Sie sind auf Seite 1von 10

This article was downloaded by: [Nipissing University]

On: 13 October 2014, At: 20:44


Publisher: Taylor & Francis
Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: Mortimer House,
37-41 Mortimer Street, London W1T 3JH, UK

Molecular Simulation
Publication details, including instructions for authors and subscription information:
http://www.tandfonline.com/loi/gmos20

Molecular dynamics study on drug resistance


mechanism of HCV NS3/4A protease inhibitor:
BI201335
a a
Jianjian Fu & Jing Wei
a
Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of
Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, P.R. China
Published online: 27 May 2014.

Click for updates

To cite this article: Jianjian Fu & Jing Wei (2014): Molecular dynamics study on drug resistance mechanism of HCV NS3/4A
protease inhibitor: BI201335, Molecular Simulation, DOI: 10.1080/08927022.2014.917298

To link to this article: http://dx.doi.org/10.1080/08927022.2014.917298

PLEASE SCROLL DOWN FOR ARTICLE

Taylor & Francis makes every effort to ensure the accuracy of all the information (the Content) contained
in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no
representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the
Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and
are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and
should be independently verified with primary sources of information. Taylor and Francis shall not be liable for
any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever
or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of
the Content.

This article may be used for research, teaching, and private study purposes. Any substantial or systematic
reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any
form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http://
www.tandfonline.com/page/terms-and-conditions
Molecular Simulation, 2014
http://dx.doi.org/10.1080/08927022.2014.917298

Molecular dynamics study on drug resistance mechanism of HCV NS3/4A protease


inhibitor: BI201335
Jianjian Fu and Jing Wei*
Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin
University, Tianjin 300072, P.R. China
(Received 28 November 2013; final version received 15 April 2014)

Hepatitis C virus (HCV) infection is a serious threat to global health. NS3 serine protease is one of the most advanced HCV
drug targets. However, the high mutation rate makes many protease inhibitors ineffective and allows viral replication to
continue. To investigate the structural basis of the molecular mechanism of HCV resistance to inhibitors, molecular
dynamics and molecular mechanics Poisson Boltzmann/surface area calculations were carried out on HCV NS3 serine
protease BI201335 complexes. The drug resistance to BI201335 is explained by the fact that seven single mutations
weaken the biological activity by lessening the sum of electrostatic interactions in the gas phase and polar solvation. The
computational results demonstrate that the mutations affect the BI201335 binding through direct and indirect mechanisms.
Downloaded by [Nipissing University] at 20:44 13 October 2014

Seven single mutations lead to significant changes in the conformation, such as the shifts of the side chain of His57 and
Lys136 and the movement of the P2 group of BI201335 towards the solvent. Furthermore, the contributions of Lys136
significantly decrease, which is the most major binding attraction. The shifts of the side chain of His57 induce the lack of
hydrogen bond between His57 with Asp81 expert for D168G mutation. Detailing the molecular mechanisms of BI201335
drug resistance provides some helpful insights into the nature of mutational effect and aid the rational design of potent
inhibitors combating HCV.
Keywords: HCV NS3 serine protease; BI201335; drug resistance mechanism; MM-PBSA

1. Introduction improved tolerance. Second-wave protease inhibitors offer


Estimates of the global prevalence of hepatitis C virus improved pharmacokinetics and tolerable side effect
(HCV) reveal that 170 million people worldwide are profiles, such as BI201335, simprevir and asunaprevir,
infected with HCV, representing , 3% of the worlds which are in phase III trials.[9,10]
population.[1] HCV is recognised as a major cause of BI201335, developed by Boehringer Ingelheim Incor-
chronic liver disease, cirrhosis, hepatocellular carcinomas, poration, is a promising drug candidate that targets the
as well as liver transplantations in developed countries. viral NS3/NS4A serine protease.[11] Recently, the crystal
[2,3] More than 350,000 people die from hepatitis C- structure of BI201335 bound to NS3/4A was reported,[12]
related liver diseases every year.[1] However until now, which shows that BI201335 adopts an extended confor-
there is no effective protective vaccine to fight HCV mation, similar to other protease inhibitors, to interact with
infection owing to its high mutation rate. the enzymes substrate-binding site (Figure 1). However,
For the past decade, the standard treatment of HCV unlike other inhibitors, BI201335 possesses a C-terminal
infection is a combination of pegylated interferon a and carboxylic acid that non-covalently binds to the active site,
ribavirin (Peg-IFN/RBV). Unfortunately, Peg-IFN/RBV and a bromo-quinoline substitution on its proline residue
produces sustained virological response in only 45% of that provides significant potency to the viral protease.[12]
genotype 1-infected patients and is limited by substantial As patients begin treatment, some mutations arise as a
side effects.[4,5] In 2011, the treatment of HCV infection consequence of the selective pressures of antiviral drugs
has been revolutionised by the approval of two ketoamide and result in drug-resistant variants. To unravel the atomic
inhibitors boceprevir and telaprevir, which are the first mechanism of drug resistance, it is essential to develop
direct-acting antiviral agents against the NS3/4A serine specific and effective drugs against HCV. Several
protease-inhibiting intracellular viral replication.[6 8] computational studies have been performed to understand
However, these first-generation drugs also have significant the drug resistance mechanism of NS3/4A mutations: the
limitations due to a thrice-daily dosing (with food), free energy perturbation method and molecular mechan-
extensive drug drug interactions and low barriers to ism Poisson Boltzmann surface area (MM-PBSA)
resistance. Thus, there is an urgent need to develop the method.[13 19] Molecular mechanics (MM) force field
virus-specific drugs that have promising efficacy and is one of the most popular approaches to estimate the

*Corresponding author. Email: jingwei_201311@163.com


q 2014 Taylor & Francis
2 J. Fu and J. Wei

Figure 1. (Colour online) The ligand binding modes of BI201335 in WT NS3/4A. For clarity, only interacting residues are displayed. (a)
Downloaded by [Nipissing University] at 20:44 13 October 2014

Molecular surface representation of the active site of the WT NS3/4A protease (PDB ID: 3P8N) with BI201335 and the selected sub-sites
are indicated (H-bond, hydrophobic and mild polar are coloured in magenta, green and blue, separately). (b) 2D chemical structure of
BI201335 and the schematic representation of the hydrogen bond interaction with NS3/4A protease (ligand exposure is coloured in
purple.

affinity of protein ligand binding in rational drug the desired residue followed by MD simulated annealing
discovery. optimisation.
The high-resolution structure of the NS3/4A
BI201335 complex can be a starting point to understand
the binding and resistance mechanism due to point 2.2 MD simulations
mutations. In this study, on the basis of the structure of MD simulations of the eight systems were performed
BI201335 in complex with the wild-type (WT) NS3/4A using the SANDER module, implemented in the Amber
and its seven single-site mutations, molecular dynamics 10.0 package.[23] The Amber parm99SB [24] force field
(MD) simulation, binding free energy calculation and free and the general AMBER force field [25] were,
energy decomposition were performed to explore the respectively, used for the protein and the ligand. The
binding and resistance mechanism of NS3/4A to force field parameters of BI201335 were prepared with the
BI201335. The binding free energies were calculated by Antechamber module.[26] Counter ions (Cl2) were added
using MM, polar solvation energy from Generalized Born to neutralise the charge of each inhibitor-bound system.
(GB)/Poisson Boltzmann (PB) continuum solvation, and The complex was solved in the TIP3P water box,[27] with
non-polar solvation energy from a surface area model, the truncated octahedron periodic box of size 8 A under the
referred to as MM-GBSA/MM-PBSA.[20,21] explicit waters. The particle mesh Ewald method [28] was
used to treat long-range electrostatic interactions. The cut-
off distance for the long-range electrostatic and the van der
2. Materials and methods Waals energy terms were set at 8.0 A. The SHAKE
algorithm [29] was applied to fix all covalent bonds
2.1 Preparation of the system
containing a hydrogen atom. The solvated system was
The initial crystal structures of the WT HCV NS3/4A energy-minimised in three steps prior to the MD
serine protease complexed with the drug BI201335 (PDB simulation. At first, the protein ligand was frozen while
code: 3P8N) with a resolution of 1.9 A [12] was extracted the solvent water molecules and counterions were allowed
from atomic coordinate entries available in the Protein to move. Next, the ligand and the side chain of the receptor
Date Bank. An Na ion was removed for the reason that it residues were allowed to move in addition to the water
is far from the active site of the protease. molecules. Finally, all atoms were permitted to move
As the experimentally determined structures of the freely. Subsequently, the complex of the minimised
mutant proteins were not available, Modeller software [22] structures was heated from 0 to 300 K for 100 ps
was used to generate seven mutant structures (R155K/Q, calculation, the temperature was maintained at a
A156V/T, D168G/A/V) in complex with BI201335 by temperature of 300 K with a coupling coefficient of
substituting specific residues using the WT structure as the 2.0 fs. After heating calculation, each system was
template. The procedure involved the mutated residue to equilibrated in the canonical ensemble of density
Molecular Simulation 3

calculation for 100 ps and thus for 0.5 ns without restraint. energies in several earlier studies applying the same
Finally, the systems were subjected to a 30- ns MD approximation.[32,33] In our calculations, the energy
simulation at 300 K and 1 atm pressure with an integration contribution from entropy changes on ligand binding was
step of 2 fs and NPT as an ensemble type. The MD not included.
simulation was performed with a periodic boundary
condition. Atom coordinates were collected to analyse the
structure in detail using the PTRAJ modules. The 2.4 Residue inhibitor interaction decomposition
convergence of energies, the H-bond, the distance and On account of the huge demand of computational
the atomic root-mean-square deviation (RMSD) of the resources for PB calculations, the interaction between
stability of the systems were analysed based on the above- inhibitors and each residue in protease was analysed using
mentioned MD simulations. the MM-GBSA module in AMBER 10.[34] Energy
decomposition calculation analyses same snapshots as
the free energy calculation. The binding interaction of
2.3 MM-PBSA calculations each inhibitor residue pair includes four terms:
MM-PBSA method in AMBER10 was implemented to
DGinhibitor2residues DGvdw DGele DGgb DGnp : 5
calculate the binding free energies between BI201335 and
Downloaded by [Nipissing University] at 20:44 13 October 2014

the protease.[20,21,23] For each system, a total number of


where van der Waals contribution (DGvdw) and electro-
4000 snapshots were extracted from the last 20-ns MD
static contribution (DGele) can be calculated by the Sander
simulation. In general, the binding free energies in
program in Amber 10.0. The polar solvation contribution
condensed phase can be calculated according to the
(DGgb) is computed using the GB module.
following equations.
The total free energy of binding was composed of the
following terms:
3. Results and discussion
DGb DEMM DGsol 2 TDS; 1 3.1 Stability and flexibility of the system simulations
To explore the effect of mutations on the conformational
DEMM DEele DEvdw ; 2 stability of NS3/4A BI201335 complexes, the RMSD of
backbone of the protease and inhibitor to their original
conformations during the 30 ns production phase were
DGsol DGpb DGnp ; 3 calculated and are depicted in Figure 2. The RMSD plots
indicate that the conformations of the WT and seven
where DGb is the binding free energy in solution; DEMM is single-mutant NS3/4A BI201335 complexes became
the MM free energy in gas phase, comprising electrostatic
(DEele) and van der Waals (DEvdw) interactions between
the ligand and the protease. The salvation free energy
(DGsol) is further divided into two components, where
DGsol is the solvation free energy, and DGpb and DGnp are
the polar and non-polar contributions to the solvation free
energy, respectively.[30,31] DGpb is determined by solving
the PB equations numerically and calculates the electro-
static energy according to the electrostatic potential,[31]
and the non-polar contribution is estimated by the solvent-
accessibility surface area (SASA) method, where the
values for g and b were set to 0.00542 kcal/(mol A2) and
0.92 kcal/mol, respectively [30]:

DGnp gSASA b: 4

The contribution of entropy (TDS) to binding free energies


is an important contribution in enzyme ligand bindings.
However, a number of studies published by Kollman and
Kuhn and Warshels group showed that entropy does not Figure 2. RMSDs, all Ca atoms o the NS3/4A BI201335
complex during the MD simulations complexes during the
contribute much to the relative binding free energies of the production phases relative to the initial minimised structures. (a)
ligands to the same protein and the good agreement Time evolution of the RMSD of all protein backbone atoms. (b)
between the calculated and experimental relative binding Time evolution of the RMSD of heavy atoms for the inhibitors.
4 J. Fu and J. Wei

Table 1. Binding free energy components for the NS3/4A BI201335 complexes by using the MM-PBSA method (kcal/mol).

DEvdw DEele DEMM DGnp DGpb DGsol

Complex DEele DGpb DGb Resistance level [35]


WT 244.50 2282.24 2 326.75 2 6.76 281.22 1.02 274.46 252.30 1
A156V 246.63 2219.09 2 265.72 2 6.91 225.25 6.16 218.35 247.38 150 ^ 10
A156T 241.19 2240.20 2 281.39 2 5.72 242.59 2.39 236.87 244.53 270 ^ 98
R155Q 247.75 2219.37 2 267.12 2 6.56 227.96 8.59 221.40 245.72 60 ^ 9
R155K 244.69 2246.14 2 290.83 2 6.09 251.13 4.99 245.04 245.79 350 ^ 14
D168G 252.33 2228.14 2 280.47 2 7.45 250.64 21.50 243.19 237.29 80 ^ 30
D168A 249.44 2225.75 2 275.19 2 7.10 243.94 18.19 236.84 238.35 690 ^ 332
D168V 237.71 2266.93 2 304.74 2 5.80 265.66 21.27 259.86 244.88 970 ^ 256

equilibrated and stable after 10 ns. The RMSD of all eight 245.72, 237.29, 247.38, 244.53, 245.79, 238.35 and
complexes exhibit low backbone RMSD values with mean 244.88 kcal/mol, respectively.
values around 0.86 1.31 A for the backbone and around Figure 3 depicts the insignificant changes in the
Downloaded by [Nipissing University] at 20:44 13 October 2014

1.80 2.66 A for inhibitor, respectively. The trajectory of energetic profile. In all eight protease inhibitor com-
A156V mutant/BI201335 complex fluctuates more than plexes, the MM free energies are the main driving force of
other trajectories, which implies that the A156V mutant energies to the inhibitor binding. In particular, electrostatic
complex undergoes larger changes in conformation than interactions in the gas phase provide extraordinarily
other seven complexes (1.31 A for the backbone and significant contribution to the binding free energies.
2.66 A for inhibitor, respectively). This result suggests that However, their favourable effects are offset by polar
the conformations are similar to the starting structures, and solvation terms that are positive and unfavourable for the
the stabilities of the dynamic equilibriums for eight binding free energies. The resulting balance of the two
complexes are reliable. Considering these, the confor- contributions, the electrostatic interactions in the gas phase
mations of the complexes after equilibrium were chosen to and the polar solvation terms, is slightly unfavourable to
assess the quality of MD simulations, energetic and binding interactions in all eight systems. In eight systems,
structural properties. van der Waals energies are about seven times stronger than
non-polar solvation energies. Thus, it can be concluded
that van der Waals energy is also the driving force for the
3.2 Analyses of binding free energies binding of BI201335 NS3/4A complexes. Non-polar
To gain insight into the forces involved in substrate binding, solvation terms, which correspond to the burial of SASA
total binding free energy and individual energy components upon binding, contribute slightly favourably.
for each system were obtained by using the MM/PBSA Compared with the WT, the binding affinity of each
method based on these trajectories. Table 1 lists the van der single mutation complex decreases by 4.92 15.01 kcal/
Waals (DEvdw), electrostatic (DEele), non-polar solvation mol, which elucidates that the mutant NS3/4A binds
(DGnp) and electrostatic solvation (DGpb) for the eight weakly to BI201335 than the WT NS3/4A. This also
proteaseinhibitor complexes. As shown in Table 1, the explains the fact that mutated NS3/4A proteases exhibit
predicted binding free energies of BI201335 to WT, R155Q, drug resistance to BI201335. It is very encouraging that
D168G, A156V, A156T, R155K, D168A and D168V the resistance levels of available experiment [35] of the
mutants of HCV NS3/4A serine protease are 252.30, BI201335 mutant NS3/4A complexes are consistent with

Figure 3. Profiles of various binding energy components (kcal/mol) for the WT and mutant NS3/4A BI201335 complexes.
Molecular Simulation 5
Downloaded by [Nipissing University] at 20:44 13 October 2014

Figure 4. Per-residue interaction spectrum of NS3/4A serine protease with BI201335. (a) WT, (b) A156T/V, (c) R155K/Q, (d) D168A/
G/V.

our predictions. The differences of the non-polar solvation mutations have obvious effect on the interaction between
energies among the eight systems are very small, which Lys136 and BI201335, where DGb values of Lys136 drop
indicates that the better cavity packing is maintained in the to some extent (ranging from 2 1.11 to 2 5.16 kcal/mol).
mutated complexes. The decrease of the sum of The decrease in the contribution of Lys136 can explain the
electrostatic interactions in the gas phase and the polar drug resistance to BI201335. For all eight systems, the
solvation terms should be essential to the resistance main force for amino acid His57 is van der Waals energy.
relative to the WT. The contribution of His57 to the binding affinity is
stronger in D168A, R155Q/K and A156V/T than WT.
Interestingly, the decompositions of DGb values of Asp81
3.3 Free energy decomposition analysis residues have unfavourable contribution of 0.69
To locate the residues, which contribute to BI201335 1.22 kcal/mol except for D168A mutation that shows a
interaction, and understand the mutation mechanisms, favourable contribution of 2 1.50 kcal/mol.
absolute binding free energies are calculated for eight
complexes by MM/GBSA method. The binding free
energy is decomposed into inhibitor residue pairs in order 3.4 Intermolecular hydrogen bonds analysis
to generate an inhibitor residue interaction spectrum. Hydrogen bonds play an essential role in stabilising
Figure 4 illustrates the decomposition of DGb values on a protein ligand complexes. To understand the effects of
per-residue basis for eight complexes. mutations on the inter-molecular interactions, we
For WT systems, the major binding attractions come investigated the geometry and stability of the hydrogen-
from the residues Lys136, Gly137, Ser138, Ser139, bond network formed by the inhibitor in the binding
Arg155 and Ala156, suggesting that they play important pocket. In Table 2, the protease BI201335 H-bond
roles in the binding. The favourable forces of these six contacts are characterised in terms of distances between
residues are electrostatic energies, which is in accordance heavy atoms and their percentage of occurrence on the
with the H-binding network from the experiment. Lys136 basis of the trajectories of the MD simulations (4000
is the most important amino acid that contributes a lot to snapshots). For the identification of hydrogen bonds, the
the binding affinities (2 6.79 kcal/mol). Seven single distance between donor and acceptor heavy atom was less
6 J. Fu and J. Wei

Table 2. The occupancy (%) of hydrogen bonds.

Donor Acceptor Acceptor WT A156V A156T R155Q R155K D168G D168A D168V
1@O18 157@H 157@N 99.56 96.22 99.83 99.90 99.68 99.23 91.16 90.78
1@O3 137@H 137@N 98.43 96.11 98.57 99.41 98.38 10.27 62.33 87.94
1@O1 139@HG 139@OG 93.68 94.22 97.65 97.05 97.59 14.45 29.44 73.27
157@O 1@H24 1@N24 98.14 95.30 99.24 98.44 99.06 89.71 89.92 89.46
81@OD2 j 57@H 57@N 50.65 0.19 2.08 1.00 2.33 47.42 8.27 3.17
80@O 155@HH11 155@NH2 64.17 0.10 72.19 60.70 30.68 16.41
155@O 1@H9 1@N9 72.84 33.70 73.02 73.40 45.24 81.40 85.16 72.92
168@OD2 155@HH21 155@NH2 87.44 78.62 85.33
168@OD1 155@HH21 155@NH2 71.29 55.40 78.95
168@OD2 155@HE 155@NE 63.100 71.00 72.76

than 3.5 A, and the D H A angle was greater than or binding affinity by only about 270 ^ 98-versus 150 ^ 10-
equal to 1208. fold change.[35]
BI201335 has a peptidic backbone so that it can form We can also observe that the non-bonded electrostatic
extended, substrate-like interactions with an exposed b- energy plays a major role in the ligand binding of
Downloaded by [Nipissing University] at 20:44 13 October 2014

barrel of the enzymes substrate-binding site. BI201335 BI201335 HCV NS3/4A WT and A156V/A156T mutant
forms five hydrogen bonds with WT protease that plays complexes. Although there is a decreased non-bonded
an important role in ligand binding (Figure 1), including electrostatic energy in both the mutant complexes, A156V
between the P1 P2 amide N9 and Arg155: O, between complex has a larger decrease than A156T mutation
the carbonyl O18 of the P2 P3 amide and Ala157: N, (2 219.09 kcal/mol vs. 2 240.20 kcal/mol). However, the
between the carbamate nitrogen of the N-terminal solvent shielding of this interaction would make its
capping group and Ala157: O, between the oxygen contribution by the displacement of P2 and P3 moieties.
atom O1 of carboxyl group and Ser139OG and between As shown in Table 1, a relatively higher de-solvation cost
the oxygen atom O3 of carboxyl group and Gly137N. was noticed for A156T (236.87 kcal/mol) than A156V
These five hydrogen bonds are preserved with occurrence (218.35 kcal/mol) which along with a decrease in DEvdw
frequencies of 72.84%, 99.56%, 98.14%, 93.68% and made it more resistant than A156V mutation
98.43%, respectively. These two H-bonds between (2 41.19 kcal/mol for A156T vs. 2 46.63 kcal/mol for
BI201335 and Ala157 are stable in the seven mutant A156V).
inhibitor complexes which have maintained about 90% Furthermore, we give a detailed picture of the effects
occupancy during the MD simulation. However, the of these changes in the binding pocket of BI201335.
hydrogen bonds between carboxyl group and Gly137 and Compared with the WT, we monitored the structural
Ser139 are weaker in D168G/A than other complexes, changes (shown in Figure 5(b)). Ala156 is mutated in a
especially D168G with lower occupancies of 10.27% and more bulky residue Val/Thr resulting in a steric hindrance
14.45%, respectively. The canonical substrate-like to BI201335. The crowding in the region makes the
intermolecular hydrogen bond between the P1 P2 quinolone-P2 substituent of BI201335 to move away from
amide N9 and Arg155: O is broken in A156V and the extended S2 sub-site. The BI201335 motion further
R155K mutations. induces the shift of the side chain of Lys136 and His57,
resulting in their contribution change.
The shift of key residue Lys136 affects severely the
3.5 The drug resistance mechanism of the studied interaction with BI201335 which its contribution drops to
mutants to BI201335 2 2.35/2 2.96 kcal/mol for A156V/A156T mutation while
3.5.1 A156V/A156T mutation 2 6.79 kcal/mol for WT, mainly from the decrease of
A156 is located on the S2 strand in close contact with both electrostatic interaction.
trans-hydroxyproline and bromo-quinoline P2 part of The shift of His57 leads to the loss of hydrogen bond
BI201335. The presence of P2-proline motif in all reported between His57 and Asp81, which makes the main chain of
inhibitors has been shown to cause increased susceptibility Gln80 and Asp81 more flexible. Especially, the hydrogen
against mutations at the A156 position.[17] bond between Gln80 and Arg155 is broken in the A156V
The total binding free energies of the mutant A156V mutant, which results in the decrease of the contribution of
and the mutant A156T are 2 47.38 and 2 44.53 kcal/mol Arg155 (2 0.75 kcal/mol). To profoundly analyse this
(Table 1), which agrees well with the experimental data phenomenon, the hydrogen bond distances (defined by the
that the A156T mutation affects the binding of BI201335 distance of Gln80O and Arg155NH2) were monitored
to a greater extent than A156V mutation, decreasing the over the all-time simulation, and the plots of distance
Molecular Simulation 7

versus the simulation time are shown in Figure 6. The plots guanidine at the bottom of the S2 pocket and adopts an
suggest that the hydrogen bond interaction between Gln80 extended conformation to bind with the large bromo-
and Arg155 disappears completely throughout the whole quinoline P2 substituent of BI201335. A salt bridge
simulation for A156V mutation. Interestingly, the formed between Asp168 with Arg155 and a hydrogen
contributions of His57 increase from 2 0.77 kcal/mol in bond between Gln80 and Arg155 play important roles in
WT NS3/4A compared with 2 3.02 kcal/mol in A156V the stabilisation of NS3/4A BI201335 complex. Thus,
mutation and 2 1.78 kcal/mol in A156T mutation, substitutions at this position are the major causes of
respectively, which indicates that the former is main clinical resistance to BI201335, often leading to a large
driving force of the van der Waals interaction and the latter decrease in ligand binding.
is main driving force of the electrostatic interaction. The main factor contributing to the resistance level of
R155Q/K was the decrease in the electrostatic interaction
(2 219.37 kcal/mol for R155Q and 2 246.14 kcal/mol for
3.5.2 R155Q/R155K mutation R155K). Both R155K and R155Q substitutions result in
To survey the changes in conformation, we superimposed the loss of salt bridge interactions with Asp168 and lack
the structure of NS3/4A BI201335 complexes based on hydrogen bond with Gln80, which makes Gln80 and
the coordinates of the protease backbone atoms (shown in Asp81 more flexible. Furthermore, the hydrogen bond
Figure 5(c)). The side chain of Arg155 locates its between His57 and Asp81 is lost and a significant shift in
Downloaded by [Nipissing University] at 20:44 13 October 2014

Figure 5. (Colour online) The superposition of the binding of BI201335 to WT and mutant NS3/4A. The proteins are shown in stick
model and the inhibitors are shown in wire model. WT NS3/4A BI201335 complex is colour-coded: magenta. (a) WT NS3/4A
BI201335 complex and NS3/4A BI201335 complex. (b) The superposition of the binding of BI201335 to WT, A156V mutant (green)
and A156T mutant (orange). (c) The superposition of the binding of BI201335 to WT and R155Q (green), R155K (orange). (d) The
superposition of the binding of BI201335 to WT and D168G (cyan), D168A (green) and D168V (orange).
8 J. Fu and J. Wei

decrease in binding of D168G/A/V variant proteases


with BI201335 is due to the indirect interaction for two
main reasons. First, the mutations of Asp168 prevent the
formation of the salt bridge between Arg155 and residue
168. Thus, Arg155 looses its S2-anchoring property
towards the P2 quinoline moiety of BI201335. Second, the
side chain of glycine/alanine/valine is shorter than the side
chain of aspartic acid, causing the shift of BI201335
towards the residue 168. In turn, the backbone of
BI201335 moves away from the original conformations
and the extended P2 group is further exposed to the
solvent.
The D168G mutation results in a strong 80-fold
resistance. However, the D168G mutation does not have
any direct impact on BI201335, which the decomposition
Figure 6. The monitoring for the hydrogen bond between Q80
and R155 during the all-time simulations. Plots show the time of the binding free energy of residue 168 has no significant
evolution of hydrogen bond distance of Gln80O and Arg155NH2 change. The Gly168 side chain is too short to provide more
Downloaded by [Nipissing University] at 20:44 13 October 2014

for WT NS3/4A BI201335 complex, A156V mutant NS3/4A interaction with the P2 group. Thus, D168G directly alters
BI201335 complex and A156T mutant NS3/4A BI201335 the conformation of the Arg155 side chain and indirectly
complex. alters the conformation of the complex between BI201335
and NS3/4A (Figure 5(d)). The P2 group adopts an
extended conformation in the solvent-exposed domain.
the positions of the side chain of His57 was observed.
The movement of BI201335 disrupts the hydrogen bonds
However, the contribution of His57 becomes stronger due with both Ser139 and Gly137 and results in the shift of the
to electrostatic and van der Waals interaction energies side chain of His57 and Lys136. The contributions of
(Supplementary Table 1). In addition, His57 bonds with Lys136, Ser138 and Ser139 are weaker in D168G than the
Lys155 through the hydrogen bond interaction with WT NS3/4A complex. Interestingly, an increase in the
occurrence frequency of 92.54% in R155K mutation. The interaction energy is observed for Arg155 with 0.83 kcal/
P2 group of the inhibitor has extended away from the S2 mol in D168G complexes compared with the WT
sub-site and more exposed to the solvent. Furthermore, the complex, which is mainly from electrostatic contribution.
side chain of Lys136 shifts to avoid crowding resulting The D168A and D168V mutations, even though with a
from the movement of P2 group. This explains the reason higher 690-fold and 970-fold resistance, share the same
for the decrease in the contribution of Lys136. molecular mechanism. However, the changes in confor-
When Arg155 is mutated to Lys155, the electrostatic mations disrupt the hydrogen bonds between His57 and
interactions between Lys155 and the P1 P2 amide N9 Asp81 and between Gln80 and Arg155. The contributions
thus weaken. Compared with WT with occupancy of of Gly137, Ser138, Ser139, Arg155 and Ala156 become
72.84%, the R155K mutation does have an obvious effect weaker in D168A mutations than in D168G mutations,
on the hydrogen bond between residue 155 and BI201335, which explains its stronger resistance. Interestingly, the
where the hydrogen bond occupancies are reduced to D168A mutation induced some additional favourable
45.24%. The above-mentioned result might be the interactions with Asp81, as compared with the unfavour-
explanation for the higher resistance level for R155K able interactions for the other seven systems.
than R155Q.

4. Conclusions
3.5.3 D168G/D168A/D168V mutations In our work, 30-ns MD simulations combined with the
During the interaction of NS3/4A and BI201335, Asp168 calculation of the binding free energy by using MM-PBSA
does not directly participate in the anchoring of BI201335. have been performed to investigate the mechanism of the
However, Asp168 is indirectly stabilising the confor- drug resistance to BI201335. The results have shown that
mation of Arg155 residue by a salt bridge, which is very the rank of calculated absolute binding free energy is in
important to orient Arg155 in a favourable conformation good agreement with the experimental result. The decrease
to BI201335 binding. in the electrostatic energy in the gas phase definitely drives
For all the three mutations, D168G/A/V, significant the drug resistance to BI201335. The mutations of A156,
efforts were focused on BI201335 because of its R155 and D168 seriously affect the conformation of
sensitivity to these mutations (80- to 970-fold resistance the side chain of Lys136 and His57, leading to the
level). Computational modelling indicates that the unfavourable induced-fit binding with BI201335. The
Molecular Simulation 9

mutations induce the movement of the large P2 moiety of [18] Courcambeck J, Bouzidi M, Perbost R, Jouirou B, Amrani N,
Cacoub P, Pepe G, Sabatier J-M, Halfon P. Resistance of hepatitis C
BI201335 away from the extended S2 sub-site. These virus to NS3-4A protease inhibitors: mechanisms of drug resistance
detailed insights into the mechanism of BI201335 induced by R155Q, A156T, D168A and D168V mutations. Antiviral
resistance will be very useful in the structure-based Ther. 2006;11:847855.
[19] Lin C, Lin K, Luong YP, Rao BG, Wei YY, Brennan DL, Fulghum
designing of novel inhibitors that will be less susceptible JR, Hsiao HM, Ma S, Maxwell JP, Cottrell KM, Perni RB, Gates
to resistance. CA, Kwong AD. In vitro resistance studies of hepatitis C virus
serine protease inhibitors, VX-950 and BILN-2061: modeling
analysis suggests that there are different mechanisms of resistance to
VX-950 and BILN-2061. J Biol Chem. 2004;17:1750817514.
References [20] Srinivasan J, Cheatham, III TE, Cieplak P, Kollman PA, Case DA.
[1] World Health Organization. Media centre fact sheet N 164. [updated Continuum solvent studies of the stability of DNA, RNA, and
2013 July] Available from: http://www.who.int/mediacentre/facts phosphoramidate-DNA helices. J Am Chem Soc. 1998;
heets/fs164/en/ 120:9401 9409.
[2] Verna EC, Brown RS. Hepatitis C virus and liver transplantation. [21] Kollman PA, Massova I, Reyes C, Kuhn B, Huo S, Chong Ll, Lee
Semin Liver Dis. 2006;10:919. M, Lee T, Duan Y, Wang W, Donini O, Cieplak P, Srinivasan J,
[3] Moradpour D, Penin F, Rice CM. Replication of hepatitis C virus. Case DA, Cheatham, III TE. Calculating structures and free energies
Nat Rev Microbiol. 2007;5:453 463. of complex molecules: combining molecular mechanics and
[4] Soriano V, Peters MG, Stefan Z. New therapies for hepatitis C virus continuum models. Acc Chem Res. 2000;33:889897.
infection. Clin Infect Dis. 2009;48:313320. [22] Eswar N, Marti-Renom MA, Webb B, Madhusudhan MS, Eramian
[5] Lemona SM, Mckeatingb JA, Pietschmannc T, Frickd DN, Glenne D, Shen M, Pieper U, Sali A. Comparative protein structure
modeling with MODELLER. Curr Protocol Bioinform. 2006;15
Downloaded by [Nipissing University] at 20:44 13 October 2014

JS, Tellinghuisenf TL, Symonsg J, Furmanh PA. Development of


novel therapies for hepatitis C. Antiviral Res. 2010;86:7992. (Supplement):5.6.15.6.30.
[6] Welsch C, Jesudian A, Zeuzem S, Jacobson I. New direct-acting [23] Case DA, Kolossvary I, Wong KF, Paesani F, Vanicek J, Wu X,
antiviral agents for the treatment of hepatitis C virus infection and Brozell SR, Steinbrecher T, Gohlke H, Yang L, Tan C, Mongan J,
perspectives. Gut. 2012;61:3646. Hornak V, Cui G, Mathews DH, Seetin MG, Sagui C, Babin V,
[7] Kim JJ, Culley CM, Mohammad RA. Telaprevir: an oral protease Kollman PA. AMBER 10. San Francisco, CA: University of
inhibitor for hepatitis C virus infection. Am J Health-Syst Pharm. California; 2008.
2012;69:19 33. [24] Hornak V, Abel R, Okur A, Strockbine B, Roitberg A, Simmerling
[8] Trembling PM, Tanwar S, Dusheiko GM. Boceprevir: an oral C. Comparison of multiple amber force fields and development of
protease inhibitor for the treatment of chronic HCV infection. improved protein backbone parameters. Proteins. 2006;65:
Expert Rev Anti Infect Ther. 2012;10:269279. 712725.
[9] Clark VC, Peter JA, Nelson DR. New therapeutic strategies in HCV: [25] Wang J, Wolf RM, Caldwell JW, Kollman PA, Case DA.
second-generation protease inhibitors. Liver Int. 2013;33:8084. Development and testing of a general AMBER force field. J
[10] Fusco DN, Chung RT. New protease inhibitors for HCV: help is on Comput Chem. 2004;25:11571174.
the way. J Hepatol. 2011;54:1087 1089. [26] Wang JM, Wang W, Kollman PA, Case DA. Antechamber, an
[11] Brunet ML, Bailey MD, Goudreau N, Bhardwaj PK, Bordeleau J, software package for molecular mechanical calculations. J Chem Inf
Bos M, Bousquet Y, Cordingley MG, Duan J, Forgione P, Garneau Comput. 2001;222:U403.
M, Ghiro E, Gorys V, Goulet S, Halmos T, Kawai SH, Naud J, [27] Jorgensen WL, Chandrasekhar J, Madura JD, Impey RW, Klein ML.
Poupart MA, White PW. Discovery of a potent and selective non- Comparison of simple potential functions for simulating liquid
covalent linear inhibitor of the hepatitis C virus NS3 protease water. J Chem Phys. 1983;79:926935.
(BI201335). J Med Chem. 2010;53:6466 6476. [28] Essmann U, Perera L, Berkowitz ML, Darden T, Lee H, Pedersen
[12] Lemke CT, Goudreau N, Zhao S, Hucke O, Thibeault D, Brunet LG. A smooth particle mesh Ewald method. J Chem Phys. 1995;
ML, White PW. Combined X-ray, NMR, and kinetic analyses reveal 103:8577 8593.
uncommon binding characteristics of the hepatitis C virus NS3- [29] Ryckaert JP, Ciccotti G, Berendsen HJC. Numerical integration of
NS4A protease inhibitor BI201335. J Biol Chem. 2011; the Cartesian equations of motion of a system with constraints:
286:1143411443. molecular dynamics of n-alkanes. J Comput Phys. 1977;23:
[13] Hotiana HA, Haider MK. Structural modeling of HCVNS3/4A 327341.
serine protease drug-resistance mutations using end-point con- [30] Sitkoff D, Sharp KA, Honig BH. Accurate calculation of hydration
tinuum solvation and side-chain flexibility calculations. J Chem Inf free energies using macroscopic solvent models. J Phys Chem.
Model. 2013;53:435451. 1994;98:1978 1988.
[14] Xue W, Pan D, Zhang W, Liu H, Yao X. Molecular modeling study [31] Gilson MK, Sharp KA, Honig BH. Calculating the electrostatic
on the resistance mechanism of HCV NS3/4A serine protease potential of molecules in solution: method and error assessment.
mutants R155K, A156V and D168A to TMC435. Antiviral Res. J Comput Chem. 1987;9:327335.
2012;93:126 137. [32] Hou T, Wang J, Li Y, Wang W. Assessing the performance of the
[15] Pan D, Xue W, Zhang W, Liu H, Yao X. Understanding the drug MM/PBSA and MM/GBSA method. 1. The accuracy of binding free
resistance mechanism of hepatitis C virus NS3/4A to ITMN-191 due energy calculations based on molecular dynamics simulations.
to R155K, A156V, D168A/E mutations: a computational study. J Chem Inf Model. 2011;51:6982.
Biochim Biophys Acta. 2012;1820:15261534. [33] Rastelli G, Del Rio A, Degliesposti G, Sgobba M. Fast and accurate
[16] Guo Z, Prongay A, Tong X, Fischmann T, Bogen S, Velazquez F, predictions of binding free energies using MM-PBSA and MM-
Venkatraman S, Njoroge FG, Madison V. Computational study of GBSA. J Comput Chem. 2010;31:797810.
the effects of mutations A156T, D168V, and D168Q on the binding [34] Onufriev A, Bashford D, Case DA. Modification of the generalized
of HCV protease inhibitors. J Chem Theory Comput. 2006;2: born model suitable for macromolecules. J Phys Chem B. 2000;104:
16571663. 37123720.
[17] Ortqvist P, Vema A, Ehrenberg AE, Dahl G, Ronn R, Akerblom E, [35] Lagace L, White PW, Bousquet C, Dansereau N, Do F, Linas-
Karlen A, Danielson UH, Sandstrom A. Structure activity Brunet M, Marquis M, Massariol M-J, Maurice R, Spickler C,
relationships of HCV NS3 protease inhibitors evaluated on the Thibeault D, Triki I, Zhao S, Kukolj G. In vitro resistance profile of
drug-resistant variants A156T and D168V. Antiviral Ther. the hepatitis C virus NS3 protease inhibitor BI201335. Antimicrob
2010;15:841 852. Agents Chemother. 2011;56:569 572.

Das könnte Ihnen auch gefallen