Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Fluorophore Labeled Kinase Detects Ligands That Bind within the MAPK Insert of p38α Kinase

  • Matthäus Getlik ,

    Contributed equally to this work with: Matthäus Getlik, Jeffrey R. Simard

    Affiliations Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany, Max-Planck-Institute of Molecular Physiology, Dortmund, Germany

  • Jeffrey R. Simard ,

    Contributed equally to this work with: Matthäus Getlik, Jeffrey R. Simard

    Affiliations Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany, Max-Planck-Institute of Molecular Physiology, Dortmund, Germany

  • Martin Termathe,

    Affiliations Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany, Max-Planck-Institute of Molecular Physiology, Dortmund, Germany

  • Christian Grütter,

    Affiliations Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany, Fakultät Chemie, Chemische Biologie, Technische Universität Dortmund, Dortmund, Germany, Max-Planck-Institute of Molecular Physiology, Dortmund, Germany

  • Matthias Rabiller,

    Affiliations Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany, Max-Planck-Institute of Molecular Physiology, Dortmund, Germany

  • Willem A. L. van Otterlo,

    Affiliation Department of Chemistry and Polymer Sciences, Stellenbosch University, Stellenbosch, South Africa

  • Daniel Rauh

    daniel.rauh@tu-dortmund.de

    Affiliations Chemical Genomics Centre of the Max Planck Society, Dortmund, Germany, Fakultät Chemie, Chemische Biologie, Technische Universität Dortmund, Dortmund, Germany, Max-Planck-Institute of Molecular Physiology, Dortmund, Germany

Abstract

The vast majority of small molecules known to modulate kinase activity, target the highly conserved ATP-pocket. Consequently, such ligands are often less specific and in case of inhibitors, this leads to the inhibition of multiple kinases. Thus, selective modulation of kinase function remains a major hurdle. One of the next great challenges in kinase research is the identification of ligands which bind to less conserved sites and target the non-catalytic functions of protein kinases. However, approaches that allow for the unambiguous identification of molecules that bind to these less conserved sites are few in number. We have previously reported the use of fluorescent labels in kinases (FLiK) to develop direct kinase binding assays that exclusively detect ligands which stabilize inactive (DFG-out) kinase conformations. Here, we present the successful application of the FLiK approach to develop a high-throughput binding assay capable of directly monitoring ligand binding to a remote site within the MAPK insert of p38α mitogen-activated protein kinase (MAPK). Guided by the crystal structure of an initially identified hit molecule in complex with p38α, we developed a tight binding ligand which may serve as an ideal starting point for further investigations of the biological function of the MAPK insert in regulating the p38α signaling pathway.

Introduction

During the last decade, a plethora of small molecule kinase modulators have been developed [1]. The majority of these molecules bind to the highly conserved ATP pocket and are often plagued by modest selectivity, which in the case of kinase inhibitors leads to the inhibition of multiple kinases [2], [3]. Thus, selective modulation of kinase function remains a major task and the identification of next generation ligands which bind to less conserved sites and specifically target non-catalytic functions of kinases such as protein-protein interactions, DNA binding and subcellular localization, are considered the next great challenges in kinase research [19]. Allosteric ligands that bind to distal binding sites and often stabilize catalytically inactive kinase conformations are prime examples of modern kinase inhibitor research and represent a promising approach towards the development of selective kinase inhibitors [4]. However, only a few kinases such as PDK1, Akt, Mek or Bcr-Abl are known to be regulated by such ligands [1], [3], [5], [6], [7] and approaches that allow for the unambiguous identification of molecules that bind to these less conserved sites are few in number.

Interestingly, several MAP kinases, cyclin dependant kinases (CDKs) and glycogen synthase kinase 3 (GSK-3) contain a hydrophobic pocket at their C terminus about 30 Å away from the ATP-pocket [8], [9]. This C-terminal insert regulates the intracellular localization of GSK-3, binds regulatory proteins in CDK2 and has been shown to bind substrates such as transcription factors and phosphatases in Erk2 [9]. Recently, Diskin et al. confirmed the existence of the MAPK insert in p38α using protein X-ray crystallography and proposed that this structural feature may “fine-tune” kinase activity.[10] This same binding site in p38α is formed between the α-helices 1L14 and 2L14, and the C-terminal domain of the kinase (Figure 1A). Structural biology studies of p38α in complex with β-octyl-glucopyranoside revealed flexibility of this region and its ability to accommodate small molecules. Binding of the detergent shifts the two α-helices 1L14 and 2L14 about 3 Å away from the C-lobe and induces a conformational change of the αEF/αF-loop (Figure 1B) [10]. Although, as yet no clear biological function can be attributed to this hydrophobic pocket in p38α, potent ligands which specifically bind to this allosteric site may offer a valuable starting point for the development of chemical biology tool compounds for the investigation of its biological function.

thumbnail
Figure 1. Crystal structure of p38α and lipid binding FLiK-assay.

(A) Crystal structures of wild type p38α (grey) in complex with ATP (red) and in complex with 1b (green, boxed). The alpha-helix C (blue), activation loop (orange), glycine-rich loop (green) and, MAPK insert (red) are highlighted. 1b binds to the lipid binding site and forms a direct hydrogen bond with Asp294. Furthermore, binding of the ligand is stabilized through a network of water-mediated hydro-gen bonds as well as π-π-interactions between the quinazoline core and the side chain of Trp197. (B) Conformational changes triggered by ligand binding to the p38α MAPK insert. Ser252 was mutated to Cys252 and was labeled with a fluorophore (large spheres) to generate a direct binding assay that detects structural changes in the conformation of the lipid binding site. (C) Acrylodan emission at 475 nm decreases upon binding of 1b to the lipid binding site of p38α. (D) Endpoint equilibrium measurements can be made to directly obtain the KD of 1b.

https://doi.org/10.1371/journal.pone.0039713.g001

Results

The lipid binding pocket of p38α has been reported to bind lipids [10] as well as a few small molecule fragments. Interestingly, these reported ligands do not bind exclusively to the MAPK insert, as structural studies have revealed secondary binding within the ATP-pocket [9], [11], [12]. To our knowledge, no high affinity compounds are known which exclusively bind to the lipid binding site of p38α. Thus, identification of new ligands with high affinity for this pocket might stimulate the development of more specific ligands which may enable future research to elucidate its function. To this end, we applied the FLiK approach to develop a fluorescent-labeled kinase assay system, which specifically takes advantage of the ligand-induced conformational change of α-helices 1L14 und 2L14 of the p38α MAPK insert. This HTS-amenable assay allowed the identification and characterization of 2-phenylquinazolines as ligands for this allosteric site. We were able to further develop higher affinity ligands and confirmed binding to this remote pocket using protein X-ray crystallography.

We initially screened a small molecule library using our previously reported FLiK assay, which detects ligand-induced conformational changes in the glycine-rich loop of p38α, [13] and weakly detected binding of the 2-phenylquinazoline 1b to p38α. To better understand the binding mode and how this ligand interacts with the glycine-rich loop, we crystallized 1b in complex with wild type p38α and were surprised to find the molecule bound exclusively within the p38α MAPK insert (Figure 1A) (PDB code 4DLI), far away from the glycine-rich loop. The bound ligand is stabilized by a complex network of interactions. The primary amine in the 7-position of the quinazoline core directly hydrogen bonds to the side chain of Asp294. The quinazoline core itself forms interactions with the peptide backbone of Lys249 and Ile250 via two mediating water molecules. Furthermore, the side chain of Trp197 stabilizes binding of the quinazoline core via π-π-interactions. Interestingly, the side chain of Ser251 forms two water-mediated hydrogen bonds to the peptide backbone of Trp197 that may further stabilize the conformation of the αEF/αF-loop and the flipped orientation of the Trp197 side chain which is adopted when the ligand is bound. The cyclopropyl substituent occupies a hydrophobic subpocket that is formed by the side chains of Leu195, Leu232, Leu236 and Leu291 in the back of this binding pocket. Finally, the secondary amine which bridges the cyclopropyl residue to the quinazoline core interacts with the side chain of Glu192 and the peptide backbone of both Leu291 and Asp292 via water-mediated hydrogen bonds. However, in order to accurately measure the affinity of these compounds and to understand structure-activity relationships, it was necessary to develop a direct binding assay to enable specific detection of ligands binding within the MAPK insert. We sought to take advantage of the flexibility of this site in response to ligand binding and applied the FLiK approach to this pocket by introducing a Cys mutation (S252C) via site-directed mutagenesis into the flexible loop bridging α-helices 1L14 and 2L14, to enable subsequent labeling of the Cys side chain with the environmentally-sensitive fluorophore acrylodan (Figure 1B). We expected that conformational changes induced by ligand binding to this site would change the conformation of the MAP insert and, consequently, the charged microenvironment in the vicinity of acrylodan. Indeed, the altered fluorescent properties resulted in a shift of the emission maxima in the ligand bound state (Figure 1C). Upon ligand binding to this site, the ratio of fluorescence intensities measured at these maxima changed in a dose-dependent manner, allowing direct determination of the binding constant KD (Figure 1D). Using this assay, the initial hit 1b bound with a KD of 2.4 μM to the p38α MAPK insert. SB203580 and BIRB-796, known p38α inhibitors which bind within the ATP binding site, were not detected up to a concentration of 10 µM (data not shown) confirming that the observed fluorescence response is due solely to binding to the p38α MAPK insert.

Guided by the crystal structure of 1b in complex with p38α we synthesized a focused set of 2-arylquinazolines designed to probe the MAP insert pocket and increase compound binding affinity within this site (Figure 2). We hypothesized that this cavity would tolerate larger and more hydrophobic side chains at the 4-position of the quinazoline core. Briefly, the quinazoline cores were generated by a Niementowski-type strategy which entailed condensing amidines with the appropriate amino carboxylic acids. Position-4 of the resultant quinazolinols was functionalized through chlorination using SOCl2, thereby enabling the subsequent introduction of primary amines to this position via nucleophilic substitution. Finally, the nitro groups on the quinazoline scaffolds were reduced utilizing Pd/C and ammonium formate as a hydrogen donor, to afford the corresponding 4,6-quinazolinediamines. The resulting SAR of this focused set of ligands followed the predicted trend with 8a being the tightest binder with a KD of 280 nM (Figure 3). As revealed by protein X-ray crystallography of the corresponding amine analog 8b (PDB code 4DLJ), the phenylethylene moiety extends further into the hydrophobic pocket when compared to 1b, suggesting that the deeper occupancy of this hydrophobic sub-pocket by the large substituent in the 4-position likely contributes to the increased affinity of 8a (Figure 4B). This also explains the slight loss of potency of 7a and 6a, which have progressively shorter moieties at this position. Interestingly, all corresponding amine analogs bound to the lipid binding pocket with lower affinities than their corresponding nitro analogs. Also, introduction of a pyridine in the 2-position of the quinazoline core decreased the binding affinity. Due to the hydrophobic nature of the MAPK insert in p38α, we speculate that the introduction of polar groups or heteroatoms might be less tolerated. Although several co-crystallization attempts were unsuccessful, we postulate that the nitro-substituent may increase affinity by forming favorable water- or ion-mediated interactions with Asp residues 292 and 294. Considering the unique binding mode of 1b, we wanted to better understand why this compound was initially detected, although weakly, and initially identified by our glycine-rich loop FLiK binding assay. We carried out a co-evolution analysis of the sequences of several mammalian CMGC kinases [14] using the analysis algorithms previously described by Lockless and Ranganathan [15]. This analysis supports the existence of a network of interdependent amino acids which link the hydrophobic pocket to the N-lobe of CMGC kinases, thus providing evidence of a clear correlation between residues in the ATP pocket of p38α and its MAPK insert (Figure 4C, D). Interestingly, this analysis is supported by NMR data recently reported by Francis et al. in which the binding of the p38α DFG-out inhibitor BIRB-796 not only perturbed chemical shifts within the ATP binding site but also affected the chemical shifts of residues in the MAPK insert in the C-terminal lobe [16]. While it is important to note that this analysis merely detects evolutionary correlations, the internal “hydrophobic spine” highlighted by this analysis, and proposed elsewhere, [17], [18] appears to be well in line with our experimental results.

thumbnail
Figure 2. General synthesis of 2-arylquinazolines.

See table in Figure 3 for identity of substituents (R).

https://doi.org/10.1371/journal.pone.0039713.g002

thumbnail
Figure 3. Structure activity relationship of 2-arylquinazolines.

Each concentration of compound was tested in triplicate. Ratiometric emission values were plotted against the concentration of compound on a logarithmic scale to generate a binding curve. KD values of various 2-arylquinazolines were determined bound to p38α.

https://doi.org/10.1371/journal.pone.0039713.g003

thumbnail
Figure 4. Co-crystal structure of p38α and co-evolution analysis.

(A) Crystal structure of wild type p38α (grey) in complex with 8b (yellow). (B) Alignment of 1b and 8b bound to the lipid binding site of MAPK p38α. The 2-phenyl quinazoline scaffolds align nicely. The phenyl ethylene moiety of 8b extends further into the hydro-phobic back pocket (brown) resulting in a higher binding affinity. (C, D) p38α in complex with 1b. Amino acids showing significant co-evolution are colored according to the strength of the cumulative effect they exert on other residues (C) and of the cumulative effect other residues have on them (D), yellow symbolizing the strongest effect on both panels.

https://doi.org/10.1371/journal.pone.0039713.g004

Discussion

Although initial detection of 1b with a binding assay based on conformational changes of the glycine-rich loop supports this “cross-talk” model of allosteric modulation, we were not able to observe any inhibition of kinase activity when testing this focused library of MAP insert binders on p38α. Additionally, 1b was profiled against 95 additional kinases in activity-based biochemical assays (Figure 5). Interestingly, there are naturally occurring isoforms of p38α which show altered signaling pathway preferences [19]. The major differences between these isoforms and wild type p38α lie at the C-terminal end of the kinase, including regions within or proximal to the MAP kinase insert. We propose that the MAP kinase insert may have a scaffolding function and somehow regulates p38α signaling independent of the activity of p38α itself [20]. Although it is beyond the scope of this work, we believe that future studies aimed at elucidating the biological function(s) of this site may rely on the use of tightly binding molecules, such as those presented, which can perturb the conformation of the MAP kinase insert, disrupt or enhance protein-protein interactions with this structural feature and provide kinase researchers with alternative mechanisms for modulating the complex MAPK network [21], [22]. In conclusion, we have developed a fluorescence-based HTS binding assay for the MAPK insert of p38α and identified scaffolds which bind exclusively to this unique pocket. Using protein X-ray crystallography, we validated the screening results and successfully guided the medicinal chemistry efforts to generate potent binders of the p38α lipid binding site. These compounds might present a valuable starting point for further investigations of the p38α MAPK insert biology. Also, the presented assay is a further proof of the general applicability of the FLiK technology and presents an advantageous strategy for the identification of yet unknown kinase modulators.

thumbnail
Figure 5. Kinase selectivity profile of 1b.

Compound 1b was profiled against a selected panel of 95 different kinases at a concentration of 10 µM employing a radioactive (33P-ATP) filter-binding assay (National Centre for Protein Kinase Profiling, Dundee). The numbers represent the remaining kinase activity in %. The profiling reveals that compound 1b does not inhibit enzymatic activity of any of the tested kinases.

https://doi.org/10.1371/journal.pone.0039713.g005

Materials and Methods

The fluorophore 6-acryloyl-2-dimethylaminonaphthalene (acrylodan) was purchased from Invitrogen GmbH (Germany). Cuvettes and mini stir bars were obtained from Carl Roth GmbH (Germany). Small volume (20 µL fill volume) black flat bottom 384-well plates were obtained from Greiner Bio-One GmbH (Solingen, Germany). Crystallization plates (EasyXtal Tool; 24-well) were obtained from Qiagen GmbH (Germany). All supplies for the p38α HTRF assay kit were purchased from CisBio (Bagnols-sur-Cèze, France).

Expression, purification and labeling of p38α

The human p38α kinase construct containing the mutations required for specific labeling (C119S/C162S/S252C) as well as wild type p38α were cloned into a pOPINF vector and transformed as an N-terminal His-tag construct with a PreScission Protease cleavage site into BL21(DE3) Codon+RIL E. coli. Protein expression, purification, fluorophore labeling and verification of mono-labeling of the kinase were performed essentially as described previously. Labeling of the mutant p38α protein with thiol-reactive acrylodan was carried out by adding a 1.5∶1 molar excess of the fluorophore (dissolved in DMSO) to the purified protein in 50 mM Hepes Buffer (pH 7.0). The mixture was allowed to react in the dark overnight at 4°C. Unreacted acrylodan was subsequently removed by three cycles of buffer exchange into FLiK Measurement Buffer (50 mM Hepes Buffer, pH 7.4+200 mM NaCl) using a 10k-MWCO Centricon. The protein was concentrated to ∼200 μM, aliquoted and frozen at −80°C. Mono-labeling of the protein was verified by ESI-MS as described previously [23], [24].

thumbnail
Table 1. Data Collection and Refinement Statistics for p38α crystal structures in complex with 1b and 8b a.

https://doi.org/10.1371/journal.pone.0039713.t001

Characterization of the p38α lipid lite FLiK assay

Characterization of the fluorescence response of MAP insert-labeled p38α was carried out as described previously for p38α labeled on the activation loop [23]. For this particular FLiK assay, we employed 1b as a reference compound since protein X-ray crystal structures revealed exclusive binding to the lipid binding site of p38α. The binding of this ligand induces movement in the MAP insert, thereby altering the microenvironment and the fluorescence properties of acrylodan. Briefly, a polystyrene cuvette containing a suspension of 50 nM acrylodan-labeled p38α in FLiK Buffer was excited at 386 nm and emission spectra were measured in the absence and presence of increasing concentrations of 1b. The compound was dissolved in 100% DMSO and delivered in sequential small doses to a single sample. The %v/v DMSO did not exceed 0.2% in cuvettes. These measurements revealed that the major emission maximum shifts from ∼475 nm to ∼515 nm as the lipid acrylodan-labeled lipid binding sites become saturated with 1b. This shift in emission maxima enabled the use of ratiometric values (R = I515/I475) for plotting binding curves and determining the KD of ligands. Ratiometric fluorescence readouts are advantageous in that they provide an internal correction for small errors in sample volume which can affect signal intensity. All measurements of the cuvettes were made with a JASCO FP-6500 fluorescence spectrophotometer (JASCO GmbH, Germany).

Adaptation to high-throughput screening formats

The assay was adapted to high-throughput screening formats which enabled KD values to be determined in black small volume assay plates. Briefly, a dilution series of each compound was first prepared in 100% DMSO at 20X the desired final concentration and 1 μL was transferred to the assay plate. Compounds were then mixed with 19 µL of FLiK Buffer (supplemented with 0.01% Triton X-100) containing acrylodan-labeled p38α (100 nM). The final concentration of compound in the dilution series ranged from 0–50 μM (5% v/v DMSO final). Plates were covered with an adhesive aluminum foil and incubated for 30 min at RT prior to measurement of emission intensities at 470 and 520 nm using a Tecan Safire2 plate reader. The adjustment in chosen wavelengths in 384-well plates was necessary to maximize the resulting fluorescence ratio as we suspect the addition of detergent to assay buffer caused a slight shift in the emission maxima. Binding curves were plotted using the ratio R = I520/I470 to determine the KD. A calculated Z'>0.7 (using negative and positive binding controls) suggests that the FLiK assay for the lipid site of p38α is robust and amenable to high-throughput screening formats.

Chemistry

Unless otherwise noted, all reagents and solvents were purchased from Acros, Fluka, Sigma, Aldrich or Merck and used without further purification. Dry solvents were purchased as anhydrous reagents from commercial suppliers. 1H and 13C NMR spectra were recorded on a Varian Mercury 400, Bruker Avance DRX 400, Bruker DRX 500 or Varian Inova 600 spectrometer. 1H NMR spectroscopy chemical shifts are reported in δ (ppm) as s (singlet), d (doublet), dd (doublet of doublet), t (triplet), q (quartet), m (multiplet) and bs (broad singlet), and are referenced to the residual solvent signal: CDCl3 (7.26), CD3OD (4.87) and, DMSO-d6 (2.50). 13C NMR spectra are referenced to the residual solvent signal: CDCl3 (77.0), CD3OD (21.4), DMSO-d6 (39.0). All final compounds were purified to >95% purity, as determined by high-performance liquid chromatography (HPLC). Purities were measured using an Agilent 1200 Series HPLC system with UV detection at 210 nm (System: Agilent Eclipse XDB-C18 4.6×150 mm, 5 µM, 10 to 100% CH3CN in H2O, with 0.1% TFA, for 15 min at 1.0 mL/min). High resolution electrospray ionization mass spectra (ESI-FTMS) were recorded on a Thermo LTQ Orbitrap (high resolution mass spectrometer from Thermo Electron) coupled to an ‘Accela’ HPLC System supplied with a ‘Hypersil GOLD’ column (Thermo Electron). Analytical TLC was carried out on Merck 60 F245 aluminum-backed silica gel plates. Compounds were purified by column chromatography using Baker silica gel (40–70 µm particle size). Preparative HPLC was conducted on a Varian HPLC system (Pro Star 215) with a VP 250/21 Nucleosil C18 PPN column from Macherey-Nagel and monitored by UV at λ = 254 nm. Microwave-assisted reactions were carried out with a ‘Discover’ system from CEM Corporation.

Synthesis of 7-Nitro-2-phenyl-4(3H)-quinazolinone 4a.

Benzamidine 3a (6.0 g, 50 mmol) and 4-nitro-2-aminobenzoic acid 2 (2.8 g, 16 mmol) were dissolved in 2-methoxyethanol (300 mL) after which acetic acid (3.6 mL, 63 mmol) was added. The solution then heated at reflux (132°C) for 20 h with stirring. The reaction mixture was then cooled, filtered and the filtrate washed several times with cold MeOH to afford the desired product 4a (2.8 g, 65%) as a green-yellow colored solid which was utilized in other reactions without further purification. 1H-NMR (400 MHz, DMSO-d6): δ 12.90 (s, 1H), 8.42 (d, J = 2.2 Hz, 1H), 8.36 (d, J = 8.7 Hz, 1H), 8.24–8.19 (m, 3H), 7.66–7.55 (m, 3H); 13C-NMR (101 MHz, DMSO-d6): δ 162.29, 155.45, 152.20, 149.93, 132.92, 132.89, 129.59, 129.06, 128.91, 126.14, 123.16, 120.96; HRMS (ESI-MS): Calculated: 268.07167 for C14H10N3O3 [M+H]+, Found: 268.07173.

Synthesis of N-Cyclopropyl-7-nitro-2-phenyl-4-quinazolinamine 1a.

7-Nitro-2-phenyl-4(3H)-quinazolinone 4a (100 mg, 0.373 mmol) was transferred to a dry flask under argon after which thionyl chloride (2 mL) and 1–2 drops DMF were added. The reaction was then heated at reflux (80°C) for 3 h after which the solvent was removed under vacuum. This afforded the respective chloride as a brown-colored solid which was used without any further purification in the following reaction. The chloride (100 mg, 0.35 mmol) was dissolved in dry DCM (4 mL) and cyclopropylamine (37 μL, 0.52 mmol) in i-PrOH (2 mL) was then added drop-wise. The reaction mixture was then stirred at rt for 24 h, after which the solvent was removed under vacuum. The resulting residue was subsequently purified by silica gel column chromatography (3% MeOH/DCM as elution solvent) to afford the desired product 1a as a yellow solid (89 mg, 83%). 1H-NMR (400 MHz, DMSO-d6) δ 8.69 (d, J = 3.6 Hz, 1H), 8.54 (dd, J = 6.6, 3.0 Hz, 2H), 8.44 (dd, J = 10.7, 5.7 Hz, 2H), 8.16 (dd, J = 9.0, 2.3 Hz, 1H), 7.56–7.48 (m, 3H), 3.27–3.21 (m, 1H), 0.95–0.86 (m, 2H), 0.77–0.68 (m, 2H); 13C-NMR (101 MHz, DMSO-d6) δ 161.22, 160.58, 149.94, 149.90, 137.82, 130.72, 128.33, 128.19, 125.23, 122.80, 118.34, 117.33, 24.48, 6.13. HRMS (ESI-MS): Calculated: 307.11895 for C17H14O2N4 [M+H]+, Found: 307.11895.

Synthesis of N-(Cyclopropylmethyl)-7-nitro-2-phenyl-4-quinazolinamine 6a.

The reaction was performed as described above for 1b, in that the required chloride (100 mg, 0.373 mmol) in dry DCM (2 mL) was reacted with cyclopropylmethylamine (44.95 μL, 0.53 mmol) i-PrOH (2 mL) to afford the desired product 6a as a yellow solid (47.0 mg, 42%) after purification by silica gel column chromatography (DCM as eluent). 1H-NMR (400 MHz, DMSO-d6): δ 8.88 (br s, 1H), 8.52 (d, J = 9.0 Hz, 1H), 8.50–8.45 (m, 2H), 8.43 (d, J = 2.4 Hz, 1H), 8.19 (dd, J = 9.0, 2.4 Hz, 1H), 7.54–7.51 (m, 3H), 3,57–3.54 (m, 2H), 1.35–1.22 (m, 1H), 0.57–0.48 (m, 2H), 0.40–0.34 (m, 2H); 13C-NMR (101 MHz, DMSO-d6): δ 161.15, 159.36, 150.05, 149.87, 137.71, 130.80, 128.42, 128.13, 125.32, 122.64, 118.45, 117.43, 45.50, 10.50, 3.71; HRMS (ESI-MS): Calculated: 321.13460 for C18H17N4O2 [M+H]+, Found: 321.13449.

Synthesis of N-Benzyl-7-nitro-2-phenyl-4-quinazolinamine 7a.

The reaction was performed as described above with 1b, in that the required chloride (100 mg, 0.373 mmol) in dry DCM (2 mL) was reacted with benzylamine (57.29 μL, 0.525 mmol) in i-PrOH (2 mL) to afford the desired product 7a as a yellow solid (104.6 mg, 84%) after silica gel column chromatography (DCM as elution solvent). 1H-NMR (400 MHz, DMSO-d6): δ 9.39 (br s, 1H), 8.57 (d, J = 9.1 Hz, 1H), 8.49–8.44 (m, 3H), 8.24 (dd, J = 9.1, 2.3 Hz, 1H), 7.53–7.45 (m, 5H), 7.36–7.32 (m, 2H), 7.26–722 (m, 1H), 4.94 (d, J = 5.7 Hz, 2H); 13C-NMR (101 MHz, DMSO-d6): δ 161.15, 159.38, 150.19, 137.61, 134.64, 130.87, 129.53, 129.20, 128.45, 128.19, 127.56, 127.03, 125.30, 122.77, 118.73, 117.47, 44.17; HRMS (ESI-MS): Calculated: 357.13460 for C21H17N4O2 [M+H]+, Found: 357.13438.

Synthesis of 7-Nitro-2-phenyl-N-(2-phenylethyl)-4-quinazolinamine 8a.

The reaction was performed as described above with 1b, in that the required chloride (100 mg, 0.373 mmol) in dry DCM (2 mL) was reacted with phenethylamine (66.13 μL, 0.525 mmol) in i-PrOH (2 mL) to afford the desired product 8a as a yellow solid (111.5 mg, 86%) after silica gel column chromatography (DCM as elution solvent). 1H-NMR (400 MHz, DMSO-d6): δ 8.94 (br s, 1H), 8.52–8.42 (m, 4H), 8.18 (dd, J = 9.0, 2.3 Hz, 1H), 7.54–7.52 (m, 3H), 7.34–7.28 (m, 4H), 7.24–7.18 (m, 1H), 3.91–3.85 (m, 2H), 3.06 (t, J = 7.5, 2H); 13C-NMR (101 MHz, DMSO-d6): δ 161.03, 159.28, 150.03, 149.53, 139.41, 137.52, 130.89, 128.76, 128.47, 128.42, 128.15, 126.24, 125.18, 122.48, 118.60, 117.38, 42.68, 34.29; HRMS (ESI-MS): Calculated: 371.15025 for C22H19N4O2 [M+H]+, Found: 371.15014.

Synthesis of N4-Cyclopropyl-2-phenyl-4,7-quinazolinediamine 1b.

N-Cyclopropyl-7-nitro-2-phenyl-4-quinazolinamine 1a (32 mg, 0.10 mmol) and ammonium formate (40 mg, 0.60 mmol) were dissolved in absolute EtOH (3 mL). A spatula tip of Pd/C (5%) was then added and the reaction mixture was heated at reflux (90°C) for 2 h. The reaction solution was cooled and filtered through celite, and the filtrate evaporated under vacuum. Purification of the resulting residue was performed by preparative HPLC (Eluent: 0.1% TFA in MECN/H2O) to afford 1b as a yellow solid (18 mg, 65%). 1H-NMR (400MHz, DMSO-d6) δ 8.48 (dd, J = 7.5, 2.0 Hz, 2H), 7.89 (d, J = 8.6 Hz, 1H), 7.81 (s, 1H), 7.56–7.42 (m, 3H), 6.83–6.68 (m, 2H), 5.87 (s, 2H), 3.19–3.12 (m, 1H), 0.89–0.76 (m, 2H), 0.70–0.60 (m, 2H); 13C-NMR (101 MHz, CD3OD) δ 161.29, 157.35, 156.29, 141.56, 133.29, 131.95, 129.15, 128.63, 125.17, 117.52, 101.94, 97.39, 25.17, 5.97. HRMS (ESI-MS): Calculated: 277.14477 for C17H17N4 [M+H]+, Found: 277.14484.

Synthesis of N4-(Cyclopropylmethyl)-2-phenyl-4,7-quinazolinediamine 6b.

The reduction was performed as described above for 1b, utilizing N–(cyclopropylmethyl)–7–nitro-2–phenyl–4–quinazolinamine 6a (46 mg, 0.14 mmol) and ammonium formate (54.5 mg, 0.864 mmol) to afford compound 6b as a yellow solid (16 mg, 43%) after silica gel column chromatography (1.5% MeOH in DCM as eluent). 1H-NMR (400 MHz, DMSO-d6): δ 8.42 (dd, J = 8.0, 1.5 Hz, 2H), 7.91 (d, J = 8.8 Hz, 1H), 7.87–7.84 (m, 1H), 7.50–7.39 (m, 3H), 6.76 (dd, J = 8.8, 2.2 Hz, 1H), 6.73 (d, J = 2.1 Hz, 1H), 5.80 (s, 2H), 3.49 (t, J = 6.2 Hz, 2H), 1.28–1.21 (m, 1H), 0.51–0.44 (m, 2H), 0.36–0.30 (m, 2H); 13C-NMR (101 MHz, DMSO-d6): δ 159.21, 159.07, 152.59, 151.83, 139.25, 129.60, 128.07, 127.70, 123.66, 115.44, 106.26, 104.65, 62.08, 11.11, 3.59; HRMS (ESI-MS): Calculated: 291.16042 for C18H19N4 [M+H]+, Found: 291.16007.

Synthesis of N4-Benzyl-2-phenyl-4,7-quinazolinediamine 7b.

The reduction was performed as described above for 1b, utilizing N–benzyl–7–nitro-2–phenyl–4–quinazolinamine 7a (40 mg, 0.11 mmol) and ammonium formate (42.4 mg, 0.67 mmol) in EtOH (8 mL) to afford compound 7b as a yellow solid (16 mg, 44%) after silica gel column chromatography (1% MeOH/DCM as eluent). 1H-NMR (400 MHz, DMSO-d6): δ 8.40–8.34 (m, 3H), 7.95 (d, J = 8.8 Hz, 1H), 7.46–7.39 (m, 5H), 7.32–7.29 (m, 2H), 7.23–7.20 (m, 1H), 6.78 (dd, J = 8.8, 2.2 Hz, 1H), 6.74 (d, J = 2.1 Hz, 1H), 5.84 (s, 2H), 4.85 (d, J = 5.8 Hz, 2H); 13C-NMR (101 MHz, DMSO-d6): δ 159.03, 158.97, 152.63, 152.06, 140.48, 139.17, 129.49, 128.17, 127.96, 127.64, 127.32, 126.56, 123.53, 115.55, 106.29, 104.63, 43.50; (ESI-MS): Calculated: 327.16042 for C21H19N4 [M+H]+, Found: 327.16018.

Synthesis of 2-Phenyl-N4-(2-phenylethyl)-4,7-quinazolinediamine 8b.

The reduction was performed as described for 1b above, with 7–nitro-2–phenyl–N–(2–phenylethyl)–4–quinazolinamine 8a (48 mg, 0.12 mmol) and ammonium formate (48.8 mg, 0.77 mmol) in EtOH (8 mL) to afford compound 8b as a yellow solid (15 mg, 34%) after silica gel column chromatography (1.5% MeOH in DCM as eluent). 1H-NMR (400 MHz, DMSO-d6): δ 8.46 (dd, J = 7.9, 1.5 Hz, 2H), 7.97–7.92 (m, 1H), 7.87 (d, J = 8.8 Hz, 1H), 7.52–7.42 (m, 3H), 7.34–7.31 (m, 4H), 7.25–7.18 (m, 1H), 6.79–6.72 (m, 2H), 5.86 (s, 2H), 3.84–3.78 (m, 2H), 3.02 (t, J = 7.5 Hz, 2H); 13C-NMR (101 MHz, DMSO-d6): δ 159.10, 159.03, 157.64, 151.82, 139.89, 139.25, 129.64, 128.74, 128.44, 128.10, 127.69, 126.10, 123.57, 115.51, 106.22, 104.68, 42.29, 35.05; HRMS (ESI-MS): Calculated: 341.17607 for C22H21N4 [M+H]+, Found: 341.17574.

Synthesis of 7-Nitro-2-(4-pyridinyl)-4(3H)-quinazolinone 4b.

Pyridine-4-carboximidamide 14 (1.50 g, 9.5 mmol), 4-nitro-2-aminobenzoic acid (0.57 g, 3.1 mmol) and acetic acid (0.97 mL, 17 mmol) were dissolved in 2-methoxyethanol (20 mL) and the solution heated at reflux (132°C) for 20 h with stirring, as described above for the synthesis of 4a. Work-up as before afforded the desired product 4b (0.479 g, 57%) as a brown-colored solid, which was used without further purification. 1H-NMR (400 MHz, DMSO-d6): δ 13.18 (s, 1H), 8.83 (d, J = 6.0 Hz, 2H), 8.49 (d, J = 2.1 Hz, 1H), 8.40 (d, J = 8.7 Hz, 1H), 8.29 (dd, J = 8.7, 2.1 Hz, 1H), 8.14 (d, J = 6.1 Hz, 2H); 13C-NMR (101 MHz, DMSO-d6): δ 161.24, 152.79, 151.39, 150.14, 149.66, 139.70, 128.30, 125.89, 122.66, 121.91, 120.93; HRMS (ESI-MS): Calculated: 269.06692 for C13H9N4O3 [M+H]+, Found: 269.06696.

Synthesis of N-Cyclopropyl-7-nitro-2-(4-pyridinyl)-4-quinazolinamine 5a.

The reaction was performed as described above for the synthesis of compound 1a, with 7-nitro-2-(4-pyridinyl)-4(3H)-quinazolinone 4b (300 mg, 1.05 mmol) being used to generated the respective chloride, after which cyclopropylamine (219 μL, 3.15 mmol) was used with the addition of an aliquot of NEt3 (218 μL, 1.57 mmol), to afford the desired product 5a as a yellow oil (80 mg, 25%) after column chromatography (1% MeOH/DCM as eluent). 1H-NMR (400 MHz, DMSO-d6): δ 8.82 (br d, J = 3.6 Hz, 1H), 8.76 (d, J = 5.7 Hz, 2H), 8.50–8.47 (m, 2H), 8.37–8.35 (m, 2H), 8.23 (dd, J = 9.0, 2.3 Hz, 1H), 3.29–3.22 (m, 1H), 0.96–0.90 (m, 2H), 0.78–0.73 (m, 2H); 13C-NMR (101 MHz, DMSO-d6): δ 160.83, 159.47, 150.17, 150.05, 149.56, 145.04, 125.32, 122.99, 121.90, 119.23, 117.73, 24.53, 6.07; HRMS (ESI-MS): Calculated: 308.11420 for C16H14N5O2 [M+H]+, Found: 308.11422.

Synthesis of N-Benzyl-7-nitro-2-(4-pyridinyl)-4-quinazolinamine 7c.

The reaction was performed as described above for the synthesis of compound 1a, with 7-nitro-2-(4-pyridinyl)-4(3H)-quinazolinone 4b (100 mg, 0.373 mmol) used to generated the respective chloride after which benzylamine (114.6 μL, 1.05 mmol) to afford the desired product 7c as a yellow solid (66 mg, 51%) after column chromatography (1% MeOH/DCM as eluent, followed by a second purification utilizing 60% EtOAc/petroleum ether as solvent). 1H-NMR (400 MHz, DMSO-d6): δ 9.47 (br t, J = 5.7 Hz, 1H), 8.74 (d, J = 5.7 Hz, 2H), 8.60 (d, J = 9.0 Hz, 1H), 8.52 (d, J = 2.2 Hz, 1H), 8.32–8.27 (m, 3H), 7.48 (d, J = 7.4 Hz, 2H), 7.36–7.33 (m, 2H), 7.27–7.24 (m, 1H), 4.95 (d, J = 5.5 Hz, 2H); 13C-NMR (101 MHz, DMSO-d6): δ 159.57, 159.42, 150.21, 149.81, 144.91, 138.86, 128.39, 127.53, 126.99, 125.33, 123.06, 121.84, 119.50, 117.85, 117.80, 44.21; HRMS (ESI-MS): Calculated: 358.12985 for C20H16N5O2 [M+H]+, Found: 358.12955.

Synthesis of 7-Nitro-N-(2-phenylethyl)-2-(4-pyridinyl)-4-quinazolinamine 8c.

The reaction was performed as described above for the synthesis of compound 1a, with 7-nitro-2-(4-pyridinyl)-4(3H)-quinazolinone 4b (100 mg, 0.373 mmol) used to generated the respective chloride after which phenylethylamine (132 μL, 1.05 mmol) was utilized to afford the desired product 8c as a yellow solid (70 mg, 54%) after column chromatography (1% MeOH in DCM, followed by a second column using 60% EtOAc: petroleum ether as solvent). 1H-NMR (400 MHz, DMSO-d6): δ 9.01–8.98 (m, 1H), 8.77 (d, J = 5.6 Hz, 2H), 8.51–8.48 (m, 2H), 8.33 (d, J = 5.4 Hz, 2H), 8.27 (dd, J = 9.0, 1.9 Hz, 1H), 7.34–7.28 (m, 4H), 7.22–7.18 (m, 1H), 3.93–3.90 (m, 2H), 3.07 (t, J = 7.4 Hz, 2H); 13C-NMR (101 MHz, DMSO-d6): δ 159.52, 159.40, 150.13, 149.69, 145.09, 139.30, 128.72, 128.38, 126.17, 125.20, 123.01, 121.85, 119.37, 117.82, 117.76, 42.56, 34.25; HRMS (ESI-MS): Calculated: 372.14550 for C21H18N5O2 [M+H]+, Found: 372.14528.

Synthesis of N4-Cyclopropyl-2-(4-pyridinyl)-4,7-quinazolinediamine 5b.

The reduction was performed as described above for the synthesis of 1b, with N-cyclopropyl-7-nitro-2-(4-pyridinyl)-4-quinazolinamine 5a (55 mg, 0.179 mmol) and ammonium formate (67.7 mg, 1.07 mmol) in EtOH (5 mL) and the reaction mixture being heated at reflux (90°C) for 3 h. After a similar work-up by filtration, purification was carried out by preparative HPLC (0.1% TFA in H2O/MeCN). The combined fractions were concentrated under vacuum and then made basic with NEt3. This was followed by an extraction of the aqueous phase with EtOAc (4 times) and subsequent drying of the organic phase with Na2SO4. After filtration and evaporation of the solvent under vacuum the desired product 5b was isolated as a yellow solid (8.0 mg, 16%). 1H-NMR (400 MHz, MeOH-d4): δ 8.65 (d, J = 4.6 Hz, 2H), 8.39 (d, J = 4.6 Hz, 2H), 7.81 (d, J = 8.8 Hz, 1H), 6.96–6.85 (m, 2H), 1.15 (br d, J = 6.1 Hz, 1H), 0.92–0.89 (m, 2H), 0.73–0.68 (m, 2H); 13C-NMR (101 MHz, MeOH-d4): δ 162.58, 159.92, 154.35, 152.96, 150.21, 149.33, 124.51, 124.09, 118.15, 107.77, 107.03, 25.05, 7.16; HRMS (ESI-MS): Calculated: 278.14002 for C16H16N5 [M+H]+, Found: 278.14004.

Protein crystallography

Various inhibitors were co-crystallized with wild type p38α using conditions similar to those described previously [23]. Briefly, protein-inhibitor complexes were prepared by mixing 40 µL p38α (10 mg/mL) with 0.4 µL of inhibitor (100 mM in DMSO) and incubating the mixture for 1 h on ice. Samples were centrifuged at 13,000 rpm for 5 min to remove excess inhibitor. In some cases, co-crystallization carried out in the presence of excess inhibitor improved occupancy of the compounds in the crystal structure. Crystals were grown in 24-well crystallization plates using the hanging drop vapor diffusion method and by mixing 1.5 µL protein-inhibitor solution with 0.5 µL reservoir (100 mM MES pH 5.6–6.2, 20–30% PEG4000 and 50 mM n-octyl-β-D-glucopyranoside). Crystallization plates (EasyXtal Tool; 24-well) were obtained from Qiagen GmbH (Hilden, Germany). Cuvettes and mini stir bars were obtained from Carl Roth GmbH (Karlsruhe, Germany).

Structure determination, refinement and crystallographic statistics

For the crystals of p38α with inhibitors 20% glycerol was used as cryo protectant before they were flash frozen in liquid nitrogen. Diffraction data of the p38α-1b complex crystals was measured at the PX10SA beam line of the Swiss Light Source (PSI, Villingen, Switzerland) using a wavelength close to 1 Å. Diffraction data of the p38α-8b complex was collected in-house. Both datasets were processed with XDS and scaled using XSCALE [25]. Thep38α-inhibitor complex structures were solved by molecular replacement with PHASER [26] using the published p38α structures (PDB code: 1ZYJ) [27] or (PDB code: 2EWA) [28] as templates. The molecules in the asymmetric unit were manually modified using the program COOT [29]. The model was first refined with CNS8 using simulated annealing to reduce model bias. The final refinement was performed with REFMAC5 [30]. Inhibitor topology files where generated using the Dundee PRODRG2 server [31]. The two refined structures were validated with PROCHECK [32]. Data collection, structure refinement statistics, PDB-ID codes and further details for the data collection as well as Ramachandran plot results are shown in Table 1. PyMOL was used to produce the figures [33].

Acknowledgments

Beate Aust is thanked for expert technical assistance during protein expression and purification.

Author Contributions

Conceived and designed the experiments: DR MG JS. Performed the experiments: MG JS MT CG MR WVO. Analyzed the data: MG JS MT CG MR WVO DR. Contributed reagents/materials/analysis tools: MG JS MT CG MR WVO DR. Wrote the paper: MG JS DR.

References

  1. 1. Eglen R, Reisine T (2011) Drug discovery and the human kinome: recent trends. Pharmacol Ther 130: 144–156.
  2. 2. Eglen RM, Reisine T (2009) The current status of drug discovery against the human kinome. Assay Drug Dev Technol 7: 22–43.
  3. 3. Johnson LN (2009) Protein kinase inhibitors: contributions from structure to clinical compounds. Q Rev Biophys 42: 1–40.
  4. 4. Rabiller M, Getlik M, Klüter S, Richters A, Tückmantel S, et al. (2010) Proteus in the world of proteins: conformational changes in protein kinases. Arch Pharm (Weinheim) 343: 193–206.
  5. 5. Fabbro D, Manley PW, Jahnke W, Liebetanz J, Szyttenholm A, et al. (2010) Inhibitors of the Abl kinase directed at either the ATP- or myristate-binding site. Biochim Biophys Acta 1804: 454–462.
  6. 6. Wu WI, Voegtli WC, Sturgis HL, Dizon FP, Vigers GP, et al. (2010) Crystal structure of human AKT1 with an allosteric inhibitor reveals a new mode of kinase inhibition. PLoS One 5: e12913.
  7. 7. Zhang J, Adrian FJ, Jahnke W, Cowan-Jacob SW, Li AG, et al. (2010) Targeting Bcr-Abl by combining allosteric with ATP-binding-site inhibitors. Nature 463: 501–506.
  8. 8. Akella R, Moon TM, Goldsmith EJ (2008) Unique MAP Kinase binding sites. Biochim Biophys Acta 1784: 48–55.
  9. 9. Perry JJ, Harris RM, Moiani D, Olson AJ, Tainer JA (2009) p38alpha MAP kinase C-terminal domain binding pocket characterized by crystallographic and computational analyses. J Mol Biol 391: 1–11.
  10. 10. Diskin R, Engelberg D, Livnah O (2008) A novel lipid binding site formed by the MAP kinase insert in p38 alpha. J Mol Biol 375: 70–79.
  11. 11. Comess KM, Sun C, Abad-Zapatero C, Goedken ER, Gum RJ, et al. (2011) Discovery and Characterization of Non-ATP Site Inhibitors of the Mitogen Activated Protein (MAP) Kinases. ACS Chem Biol 6: 234–244.
  12. 12. Tong L, Pav S, White DM, Rogers S, Crane KM, et al. (1997) A highly specific inhibitor of human p38 MAP kinase binds in the ATP pocket. Nat Struct Biol 4: 311–316.
  13. 13. Simard JR, Getlik M, Grütter C, Schneider R, Wulfert S, et al. (2010) Fluorophore labeling of the glycine-rich loop as a method of identifying inhibitors that bind to active and inactive kinase conformations. J Am Chem Soc 132: 4152–4160.
  14. 14. University of Dundee, The Barton Group website. Accessed 2012 June 3.
  15. 15. Lockless SW, Ranganathan R (1999) Evolutionarily conserved pathways of energetic connectivity in protein families. Science 286: 295–299.
  16. 16. Francis DM, Rozycki B, Koveal D, Hummer G, Page R, et al. (2011) Structural basis of p38alpha regulation by hematopoietic tyrosine phosphatase. Nat Chem Biol 7: 916–924.
  17. 17. Kornev AP, Haste NM, Taylor SS, Eyck LF (2006) Surface comparison of active and inactive protein kinases identifies a conserved activation mechanism. Proc Natl Acad Sci U S A 103: 17783–17788.
  18. 18. Taylor SS, Kornev AP (2011) Protein kinases: evolution of dynamic regulatory proteins. Trends Biochem Sci 36: 65–77.
  19. 19. Sanz V, Arozarena I, Crespo P (2000) Distinct carboxy-termini confer divergent characteristics to the mitogen-activated protein kinase p38alpha and its splice isoform Mxi2. FEBS Lett 474: 169–174.
  20. 20. Rauch J, Volinsky N, Romano D, Kolch W (2011) The secret life of kinases: functions beyond catalysis. Cell Commun Signal 9: 23.
  21. 21. Cuadrado A, Nebreda AR (2010) Mechanisms and functions of p38 MAPK signalling. Biochem J 429: 403–417.
  22. 22. Kim EK, Choi EJ (2010) Pathological roles of MAPK signaling pathways in human diseases. Biochim Biophys Acta 1802: 396–405.
  23. 23. Simard JR, Getlik M, Grütter C, Pawar V, Wulfert S, et al. (2009) Development of a fluorescent-tagged kinase assay system for the detection and characterization of allosteric kinase inhibitors. J Am Chem Soc 131: 13286–13296.
  24. 24. Simard JR, Klüter S, Grütter C, Getlik M, Rabiller M, et al. (2009) A new screening assay for allosteric inhibitors of cSrc. Nat Chem Biol 5: 394–396.
  25. 25. Kabsch W (1993) Automatic processing of rotation diffraction data from crystals of initially unknown symmetry and cell constants. J Appl Cryst 26: 795–800.
  26. 26. Read RJ (2001) Pushing the boundaries of molecular replacement with maximum likelihood. Acta Crystallogr D Biol Crystallogr 57: 1373–1382.
  27. 27. Michelotti EL, Moffett KK, Nguyen D, Kelly MJ, Shetty R, et al. (2005) Two classes of p38alpha MAP kinase inhibitors having a common diphenylether core but exhibiting divergent binding modes. Bioorg Med Chem Lett 15: 5274–5279.
  28. 28. Vogtherr M, Saxena K, Hoelder S, Grimme S, Betz M, et al. (2006) NMR characterization of kinase p38 dynamics in free and ligand-bound forms. Angew Chem Int Ed Engl 45: 993–997.
  29. 29. Emsley P, Cowtan K (2004) Coot: model-building tools for molecular graphics. Acta Crystallogr D 60: 2126–2132.
  30. 30. Murshudov GN, Vagin AA, Dodson EJ (1997) Refinement of macromolecular structures by the maximum-likelihood method. Acta Crystallogr D 53: 240–255.
  31. 31. Schuttelkopf AW, van Aalten DM (2004) PRODRG: a tool for high-throughput crystallography of protein-ligand complexes. Acta Crystallogr D Biol Crystallogr 60: 1355–1363.
  32. 32. Laskowski RA, McArthur MW, Moss DS, Thornton JM (1993) PROCHECK: a program to check the stereochemical quality of protein structures. Journal of Applied Crystallography 26: 263–291.
  33. 33. DeLano WL (2002) The PyMOL Molecular Graphics System. http://wwwpymolorg.