155
Views
1
CrossRef citations to date
0
Altmetric
Review

Computational Approaches for Designing Potent and Selective Analogs of Peptide Toxins as Novel Therapeutics

&
Pages 1645-1658 | Published online: 18 Nov 2014

References

  • Ashcroft FM . Ion Channels and Disease: Channelopathies.Academic Press, CA, USA (2000). 
  • Wulff H , ZhorovBS. K+ channel modulators for the treatment of neurological disorders and autoimmune dieseases. Chem. Rev.108, 1744–1773 (2008).
  • Wulff H , CastleNA, PardoLA. Voltage-gated potassium channels as therapeutic targets. Nat. Rev. Drug Disc.8, 982–1001 (2009).
  • Saez NJ , SenffS, JensenJEet al. Spider-venom peptides as therapeutics. Toxins2, 2851–2871 (2010).
  • Knapp O , McArthurJR, AdamsDJ. Conotoxins targeting neuronal voltage-gated sodium channel subtypes: potential analgesics. Toxins4, 1236–1260 (2012).
  • Chi V , PenningtonMW, NortonRSet al. Development of a sea anemone toxin as an immunomodulator for therapy of autoimmune diseases. Toxicon59, 529–546 (2012).
  • Beeton C , WulffH, StandiferNEet al. Kv1.3 channels are a therapeutic target for T cell-mediated autoimmune diseases. Proc. Natl. Acad. Sci. USA103, 17414–17419 (2006).
  • Beeton C , PenningtonM, WulfHet al. Targeting effector memory T cells with a selective peptide inhibitor of Kv1.3 channels for therapy of autoimmune diseases. Mol. Pharmacol.67, 1369–1381 (2005).
  • Pennington MW , BeetonC, GaleaCAet al. Engineering a stable and selective peptide blocker of the Kv1.3 channel in T lymphocytes. Mol. Pharmacol.75, 762–773 (2009).
  • Alonso H , BliznyukAA, GreadyJE. Combining docking and molecular dynamic simulations in drug design. Med. Res. Rev.26, 531–568 (2006).
  • Gordon D , ChenR, ChungSH. Computational methods of studying the binding of toxins from venomous animals to biological ion channels. Physiol. Rev.93, 767–802 (2013).
  • Rashid MH , MahdaviS, KuyucakS. Computational studies of marine toxins targeting ion channels. Mar. Drugs11, 848–869 (2013).
  • Gilson MK , ZhouHX. Calculation of protein-ligand binding energies. Ann. Rev. Biophys. Biomol. Struct.36, 21–42 (2007).
  • Deng Y , RouxB. Computations of standard binding free energies with molecular dynamics simulations. J. Phys. Chem. B113, 2234–2246 (2009).
  • Christ CD , MarkAE, van GunsterenWF. Basic ingredients of free energy calculations. J. Comput. Chem.31, 1569–1582 (2010).
  • Steinbrecher T , LabahnA. Towards Accurate Free Energy Calculations in Ligand-Protein Binding Studies. Curr. Med. Chem.17, 767–785 (2010).
  • Osborne R . Fresh from the biotech pipeline–2012. Nat. Biotechnol.31, 100–103 (2013).
  • Norton RS , PenningtonMW, BeetonC. Transforming a toxin into a therapeutic: the sea anemone potassium channel blocker ShK toxin for treatment of autoimmune diseases.. In:In Venoms to Drugs: Venom as a Source for the Development of Human Therapeutics.KingGF (Ed.). Royal Society of Chemistry, London, UK (2014) (In press).
  • Long SB , TaoX, CampbellEB, MacKinnonR. Atomic structure of a voltage-dependent K+channel in a lipid membrane-like environment. Nature450, 376–382 (2007).
  • Payandeh J , ScheuerT, ZhengN, CatterallWA. The crystal structure of a voltage-gated sodium channel. Nature475, 353–358 (2011).
  • Payandeh J , ScheuerT, ZhengN, CatterallWA. Crystal structure of a voltage-gated sodium channel in two potentially inactivated states. Nature486, 135–139 (2012).
  • Zhang X , RenW, DeCaenPet al. Crystal structure of an orthologue of the NaChBac voltage-gated sodium channel. Nature486, 130–134 (2012).
  • Halperin I , MaB, WolfsonH, NussinovR. Principles of docking: an overview of search algorithms and a guide to scoring functions. Proteins47, 409–443 (2002).
  • Brooijmans N , KuntzID. Molecular recognition and docking algorithms. Ann. Rev. Biophys. Biomol. Struct.32, 335–373 (2003).
  • Morris GM , Goodsell, DS, HallidayRSet al. Automated docking using a Lamarckian genetic algorithm and empirical binding free energy function. J. Comput. Chem.19, 1639–1662 (1998).
  • Mintseris J , PierceB, WieheKet al. Integrating statistical pair potentials into protein complex prediction. Proteins69, 511–520 (2007).
  • Dominguez C , BoelensR, BonvinAM. HADDOCK: a protein–protein docking approach based on biochemical or biophysical information. J. Am. Chem. Soc.125, 1731–1737 (2003).
  • De Vries SJ , van DijkAD, KrzeminskiMet al. HADDOCK versus HADDOCK: new features and performance of HADDOCK2.0 on the CAPRI targets. Proteins69, 726–733 (2007).
  • Warren GL , AndrewsCW, Capelli, AMet al. A critical assessment of docking programs and scoring functions J. Med. Chem. 49, 5912–5931 (2006).
  • Patra SM , BastugT, KuyucakS. Binding of organic cations to gramicidin: a channel studied with AutoDock and molecular dynamics simulations. J. Phys. Chem.B111, 11303–11311 (2007).
  • Ander M , LuzhkovVB, AqvistJ. Ligand binding to the voltage-gated Kv1.5 potassium channel in the open state–Docking and computer simulations of a homology model. Biophys. J.94, 820–831 (2007).
  • Yi, H, Qiu, S, Cao, Z.J, Wu, Y.L, Li, W.X. Molecular basis of inhibitory peptide maurotoxin recognizing Kv1.2 channel explored by ZDOCK and molecular dynamic simulations. Proteins70, 844–854 (2008).
  • Chen PC , KuyucakS. Mechanism and energetics of charybdotoxin unbinding from a potassium channel from molecular dynamics simulations. Biophys. J.96, 2577–2588 (2009).
  • Yu LP , SunCH, SongDYet al. Nuclear magnetic resonance structural studies of a potassium channel–charybdotoxin complex. Biochemistry44, 15834–15841 (2005).
  • Chen PC , KuyucakS. Developing a comparative docking protocol for the prediction of peptide selectivity profiles: investigation of potassium channel toxins. Toxins4, 110–138 (2012).
  • Humphrey W , DalkeA, SchultenK. VMD–visual molecular dynamics. J. Mol. Graph.14, 33–38 (1996).
  • Bastug T , KuyucakS. Importance of the peptide backbone description in modeling the selectivity filter in potassium channels. Biophys. J.96, 4006–4012 (2009).
  • Phillips JC , BraunR, WangWet al. Scalable molecular dynamics with NAMD. J. Comput. Chem.26, 1781–1802 (2005).
  • Brooks BR , BrooksCL, MacKerellADet al. CHARMM: the biomolecular simulation program. J. Comput. Chem.30, 1545–1614 (2009).
  • Frenkel D , SmitB. Understanding Molecular Simulation: From Algorithms to Applications.Academic Press, CA, USA (1996).
  • Leach AR . Molecular Modelling, Principles, Applications.Prentice Hall, NY, USA (2001).
  • Stansfeld PJ , SansomMSP. Molecular simulation approaches to membrane proteins. Structure19, 1562–1572 (2011).
  • Dror RO , DirksRM, GrossmanJPet al. Biomolecular simulations: a computational microscope for molecular biology. Annu. Rev. Biophys.41, 429–452 (2012).
  • Bastug T , KuyucakS. Molecular dynamics simulations of membrane proteins. Biophys. Rev.4, 271–282 (2012).
  • Kollman PA , MassovaI, ReyesCet al. Calculating structures and free energies of complex molecules: combining molecular mechanic and continuum models. Acc. Chem. Res.33, 889–897 (2000).
  • Enyedy IJ , EganWJ. Can we use docking and scoring for hit-to-lead optimization?J. Comput. Aided Mol. Des.22, 161–168 (2008).
  • Schneider G . Virtual screening: an endless staircase?Nat. Rev. Drug Discov.9, 273–276 (2010).
  • Brown SP , MuchmoreSW. Large scale application of high-throughput molecular mechanics with Poisson-Boltzmann surface area for routine physics-based scoring of protein-ligand complexes. J. Med. Chem.52, 3159–3165 (2009).
  • Singh N , WarshelA. Absolute binding free energy calculations: on the accuracy of computational scoring of protein-ligand interactions. Proteins78, 1705–17123 (2010).
  • Michel J , EssexJW. Prediction of protein-ligand affinity by free energy simulations: assumptions, pitfalls and expectations. J. Comput. Aided Mol. Des.24, 639–658 (2010).
  • Chodera JD , MobleyDL, ShirtsMRet al. Alchemical free energy methods for drug discovery: progress and challenges. Curr. Opin. Struct. Biol.21, 150–160 (2011).
  • deRuiter A , OostenbrinkC. Free energy calculations of protein–ligand interactions. Curr. Opin. Chem. Biol.15, 547–552 (2011).
  • Mobley DL , KlimovPV. Perspective: alchemical free energy calculations in drug discovery. J. Chem. Phys.137, 230901 (2012).
  • Kumar S , BouzidaSD, SwensenRHet al. The weighted histogram analysis method for free-energy calculations on biomolecules. J. Comput. Chem.13, 1011–1021 (1992).
  • Chen PC , KuyucakS. Accurate determination of the binding free energy for KcsA-charybdotoxin complex from the potential of mean force calculations. Biophys. J.100, 2466–2474 (2011).
  • Park S , SchultenK. Calculating potentials of mean force from steered molecular dynamics simulations. J. Chem. Phys.120, 5946–5961 (2004).
  • Bastug T , ChenPC, PatraSM, KuyucakS. Potential of mean force calculations of ligand binding to ion channels from Jarzynski's equality and umbrella sampling. J. Chem. Phys.128, 104–112 (2008).
  • Heinzelmann G , BastugT, KuyucakS. Free energy simulations of ligand binding to the aspartate transporter GltPh. Biophys. J.101, 2380–2388 (2011).
  • Heinzelmann G , BastugT, KuyucakS. Mechanism and energetics of ligand release in the aspartate transporter GltPh. J. Phys. Chem B117, 5486–5496 (2013).
  • Heinzelmann G , ChenPC, KuyucakS. Computation of standard binding free energies of polar and charged ligands to the glutamate receptor GluA2. J. Phys. Chem B118, 1813–1824 (2014).
  • Rashid MH , HeinzelmannG, HuqRet al. A potent and selective peptide blocker of the Kv1.3 channel: prediction from free-energy simulations and experimental confirmation. PLoS One8, e78712 (2013).
  • Doyle DA , CabralJM, PfuetznerRAet al. The structure of the potassium channel: molecular basis of K+ conduction and selectivity. Science280, 69–77 (1998).
  • Rashid MH , KuyucakS. Affinity and selectivity of ShK toxin for the Kv1 potassium channels from free energy simulations. J. Phys. Chem. B116, 4812–4822 (2012).
  • Khabiri M , NikoueeA, CwiklikLet al. Charybdotoxin unbinding from the mKv1.3 potassium channel: a combined computational and experimental study. J. Phys. Chem. B115, 11490–11500 (2011).
  • Chen R , Robinson, A, GordonD, ChungSH. Modeling the binding of three toxins to the voltage-gated potassium channel (Kv1.3). Biophys. J.101, 2652–2660 (2011).
  • Tudor JE , PallaghyPK, PenningtonMW, NortonRS. Solution structure of ShK toxin, a novel potassium channel inhibitor from a sea anemone. Nat. Struct. Biol.3, 317–320 (1996).
  • Rauer H , PenningtonM, CahalanM, ChandyKG. Structural conservation of the pores of calcium-activated and voltage-gated potassium channels determined by a sea anemone toxin. J. Biol. Chem.274, 21885–21892 (1999).
  • Pennington MW , RashidMH, TajhyaRBet al. A C-terminally amidated analogue of ShK is a potent and selective blocker of the voltage-gated potassium channel Kv1.3. FEBS Lett.586, 3996–4001 (2012).
  • Regaya L , BeetonC, FerratGet al. Evidence for domain specific recognition of SK and Kv channels by MTX and HsTx1 scorpion toxins. J. Biol. Chem.279, 55690–55696 (2004).
  • Savarin P , Romi-LebrunR, Zinn-JustinSet al. Structural and functional consequences of the presence of a forth disulfide bridge in the scorpion short toxins: solution structure of the potassium channel inhibitor HsTx1. Protein Sci.8, 2672–2685 (1999).
  • Rashid MH , KuyucakS. Free energy simulations of binding of HsTx1 toxin to Kv1 potassium channels: the basis of Kv1.3/Kv1.1 selectivity. J. Phys. Chem. B118, 707–716 (2014).
  • Rashid MH , HuqR, TannerMet al. A potent and Kv1.3-selective analogue of the scorpion toxin HsTX1 as a potential therapeutic for autoimmune diseases. Sci. Rep.4, 4509 (2014).
  • Billen B , BosmansF, TytgatJ. Animal peptides targeting voltage-activated sodium channels. Curr. Pharm. Des.14, 2492–2502 (2008).
  • Clare JC . Targeting voltage-gated sodium channels for pain therapy. Expert Opin. Investig. Drugs19, 45–62 (2010).
  • Twede VD , MiljanichG, OliveraBM, BulajG. Neuroprotective and cardioprotectiveconopeptides: an emerging class of drug leads. Curr. Opin. Drug Discov. Devel.12, 231–239 (2009).
  • Norton Rs . µ-conotoxins as leads in the development of new analgesics. Molecules15, 2825–2844 (2010).
  • Cruz LJ , GrayWR, OliveraBMet al. Conusgeographus toxins that discriminate between neuronal and muscle sodium channels. J. Biol. Chem.260, 9280–9288 (1985).
  • Li RA , Ennis, IL, FrenchRJet al. Clockwise domain arrangement of the sodium channel revealed by µ-conotoxin (GIIIA) docking orientation. J. Biol. Chem.276, 11072–11077 (2001).
  • Hui K , LipkindG, FozzardHA, FrenchRJ. Electrostatic and steric contributions to block of the skeletal muscle sodium channel by µ-conotoxin. J. Gen. Physiol.119, 45–54 (2002).
  • Xue T , EnnisIL, SatoKet al. Novel interactions identified between µ-conotoxin and the Na+channel domain I P-loop: implications for toxin-pore binding geometry. Biophys. J.85, 2299–2310 (2003).
  • Chen R , RobinsonA, ChungSH. Mechanism of µ-conotoxin PIIIA binding to the voltage-gated Na+ channel NaV1.4. PLoS One9, e93267 (2014).
  • McArthur JR , OstroumovV, Al-SabiAet al. Multiple, distributed interactions of µ-conotoxin PIIIA associated with broad targeting among voltage-gated sodium channels. Biochemistry50, 116–124 (2011).
  • McArthur JR , SinghG, O'MaraMLet al. Orientation of µ-conotoxin PIIIA in a sodium channel vestibule, based on voltage dependence of its binding. Mol. Pharmacol.80, 219–227 (2011).
  • Tikhonov DB , ZhorovBS. Architecture and pore block of eukaryotic voltage-gated sodium channels in view of NaVAb bacterial sodium channel structure. Mol. Pharmacol.82, 97–104 (2012).
  • Chen R , ChungSH. Mechanism of tetrodotoxin block and resistance in sodium channels. Biochem. Biophys. Res. Commun.446, 370–374 (2014).
  • Chang NC , FrenchRJ, LipkindGMet al. Predominant interactions between µ-conotoxin Arg-13 and the skeletal muscle Na+channel localized by mutant cycle analysis. Biochemistry37, 4407–4419 (1998).
  • Wilson MJ , YoshikamiD, AzamLet al. µ-conotoxins that differentially block sodium channels NaV1.1 through NaV1.8 identify those responsible for action potentials in sciatic nerve. Proc. Natl. Acad. Sci. USA108, 10302–10307 (2011).
  • Catterall WA . Ion channel voltage sensors: structure, function, and pathophysiology. Neuron67, 915–928 (2010).
  • Middleton RE , WarrenVA, KrausRLet al. Two tarantula peptides inhibit activation of multiple sodium channels. Biochemistry41, 14734–14747 (2002).
  • Schmalhofer WA , CalhounJ, BurrowsRet al. ProTx-II, a selective inhibitor of NaV1.7 sodium channels, blocks action potential propagation in nociceptors. Mol. Pharmacol.74, 1476–1784 (2008).
  • Xiao, Y et al. Tarantula huwentoxin-IV inhibits neuronal sodium channels by binding to receptor site 4 and trapping the domain II voltage sensor in the closed configuration. J. Biol. Chem.283, 27300–27313 (2008).
  • Sokolov S , KrausRL, ScheuerT, CatterallWA. Inhibition of sodium channel gating by trapping the domain II voltage sensor with protoxin II. Mol. Pharmacol.73, 1020–1028 (2008).
  • Xiao Y , BlumenthalK, JacksonJOet al. The tarantula toxins ProTx-II and Huwentoxin-IV differentially interact with human NaV1.7 voltage sensors to inhibit channel activation and inactivation. Mol. Pharmacol.78, 1124–1134 (2010).
  • Xiao Y , JacksonJO, LiangS, CumminsTR. Common molecular determinants of tarantula huwentoxin-IV inhibition of Na+ channel voltage sensors in domains II and IV. J. Biol. Chem.286, 27301–27310 (2011).
  • Chen R , ChungSH. Conserved functional surface of anti-mammalian scorpion β-toxins. J. Phys. Chem. B116, 4796–4800 (2012).
  • Chen R , ChungSH. Binding modes and functional surface of anti-mammalian scorpion α-toxins to sodium channels. Biochemistry51, 77775–77782 (2012).
  • Muddana HS , FenleyAT, MobleyDL, GilsonMK. The SAMPL4 host-guest blind prediction challenge: an overview. J. Comput. Aided Mol. Des.28, 305–317 (2014).
  • Eriksson MAL , RouxB. Modeling the structure of agitoxinin complex with the Shaker K+ channel: a computational approach based on experimental distance restraints extracted from thermodynamic mutant cycles. Biophys. J.83, 2595–2609 (2002).

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.