Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Microbial Pathogenesis 118 (2018) 365–377

Contents lists available at ScienceDirect

Microbial Pathogenesis
journal homepage: www.elsevier.com/locate/micpath

Identification of inhibitor against H. pylori HtrA protease using structure- T


based virtual screening and molecular dynamics simulations approaches
Nivedita Rai, R. Muthukumaran, R. Amutha∗
Centre for Bioinformatics, Pondicherry University, Pondicherry, 605014, India

A R T I C LE I N FO A B S T R A C T

Keywords: The HtrA protease of Helicobacter pylori, which efficiently colonizes at the gastric epithelial of host cells, disrupts
HpHtrA the mucosal integrity of E-cadherin and spreads inflammatory diseases including gastric cancer by cleaving the
Homology modeling cell-cell adhesion of the host. The lack of knowledge on the molecular diversity, structural and functional be-
Molecular dynamics havior of HpHtrA necessitated the present study to explore its inhibition mechanism. At first, the similarity of
Virtual screening
HpHtrA with other gastro-intestinal pathogenic HtrA bacteria and its remote relationship with the Human HtrA
Binding energy
Hydroxyl-piperidine
homologs were ensured by the phylogenetic analysis and hence was identified as a novel therapeutic target for
4-aminopiperidine scaffold further design of inhibitors. The three dimensional structure of HpHtrA was modeled and simulated to achieve
its stable conformation and was used as a receptor to screen for the possible lead compound through virtual
screening (using ∼ 1.3 million compounds). Molecular dynamics simulations followed by the binding energy
analysis revealed the affinity of the compound 300040 in forming a stable complex with HpHtrA and thereby
revealed its potent role in inhibiting HpHtrA. It is also worthy to mention that, structurally, the ligand binding at
the catalytic site of HpHtrA is mainly facilitated by the significant dynamics of L2 loop. Based on the present
study, the hydroxyl-piperidine with 4-aminopiperidine scaffold is proposed to be one of the best possible lead
compounds for the inhibition of H. pylori.

1. Introduction of the toxins and pathogens. Similarly, in gastric epithelial cells, the E-
cadherin plays a very crucial role in cell-cell adhesion, and also pre-
The beneficial bacterial flora influences the anatomy, physiology of vents bacterial invasions and metastasis of many tumor cells [5,6]. The
the host and also provides protection against pathogenic susceptibility. E-cadherin is considered as an attractive target by the bacterial or-
Whereas, the pathogenic bacteria possesses inherent ability to breach ganism like H. pylori which colonizes in the gastric epithelial lining and
host defense mechanism and cause infection in specific area of the body involves in the metastasis of gastric cancer-related diseases. Recent
like skin, urinary tract, gastroenteritis, etc. [1,2]. The bacterial gas- reports revealed that High-temperature requirement A (HtrA, a secre-
troenteritis, a gastrointestinal infection, is caused by Escherichia coli tary protease) [7,8] preferentially recognizes and cleaves the signature
pathogenic strains, Shigella dysenteriae, Salmonella typhimurium, Yersinia motif, containing the [VITA]-[VITA]-x-x-D-[DN] consensus sequence of
pseudotuberculosis, Vibrio cholera, Clostridium perfringens, Bacillus cereus, E-cadherin, in which ‘x’ could be any amino acids [9]. Thus HtrA is
Staphylococcus aureus, Campylobacter jejuni, and Helicobacter pylori, etc. crucial for the pathogenic invasiveness of the host cells. The inhibition
According to World Health Organization (WHO), the H. pylori is rated of HtrA-mediated cleavage of E-cadherin significantly reduces the mi-
as the class-I carcinogen causing gastric cancer [5–8]. The H. pylori gration of H. pylori infections [10]. HtrA protease and their homologs
efficiently colonizes in the gastric epithelial of host cells for more than a are evolutionarily conserved in all living organisms. E.coli contains
decade, and disrupts the mucosal integrity and hence involves in sev- three differentially regulating HtrAs proteases such as DegP, DegQ, and
eral inflammatory diseases such as chronic gastritis, ulceration of gas- DegS. The DegP is well-characterized as a serine protease which de-
tric and duodenum, gastric malignancies and gastric cancer [3,4]. The grades the unfolded protein in an ATP independent manner. It mainly
antibiotic resistant nature of H. pylori encourages finding novel in- consists of a conserved N-terminal serine protease and two PDZ (Post-
hibition mechanisms that control disease progression. Hence, it is very synaptic density protein/Dlg1/Zo1) domains such as PDZ1 and PDZ2
important to identify the novel targets in H. pylori. [11]. Under various stress conditions, the bacterial cells would establish
The host epithelial surface is the first protective barrier against most an effective mechanism of protein quality control to monitor the folded


Corresponding author.
E-mail addresses: amutha_ramu@yahoo.com, ramutha@bicpu.edu.in (R. Amutha).

https://doi.org/10.1016/j.micpath.2018.03.027
Received 16 October 2017; Received in revised form 21 January 2018; Accepted 15 March 2018
Available online 16 March 2018
0882-4010/ © 2018 Elsevier Ltd. All rights reserved.
N. Rai et al. Microbial Pathogenesis 118 (2018) 365–377

state of the proteins and support further protein folding, repair and
degradations in both cytoplasmic and periplasmic locations [12,13].
The HtrA protein is implicated as a virulence factor in several patho-
genic bacteria such as E. coli (Entero-pathogenic strain), S. typhimurium,
B. cereus, S. aureus, C. jejuni, H. pylori, etc [14,15]. In this study, the
HtrA from H. pylori is considered as a target for the inhibition of H.
pylori (and will be referred as HpHtrA throughout the text). The three-
dimensional crystal structure of HpHtrA remains to be solved.
In the drug discovery field, the High throughput screening (HTS)
method remains the most preferred choice by pharma industry, despite
having its drawbacks such as high cost, time consuming, and low spe-
cificity as well as sensitivity. Hence, an alternative in silico computa-
tional method named as Virtual screening (VS) has been developed for
faster screening process. In in silico drug design, small molecular in-
hibitors are identified through VS method [16]. The VS is a computa-
tional approach, in which a large collection of compounds is screened
Fig. 1. Phylogenetic tree diagram of Helicobacter pylori HtrA (HpHtrA).
against a particular target to identify a potential lead. For example,
rhodanine scaffold (a competitive inhibitor at low micromolar con-
centration) was identified by structure-based virtual screening method BioNJ algorithms to a pairwise distance estimation matrix.
for the inhibition of arylalkylamine N-acetyltransferase (AANAT) [17].
The Captopril (an anti-hypertensive) drug, which is a potent inhibitor of 2.1.2. Homology modeling and validation of HpHtrA
Angiotensin-converting enzyme, was successfully identified by struc- The three-dimensional structure of HpHtrA is not yet solved and
ture-based virtual screening. Similarly, Zanamivir is a neuraminidase hence the homology modeling approach is employed to generate
targeted anti-influenza drug designed by using the structure-based structure of HpHtrA. The template for homology modeling was identi-
virtual screening approach [18]. Similarly, some inhibitors are pro- fied by performing BLAST (blastp) for HpHtrA protein sequence (target)
posed for inhibiting HpHtrA, but there is no “on-shelf” drug or vaccine against protein data bank (PDB) with default parameters [25]. The
available so far that passed the clinical trials. Hoy, B. et al., have de- sequence (UniProt ID: G2J5T2) showing highest similarity with the
signed inhibitor against HtrA (H. pylori HtrA inhibitor, HHI) using target is selected as template for homology modeling using Modeller
virtual screening combined with comparative homology modeling [8]. 9v11 software [26]. The alignment of both target and template se-
In 2015, Perna, A.M et al. used fragment based approach and identified quences generated about ten optimized structures. From the generated
a lead compound with a higher binding affinity towards HpHtrA [19]. structures, the most reliable model was screened based on the least
With the advantages of virtual screening, the present work also adopts Discrete Optimized Protein Energy (DOPE) score. The modeled struc-
virtual screening approach to identify lead compound against HpHtrA ture of HpHtrA was validated using SAVES server [27] in which the
(Helicobacter pylori secreted HtrA) inhibition. Even though HtrA pro- stereochemical properties as well as the model quality were evaluated
tease is mostly conserved among all living organisms (particularly in by PROCHECK and ERRAT programs, respectively. The significance of
bacteria), the HpHtrA protease is yet targeted by considering the facts consistency between modeled and template HtrA was evaluated using
such as: (i) accessibility in the extracellular region of the host epithelial ProSA (Protein Structure Analysis) server [28]. Further, the secondary
cell, (ii) poor homology (∼33% sequence identity) at protein level with structure information of HpHtrA was predicted from the aligned se-
the human homolog of HtrA (like HtrA1, HtrA2, HtrA3, and HtrA4) and quences using ESPript (Easy Sequencing in PostScript) server [29].
also (iii) the different site specificity due to the active site polarity and
sub-pocket shape of human HtrA which differs with HpHtrA [8]. Hence 2.2. Screening of novel inhibitors against HpHtrA
the HpHtrA showing no cross-reactivity with the human HtrA, is con-
sidered as a novel therapeutic target [20]. 2.2.1. Receptor preparation
Modeled structure of the receptor, HpHtrA was simulated for a
2. Materials and methods period of 50 ns and the stable structure corresponding to the least en-
ergy was selected for further virtual screening studies. The receptor
2.1. Structure prediction of HpHtrA structure was prepared using the protein preparation wizard of Maestro
v9.2, Schrödinger, in which the structure of HpHtrA was corrected to
2.1.1. Sequence, alignment and phylogenetic analysis of HpHtrA have appropriate bond orders. Sufficient hydrogen atoms were added to
The gastrointestinal bacterial HtrAs (such as E. coli strains, S. dys- reproduce the pH of 7.0. Finally, the structure was minimized using the
enteriae, S. typhimurium, Y. pseudotuberculosis, V. cholera, C. perfringens, OPLS-2005 force field to void off the steric clashes.
B. cereus, S. aureus, C. jejuni, and H. pylori) and human homologs of HtrA
sequences (HtrA1, HtrA2, HtrA3 and HtrA4) were retrieved from 2.2.1.1. Binding site identification and receptor grid
UniProt. The conserved regions among HtrA of the selected pathogenic generation. Identification of hot spots is the most important step in
bacteria are identified through multiple sequence alignment (MSA). the design of inhibitor against protein-ligand interactions. Here, the
The MSA was performed by ClustalW [21] algorithm using the default binding site of HpHtrA was predicted using FTMap server [30].The
parameters. The evolutionary analysis was performed by using MEGA7 missing atoms in the structure/receptor were added prior to the
tool [22] to identify the evolutionary relationship of HpHtrA with other calculation of Poisson–Boltzmann potential. To identify the hot spot,
gastrointestinal pathogenic bacteria as well as human HtrA homologs the receptor is assumed to be wrapped by a dense grid over its surface
(Fig. 1). The phylogenetic tree was constructed for these selected se- and small molecular probes such as organic solvents are allowed to
quences using maximum likelihood statistical method based on JTT generate bound poses as much as possible within the receptor surface.
(Jones-Taylor-Thornton) matrix [23]. The phylogenetic tree topology This is achieved using the rigid body docking with fast Fourier
was obtained by Neighbor-joining algorithm [24]. In tree topology, all transform correlation approach.
positions containing gaps or missing data were completely eliminated. Each probe samples millions of docked poses and the generated
Overall 315 positions were found in the final dataset. The initial tree bound conformations were minimized using Chemistry at Harvard
was obtained automatically by applying default Neighbor-Join and Macromolecular Mechanics (CHARMM) potential. Finally, the cluster

366
N. Rai et al. Microbial Pathogenesis 118 (2018) 365–377

was ranked based on the average Boltzmann energy. The predicted when the force value is lesser than 1000.0 kJ/mol/nm. The long-range
binding site residues are used by the receptor grid generation panel of electrostatic interactions were evaluated using Particle Mesh Ewald
Glide module. A grid box of dimension 10 × 10 × 10 Å was generated method [36] with a cut-off of 10 Å. All non-bonded interactions were
by fixing the predicted binding site residues at the centroid of the grid treated using Lennard–Jones interaction with a cut-off of 14 Å. The
box. The non-polar part of the receptor atom is scaled using the van der LINCS algorithm was used to constraint the bonds involving hydrogen
Waals radii of 1.0 Å and a partial charge cutoff value of 0.25 Å. atoms [37]. The constant temperature was maintained using V-rescale
thermostat for NVT ensemble with a coupling constant of 0.1 ps. The
2.2.1.2. Ligand preparation and virtual screening. The drug-like small Parrinello–Rahman Barostat for NPT ensemble was used at a constant
molecule library of ∼1.3 million compounds was prepared by pressure of 1 bar, coupling constant of 2.0 ps and compressibility of
retrieving compounds from various databases such as Specs, Zinc 4.5e−5/bar. The entire system was equilibrated using NVT and NPT
(clear Drug-Like), Asinex (BioDesign and Antibacterial), and ChemDiv ensembles for 500 and 500 ps, respectively [38,39]. The final produc-
(peptidomimetic). The LigPrep module implemented in Schrödinger tion run for all equilibrated systems was performed for a period of 50 ns
package was used for ligand preparation. The LigPrep module will filter and the trajectories were saved at every 2 ps. The stable trajectory has
the ligands based on several descriptors such as force field, been used to explore the free energy landscape as well as binding free
stereochemical information, ionization states, tautomer, low energy energy analysis. All binding interactions were visualized using Dis-
ring conformations, and also by retaining their native chirality. The covery Studio 3.1 (Biovia, http://www.accelrys.com).
ionization states at the physiological pH (7.0 ± 2.0) were set for all
ligands using Epik state penalty. The structure of filtered ligands was 2.4. Principal component analysis and free energy landscape
minimized using OPLS-2005 force field.
After completing both receptor and ligand preparations and grid The principal component analysis (PCA) reduces the dimensionality
generation, the ligands were screened using the Glide docking program of the data obtained from the stable MD trajectory. PC represents the
implemented in Maestro v9.2 suite [31]. For docking, a semi-flexible collective motion of the molecule based on their mass weighted cov-
docking method, which applies some flexibilities to the bonds in the ariance matrix [40]. The covariance matrix (Cij ) is calculated from the
ligand, was adopted. The ligands were systematically screened based on mass-weighted Cartesian coordinates (i and j) of the N-particle system
a hierarchal screening approach to find the best possible binding poses sampled over the simulated trajectory and is given
in the receptor grid space. Based on scoring function and sampling
Cij = (x i − x i )(x j − x j ) (1.1)
criteria, a three-stage filter process using (i) High Throughput Virtual
Screening (HTVS), (ii) Standard Precision (SP), and (iii) Extra precision First two PC modes i.e., PC1 and PC2 are taken further for con-
(XP) with 50, 30 and 10% of cutoff criteria has been used for docking. formational sampling.
The ring conformers and nitrogen inversions were sampled during The free energy landscape (FEL) is a conformational sampling pro-
flexible docking. The best poses were generated by Glide XP and ranked cedure used to explore all possible conformational changes of HpHtrA
according to both Glide score and energy values [32]. A set of top- after ligand binding [41,42]. The principal modes PC1 and PC2, gen-
ranked three ligands was subjected to molecular dynamics simulations erated using the stable trajectory (between 30 and 50 ns), were used to
for a period of 50 ns, to study the behavior of HpHtrA protein in the calculate the free energy of HpHtrA in different complex states. The
presence of inhibitor. two-dimensional FEL plot shows the joint probability of finding HpHtrA
complex in given conformational states during dynamics. The FEL is
2.2.2. ADME properties defined as:
In-silico study of the absorption, distribution, metabolism and ex-
cretion (ADME) properties of all selected compounds were predicted G (PC1, PC 2) = −kb T ln P(PC1, PC 2) (1.2)
using QikProp Version3.4 module. Qikprop provides a rapid prediction Where kb is Boltzmann constant, T is temperature, and P(PC1, PC2) is the
of ADME properties for drug candidate molecules. For each successfully normalized joint probability distribution.
processed molecule, the Qikprop predicts the pharmaceutically relevant
descriptors. It evaluates the acceptability of the filtered compounds
2.5. Binding free energy calculation
based on Lipinski’ rule of 5, log IC50, logP values, etc. [33].

The MM/PBSA approach was used to compute the binding free


2.3. Molecular dynamics simulations
energy of HpHtrA complexes, which quantify the molecular interaction
strength. A total of 100 frames from stable MD trajectory (between 30
At first, the modeled structure of HpHtrA was subjected to mole-
and 50 ns) were extracted and subjected to GMXPBSA tool [43]. In
cular dynamics simulations to analyze its conformational stability. The
MM/PBSA calculation, the binding energy of ligand in binding cavity of
stable conformation of HpHtrA was used in virtual screening (VS) to
top three HpHtrA complexes was computed as:
identify the lead compounds. From a set of 10 ligands, screened from
VS, the top three ligands (compound IDs: 402721, 402751 and 300040, ΔGbinding = Gcomplex − (Gprotein + Gligand ) (1.3)
which are listed in Table 2) and a previously reported lead compound
by Perna, A.M. et al., showing a ligand efficiency constant, KD value of Where, Gcomplex is the total binding free energy of the HpHtrA-ligand
13 ± 2 μm, which is considered as reference compound [19], were used complex. Gprotein and Gligand are the individual binding free energy of
for further simulations. The general protocol adopted in MD simulation HpHtrA and ligand molecule, respectively. The free energy per residue
is briefed below. contribution was calculated as
The molecular dynamic simulation was carried out using GROMACS Gx = ΔEMM − TΔS + ΔGsolvation (1.4)
v4.5 tool for a period of 50 ns by applying GROMOS43a1 force field
[34]. The ligand topology was generated using PRODRG server [35]. Where, x denotes the HpHtrA, ligand and complex forms as well. EMM is
The steepest descent algorithm is used to remove the steric clashes the average molecular mechanics potential energy in vacuum. T and S
present in the modeled structure. The modeled HpHtrA structure was are temperature and entropy, respectively and Gsolvation is a free energy
solvated explicitly using SPC water model in a cubic cell of dimension of solvation. The EMM is calculated by
12 × 12 × 12 Å. The solvated system was neutralized by adding suffi- ΔEMM = ΔEbonded + ΔEnonbonded (1.5)
cient counter-ions and was subjected to energy minimization (using
steepest descent algorithm), which will automatically be truncated ΔEMM = ΔEbonded + EElectrostatic + Evdw (1.6)

367
N. Rai et al. Microbial Pathogenesis 118 (2018) 365–377

EElectrostatic and Evdw were obtained using Coulomb and Lennard-jones


(LJ) potential functions, respectively. For single trajectory approach,
Ebonded was set to zero. In an implicit solvent model, the free energy of
solvation was derived from Gpolar and Gnonpolar contributions using
Adaptive Poisson-Boltzmann (APBS) program [44].The polar con-
tribution refers to the energy required to transfer the solute from a low
dielectric continuum medium (ε = 1.5) to a solvent having the di-
electric constant for water (ε = 80).
The non-electrostatic term Gnonpolar was calculated by SASA-only
non-polar model such as:
Gnonpolar = γA + β (1.7)
−1 −2
Where, γ = 0.0227 kJ mol Å is a surface tension coefficient of
the solvent, A is the SASA and b is the fitting parameter. For non-polar
solvation energy, the dielectric boundary is defined by the probe of
radius 1.4 Å.

3. Results and discussion Fig. 2. Superimposition of the modeled (blue) and template (grey) structures. The pro-
tease, PDZ1 and PDZ2 domains of HpHtrA are labeled and the catalytic triad residues are
represented in stick model. (For interpretation of the references to color in this figure
3.1. Sequence and phylogenetic analysis of HpHtrA legend, the reader is referred to the Web version of this article.)

The HtrA sequences of gastro pathogenic bacteria like H. pylori


(UniProt ID: G2J5T2), E. coli (UniProt ID: P0C0V0), S. dysenteriae (UniProt modeled structure superimposes well over the template with a least root
mean square deviation (rmsd) of 0.39 Å (Fig. 2). The HpHtrA structure
ID: Q32JU7), S. typhimurium (UniProt ID: P26982), Y. pseudotuberculosis
consists of Protease, PDZ1 and PDZ2 domains and in overall it is formed
(UniProt ID: Q66EE4), V.cholera (UniProt ID: C3LS56), C. perfringens
by eight α-helices, three 310-helices, and twenty four β-sheets (Fig. S1).
(UniProt ID: A0A0N7BTU8), B. cereus (UniProt ID: B7IRP7), S. aureus
The MSA performed on the pathogens classified under group I reveals
(UniProt ID: Q2FZP2), and human homologs of HtrA1 (UniProt ID:
that the protease domain consists of highly conserved motifs formed by
Q92743), HtrA2 (UniProt ID: O43464), HtrA3 (UniProt ID: P83110), and
residues Leu109-Ile115, Glu207-Ala214, Gly219-Gly225, Gly248-
HtrA4 (UniProt ID: P83105) were retrieved from UniProt database. The
Met257 and the active site residues His116, Asp147, Ser221 (forming
conserved regions were identified using MSA and the evolutionary re-
the catalytic triad) and is shown in Fig. S1.
lationship between HpHtrA and other selected pathogens as well as human
Details on the evaluation of modeled HpHtrA structure is given in
homologs was studied using phylogenetic analysis.
Table 1. The modeled structure of HpHtrA, validated using Ra-
The molecular evolution and sequence diversity between the gastro
machandran plot, reveals the presence of about 99.7% of amino acid
pathogenic bacteria and human HtrAs were explored using the seed tree
residues in favored as well as allowed regions and only 0.3% of residues
with a highest log-likelihood score of −7234.85. All nodes in the
in the disallowed region (Fig. S2a). In addition, the Verify3D and
phylogenetic tree, showing high bootstrap scores, reveal the reliability
ERRAT scores (79.44 and 74.94%, respectively), reflect the reliability of
of the observed phylogenetic relation. The phylogenetic tree consists of
the generated model (Figs. S2b and c). The protein structure analysis
three major taxons (group 1, II and III encircled in Fig. 1) with boot-
performed using ProSA server reveals the Z-score values −7.21 and
strap values 92, 100 and 82, respectively. The target sequence of
−8.86 for the modeled HpHtrA and template (PDB ID: 3CS0) struc-
HpHtrA (UniProt ID: G2J5T2 which is underlined in Fig. 1) is classified
tures, respectively (Fig. S2d). All these analyses confirm the reliability
in group I. This group 1 is characterized by two subgroups in which the
of the modeled HpHtrA structure.
target HpHtrA and C. jejuni share a common ancestor with a bootstrap
value of 100, whereas the remaining pathogens such as E. coli, S. ty-
phimurium, Y. pseudotuberculosis, V. cholera, form the major sub group. 3.3. Structure dynamics of HpHtrA
In group II, the closely related human homologs such as HtrA1, HtrA3
and HtrA4 are grouped together with a bootstrap value of 92 and the The modeled HpHtrA structure was simulated for a period of 50 ns.
HtrA2 shares a common ancestor (a bootstrap value of 100) with other During simulation, the backbone rmsd of HpHtrA is stabilized after 20
group II members. The members of group III also ensure their evolu- ns with an average value of 5.51 ± 0.3 Å (Fig. 4a). The residual fluc-
tionarily distant relationship with human HtrAs. The HpHtrA shares tuations (rmsf) calculated using Cα atoms of HpHtrA is shown in Fig. 5.
similarity with E.coli (Fig. 1), and is distantly related to human HtrA. In HpHtrA, mainly the loop regions of protease and PDZ domains
The separation of human homologs with other HtrA homologs clearly show higher fluctuations while the rest of the residues are significantly
depicts the diversion of HpHtrA with all other human homologs. It stable. In particular, the protease domain residues such as (i) Gly68-
indicates that even though all these sequences belong to the same Phe83 (from LA loop), (ii) Pro129-Ser131 (loop in between β4-β5
protein family (HtrA), they clearly disclose a separate evolutionary sheets), (iii) Ser166-Asp168 (310-helix) and (iv) Ser241-Gly245, (in L2
lineage from others. Hence, the HpHtrA can be used as a novel ther- loop region) display higher fluctuations above 3.9 Å. Similarly, (i) the
apeutic target.
Table 1
3.2. Structure of HpHtrA Evaluation of modeled HpHtrA.

DOPE score −38902.08 kcal/mol


The HpHtrA discloses about 43% of sequence similarity as well as PROCHECK 87.5% in core
82% of query coverage (e-value of 2e−80) with HtrA of E. coli (PDB ID: 10.6% in allowed
3CS0). As the crystal structure of HpHtrA is not yet reported experi- 1.6% generously allowed
mentally, a comparative homology modeling is performed by Modeller 0.3% in disallowed
Verify3D 79.44%
software to model the structure of HpHtrA using 3CS0 as a template.
ERRAT 74.94%
Among the generated 10 models, the structure with the least DOPE Z-score −7.21
score (−38902.08 kcal/mol) is selected for further analysis. The

368
N. Rai et al. Microbial Pathogenesis 118 (2018) 365–377

Table 2
Top ten compounds with Glide score and Glide energy (kcal/mol).

No Database Comp. ID Glide score Glide energy Interacting residues

1 402721 −8.13 −48.65 Asn114, His116, Ser144, Glu145, Ser146, Asp147, Ile203, Asn204, Thr237 Ser241,
Thr243, Lys242

2 402751 −7.32 −46.51 His116, Ser144, Glu145, Asp147, Ile203, Asn204, Ser205, Ile239, Thr243, Arg334

3 300040 −6.95 −41.43 Lys62, Glu96, Asn114, Asn115, His116, Asp119, Ser144, Asp147, Ile203, Asn204,
Thr237, Ile239, Thr243, Gln382

4 180786 −6.92 −35.60 Lys62, Glu96, His116, Asp119, Asp147, Asn204, Ile239

5 110 −6.91 −58.06 Asn114, Asn115, His116, Asp119, Ser144, Asp147, Asn204, Ser221, Thr237, Ile239,
Ala381, Gln382, Gln389, Asp392, Gln414, Asn416

6 3056 −6.89 −48.17 Lys62, Asn115, His116, Ser144, Glu145, Asp147, Ile203, Asn204, Thr237, Ile239,
Thr243, Gly244

7 92950 −6.87 −41.3 Arg97, Asn114, His116, Asp147, Ile203, Asn204, Thr237, Thr243, Gly244

8 18760 −6.87 −47.23 Lys62, Glu96, Asn114, His116, Asp119, Asp147, Ile203, Asn204, Ser221, Thr237

9 177468 −6.85 −45.75 Lys62, Asn114, His116, Asn115, Ser144, Glu145, Ser146, Asp147, Ile203, Ser204,
Ala381, Gly244, Thr237, Ile239, Thr243

10 4871 −6.83 −38.99 Lys62, Glu96, Arg97, Asn114, His116, Asp147, Asn204, Ser221, Ile239, Gly244,
Thr243

11 reference −4.5 −32.37 His 116, Asp147, Glu145, Ile203, Asn204, Ser205, Ala238, Ile239, Thr243, Gly244,
Arg334

369
N. Rai et al. Microbial Pathogenesis 118 (2018) 365–377

Fig. 3. Representation of the electrostatic surface of HpHtrA-ligand complex (a). The compounds 402721, 402751 300040 and reference in stick model are represented in magenta, green,
yellow and blue, respectively. The subsets b, c, d and e, depicts a close view of ligand bound at the binding pocket of HpHtrA in HpHtrA1, HpHtrA2, HpHtrA3 and HpHtrAref complexes,
respectively. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

310-helix of PDZ1 (Leu283-Lys294), (ii) loop in between β19-β20 sheets probes and performs a global search over a dense grid around HpHtrA,
(Met401-Asp405) and (iii) α7-helix of PDZ2 (Pro420-Gln423) show and finds favorable positions as the possible binding pockets using
higher fluctuations above 3.6 Å. The compactness of HpHtrA, measured simple energy function.
from the radius of gyration (Rg), is stabilized after 20 ns with an The best ranked binding pocket, identified using FTMap server, is
average value of 21.52 ± 0.10 Å (Fig. 4c). The least energy structure of located inside the protease domain. This binding pocket is formed by
HpHtrA (extracted at 20.9 ns) is used further for virtual screening. Asn114-His116, Ser144-Asp147, Ile203-Tyr206, Ser221, and Thr237-
Gly244 residues and are considered for the VS studies. It is interesting
3.3.1. Active site prediction to observe that the binding pocket identified by FTMap includes the
The FTMap server is used to predict the active site of HpHtrA. It conserved catalytic triad residues (His116, Asp147 and Ser221) of
employs the small molecules (which vary in size, shape and polarity) as serine protease family as well.

Fig. 4. Variation in the rmsd (a), Rg(c), and SASA (d) of HpHtrA in native (blue), HpHtrA1 (black), HpHtrA2 (red), HpHtrA3 (green), and HpHtrAref (yellow) structures. The subset b
depicts only the rmsd of the ligands present in the complex. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

370
N. Rai et al. Microbial Pathogenesis 118 (2018) 365–377

3.4. Molecular dynamics of complexes

The nature of interactions between the ligand and HpHtrA is ana-


lyzed by performing molecular dynamics simulations on the docked
complexes for a period of 50 ns. The HpHtrA complexes are stabilized
well with respect to native HpHtrA. The average backbone rmsd of
HpHtrA1, HpHtrA2, HpHtrA3 and HpHtrAref decreases by about 30.6,
26.3, 42.3 and 51.8% respectively, when compared to the native
HpHtrA. Simulations of the docked complexes such as HpHtrA1,
HpHtrA2, HpHtrA3 and HpHtrAref reveal a highly stable dynamics after
20 ns with an average backbone rmsd value of 3.83 ± 0.3 Å,
4.06 ± 0.4 Å, 3.2 ± 0.2 Å and 2.65 ± 0.2 Å, respectively. Both HpHtrA1
and HpHtrA2 complexes show similar pattern of rmsd variation,
Fig. 5. Variation in the rmsf (a) of HpHtrA in native (blue), HpHtrA1 (black), HpHtrA2
whereas the deviation is significantly lesser in HpHtrA3 and HpHtrAref
(red), HpHtrA3 (green) and HpHtrAref (yellow) structures. The regions of LA, L2 loops and
310-helix are labeled. (For interpretation of the references to color in this figure legend, complexes. In general, the rmsd profile of all four complexes including
the reader is referred to the Web version of this article.) HpHtrAref reveals a favorable interaction with HpHtrA and hence are
stabilized during dynamics (Fig. 4a).
Similarly, the rmsd of ligands is also calculated separately (Fig. 4b).
3.3.2. Screening of potential lead molecule against HpHtrA
The observed consistent rmsd pattern reflects the stability of ligand in
Structure-based virtual screening (SBVS) involves the screening of a
the binding cavity. It is interesting to observe that the ligands in
large collection of small molecules present in the database using
HpHtrA1, HpHtrA2, HpHtrA3 and HpHtrAref complexes show a stable
docking protocols. In this study, the compounds from Specs, Asinex
rmsd profile with marginal deviations 2.49 ± 0.25, 2.78 ± 0.11,
(BioDesign, Antibacterial), ChemDiv (peptidomimetic), and Zinc (clean
2.4 ± 0.44 and 1.3 ± 0.25 Å, respectively. The average Rg of HpHtrA1,
Drug-Like compound) databases are used to generate the library
HpHtrA3 and HpHtrAref complexes are more compact with an average
(having ∼1.3 million compounds) to screen against HpHtrA.
of 21.86 ± 0.09, 21.72 ± 0.20 and 21.29 ± 0.1 Å, respectively, when
As per the standard protocol, the screening begins with different
compared to the HpHtrA2 complex for which the Rg increases to
scoring system HTVS and followed by SP as well as XP, to narrow down
22.31 ± 0.16 Å (Fig. 4c). The slightly enhanced Rg values of the com-
the compounds with desired features and also to select the lead com-
plexes indicate the structural relaxation of the binding pocket, after the
pounds, finally. After the completion of every screening stage, a certain
ligand binding.
percentage of screened compounds is used as the input to the next
The solvent accessible surface area (SASA) is calculated to measure
scoring scheme. In this study, about 50% of the best conformational
the solvent exposure of HpHtrA in native and complex structures. The
states screened by HTVS are fed as the input for SP and about 30% of
native HpHtrA shows an average area of 10905 ± 248 Å2, whereas the
the best output from SP is provided to the XP scoring scheme. In XP HpHtrA-ligand complexes namely HpHtrA1, HpHtrA2, HpHtrA3 and
protocol, only 10% of compounds with best scoring states are filtered
HpHtrAref reveal a comparatively increased solvent exposure with
out and are ranked based on the docking score (GLIDE score) shown in average values of 11323.4 ± 221, 10942.8 ± 232, 10993 ± 214 and
Table 2. From Table 2, the first three compounds with IDs 402721, 10987 ± 184 Å2, respectively (Fig. 4d). As the L2 loop is observed to
402751, and 300040 (shown in Fig. 3, will be referred as HpHtrA1, play a significant role in localizing the ligand inside the binding pocket
HpHtrA2, and HpHtrA3, respectively, in the text) show higher glide of HtrA, the SASA of the receptor binding pocket (formed by residues
score (varying from −8.13 to −6.95 kcal/mol) and hence are subjected such as Asn114-His116, Ser144-Asp147, Ile203-Tyr206, Ser221,
to molecular dynamics simulations. For comparison purpose, one of the Thr237-Gly244) including the L2 loop (only the residues involving in
reported lead compound with good binding affinity in an in vitro assay, ligand binding such as Thr237-Gly244) has been examined. In Fig. S3,
is also docked to HpHtrA structure (HpHtrAref) and docking score is the variation shown in Set 1 is the SASA calculated for the binding
shown in Table 2. This docked complex is further simulated for 50 ns to pocket with L2 loop, which is increased from 912 ± 45 Å2 (native
understand the interaction pattern of this reported lead compound. structure) to 992.5 ± 37 Å2 (HtrA complexes) during the dynamics due
to ligand binding. Whereas this calculated SASA is dropped to
526 ± 22 Å2, when the L2 loop residues are not considered (Set 2 in Fig.
3.3.3. ADME properties of predicted compounds S3). Such increased solvent exposure of binding pocket with L2 loop
The drug-like properties of these top three compounds are evaluated (from 526 to 992.5 Å2) discloses its active participation during complex
for Absorption, Distribution, Metabolism, and Excretion (ADME) formation.
properties using the Lipinski's rule of five such as, (i) molecular In order to identify the fluctuations of key residues, rmsf were cal-
weight < 500 Da, (ii) hydrogen bond donors < 5, (iii) hydrogen bond culated using Cα atoms for all complexes and are shown in Fig. 5. All
acceptors < 10 and (iv) logP < 5. The physiochemical and pharma- these complexes show less amplitude fluctuations except the LA loop
ceutical properties such as the partition coefficient (QPlogPo/w) and (Ile64-Glu93) which is present in the protease domain [45] and the 310-
water solubility (QPlogS) is predicted in the range of −1.066 to 2.666, helix in PDZ1 domain. It is interesting to observe that the binding of
and −3.465 to −0.304, respectively. ligand in HpHtrA1 and HpHtrA3 increases the fluctuation of L2 loop
The estimated number of H-bonds formed by the solute with the (region Ser241-Gly245) by 4.13 and 4.73 Å, respectively. This pro-
solvent (H2O) in aqueous solution is shown in Table 3. The values for nounced fluctuation is presumed by the rotation of L2 loop that facil-
the donors and acceptors are averaged over each configuration and are itates the passage of the ligand molecule into the catalytic site.
observed to vary from 3.0 to 4.0, and 8.9–9.9, respectively. The polar Whereas, the same L2 loop fluctuation is arrested in HpHtrA2 and as
surface area ranges from ∼82.54 to 147.4 Å2. The bioavailability and well as HpHtrAref complex with rms fluctuation of 2.58 and 1.92 Å,
toxicity values range between −4.6 and −5.96. Overall, ∼34–80% of respectively. In addition, the observed free dynamics of 310-helix of the
oral absorption in human body is revealed by these compounds. All PDZ1 domain with rmsf fluctuation of 4.4 Å in HpHtrA1 is gradually
these physiochemical parameters, predicted by Qikprop module [33], decreasing in HpHtrA2, HpHtrA3 and HpHtrAref complexes with an
are within the acceptable range for these selected compounds and average rms fluctuation of 3.0, 2.7, and 2.3 Å, respectively. In com-
hence indicate their potential to act as drug-like molecules (Table 3). parison to all other complexes, the dynamic fluctuation of HpHtrA was
arrested in HpHtrAref complex.

371
N. Rai et al. Microbial Pathogenesis 118 (2018) 365–377

Table 3
ADME properties of top three compounds.

Compound ID Molecular Weighta QPlogPo/wb QPlogSc PSAd hERGe % of human Oral Absorptionf DonorHBg AcceptorHBh

402721 343.39 −1.06 −0.30 147.43 −4.605 34.02 4 9.9


402751 444.52 2.66 −3.46 101.40 −5.960 80.39 3 9.2
300040 395.93 1.44 −2.08 82.54 −5.677 65.09 3 8.9

a
Molecular Weight (Weight < 500).
b
Predicted octanol/water partition coefficient (from −2.0 to 6.5).
c
Predicted aqueous solubility; S in mol/L (acceptable range: −6.5 to 0.5).
d
Polar surface area.
e
Predicted blockage of hERG K + channel (reasonable value < −6).
f
% of human oral absorption (<25% is weak and >80% is strong).
g
H-bond Donor (range for 95% of drugs: 0–6).
h
H-bond Acceptor (range for 95% of drugs: 2–20).

3.4.1. Principal component analysis and free energy landscape of FEL with two prominent clusters of least energy basins (Fig. 6b–c).
Principal component analysis (PCA) is performed on the stable MD Whereas the HpHtrA1 and HpHtrAref complex samples a comparatively
trajectories (between 30 and 50 ns) to explore the essential dynamics of narrow FEL space with two clusters separated by high energy barriers.
HpHtrA complexes over the two-dimensional free energy landscape. Similarly, the HpHtrAref compound spans a single distinct low energy
The dynamics of HpHtrA1, HpHtrA2, HpHtrA3, and HpHtrAref complex cluster surrounded by high energy barriers (Fig. 6a, d). In HpHtrA2
are mainly governed by the first 10 PCs as evidenced by their cumu- complex, the two most prominent clusters are located significantly
lative contributions 64.38, 69.54, 70.37 and 62.7%, respectively (Fig. closer to each other. The representative structures from the clusters of
S4). least energy was extracted and superimposed over the initial structure
The direction of conformational evolution of HpHtrA complexes in using modevectors module of PyMOL and are shown in Fig. 7a–d. In
the free energy landscape was derived using the first two PCs. The re- HpHtrA complexes, the α5 helix in PDZ1 domain moves closer to the
gion colored in purple indicates the clustered least free energy (ΔG = 0) protease domain and hence this α5 helix (Asn335) interacts with the L3
states with the highest probability of occurrence in FEL. The con- loop (Tyr206) of protease domain. The present analysis discloses the
formation of HpHtrA2 and HpHtrA3 is observed to sample a wider area interactions between PDZ1 and protease domains as reported in the

Fig. 6. Free Energy Landscape calculted using PC1 (nm) and PC2 (nm) modes of HpHtrA in HpHtrA1 (a), HpHtrA2 (b), HpHtrA3 (c), and HpHtrAref (d) complexes. The least energy basins
are indicated using yellow triangle. The FEL energy scale is indicated in kJ/mol. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version
of this article.)

372
N. Rai et al. Microbial Pathogenesis 118 (2018) 365–377

Fig. 7. Superimposition of the initial (grey) over the structures extracted from the two least energy basins of FEL (intermediate (blue) I, and final (green) structure II) in HpHtrA1 (a),
HpHtrA2 (b), HpHtrA3 (c), and HpHtrAref (d) complexes. The arrow direction indicates the change in position of lower energy structure over initial structure. The ligand motion are shown
for all the complexes. (For interpretation of the references to color in this figure legend, the reader is referred to the Web version of this article.)

crystal structure (PDB ID: 3CS0). In addition, the LA loop of protease propanoic group away from the binding site to a distance of 6 Å
domain reorients toward the PDZ2 domain, which enhances the ac- (Fig. 7d). The amplitude of upward lifting of L2 loop is higher in
cessibility of the ligand into the binding cavity. HpHtrA1 and HpHtrA3 when compared to HpHtrA2. Whereas in case of
In HpHtrA1, the 310-helix of PDZ1 domain moves in the direction of HpHtrAref complex, the L2 loop motion is less pronounced and re-
L3 loop of protease. In addition, the upward motion of L2 loop provides inforces similar rmsf pattern (Fig. 7dI, dII). This could be one of the
the accessibility for the ligand binding and hence, the benzenesulfonyl facts of observing lesser affinity of these compounds (402751 and re-
group of ligand molecule is re-located towards the binding site ference compounds) towards HpHtrA when compared to the rest of the
(Fig. 7aII). The benzenesulfonyl group is gradually translocated to- 402721 and 300040 compounds (Fig. 7b, d).
wards the catalytic site with an angular rotation of 161.1° and a dis-
tance of 2.8 Å. Similarly, in case of HpHtrA3, the upward movement of
L2 loop results in the translocation of chlorophenyl group towards the 3.4.2. Interaction analysis of HpHtrA complexes
catalytic site with the angular rotation of 88.6° and distance of 3.2 Å The top two hit compounds, such as 402721 and 402751 are
(Fig. 7cII). Due to this rotation of chlorophenyl group in HpHtrA3 and identified with pyrrolidine group (core moiety) in which, the two hy-
subsequently the carbonyl group is forming interaction with catalytic droxyl groups are attached in cis conformation. These hydroxyl groups
Ser221 residue. Whereas in HpHtrA2, the L2 loop is partially lifted of pyrrolidine are responsible for the major interactions with the
upwards. Hence, the hydrophobic benzyl group is not able to accom- HpHtrA binding site. The compound 300040 present in the HpHtrA3
modate itself inside the binding cavity and thus results in a rotational complex, is the piperidine derivative and forms interactions with the
shift of 131.5° and moves away from the catalytic site by a distance of catalytic triad. Whereas, in reference compound, the carboxylic group
3.3 Å (Fig. 7bII). Similar to HpHtrA2, the movement of L2 is restricted in of pyrrole derivative interacts with one of the catalytic triad residue.
HpHtrAref which results in relocating the pyrrole group along with Overall, all simulated compounds are having nitrogen based

373
N. Rai et al. Microbial Pathogenesis 118 (2018) 365–377

heterocyclic group. The electrostatic surface for all complexes is shown 1.7, 1.7 and 2.6 Å, respectively. Apart from these H-bonding
in Fig. 3a which clearly depict the nature of the binding cavity. interactions, the residues such as Glu96, Asn114, Asn115, His116,
Asp119, Glu145-Ser146, Ile203-Ser205 and Arg366 are involved in the
3.4.2.1. Compound 402721. The top hit compound 402721 possesses a non-bonded interactions (Fig. 8e). During MD simulations, the
higher glide score of −8.134 kcal/mol and forms H-bonding rotational shift of benzyl chloride towards the catalytic center is
interactions with residues Ser144, Glu145, Asp147, Ile203 and responsible for the L2 loop uplift motion and hence both π-π and π-
Asn204 of HpHtrA. One of the active site residues, Asp147 forms H- cation interactions have vanished. This π interaction is replaced by two
bonding interactions with the hydroxyl group (–OH group) of π-cation interactions which are formed between piperidine nitrogen
pyrrolidine and amine nitrogen (-NH2+) at distances 2.1 and 1.8 Å, and –NH2+ group of ligand with the imidazole ring of His116 (Fig. 8f).
respectively (Fig. 8a). It is also forming charge interaction (2.8 Å) with The H-bond interaction of Ile203, Asn204 and Ile239 residues with the
-NH2+ group of ligand. The hydroxyl group of pyrrolidine derivative trans-OH group of piperidine has been replaced by (i) Asp147
forms H-bonding interactions with the amide group of Asn204 and the interacting with piperidine nitrogen and (ii) Asn204 interacting with
carbonyl oxygen of Ile203 at distances 2.1 and 1.7 Å, respectively. The –NH2+ group of ligand (Fig. 8e and f).
carbonyl oxygen of Ser144 and Glu145 interact with the amide group of Two lower energy conformations (at 37.37 and 48.31 ns) were ex-
the compound (1.8 and 1.9 Å, respectively). tracted from the FEL and were superimposed over the initial con-
In addition to these H-bonding interactions, other residues such as formation of HpHtrA3. Superimpositions of these conformations clearly
Ser205, Asn114, His116, Thr237-Ser241, Thr243, and Gly244, etc. are depict the conformational changes in both LA and L2 loops as well as
involved in non-bonded interactions with the compound. Two lower ligand molecule (Fig. 7c1, c2). The H-bonds formed in HpHtrA3-ligand
energy conformations (observed at 33.68 and 47.23 ns) were extracted complex during simulation are consistent and similar to HpHtrA1 is
from the FEL generated using PC1 and PC2 modes. The superimposition shown in Fig. S5. In the least energy structure, the ligand forms the H-
of these two FEL structures over the initial conformation of HpHtrA1 bond interactions (with the catalytic triad residues such as Ser221 and
shows the dynamics of both LA and L2 loops of protease and also the Asp147) and π-cation interactions (with His116) with HpHtrA (Fig. 8f).
310-helix of the PDZ1 domain (Fig. 7a). The ligand binding is con- These interactions clearly depict that this compound is capable of in-
sidered responsible for the cavity opening mediated by the upward teracting with all the catalytic triad residues and hence form a sig-
motion of L2 loop (shown by an arrow in Fig. 7aI, aII). The ligand is nificantly stable complex.
translocated inside the binding cleft with a conformational tilt of ben-
zenesulfonyl group towards the cleavage site near Ser221 (by a distance 3.4.2.4. Reference compound. To understand the binding affinity of the
of 2.8 Å as shown in Fig. 7aII). This movement of the ligand releases the identified compounds in comparison with the known inhibitor, a
interactions of hydroxyl pyrrolidine with Asp147 and Ile203 residues previously known inhibitor with KD value of (13 ± 2) μM [19],
and reforms interactions between Ser205 and carbonyl oxygen of li- referred as HpHtrAref compound was used. This reported compound
gand. The number of H-bonds, polar and nonpolar interactions in was docked and simulated for 50 ns to understand the interacting
HpHtrA1 complex before and after simulations are shown (Fig. 8a and pattern. The reference compound interacts with the residues present in
b). Fig. S5 depicts the number of H-bonds observed throughout the si- the binding cavity of HpHtrA with a glide score of −4.5 kcal/mol. The
mulation time and the specific atoms involving in H-bonds are tabu- furan ring of the ligand forms π-cation interaction with the guanidine
lated as well (Table S1). moiety of Arg334 residue. The carboxyl group forms H-bond with
Asn204 and electrostatic interaction with Asp147 (with a distance of
3.4.2.2. Compound 402751. This ligand interacts well with the residues 1.8 and 3.7 Å, respectively). Apart from these interactions, some other
present in the binding cavity of HpHtrA with a glide score of non-bonded interactions are formed by His116, Glu145, Ile203, Ser205,
−7.325 kcal/mol. The ligand benzene ring forms π-π and π-cation Ala238, Ile239, Thr243 and Gly244 residues (Fig. 8g).
interactions with the imidazole ring of His116 (in the catalytic triad) Superimposition of the lowest energy structures extracted at 37.9
and ε-amino group of residue Lys62, respectively. The amino group and 46.5 ns over the initial structure clearly depicts the overall motion
(–NH (CH2)2+) forms H-bond (1.9 Å) with Asp147. The hydroxyl group of LA loop (protease domain) and 310 helix (PDZ1 domain). In addition,
of pyrrolidine forms H-bonding interactions with the carbonyl oxygen the L2 loop residue, mainly Ile240 residue, moves toward the pyrrole
group of Ile203 (1.9 and 1.8 Å) and side chain -NH2 group of Asn204 group of ligand (Fig. 7d). The representative structure extracted from
(1.8 Å). The carbonyl group of Asp147 forms two H-bonds with -NH2+ FEL shows that the outward translocation of the ligand releases the H-
group of the ligand with a distance of 2.6 and 1.7 Å, respectively. The bond and π-cation interactions due to which, the imidazole side chain
carbonyl oxygen of the ligand forms H-bond (1.9 Å) with amino group of His116 residue forms H-bond (2.2 Å) with the carbonyl group of li-
of Ile239 residue (Fig. 8c). To examine the receptor-ligand interaction gand molecule (Fig. 8h). Fig. S5 clearly depicts the number of H-bonds
stability during MD simulations, the representative FEL structures formed by the ligand inside the cavity in HpHtrAref, which is highly
extracted at 37.0 and 44.94 ns were superimposed on the initial flexible during simulation.
structure of HpHtrA2 (Fig. 7bI, bII). The rotational shift of benzyl
group towards the proximity of L2 loop forms various non bonded 3.4.3. Binding free energy of docked complexes
interactions with Ile239-Gly244 residues. Due to which, the L2 loop The binding energy is computed to quantify the efficiency of ligand to
fluctuation is arrested and, the π-cation and π-π interactions are lost in interact with the binding pocket of HpHtrA. The binding energy of each
the HpHtrA2 complex. Another H-bond, between carbonyl oxygen and complex is calculated by extracting the frames at an interval of 200 ps from
Gly244 (L2 loop residue), is formed at a distance of 1.6 Å (Fig. 8d). the trajectory recorded between 30 and 50 ns. The energy components such
Overall, four H-bonds (Table S1) are formed in HpHtrA2 complex and as van der Waal, electrostatic, polar and nonpolar solvation and the cal-
are consistent throughout the simulation (Fig. S5). culated contributions are listed in Table 4. The electrostatic energy for
HpHtrA1, HpHtrA2, HpHtrA3 and HpHtrAref complexes are −144.87 ± 1.3,
3.4.2.3. Compound 300040. Compound 300040 is bound well inside −112.37 ± 1.5, −233.84 ± 2.7 kcal/mol and −21.11 ± 1.71, respec-
the binding cavity of HpHtrA with glide score of −6.95 kcal/mol. In tively. Among all these complexes, only the HpHtrA3 complex possesses
HpHtrA3 complex, the benzyl group forms π-π and π-cation interactions highly negative electrostatic energy and the favorable contribution of polar
with His116 and Lys62 residues. The trans-OH group of piperidine energy. As a result, the overall binding free energy of HpHtrA3 is higher
derivative forms H-bonding interactions with Asn204, Ile239, and (−57.3 ± 1.0 kcal/mol) when compared to HpHtrA2 (−49.66 ± 0.6 kcal/
Ile203 residues, while the –NH2+ group of ligand forms H-bond mol), HpHtrA1 (−45.63 ± 0.4 kcal/mol) and HpHtrAref
interaction with Asp147 (in the catalytic triad) at a distance of 1.8, (−15.84 ± 0.54 kcal/mol) complexes. Overall, the compound 300040

374
N. Rai et al. Microbial Pathogenesis 118 (2018) 365–377

Fig. 8. The 2-D representation of the HpHtrA-ligand interactions observed in the docked (I) and simulated (II) complexes such as HpHtrA1 (a, b), HpHtrA2 (c, d), HpHtrA3 (e, f) and
HpHtrAref (g, h).

375
N. Rai et al. Microbial Pathogenesis 118 (2018) 365–377

Table 4
Binding Energy (in kcal/mol) calculated for HpHtrA-complexes using MM/PBSA method.

Name of the complex vdW Energy Electrostatic Energy Polar Energy Nonpolar Energy Polarc Nonpolarc Binding Energy

HpHtrA1 −42.29 ± 0.4 −144.87 ± 1.3 145.25 ± 1.0 −3.70 ± 0.01 0.37 −46.00 −45.63 ± 0.4
HpHtrA2 −53.24 ± 0.4 −112.37 ± 1.5 120.34 ± 1.1 −4.39 ± 0.01 7.97 −57.64 −49.66 ± 0.6
HpHtrA3 −37.83 ± 0.43 −233.84 ± 2.7 218.52 ± 2.1 −4.15 ± 0.0 −15.31 −41.98 −57.3 ± 1.0
HpHtrAref. −34.43 ± 0.38 −21.11 ± 1.71 42.78 ± 1.47 −3.07 ± 0.02 21.66 −37.5 −15.84 ± 0.54

Polarc: Polar contribution.


Nonopolarc: Nonpolar contribution.

expresses a significantly higher affinity towards HpHtrA and hence could be Acknowledgement
considered as a potential lead molecule to inhibit HpHtrA.
One of the authors, Amutha Ramaswamy acknowledges the Science
and Engineering Research Board, India for providing computational
4. Conclusion facilities under Fast Track Research Project for Young Scientists
Scheme. Nivedita Rai, and R. Muthukumaran acknowledges
The HtrA of Helicobacter pylori cleaves the extracellular domain of E- Pondicherry University for providing research fellowship. The authors
cadherin present in the gastric epithelial of the host cell to break the declare no conflict of interest.
cell-cell adhesion and spread infections. As a result, it is associated with
several gastric diseases like mucosa-associated lymphoid tissue (MALT) Appendix A. Supplementary data
lymphoma, chronic gastritis, gastric cancer, etc. It is also up-regulated
during stress conditions caused by an oxidative agent, H2O2 [46] and Supplementary data related to this article can be found at http://dx.
low pH in gastric epithelial. The inhibitor design for the active site of doi.org/10.1016/j.micpath.2018.03.027.
HpHtrA is required to prevent the fragmentation of E-cadherin. This
study mainly focuses on the (i) molecular evolution of HpHtrA, (ii) References
identification of lead molecules to inhibit HpHtrA protease and (iii) the
structural dynamics of HpHtrA in free and complex forms. [1] D.E. Neal Jr., Host defense mechanisms in urinary tract infections, Urol. Clin. North
The phylogenetic analysis reveals the similarity of HpHtrA with E. Am. 26 (4) (1999) 677–686.
[2] S. Schubert, Urinary tract immunology, Urologe 49 (5) (2010) 606–611, http://dx.
coli and other gastrointestinal bacteria. The distant relationship of doi.org/10.1007/s00120-010-2252-5.
HpHtrA with human HtrA homologs clearly witnesses that the human [3] M.J. Blaser, J.C. Atherton, Helicobacter pylori persistence: biology and disease, J.
HtrA protease shares no cross-reactivity with other pathogenic HtrA. Clin. Invest. 113 (3) (2004) 321–333, http://dx.doi.org/10.1172/JCI20925.
[4] R.M. Peek Jr., J.E. Crabtree, Helicobacter infection and gastric neoplasia, J. Pathol.
The homology modeling and validation of HpHtrA revealed a good 208 (2) (2006) 233–248, http://dx.doi.org/10.1002/path.1868.
agreement with the template structure of E. coli (PDB ID: 3CS0) with a [5] F. van Roy, Beyond E-cadherin: roles of other cadherin superfamily members in
rmsd of 0.39 Å. It is also observed that both HpHtrA and E. coli share cancer, Nat. Rev. Canc. 14 (2) (2014) 121–134, http://dx.doi.org/10.1038/nrc3647
nrc3647.
common conserved catalytic triad residues. The modeled structure of
[6] C.A. Janeway Jr., How the immune system works to protect the host from infection:
HpHtrA is simulated for a period of 50 ns and the least energy con- a personal view, Proc. Natl. Acad. Sci. U. S. A. 98 (13) (2001) 7461–7468, http://
formation was used for further screening process. Based on the struc- dx.doi.org/10.1073/pnas.131202998 131202998.
ture-based virtual screening approach, complexes of HpHtrA with [7] B. Hoy, T. Geppert, M. Boehm, F. Reisen, P. Plattner, G. Gadermaier, N. Sewald,
F. Ferreira, P. Briza, G. Schneider, S. Backert, S. Wessler, Distinct roles of secreted
402721, 402751 and 300040 compounds were simulated further to HtrA proteases from gram-negative pathogens in cleaving the junctional protein
identify the possible potential lead compound. For a comparative and tumor suppressor E-cadherin, J. Biol. Chem. 287 (13) (2012) 10115–10120,
analysis, a reported lead compound is also been simulated for 50 ns. http://dx.doi.org/10.1074/jbc.C111.333419 C111.333419.
[8] B. Hoy, M. Lower, C. Weydig, G. Carra, N. Tegtmeyer, T. Geppert, P. Schroder,
The dynamic LA, L2 loop regions of protease and 310-helix of PDZ1 N. Sewald, S. Backert, G. Schneider, S. Wessler, Helicobacter pylori HtrA is a new
domains of HpHtrA accommodate the ligand inside the binding pocket secreted virulence factor that cleaves E-cadherin to disrupt intercellular adhesion,
and further stabilize the complexes. The interaction between HpHtrA EMBO Rep. 11 (10) (2010) 798–804, http://dx.doi.org/10.1038/embor.2010.114
embor2010114.
and ligand is monitored using both H-bond and binding energy ana- [9] T.P. Schmidt, A.M. Perna, T. Fugmann, M. Bohm, H. Jan, S. Haller, C. Gotz,
lyses. The L2 loop motion is mainly regulating the ligand binding to- N. Tegtmeyer, B. Hoy, T.T. Rau, D. Neri, S. Backert, G. Schneider, S. Wessler,
wards the catalytic site. The higher amplitude fluctuation of an L2 loop Identification of E-cadherin signature motifs functioning as cleavage sites for
Helicobacter pylori HtrA, Sci. Rep. 6 (2016) 23264, http://dx.doi.org/10.1038/
is significantly observed in complex HpHtrA1 and HpHtrA3, wherein
srep23264 srep23264.
HpHtrA3, the ligand is interacting with the catalytic residue Ser221. [10] C.M. Abfalter, M. Schubert, C. Gotz, T.P. Schmidt, G. Posselt, S. Wessler, HtrA-
The complex of HpHtrA with ligand 300040 (HpHtrA3) is observed to mediated E-cadherin cleavage is limited to DegP and DegQ homologs expressed by
gram-negative pathogens, Cell Commun. Signal. 14 (1) (2016) 30, http://dx.doi.
express a higher binding energy (−57.3 ± 1.0 kcal/mol) than other
org/10.1186/s12964-016-0153-y 10.1186/s12964-016-0153-y.
complexes such as HpHtrA1 (−45.63 ± 0.4 kcal/mol) and HpHtrA2 [11] T. Krojer, M. Garrido-Franco, R. Huber, M. Ehrmann, T. Clausen, Crystal structure
(−49.66 ± 0.6 kcal/mol). Overall, the identified lead molecule, from of DegP (HtrA) reveals a new protease-chaperone machine, Nature 416 (6879)
the present analysis, is having improved binding energy when com- (2002) 455–459, http://dx.doi.org/10.1038/416455a 416455a.
[12] K.L. Strauch, J. Beckwith, An Escherichia coli mutation preventing degradation of
pared to the reference compound (−15.84 ± 0.54 kcal/mol). Hence, abnormal periplasmic proteins, Proc. Natl. Acad. Sci. U. S. A. 85 (5) (1988)
the compound 300040 that expresses significantly higher affinity to- 1576–1580.
wards HpHtrA could be considered as a potential lead molecule to in- [13] B. Lipinska, O. Fayet, L. Baird, C. Georgopoulos, Identification, characterization,
and mapping of the Escherichia coli htrA gene, whose product is essential for
hibit HpHtrA. In other words, the ligand with hydroxyl-piperidine and bacterial growth only at elevated temperatures, J. Bacteriol. 171 (3) (1989)
4-aminopiperidine scaffold could be considered for the designing and 1574–1584.
development of novel therapeutic inhibitors against HpHtrA induced [14] J. Skorko-Glonek, D. Zurawa-Janicka, T. Koper, M. Jarzab, D. Figaj, P. Glaza,
B. Lipinska, HtrA protease family as therapeutic targets, Curr. Pharmaceut. Des. 19
diseases. It is also worth mentioning that the compound containing (6) (2013) 977–1009 CPD-EPUB-20120918-22.
hydroxyl-piperidine and 4-aminopiperidine scaffold identified from this [15] M. Boehm, B. Hoy, M. Rohde, N. Tegtmeyer, K.T. Baek, O.A. Oyarzabal,
study is also known for its anti-cancerous property [47–49]. L. Brondsted, S. Wessler, S. Backert, Rapid paracellular transmigration of
Campylobacter jejuni across polarized epithelial cells without affecting TER: role of
proteolytic-active HtrA cleaving E-cadherin but not fibronectin, Gut Pathog. 4 (1)

376
N. Rai et al. Microbial Pathogenesis 118 (2018) 365–377

(2012) 3, http://dx.doi.org/10.1186/1757-4749-4-3 1757-4749-4-3. Med. Chem. 49 (21) (2006) 6177–6196, http://dx.doi.org/10.1021/jm051256o.


[16] E. Lionta, G. Spyrou, D.K. Vassilatis, Z. Cournia, Structure-based virtual screening [33] F. Yamashita, M. Hashida, In silico approaches for predicting ADME properties of
for drug discovery: principles, applications and recent advances, Curr. Top. Med. drugs, Drug Metabol. Pharmacokinet. 19 (5) (2004) 327–338 JST.JSTAGE/dmpk/
Chem. 14 (16) (2014) 1923–1938 CTMC-EPUB-62572. 19.327.
[17] L.M. Szewczuk, S.A. Saldanha, S. Ganguly, E.M. Bowers, M. Javoroncov, [34] W.R.P. Scott, P.H. Hünenberger, I.G. Tironi, A.E. Mark, S.R. Billeter, J. Fennen,
B. Karanam, J.C. Culhane, M.A. Holbert, D.C. Klein, R. Abagyan, P.A. Cole, De novo A.E. Torda, T. Huber, P. Krüger, W.F. van Gunsteren, The GROMOS biomolecular
discovery of serotonin N-acetyltransferase inhibitors, J. Med. Chem. 50 (22) (2007) simulation program package, J. Phys. Chem. 103 (19) (1999) 3596–3607, http://
5330–5338, http://dx.doi.org/10.1021/jm0706463. dx.doi.org/10.1021/jp984217f.
[18] T.T. Talele, S.A. Khedkar, A.C. Rigby, Successful applications of computer aided [35] A.W. Schuttelkopf, D.M. van Aalten, PRODRG: a tool for high-throughput crystal-
drug discovery: moving drugs from concept to the clinic, Curr. Top. Med. Chem. 10 lography of protein-ligand complexes, Acta. Crystallogr. D Biol. Crystallogr. 60 (Pt
(1) (2010) 127–141 CTMC-Abs-0010-10-1. 8) (2004) 1355–1363, http://dx.doi.org/10.1107/S0907444904011679
[19] A.M. Perna, T. Rodrigues, T.P. Schmidt, M. Bohm, K. Stutz, D. Reker, B. Pfeiffer, S0907444904011679.
K.H. Altmann, S. Backert, S. Wessler, G. Schneider, Fragment-based de novo design [36] D. Tom, Y. Darrin, P. Lee, Particle mesh Ewald: an Nâ‹…log(N) method for Ewald
reveals a small-molecule inhibitor of Helicobacter Pylori HtrA, Angew Chem. Int. sums in large systems, J. Chem. Phys. 98 (12) (1993) 10089–10092, http://dx.doi.
Ed. Engl. 54 (35) (2015) 10244–10248, http://dx.doi.org/10.1002/anie. org/10.1063/1.464397.
201504035. [37] B. Hess, H. Bekker, H.J.C. Berendsen, J.G.E.M. Fraaije, LINCS: a linear constraint
[20] R.M. Raju, A.L. Goldberg, E.J. Rubin, Bacterial proteolytic complexes as therapeutic solver for molecular simulations, J. Comput. Chem. 18 (12) (1997) 1463–1472,
targets, Nat. Rev. Drug Discov. 11 (10) (2012) 777–789, http://dx.doi.org/10. http://dx.doi.org/10.1002/(sici)1096-987x(199709)18:12<1463::aid-jcc4>3.0.
1038/nrd3846 nrd3846. co;2-h.
[21] M.A. Larkin, G. Blackshields, N.P. Brown, R. Chenna, P.A. McGettigan, [38] G. Bussi, D. Donadio, M. Parrinello, Canonical sampling through velocity rescaling,
H. McWilliam, F. Valentin, I.M. Wallace, A. Wilm, R. Lopez, J.D. Thompson, J. Chem. Phys. 126 (1) (2007) 014101, http://dx.doi.org/10.1063/1.2408420.
T.J. Gibson, D.G. Higgins, Clustal W and clustal x version 2.0, Bioinformatics 23 [39] M. Parrinello, A. Rahman, Polymorphic transitions in single crystals: a new mole-
(21) (2007) 2947–2948, http://dx.doi.org/10.1093/bioinformatics/btm404 cular dynamics method, J. Appl. Phys. 52 (12) (1981) 7182–7190, http://dx.doi.
btm404. org/10.1063/1.328693.
[22] K. Tamura, D. Peterson, N. Peterson, G. Stecher, M. Nei, S. Kumar, MEGA5: mole- [40] B. Hess, Similarities between principal components of protein dynamics and
cular evolutionary genetics analysis using maximum likelihood, evolutionary dis- random diffusion, Phys. Rev. 62 (6) (2000) 8438–8448.
tance, and maximum parsimony methods, Mol. Biol. Evol. 28 (10) (2011) [41] H. Frauenfelder, S.G. Sligar, P.G. Wolynes, The energy landscapes and motions of
2731–2739, http://dx.doi.org/10.1093/molbev/msr121 msr121. proteins, Science 254 (5038) (1991) 1598–1603.
[23] D.T. Jones, W.R. Taylor, J.M. Thornton, The rapid generation of mutation data [42] I. Tavernelli, S. Cotesta, E.E. Di Iorio, Protein dynamics, thermal stability, and free-
matrices from protein sequences, Comput. Appl. Biosci. 8 (3) (1992) 275–282. energy landscapes: a molecular dynamics investigation, Biophys. J. 85 (4) (2003)
[24] N. Saitou, M. Nei, The neighbor-joining method: a new method for reconstructing 2641–2649, http://dx.doi.org/10.1016/S0006-3495(03)74687-6 S0006-3495(03)
phylogenetic trees, Mol. Biol. Evol. 4 (4) (1987) 406–425. 74687-6.
[25] S.F. Altschul, W. Gish, W. Miller, E.W. Myers, D.J. Lipman, Basic local alignment [43] C. Paissoni, D. Spiliotopoulos, G. Musco, A. Spitaleri, GMXPBSA 2.1: a GROMACS
search tool, J. Mol. Biol. 215 (3) (1990) 403–410, http://dx.doi.org/10.1016/ tool to perform MM/PBSA and computational alanine scanning, Comput. Phys.
S0022-2836(05)80360-2 S0022-2836(05)80360-2. Commun. 186 (Supplement C) (2015) 105–107 https://doi.org/10.1016/j.cpc.
[26] A. Fiser, A. Sali, Modeller: generation and refinement of homology-based protein 2014.09.010.
structure models, Methods Enzymol. 374 (2003) 461–491, http://dx.doi.org/10. [44] N.A. Baker, D. Sept, S. Joseph, M.J. Holst, J.A. McCammon, Electrostatics of na-
1016/S0076-6879(03)74020-8 S0076687903740208. nosystems: application to microtubules and the ribosome, Proc. Natl. Acad. Sci. U.
[27] R.A. Laskowski, M.W. MacArthur, D.S. Moss, J.M. Thornton, PROCHECK: a pro- S. A. 98 (18) (2001) 10037–10041, http://dx.doi.org/10.1073/pnas.181342398
gram to check the stereochemical quality of protein structures, J. Appl. Crystallogr. 181342398.
26 (2) (1993) 283–291, http://dx.doi.org/10.1107/S0021889892009944. [45] N. Rai, A. Ramaswamy, Temperature dependent dynamics of DegP-trimer: a mo-
[28] M. Wiederstein, M.J. Sippl, ProSA-web: interactive web service for the recognition lecular dynamics study, Comput. Struct. Biotechnol. J. 13 (2015) 329–338, http://
of errors in three-dimensional structures of proteins, Nucleic Acids Res. 35 (Web dx.doi.org/10.1016/j.csbj.2015.04.004 S2001-0370(15)00023-9.
Server issue) (2007) W407–W410, http://dx.doi.org/10.1093/nar/gkm290 [46] C.H. Huang, S.H. Chiou, Proteomic analysis of upregulated proteins in Helicobacter
gkm290. pylori under oxidative stress induced by hydrogen peroxide, Kaohsiung J. Med. Sci.
[29] X. Robert, P. Gouet, Deciphering key features in protein structures with the new 27 (12) (2011) 544–553, http://dx.doi.org/10.1016/j.kjms.2011.06.019 S1607-
ENDscript server, Nucleic Acids Res. 42 (Web Server issue) (2014) W320–W324, 551X(11)00140-9.
http://dx.doi.org/10.1093/nar/gku316 gku316. [47] C.Y. Hong, Y.K. Kim, J.H. Chang, S.H. Kim, H. Choi, D.H. Nam, Y.Z. Kim, J.H. Kwak,
[30] R. Brenke, D. Kozakov, G.Y. Chuang, D. Beglov, D. Hall, M.R. Landon, C. Mattos, Novel fluoroquinolone antibacterial agents containing oxime-substituted
S. Vajda, Fragment-based identification of druggable 'hot spots' of proteins using (Aminomethyl)pyrrolidines:  synthesis and antibacterial activity of 7-(4-
Fourier domain correlation techniques, Bioinformatics 25 (5) (2009) 621–627, (Aminomethyl)-3-(methoxyimino)pyrrolidin-1-yl)-1-cyclopropyl-6-fluoro- 4-oxo-
http://dx.doi.org/10.1093/bioinformatics/btp036 btp036. 1,4-dihydro[1,8]naphthyridine-3-carboxylic acid (LB20304),1, J. Med. Chem. 40
[31] R.A. Friesner, J.L. Banks, R.B. Murphy, T.A. Halgren, J.J. Klicic, D.T. Mainz, (22) (1997) 3584–3593, http://dx.doi.org/10.1021/jm970202e.
M.P. Repasky, E.H. Knoll, M. Shelley, J.K. Perry, D.E. Shaw, P. Francis, [48] B. Patil, S. Banerjee, Importance of piperidine moiety in medicinal Chemistry re-
P.S. Shenkin, Glide: a new approach for rapid, accurate docking and scoring. 1. search: a review, J. Pharm. Res. 5 (12) (2012) 5493–5509.
Method and assessment of docking accuracy, J. Med. Chem. 47 (7) (2004) [49] M. Vilums, A.J. Zweemer, S. Dekkers, Y. Askar, H. de Vries, J. Saunders, D. Stamos,
1739–1749, http://dx.doi.org/10.1021/jm0306430. J. Brussee, L.H. Heitman, I.J. AP, Design and synthesis of novel small molecule
[32] R.A. Friesner, R.B. Murphy, M.P. Repasky, L.L. Frye, J.R. Greenwood, T.A. Halgren, CCR2 antagonists: evaluation of 4-aminopiperidine derivatives, Bioorg. Med. Chem.
P.C. Sanschagrin, D.T. Mainz, Extra precision glide: docking and scoring in- Lett 24 (23) (2014) 5377–5380, http://dx.doi.org/10.1016/j.bmcl.2014.10.060.
corporating a model of hydrophobic enclosure for protein-ligand complexes, J.

377

You might also like