2,237
Views
251
CrossRef citations to date
0
Altmetric
Review

Molecular dynamics simulations and novel drug discovery

, , , , &
Pages 23-37 | Received 01 Jul 2017, Accepted 07 Nov 2017, Published online: 15 Nov 2017

References

  • Ye W, Wang W, Jiang C, et al. Molecular dynamics simulations of amyloid fibrils: an in silico approach. Acta Biochim Biophys Sin (Shanghai). 2013;45:503–508.
  • Zhang Z, Chen H, Bai H, et al. Molecular dynamics simulations on the oligomer-formation process of the GNNQQNY peptide from yeast prion protein Sup35. Biophy J. 2007;93:1484–1492.
  • Barducci A, Chelli R, Procacci P, et al. Misfolding pathways of the prion protein probed by molecular dynamics simulations. Biophy J. 2005;88:1334–1343.
  • Urbanc B, Betnel M, Cruz L, et al. Elucidation of amyloid β-protein oligomerization mechanisms: discrete molecular dynamics study. J Am Chem Soc. 2010;132:4266–4280.
  • Campos SRR, Machuqueiro M, Baptista AM. Constant-pH molecular dynamics simulations reveal a β-rich form of the human prion protein. J Phys Chem B. 2010;114:12692–12700.
  • Chen W, Kamp MW, Daggett V. Structural and dynamic properties of the human prion protein. Biophys J. 2014;106:1152–1163.
  • Santini S, Derreumaux P. Helix H1 of the prion protein is rather stable against environmental perturbations: molecular dynamics of mutation and deletion variants of PrP (90–231). Cell Mol Life Sci. 2014;61:951–960.
  • DeMarco ML, Daggett V. From conversion to aggregation: protofibril formation of the prion protein. Proc Natl Acad Sci USA. 2004;101:2293.
  • Barducci A, Chelli R, Procacci P, et al. Metadynamics simulation of prion protein: β-structure stability and the early stages of misfolding. J Am Chem Soc. 2006;128:2705–2710.
  • Benetti F, Biarnés X, Attanasio F, et al. Structural determinants in prion protein folding and stability. J Mol Biol. 2014;426:3796–3810.
  • Karplus M, McCammon JA. Molecular dynamics simulations of biomolecules. Nat Struct Mol Biol. 2002;9:646–652.
  • Moore DJ, West AB, Dawson VL, et al. Molecular pathophysiology of Parkinson’s disease. Annu Rev Neurosci. 2005;28:57–87.
  • Kitchen DB, Decornez H, Furr JR, et al. Docking and scoring in virtual screening for drug discovery: methods and applications. Nat Rev Drug Discov. 2004;3:935–949.
  • McInnes C. Virtual screening strategies in drug discovery. Curr Opin Chem Boil. 2007;11:494–502.
  • Cheng T, Li Q, Zhou Z, et al. Structure-based virtual screening for drug discovery: a problem-centric review. Aaps J. 2012;14:133–141.
  • Rastelli G, Rio AD, Degliesposti G, et al. Fast and accurate predictions of binding free energies using MM-PBSA and MM-GBSA. J Comput Chem. 2010;31:797–810.
  • Alonso H, Bliznyuk AA, Gready JE. Combining docking and molecular dynamic simulations in drug design. Med Res Rev. 2006;26:531–568.
  • Yang Y, Shen Y, Liu H, et al. Molecular dynamics simulation and free energy calculation studies of the binding mechanism of allosteric inhibitors with p38α MAP kinase. J Chem Inf Model. 2011;51:3235–3246.
  • Xue W, Pan D, Yang Y, et al. Molecular modeling study on the resistance mechanism of HCV NS3/4A serine protease mutants R155K, A156V and D168A to TMC435. Antivir Res. 2012;93:126–137.
  • Zheng G, Xue W, Wang P, et al. Exploring the inhibitory mechanism of approved selective norepinephrine reuptake inhibitors and reboxetine enantiomers by molecular dynamics study. Sci Rep-UK. 2016;6:26883.
  • Haq FU, Abro A, Raza S, et al. Molecular dynamics simulation studies of novel β-lactamase inhibitor. J Mol Graph Model. 2017;74:143–152.
  • Haile JM. Molecular dynamics simulation. New York: Wiley; 1992.
  • Spellings M, Marson RL, Anderson JA, et al. GPU accelerated Discrete Element Method (DEM) molecular dynamics for conservative, faceted particle simulations. J Comput Phys. 2017;334:460–467.
  • Pronk S, Páll S, Schulz R, et al. GROMACS 4.5: a high-throughput and highly parallel open source molecular simulation toolkit. Bioinformatics. 2013;29:845–854.
  • Adcock SA, McCammon JA. Molecular dynamics: survey of methods for simulating the activity of proteins. Chem Rev. 2006;106:1589–1615.
  • Klepeis JL, Lindorff-Larsen K, Dror RO, et al. Long-timescale molecular dynamics simulations of protein structure and function. Curr Opin Struc Biol. 2009;19:120–127.
  • Phillips JC, Braun R, Wang W, et al. Scalable molecular dynamics with NAMD. J Comput Chem. 2005;26:1781–1802.
  • Chiti F, Dobson CM. Protein misfolding, functional amyloid, and human disease. Annu Rev Biochem. 2006;75:333–366.
  • Gsponer J, Haberthür U, Caflisch A. The role of side-chain interactions in the early steps of aggregation: molecular dynamics simulations of an amyloid-forming peptide from the yeast prion Sup35. Proc Natl Acad Sci USA. 2003;100:5154–5159.
  • Lemkul JA, Bevan DR. Assessing the stability of Alzheimer’s amyloid protofibrils using molecular dynamics. J Phys Chem B. 2010;114:1652–1660.
  • Eisenberg D, Jucker M. The amyloid state of proteins in human diseases. Cell. 2012;148:1188–1203.
  • Levine H. Thioflavine T interaction with synthetic Alzheimer’s disease β‐amyloid peptides: detection of amyloid aggregation in solution. Protein Sci. 1993;2:404–410.
  • Spillantini MG, Schmidt ML, Lee VMY, et al. α-Synuclein in lewy bodies. Nature. 1997;388:839–840.
  • Cooper GJ, Willis AC, Clark A, et al. Purification and characterization of a peptide from amyloid-rich pancreases of type 2 diabetic patients. Proc Natl Acad Sci USA. 1987;84:8628–8632.
  • Hsich G, Kenney K, Gibbs CJ Jr, et al. The 14-3-3 brain protein in cerebrospinal fluid as a marker for transmissible spongiform encephalopathies. New Engl J Med. 1996;335:924–930.
  • Boellaard JW, Schlote W, Tateishi J. Neuronal autophagy in experimental Creutzfeldt-Jakob’s disease. Acta Neuropathol. 1989;78:410–418.
  • Rowe CC, Ng S, Ackermann U, et al. Imaging β-amyloid burden in aging and dementia. Neurology. 2007;68:1718–1725.
  • Selkoe DJ. Alzheimer’s disease: genes, proteins, and therapy. Physiol Rev. 2001;81:741–766.
  • Gessel MM, Bernstein S, Kemper M, et al. Familial Alzheimer’s disease mutations differentially alter amyloid Β-protein oligomerization. ACS Chem Neurosci. 2012;3:909–918.
  • Nasica-Labouze J, Nguyen PH, Sterpone F, et al. Amyloid Β protein and Alzheimer’s disease: when computer simulations complement experimental studies. Chem Rev. 2015;115:3518–3563.
  • Bitan G, Vollers SS, Teplow DB. Elucidation of primary structure elements controlling early amyloid Β-protein oligomerization. J Biol Chem. 2003;278:34882–34889.
  • CôTé S, Laghaei R, Derreumaux P, et al. Distinct dimerization for various alloforms of the amyloid-beta protein: aβ1–40, Aβ1–42, and Aβ1–40 (D23n). J Phys Chem B. 2012;116:4043–4055.
  • Lin Y-S, Pande VS. Effects of familial mutations on the monomer structure of Aβ 42. Biophys J. 2012;103:47–49.
  • Lin Y-S, Bowman GR, Beauchamp KA, et al. Investigating how peptide length and a pathogenic mutation modify the structural ensemble of amyloid beta monomer. Biophys J. 2012;102:315–324.
  • Baumketner A, Krone MG, Shea J-E. Role of the familial Dutch mutation E22q in the folding and aggregation of the 15–28 fragment of the Alzheimer amyloid-Β protein. Proc Natl Acad Sci USA. 2008;105:6027–6032.
  • Huet A, Derreumaux P. Impact of the mutation A21g (Flemish Variant) on Alzheimer’s Β-amyloid dimers by molecular dynamics simulations. Biophys J. 2006;91:3829–3840.
  • Awasthi M, Singh S, Pandey VP, et al. Modulation in the conformational and stability attributes of the Alzheimer’s disease associated amyloid-beta mutants and their favorable stabilization by curcumin: molecular dynamics simulation analysis. J Biomol Struct Dyn. 2017;1–16.
  • Benilova I, Gallardo R, Ungureanu AA, et al. The Alzheimer disease protective mutation A2T modulates kinetic and thermodynamic properties of amyloid-β (Aβ) aggregation. J Biol Chem. 2014;289:30977–30989.
  • Das P, Murray B, Belfort G. Alzheimer’s protective A2T mutation changes the conformational landscape of the Aβ1-42 monomer differently than does the A2V mutation. Biophys J. 2015;108:738–747.
  • Zheng X, Liu D, Roychaudhuri R, et al. Amyloid β-protein assembly: differential effects of the protective A2T mutation and the recessive A2V familial Alzheimer’s disease mutation. ACS Chem Neurosci. 2015;6:1732–1740.
  • Nguyen PH, Sterpone F, Pouplana R, et al. Dimerization mechanism of Alzheimer Aβ40 peptides: the high content of intrapeptide-stabilized conformations in A2V and A2T heterozygous dimers retards amyloid fibril formation. J Phys Chem B. 2016;120:12111.
  • Viet MH, Nguyen PH, Derreumaux P, et al. Effect of the English familial disease mutation (H6r) on the monomers and dimers of Aβ40 and Aβ42. ACS Chem Neurosci. 2014;5:646–657.
  • Xu L, Chen Y, Wang X. Dual effects of familial Alzheimer’s disease mutations (D7h, D7n, and H6r) on amyloid Β peptide: correlation dynamics and zinc binding. Proteins. 2014;82:3286–3297.
  • Polymeropoulos MH, Lavedan C, Leroy E, et al. Mutation in the Α-synuclein gene identified in families with Parkinson’s disease. Science. 1997;276:2045–2047.
  • Zarranz JJ, Alegre J, Gómez‐Esteban JC, et al. The new mutation, E46k, of Α‐synuclein causes Parkinson and lewy body dementia. Ann Neurol. 2004;55:164–173.
  • Lesage S, Anheim M, Letournel F, et al. G51d Α‐synuclein mutation causes a novel Parkinsonian–pyramidal syndrome. Ann Neurol. 2013;73:459–471.
  • Xu L, Ma B, Nussinov R, et al. Familial mutations may switch conformational preferences in Α-synuclein fibrils. ACS Chem Neurosci. 2017;8:837–849.
  • Yu H, Han W, Ma W, et al. Transient Β-hairpin formation in Α-synuclein monomer revealed by coarse-grained molecular dynamics simulation. J Chem Phys. 2015;143:243142.
  • Wise-Scira O, Aloglu AK, Dunn A, et al. Structures and free energy landscapes of the wild-type and A30p mutant-type Α-synuclein proteins with dynamics. ACS Chem Neurosci. 2013;4:486–497.
  • Coskuner O, Wise-Scira O. Structures and free energy landscapes of the A53t mutant-type Α-synuclein protein and impact of A53t mutation on the structures of the wild-type Α-synuclein protein with dynamics. ACS Chem Neurosci. 2013;4:1101–1113.
  • Hazy E, Bokor M, Kalmar L, et al. Distinct hydration properties of wild-type and familial point mutant A53t of Α-synuclein associated with Parkinson’s disease. Biophys J. 2011;101:2260–2266.
  • Wise-Scira O, Dunn A, Aloglu AK, et al. Structures of the E46k mutant-type Α-synuclein protein and impact of E46k mutation on the structures of the Wild-Type Α-synuclein protein. ACS Chemi Neurosci. 2013;4:498–508.
  • Perlmutter JD, Braun AR, Sachs JN. Curvature dynamics of Α-synuclein familial Parkinson disease mutants molecular simulations of the micelle-and bilayer-bound forms. J Biol Chem. 2009;284:7177–7189.
  • Tsigelny IF, Sharikov Y, Kouznetsova VL, et al. Molecular determinants of Α-synuclein mutants’ oligomerization and membrane interactions. ACS Chem Neurosci. 2015;6:403–416.
  • Dlouhy SR, Hsiao K, Farlow MR, et al. Linkage of the Indiana Kindred of Gerstmann-Sträussler-Scheinker disease to the prion protein gene. Nat Genet. 1992;1:64–67.
  • Wilesmith JW, Wells G, Cranwell MP, et al. Bovine spongiform encephalopathy: epidemiological studies. Vet Rec. 1988;123:638–644.
  • Pan K-M, Baldwin M, Nguyen J, et al. Conversion of alpha-helices into beta-sheets features in the formation of the scrapie prion proteins. Proc Natl Acad Sci USA. 1993;90:10962–10966.
  • Takada LT, Kim MO, Cleveland RW, et al. Genetic prion disease: experience of a rapidly progressive dementia center in the United States and a review of the literature. Am J Med Genet B. 2017;174:36–69.
  • Doss CGP, Rajith B, Rajasekaran R, et al. In silico analysis of prion protein mutants: a comparative study by molecular dynamics approach. Cell Biochem Biophys. 2013;67:1307–1318.
  • Borgohain G, Dan N, Paul S. Use of molecular dynamics simulation to explore structural facets of human prion protein with pathogenic mutations. Biophys Chem. 2016;213:32–39.
  • Ning L, Pan D, Zhang Y, et al. Effects of the pathogenic mutation A117V and the protective mutation H111S on the folding and aggregation of Prp106-126: insights from replica exchange molecular dynamics simulations. PloS One. 2015;10:e0125899.
  • Zhang J, Wang F, Zhang Y. Molecular dynamics studies on the NMR structures of rabbit prion protein wild type and mutants: surface electrostatic charge distributions. J Biomol Struct Dyn. 2015;33:1326–1335.
  • van der Kamp MW, Daggett V. Pathogenic mutations in the hydrophobic core of the human prion protein can promote structural instability and misfolding. J Mol Biol. 2010;404:732–748.
  • Chen W, Van Der Kamp MW, Daggett V. Diverse effects on the native Β-sheet of the human prion protein due to disease-associated mutations. Biochemistry. 2010;49:9874–9881.
  • Guo J, Ren H, Ning L, et al. Exploring structural and thermodynamic stabilities of human prion protein pathogenic mutants D202n, E211q and Q217r. J Struct Biol. 2012;178:225–232.
  • Guo J, Ning L, Ren H, et al. Influence of the pathogenic mutations T188k/R/a on the structural stability and misfolding of human prion protein: insight from molecular dynamics simulations. BBA- Gen Subjects. 2012;1820:116–123.
  • Kittner M. Folding and aggregation of amyloid peptides [ Doctoral dissertation]. Potsdam: Universität Potsdam, Diss.; 2011.
  • Serpell LC. Alzheimer’s amyloid fibrils: structure and assembly. BBA-Mol Basis Dis. 2000;1502:16–30.
  • Grabenauer M, Wu C, Soto P, et al. Oligomers of the prion protein fragment 106−126 are likely assembled from Β-hairpins in solution, and methionine oxidation inhibits assembly without altering the peptide’s monomeric conformation. J Am Chem Soc. 2009;132:532–539.
  • Zhou S, Wang Q, Wang Y, et al. The folding mechanism and key metastable state identification of the Prp127–147 monomer studied by molecular dynamics simulations and Markov state model analysis. Phys Chem Chem Phys. 2017;19:11249–11259.
  • Chakraborty S, Chatterjee B, Basu S. A mechanistic insight into the amyloidogenic structure of hiapp peptide revealed from sequence analysis and molecular dynamics simulation. Biophys Chem. 2012;168:1–9.
  • Ning L, Wang Q, Zheng Y, et al. Effects of the A117V mutation on the folding and aggregation of palindromic sequences (PrP113-120) in prion: insights from replica exchange molecular dynamics simulations. Mol Biosyst. 2015;11:647–655.
  • Ning L, Guo J, Bai Q, et al. Structural diversity and initial oligomerization of PrP106–126 studied by replica-exchange and conventional molecular dynamics simulations. PloS One. 2014;9:e87266.
  • De Simone A, Pedone C, Vitagliano L. Structure, dynamics, and stability of assemblies of the human prion fragment SNQNNF. Biochem Bioph Res Co. 2008;366:800–806.
  • Xu W, Jiang P, Mu Y. Conformation preorganization: effects of S20G mutation on the structure of human islet amyloid polypeptide segment. J Phys Chem B. 2009;113:7308–7314.
  • Kundu S. Effects of different force fields on the structural character of Α synuclein Β-hairpin peptide (35-56) in aqueous environment. J Biomol Struct Dyn. 2017;1–16.
  • Sugita Y, Okamoto Y. Replica-exchange molecular dynamics method for protein folding. Chem Phys Lett. 1999;314:141–151.
  • Kästner J. Umbrella sampling. WIRES Comput Mol Sci. 2011;1:932–942.
  • Laio A, Gervasio FL. Metadynamics: a method to simulate rare events and reconstruct the free energy in biophysics, chemistry and material science. Rep Prog Phys. 2008;71:126601.
  • Pande VS. An introduction to Markov state models and their application to long timescale molecular simulation. Stanford: Advances in Experimental Medicine and Biology; 2013.
  • Freddolino PL, Harrison CB, Liu Y, et al. Challenges in protein folding simulations: timescale, representation, and analysis. Nat Phys. 2010;6:751.
  • Mo Y, Lu Y, Wei G, et al. Structural diversity of the soluble trimers of the human amylin (20–29) peptide revealed by molecular dynamics simulations. J Chem Phys. 2009;130:125101.
  • Nguyen PH, Derreumaux P. Conformational ensemble and polymorphism of the all-atom Alzheimer’s Aβ37–42 amyloid peptide oligomers. J Phys Chem B. 2013;117:5831–5840.
  • Guo J, Zhang Y, Ning L, et al. Stabilities and structures of islet amyloid polypeptide (IAPP22-28) oligomers: from dimer to 16-mer. BBA-Gen Subjects. 2014;1840:357–366.
  • Chua KP, Chew LY, Mu Y. Replica exchange molecular dynamics simulation of cross‐fibrillation of Iapp and Prp106‐126. Proteins. 2016;84:1134–1146.
  • Hu R, Zhang M, Chen H, et al. Cross-seeding interaction between Β-amyloid and human islet amyloid polypeptide. ACS Chem Neurosci. 2015;6:1759–1768.
  • Zhang M, Hu R, Liang G, et al. Structural and energetic insight into the cross-seeding amyloid assemblies of human Iapp and Rat Iapp. J Phys Chem B. 2014;118:7026–7036.
  • Baram M, Atsmon-Raz Y, Ma B, et al. Amylin–aβ oligomers at atomic resolution using molecular dynamics simulations: a link between Type 2 diabetes and Alzheimer’s disease. Phys Chem Chem Phys. 2016;18:2330–2338.
  • Jose JC, Chatterjee P, Sengupta N. Cross dimerization of amyloid-Β and αsynuclein proteins in aqueous environment: a molecular dynamics simulations study. PloS One. 2014;9:e106883.
  • William PE, Evelyn RS, Joan MJ, et al. Alzheimer’s disease amyloid propagation by a template-dependent dock-lock mechanism. Biochemistry. 2000;39:6288–6295.
  • Nguyen PH, Mai SL, Stock G, et al. Monomer adds to preformed structured oligomers of Aβ-peptides by a two-stage dock–lock mechanism. Proc Natl Acad Sci USA. 2007;104:111.
  • Schor M, Vreede J, Bolhuis PG. Elucidating the locking mechanism of peptides onto growing amyloid fibrils through transition path sampling. Biophys J. 2012;103:1296–1304.
  • Meng X-Y, Zhang H-X, Mezei M, et al. Molecular docking: a powerful approach for structure-based drug discovery. Curr Comput Aided Drug Des. 2011;7:146–157.
  • Lee H, Mittal A, Patel K, et al. Identification of novel drug scaffolds for inhibition of SARS-CoV 3-Chymotrypsin-like protease using virtual and high-throughput screenings. Bioorg Med Chem. 2014;22:167–177.
  • Hu X, Vujanac M, Southall N, et al. Inhibitors of the Yersinia protein tyrosine phosphatase through high throughput and virtual screening approaches. Bioorg Med Chem Lett. 2013;23:1056–1062.
  • Polgar T, Keseru GM. Integration of virtual and high throughput screening in lead discovery settings. Comb Chem High Throughput Screen. 2011;14:889–897.
  • Gong LL, Fang LH, Peng JH, et al. Integration of virtual screening with high-throughput screening for the identification of novel Rho-kinase I inhibitors. J Biotechnol. 2010;145:295–303.
  • Shoichet BK. Virtual screening of chemical libraries. Nature. 2004;432:862–865.
  • Irwin JJ, Shoichet BK. ZINC - a free database of commercially available compounds for virtual screening. J Chem Inf Model. 2005;45:177–182.
  • Sastry GM, Adzhigirey M, Day T, et al. Protein and ligand preparation: parameters, protocols, and influence on virtual screening enrichments. J Comput Aided Mol Des. 2013;27:221–234.
  • Lyne PD. Structure-based virtual screening: an overview. Drug Discovery Today. 2002;7:1047–1055.
  • Sliwoski G, Kothiwale S, Meiler J, et al. Computational methods in drug discov. Pharmacol Rev. 2014;66:334–395.
  • Seeliger D, de Groot BL. Ligand docking and binding site analysis with PyMOL and Autodock/Vina. J Comput Aided Mol Des. 2010;24:417–422.
  • Cavasotto CN, Orry AJW. Ligand docking and structure-based virtual screening in drug discovery. Curr Top Med Chem. 2007;7:1006–1014.
  • Cross JB, Thompson DC, Rai BK, et al. Comparison of several molecular docking programs: pose prediction and virtual screening accuracy. J Chem Inf Model. 2009;49:1455–1474.
  • Kontoyianni M, McClellan LM, Sokol GS. Evaluation of docking performance: comparative data on docking algorithms. J Med Chem. 2004;47:558–565.
  • Schulz-Gasch T, Stahl M. Binding site characteristics in structure-based virtual screening: evaluation of current docking tools. J Mol Model. 2003;9:47–57.
  • Taylor RD, Jewsbury PJ, Essex JW. A review of protein-small molecule docking methods. J Comput Aided Mol Des. 2002;16:151–166.
  • Zhou H-X. From induced fit to conformational selection: a continuum of binding mechanism controlled by the timescale of conformational transitions. Biophys J. 2010;98:L15–L7.
  • Wlodarski T, Zagrovic B. Conformational selection and induced fit mechanism underlie specificity in noncovalent interactions with ubiquitin. Proc Natl Acad Sci USA. 2009;106:19346–19351.
  • Hammes GG, Chang Y-C, Oas TG. Conformational selection or induced fit: a flux description of reaction mechanism. Proc Natl Acad Sci USA. 2009;106:13737–13741.
  • Boehr DD, Nussinov R, Wright PE. The role of dynamic conformational ensembles i biomolecular recognition. Nat Chem Biol. 2009;5:789–796.
  • Antunes DA, Devaurs D, Kavraki LE. Understanding the challenges of protein flexibility in drug design. Expert Opin Drug Discov. 2015;10:1301–1313.
  • Fukunishi Y. Structural ensemble in computational drug screening. Expert Opin Drug Metab Toxicol. 2010;6:835–849.
  • Miao Y, Goldfeld DA, Von Moo E, et al. Accelerated structure-based design of chemically diverse allosteric modulators of a muscarinic G protein-coupled receptor. Proc Natl Acad Sci USA. 2016;113:E5675–E5684.
  • Vass M, Schmidt E, Horti F, et al. Virtual fragment screening on GPCRs: a case study on dopamine D3 and histamine H4 receptors. Eur J Med Chem. 2014;77:38–46.
  • Wada M, Kanamori E, Nakamura H, et al. Selection of in silico drug screening results for G-protein-coupled receptors by using universal active probes. J Chem Inf Model. 2011;51:2398–2407.
  • Han M, Zhang JZH. Class I phospho-inositide-3-kinases (PI3Ks) isoform-specific inhibition study by the combination of docking and molecular dynamics simulation. J Chem Inf Model. 2010;50:136–145.
  • Barakat KH, Huzil JT, Jordan KE, et al. A computational model for overcoming drug resistance using selective dual-inhibitors for aurora kinase A and its T217D variant. Mol Pharm. 2013;10:4572–4589.
  • Offutt TL, Swift RV, Amaro RE. Enhancing virtual screening performance of protein kinases with molecular dynamics simulations. J Chem Inf Model. 2016;56:1923–1935.
  • Spyrakis F, Benedetti P, Decherchi S, et al. A pipeline to enhance ligand virtual screening: integrating molecular dynamics and fingerprints for ligand and proteins. J Chem Inf Model. 2015;55:2256–2274.
  • Swift RV, Jusoh SA, Offutt TL, et al. Knowledge-based methods to train and optimize virtual screening ensembles. J Chem Inf Model. 2016;56:830–842.
  • Tian S, Sun H, Pan P, et al. Assessing an ensemble docking-based virtual screening strategy for kinase targets by considering protein flexibility. J Chem Inf Model. 2014;54:2664–2679.
  • Kong X, Pan P, Li D, et al. Importance of protein flexibility in ranking inhibitor affinities: modeling the binding mechanisms of piperidine carboxamides as Type I1/2 ALK inhibitors. Phys Chem Chem Phys. 2015;17:6098–6113.
  • Genheden S, Ryde U. The MM/PBSA and MM/GBSA methods to estimate ligand-binding affinities. Expert Opin Drug Discov. 2015;10:449–461.
  • Greenidge PA, Kramer C, Mozziconacci JC, et al. Improving docking results via reranking of ensembles of ligand poses in multiple X-ray protein conformations with MM-GBSA. J Chem Inf Model. 2014;54:2697–2717.
  • Hou T, Wang J, Li Y, et al. Assessing the performance of the MM/PBSA and MM/GBSA methods: I. The accuracy of binding free energy calculations based on molecular dynamics simulations. J Chem Inf Model. 2011;51:69–82.
  • Shen J, Tan C, Zhang Y, et al. Discovery of potent ligands for estrogen receptor beta by structure-based virtual screening. J Med Chem. 2010;53:5361–5365.
  • Ge H, Wang Y, Li C, et al. Molecular dynamics-based virtual screening: accelerating the drug discovery process by high-performance computing. J Chem Inf Model. 2013;53:2757–2764.
  • Cavalli A, Spitaleri A, Saladino G, et al. Investigating drug-target association and dissociation mechanisms using metadynamics-based algorithms. Acc Chem Res. 2015;48:277–285.
  • Gervasio FL, Laio A, Parrinello M. Flexible docking in solution using metadynamics. J Am Chem Soc. 2005;127:2600–2607.
  • Limongelli V, Bonomi M, Parrinello M. Funnel metadynamics as accurate binding free-energy method. Proc Natl Acad Sci U S A. 2013;110:6358–6363.
  • Freire E. Do enthalpy and entropy distinguish first in class from best in class? Drug Discov Today. 2008;13:869–874.
  • Olsson TS, Williams MA, Pitt WR, et al. The thermodynamics of protein-ligand interaction and solvation: insights for ligand design. J Mol Biol. 2008;384:1002–1017.
  • Bleicher KH, Bohm HJ, Muller K, et al. Hit and lead generation: beyond high-throughput screening. Nat Rev Drug Discov. 2003;2:369–378.
  • Zhang C, Feng LJ, Huang YY, et al. Discovery of novel phosphodiesterase-2A inhibitors by structure-based virtual screening, structural optimization, and bioassay. J Chem Inf Model. 2017;57:355–364.
  • Hucke O, Coulombe R, Bonneau P, et al. Molecular dynamics simulations and structure-based rational design lead to allosteric HCV NS5B polymerase thumb pocket 2 inhibitor with picomolar cellular replicon potency. J Med Chem. 2014;57:1932–1943.
  • Liu H, Yao X. Molecular basis of the interaction for an essential subunit PA−PB1 in influenza virus RNA polymerase: insights from molecular dynamics simulation and free energy calculation. Mol Pharm. 2010;7:75–85.
  • Sharon A, Balaraju T, Bal C. A catalytic 3D model development of HIV-integrase and drug resistance understanding by molecular dynamics simulation. Antiviral Res. 2011;90:A43–A44.
  • Hou T, W A M, Wang W. Evaluating the potency of HIV‐1 protease drugs to combat resistance. Proteins-Structure Funct Bioinform. 2008;71:1163–1174.
  • Jiao P, Xue W, Shen Y, et al. Understanding the drug resistance mechanism of hepatitis C virus NS5B to PF-00868554 due to mutations of the 423 site: a computational study. Mol Biosyst. 2014;10:767–777.
  • Pan D, Sun H, Bai C, et al. Prediction of zanamivir efficiency over the possible 2009 Influenza A (H1N1) mutants by multiple molecular dynamics simulations and free energy calculations. J Mol Model. 2011;17:2465–2473.
  • Pan D, Xue W, Zhang W, et al. Understanding the drug resistance mechanism of hepatitis C virus NS3/4A to ITMN-191 due to R155K, A156V, D168A/E mutations: a computational study. Biochimica et Biophysica Acta. 2012;1820:1526–1534.
  • Xue W, Jin X, Ning L, et al. Exploring the molecular mechanism of cross-resistance to HIV-1 integrase strand transfer inhibitors by molecular dynamics simulation and residue interaction network analysis. J Chem Inf Modeling. 2013;53:210–222.
  • Xue W, Liu H, Yao X. Molecular mechanism of HIV-1 integrase-vDNA interactions and strand transfer inhibitor action: a molecular modeling perspective. J Comput Chem. 2012;33:527–536.
  • Pan P, Li L, Li Y, et al. Insights into susceptibility of antiviral drugs against the E119G mutant of 2009 influenza A (H1N1) neuraminidase by molecular dynamics simulations and free energy calculations. Antiviral Res. 2013;100:356–364.
  • Phanich J, Rungrotmongkol T, Kungwan N, et al. Role of R292K mutation in influenza H7N9 neuraminidase toward oseltamivir susceptibility: MD and MM/PB(GB)SA study. J Comput Aided Mol Des. 2016;30:917–926.
  • Woods CJ, Malaisree M, Pattarapongdilok N, et al. Long time scale GPU dynamics reveal the mechanism of drug resistance of the dual mutant I223R/H275Y neuraminidase from H1N1-2009 influenza virus. Biochemistry. 2012;51:4364.
  • Pawlotsky JM, Gish RG. Future therapies for hepatitis C. Antivir Ther. 2006;11:397–408.
  • Malcolm BA, Liu R, Lahser F, et al. SCH 503034, a mechanism-based inhibitor of hepatitis C virus NS3 protease, suppresses polyprotein maturation and enhances the antiviral activity of alpha interferon in replicon cells. Antimicrob Agents Chemother. 2006;50:1013–1020.
  • Reed KE, Gorbalenya AE, Rice CM. The NS5A/NS5 proteins of viruses from three genera of the family flaviviridae are phosphorylated by associated serine/threonine kinases. J Virol. 1998;72:6199–6206.
  • Pan D, Niu Y, Xue W, et al. Computational study on the drug resistance mechanism of hepatitis C virus NS5B RNA-dependent RNA polymerase mutants to BMS-791325 by molecular dynamics simulation and binding free energy calculations. Chemometrics Intell Lab Syst. 2016;154:185–193.
  • Xue W, Qi J, Yang Y, et al. Understanding the effect of drug-resistant mutations of HIV-1 intasome on raltegravir action through molecular modeling study. Mol Biosyst. 2012;8:2135–2144.
  • Qi C, Cheng X, Wei D, et al. Molecular dynamics simulation studies of the wild type and E92Q/N155H mutant of Elvitegravir-resistance HIV-1 integrase. Interdiscip Sci Comput Life Sci. 2015;7:36–42.
  • Chen Q, Buolamwini JK, Smith JC, et al. Impact of resistance mutations on inhibitor binding to HIV-1 integrase. J Chem Inf Modeling. 2013;53:3297.
  • Durrant JD, McCammon JA. Molecular dynamics simulations and drug discovery. BMC Boil. 2011;9:71.
  • Salomon-Ferrer R, Götz AW, Poole D, et al. Routine microsecond molecular dynamics simulations with AMBER on GPUs. 2. Explicit Solvent Particle Mesh Ewald J Chem Theor Comput. 2013;9:3878–3888.
  • Böde C, Kovács IA, Szalay MS, et al. Network analysis of protein dynamics. Febs Lett. 2007;581:2776–2782.
  • Marinari E, Parisi G. Simulated tempering: a new Monte Carlo scheme. Europhys Lett. 1992;19:451.
  • Nguyen PH, Okamoto Y, Derreumaux P. Communication: simulated tempering with fast on-the-fly weight determination. J Chem Phys. 2013;138:061102.
  • Zhang C, Ma J. Comparison of sampling efficiency between simulated tempering and replica exchange. J Chem Phys. 2008;129:134112.
  • Zhang T, Nguyen PH, Nasica-Labouze J, et al. Folding atomistic proteins in explicit solvent using simulated tempering. J Phys Chem B. 2015;119:6941–6951.
  • Man VH, Nguyen PH, Derreumaux P. High-Resolution Structures of the amyloid-β 1-42 dimers from the comparison of four atomistic force fields. J Phys Chem B. 2017;121:5977–5987.
  • Nguyen PH, Li MS, Derreumaux P. Effects of all-atom force fields on amyloid oligomerization: replica exchange molecular dynamics simulations of the Aβ 16–22 dimer and trimer. Phys Chem Chem Phys. 2011;13:9778–9788.
  • Freddolino PL, Harrison CB, Liu Y, et al. Challenges in protein folding simulations: timescale, representation, and analysis. Nature Phys. 2010;6:751–758.
  • Lane TJ, Shukla D, Beauchamp KA, et al. To milliseconds and beyond: challenges in the simulation of protein folding. Curr Opin Struc Boil. 2013;23:58–65.
  • Kim PM, Lu LJ, Xia Y, et al. Relating three-dimensional structures to protein networks provides evolutionary insights. Science. 2006;314:1938–1941.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.