, Smart, M. L.; Fuller, S. J.; Barden, J. A.; Petrou, S.; Sluyter, R. J.
147, C571-C581. Biol. Chem., 2003, 278, 17108-17113.
[6] Mager, P. P.; Weber, A.; Illes, P. Curr. Topics Med. Chem., 2004, [38] Sluyter, R.; Dalitz, J. G.; Wiley, J. S. Genes Immun., 2004, 5, 588-
4, 1657-1705. 591.
[7] Pollastri, G.; Przybylski, D.; Rost, B.; Baldi, P. Proteins, 2002, 47, [39] Denlinger, L. C.; Sommer, J. A.; Parker, K.; Gudipaty, L.;
228-235. Fisette, P. L.; Watters, J. W.; Proctor, R. A.; Dubyak, G. R.;
[8] Chen, C. P.; Rost, B. Protein Sci., 2002, 11, 2766-2773. Bertics, P. J. J. Immunol., 2003, 171, 1304-1311.
[9] Chen, C. P.; Rost, B. Appl. Bioinformatics, 2002, 1, 21-35. [40] Lu, K. P.; Liou, Y. C.; Zhou, X. Z. Trends Cell Biol. 2002, 12,
[10] Rost, B. J. Struct. Biol., 2001, 134, 204-218. 164-172.
[11] Rost, B.; Sander, C. Proteins, 1994, 19, 55-72. [41] Lu, K. P. Trend Biochem. Sci., 2004, 29, 200-209.
[12] Fariselli, P.; Riccobelli, P.; Casadio, R. Proteins, 1999, 36, 340- [42] Andreotti, A. H. Biochemistry, 2003, 42, 9515-9524.
346. [43] Schutkowski, M.; Bernhard, A.; Zhou, X. Z.; Shen, M.; Reimer,
[13] Dayhoff, M. O.; Barker, W. C.; Hunt, L. T. Meth Enzymol., 1983, U.; Rahfeld, J.-U.; Lu, K. P.; Fischer, G. Biochemistry, 1998, 37,
91, 524-545. 5566-5575.
[14] Corpet, F. Nucl. Acids Res., 1988, 16, 10881-10890. [44] Khakh, B. S. Nature Rev.Neurosci ., 2001, 2, 165-174.
[15] Mager, P. P.; Weber, A. Drug Des. Discov., 2003, 18, 127-150. [45] Ennion S. J.; Evans, R. J. Mol. Pharmacol., 2002, 61, 303-311.
[16] Falquet, L.; Pagni, M.; Bucher, P.; Hulo, N.; Sigrist, C. J.; [46] Clyne, J. D.; Wang, L.-F.; Hume, R. I. J. Neurosci., 2002, 22,
Hofmann, K.; Bairoch, A. Nucl. Acids Res., 2002, 30, 235-238. 3873-3880.
[17] Mager, P. P.; Weber, A.; Sanchez, L.; Wirkner, K.; Illes, P. [47] Kim, M.; Jiang, L.-H.; Wilson, H. L.; North, R. A.; Surprenant,
Protein Pept. Lett., 2006, 13, 77-81. A. EMBO J., 2001, 20, 6347-6358.
[18] Mager, P. P.; Weber, A.; Walther, H.; Wirkner, K.; Illes, P. [48] Gargett, C. E.; Wiley, J. S. Br. J. Pharmacol., 1997, 120, 1483-
Curr. Proteomics, 2005, 2, 319-324. 1490.
[19] Mager, P. P.; Weber A.; Illes, P. Med. Chem., 2005, 1, 109-115. [49] Baraldi, P. G.; Del Carmen Nunez, M.; Morelli, A.; Falzoni, S.;
[20] Wirkner, K.; Stanchev, D.; Köles, L.; Klebinger, M.; Dihazi, Di Virgilio, F.; Romagnoli, R. J. Med. Chem., 2003, 46, 1318-
H.; Fleming, V.; Evans, R. J.; Fürst, S.; Mager, P. P.; Eschrich, 1329.
K.; Illes, P. J. Neurosci., 2005, 25, 7734. [50] Baraldi, P. G.; Di Virgilio, F.; Romagnoli, R. Curr. Topics Med.
[21] Torres, G. E.; Egan, T. M.; Voigt, M. M. Biochemistry, 1998, 37, Chem., 2004, 4, 1707-1717.
14845-14851. [51] Baxter, A.; Bent, J.; Bowers, K.; Braddock, M.; Brough, S.;
[22] Mager, P. P.; Illes, P. Expert Opin. Drug Discov., 2006, in press. Fagura, M.; Lawson, M.; McInally, T.; Mortimore, M.;
[23] Gu, B. J.; Sluyter, R.; Skarratt, K. K.; Shemon, A. N.; Dao-Ung, Robertson, M.; Weaver, R.; Webborn, P. Bioorg. Med. Chem.
L. P.; Fuller, S. J.; Barden, J. A.; Clarke, A. L.; Petrou, S.; Lett., 2003, 13, 4047-4050.
Wiley, J. S. J. Biol. Chem., 2004, 279, 31287-31295. [52] Merriman, G. H.; Ma, L.; Shum, P.; McGarry, D.; Volz, F.;
[24] Smith, F. M.; Humphrey, P. A.; Murrell-Lagnado, R. D. J. Sabol, J. S.; Gross, A.; Zhao, Z.; Rampe, D.; Wang, L.; Wirtz-
Physiol., 1999, 520, 91-99. Brugger, F.; Harris, B. A.; Macdonald, D. Bioorg. Med. Chem.
[25] Feng, Y.-H.; Wang, L.; Wang, Q.; Li, X.; Gorodeski, G. I. Am. Lett., 2005, 15, 435-438.
J. Physiol. Cell. Physiol., 2005, 288, C1342-C1356. [53] Mager, P. P.; Sanchez, L. Curr. Comput.-Aided Drug Design,
[26] Jensik, P.; Thomas, C. Pflügers Arch. Eur. J. Physiol., 2002, 444, 2005, 1, 163-177.
795-800. [54] Mager, P. P. John Wiley & Sons Inc., New York, 1991.
[27] Hiken, J. F.; Steinberg, T. H. Am. J. Physiol. Cell Physiol., 2004, [55] Halperin, I.; Ma, B.; Wolfson, H.; Nussinov, R. Proteins Struct.
287, C403-C412. Funct. Genetics, 2002, 47, 409-443.

You might also like