kinome-wide spectroscopic study of drug binding site electrostatics

2
more rigorous physical model combined with effective sampling of molecular configurations is critical for binding affinity prediction to chemical accuracy, which is defined as within one order of magnitude of the true equilibrium dis- sociation constant. We have demonstrated that electrostatic interactions, espe- cially electronic polarization, are critical for protein-ligand recognition due to the significant change in electrostatic environments between bulk water and protein pockets and have achieved encouraging success in treating charged spe- cies using the polarizable Atomic Multipole Optimized Energetics for Biomo- lecular Applications (AMOEBA) force field. To maintain accuracy while also achieving efficiency, AMOEBA has been combined with the Orthogonal Space Random Walk en- hanced alchemical free energy algo- rithm. Here we pres- ent applications of this strategy for the computation of pro- tein-ligand binding affinities and, for the first time, drug solu- bility from alchemi- cal simulations using the Force Field X software. 2083-Plat MloK1 Ligand Binding Simulations Indicate an Induced-Fit Mechanism Be ´la Voß 1 , Sebastian Peuker 2 , U. Benjamin Kaupp 2 , Helmut Grubmu ¨ller 1 . 1 Max Planck Institute for Biophysical Chemistry, Go ¨ttingen, Germany, 2 Center of Advanced European Studies and Research, Bonn, Germany. Many ion channels such as the MloK1 channel are steered by ligand binding via conformational changes. Mainly two binding mechanisms have been proposed, induced fit and conformational selection. Using molecular dynamics simula- tions, we studied ligand binding of cyclic adeonise monophosphate at the cyclic nucleotide binding domain of the MloK1 ion channel of Mesorizobium loti. For this binding domain, both crystal and NMR structures have been determined for both the ligand free as well as for the ligand bound conformation. In the simulations, spontaneous binding was observed, which enabled us to de- termine reaction coordinates for the ligand binding as well as for the associated conformational change of the protein. We used a combination of force probe simulations, umbrella sampling, and unbiased simulations to determine poten- tials of mean force along these reaction coordinates, transition rates, as well as free energy differences and barriers between the most relevant substates. Our results are compared with measured affinities and kinetics, and suggest an induced fit-mechanism. 2084-Plat Pathway and Mechanism of Drug Binding to G-Protein-Coupled Recep- tors Ron O. Dror 1 , Albert C. Pan 1 , Daniel H. Arlow 1 , David W. Borhani 1 , Paul Maragakis 1 , Yibing Shan 1 , Huafeng Xu 1 , David E. Shaw 1,2 . 1 D E Shaw Research, New York, NY, USA, 2 Center for Biology and Bioinformatics, Columbia University, New York, NY, USA. How drugs bind to their receptors—from initial association, through entry into the binding pocket, to adoption of the final bound pose—has remained un- known, even for G-protein-coupled receptor modulators, which constitute one-third of all drugs. We captured this pharmaceutically critical process in atomic detail using the first unbiased molecular dynamics simulations in which drug molecules spontaneously associate with G-protein-coupled receptors to achieve final poses matching those determined crystallographically (PNAS 108:13118 (2011)). We found that several beta blockers and a beta agonist all traverse the same dominant pathway as they bind to the b 1 - and b 2 -adrener- gic receptors, initially associating with a vestibule on each receptor’s extracellular surface. Surprisingly, this association, at a distance of 15 A ˚ from the binding pocket, often presents the largest energetic barrier to binding, despite the fact that subsequent entry into the pocket requires the receptor to deform and the drug to squeeze through a narrow passage. The early barrier may reflect the substantial dehydration that occurs as the drug associates with the vestibule. Our atomic-level description of the binding process suggests opportunities for allosteric modulation and provides a structural foun- dation for future optimization of binding and unbinding rates. 2085-Plat A General Prediction Method of Scorpion Toxins’ Kv-Channel Selectivity Profiles using Haddock Po-chia Chen, Serdar Kuyucak. School of Physics, University of Sydney, Australia. The active components of animal venoms are potentially useful in many elec- trophysiological and pharmacological applications due to their highly selective nature. Of this rich concoction, the binding mechanisms of many toxin types remain to be elucidated. We therefore present a preliminary method to deduce the selectivity profile of a peptide toxin against related channels by means of docking simulations. This is tested on the family of a-KTx scorpion toxins, for which structural and limited affinity data are available for over 20 toxins across seven sub-families. Docking simulations against Kv1.1, Kv1.2 and Kv1.3 were carried out under both blind-docking trials and common-lysine- motif trials, using the program HADDOCK. This study reports on a selection of toxins for which validation can be best pro- vided, given current limitations of docking accuracy. The general selectivity profiles of toxin-subfamilies can be deduced via consensus between closely re- lated toxin-channel pairings. In particular, HADDOCK was able to classify a-KTX2 toxins as universal binders and a-KTX3 toxins as Kv1.3-selective binders. An estima- tion of individual se- lectivity profiles can be further deduced by program performance. This method is ex- pected to be useful in the refinement of toxins for channel- subtype targetting. 2086-Plat Design and Development of Drugs that Target Virus Ion Channels Matthew R. Rosenberg, Nicole C. Norris, Llara M. Weaver, Marco G. Casarotto. Australian National University, Canberra, Australia. Virus ion channels are small (3-15 kD) peptides that aggregate to form ion channels that are important for viral infection1. As viruses continue to pose a major worldwide health problem, these ion channels represent an exciting new target for therapeutic intervention. Viral ion channels that have been pre- viously identified include Vpu of the human immunodeficiency virus (HIV) and P7 of Hepatitis C however it is the M2 influenza A protein that represents the best exploited ion channel drug target so far. The proton-selective M2 ion channel is the target of the adamantane family of drug inhibitors. Use of the two most common adamantane inhibitors, amanta- dine and rimantadine have declined steadily over recent years due to the emer- gence of adamantane-resistant flu strains. We have conducted a series of surface plasmon resonance experiments designed to measure the affinity be- tween several ion channel inhibitors and M22. By examining drug binding to a number of mutant M2 constructs (derived from adamantane-resistant strains), it was possible to establish the location of the drug binding sites and to ratio- nalise the effect on drug binding of specific mutant residues. In light of these results, the prospect for future development of a new generation of M2 inhib- itors will be discussed. Moreover, we explore the possibility of expanding this field of research to incorporate ion channel proteins from other viruses. 1. Gonzalez, M and Carrasco, L (2003). FEBS Lett. (2003) 552(1):28-34. 2. Rosenberg, M and Casarotto, M (2010) PNAS 107(31):13866-71. 2087-Plat Kinome-Wide Spectroscopic Study of Drug Binding Site Electrostatics Nick Levinson, Steven G. Boxer. Stanford University, Stanford, CA, USA. Small molecule kinase inhibitors have recently demonstrated dramatic po- tential for treating cancers caused by dysregulated protein kinases. The ef- ficacy of these compounds is due to their ability to selectively target particular protein kinases, and this selectivity is remarkable given the fact that they bind to the ATP-binding site of the kinase domain, which is highly conserved in sequence across this large protein family. The origin of this selectivity is unknown, but must relate to differences in physical properties of the ATP-binding site among members of this protein family. The goal of this project is to assess how the evolutionary divergence of sequence and structure in the human kinome translates into variation in ATP-binding site electrostatics, and how this variation can be exploited to design highly selective inhibitors. I am using a clinically important class of kinase 410a Tuesday, February 28, 2012

Upload: steven-g

Post on 30-Dec-2016

214 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Kinome-Wide Spectroscopic Study of Drug Binding Site Electrostatics

410a Tuesday, February 28, 2012

more rigorous physical model combined with effective sampling of molecularconfigurations is critical for binding affinity prediction to chemical accuracy,which is defined as within one order of magnitude of the true equilibrium dis-sociation constant. We have demonstrated that electrostatic interactions, espe-cially electronic polarization, are critical for protein-ligand recognition due tothe significant change in electrostatic environments between bulk water andprotein pockets and have achieved encouraging success in treating charged spe-cies using the polarizable Atomic Multipole Optimized Energetics for Biomo-lecular Applications (AMOEBA) force field. To maintain accuracy while alsoachieving efficiency, AMOEBA has been combined with the Orthogonal Space

Random Walk en-hanced alchemicalfree energy algo-rithm. Here we pres-ent applications ofthis strategy for thecomputation of pro-tein-ligand bindingaffinities and, for thefirst time, drug solu-bility from alchemi-cal simulations usingthe Force Field Xsoftware.

2083-PlatMloK1 Ligand Binding Simulations Indicate an Induced-Fit MechanismBela Voß1, Sebastian Peuker2, U. Benjamin Kaupp2, Helmut Grubmuller1.1Max Planck Institute for Biophysical Chemistry, Gottingen, Germany,2Center of Advanced European Studies and Research, Bonn, Germany.Many ion channels such as the MloK1 channel are steered by ligand binding viaconformational changes. Mainly two binding mechanisms have been proposed,induced fit and conformational selection. Using molecular dynamics simula-tions, we studied ligand binding of cyclic adeonise monophosphate at the cyclicnucleotide binding domain of the MloK1 ion channel of Mesorizobium loti. Forthis binding domain, both crystal and NMR structures have been determined forboth the ligand free as well as for the ligand bound conformation.In the simulations, spontaneous binding was observed, which enabled us to de-termine reaction coordinates for the ligand binding as well as for the associatedconformational change of the protein. We used a combination of force probesimulations, umbrella sampling, and unbiased simulations to determine poten-tials of mean force along these reaction coordinates, transition rates, as well asfree energy differences and barriers between the most relevant substates.Our results are compared with measured affinities and kinetics, and suggest aninduced fit-mechanism.

2084-PlatPathway and Mechanism of Drug Binding to G-Protein-Coupled Recep-torsRon O. Dror1, Albert C. Pan1, Daniel H. Arlow1, David W. Borhani1,Paul Maragakis1, Yibing Shan1, Huafeng Xu1, David E. Shaw1,2.1D E Shaw Research, New York, NY, USA, 2Center for Biology andBioinformatics, Columbia University, New York, NY, USA.How drugs bind to their receptors—from initial association, through entry intothe binding pocket, to adoption of the final bound pose—has remained un-known, even for G-protein-coupled receptor modulators, which constituteone-third of all drugs. We captured this pharmaceutically critical process inatomic detail using the first unbiased molecular dynamics simulations in whichdrug molecules spontaneously associate with G-protein-coupled receptors toachieve final poses matching those determined crystallographically (PNAS108:13118 (2011)). We found that several beta blockers and a beta agonistall traverse the same dominant pathway as they bind to the b1- and b2-adrener-

gic receptors, initially associating with a vestibule oneach receptor’s extracellular surface. Surprisingly, thisassociation, at a distance of 15 A from the bindingpocket, often presents the largest energetic barrier tobinding, despite the fact that subsequent entry into thepocket requires the receptor to deform and the drug tosqueeze through a narrow passage. The early barriermay reflect the substantial dehydration that occurs asthe drug associates with the vestibule. Our atomic-leveldescription of the binding process suggests opportunitiesfor allosteric modulation and provides a structural foun-dation for future optimization of binding and unbindingrates.

2085-PlatA General Prediction Method of Scorpion Toxins’ Kv-Channel SelectivityProfiles using HaddockPo-chia Chen, Serdar Kuyucak.School of Physics, University of Sydney, Australia.The active components of animal venoms are potentially useful in many elec-trophysiological and pharmacological applications due to their highly selectivenature. Of this rich concoction, the binding mechanisms of many toxin typesremain to be elucidated. We therefore present a preliminary method to deducethe selectivity profile of a peptide toxin against related channels by means ofdocking simulations. This is tested on the family of a-KTx scorpion toxins,for which structural and limited affinity data are available for over 20 toxinsacross seven sub-families. Docking simulations against Kv1.1, Kv1.2 andKv1.3 were carried out under both blind-docking trials and common-lysine-motif trials, using the program HADDOCK.This study reports on a selection of toxins for which validation can be best pro-vided, given current limitations of docking accuracy. The general selectivityprofiles of toxin-subfamilies can be deduced via consensus between closely re-lated toxin-channel pairings. In particular, HADDOCK was able to classifya-KTX2 toxins as universal binders and a-KTX3 toxins as Kv1.3-selective

binders. An estima-tion of individual se-lectivity profiles canbe further deduced byprogram performance.This method is ex-pected to be useful inthe refinement oftoxins for channel-subtype targetting.

2086-PlatDesign and Development of Drugs that Target Virus Ion ChannelsMatthew R. Rosenberg, Nicole C. Norris, Llara M. Weaver,Marco G. Casarotto.Australian National University, Canberra, Australia.Virus ion channels are small (3-15 kD) peptides that aggregate to form ionchannels that are important for viral infection1. As viruses continue to posea major worldwide health problem, these ion channels represent an excitingnew target for therapeutic intervention. Viral ion channels that have been pre-viously identified include Vpu of the human immunodeficiency virus (HIV) andP7 of Hepatitis C however it is the M2 influenza A protein that represents thebest exploited ion channel drug target so far.The proton-selective M2 ion channel is the target of the adamantane family ofdrug inhibitors. Use of the two most common adamantane inhibitors, amanta-dine and rimantadine have declined steadily over recent years due to the emer-gence of adamantane-resistant flu strains. We have conducted a series ofsurface plasmon resonance experiments designed to measure the affinity be-tween several ion channel inhibitors and M22. By examining drug binding toa number of mutant M2 constructs (derived from adamantane-resistant strains),it was possible to establish the location of the drug binding sites and to ratio-nalise the effect on drug binding of specific mutant residues. In light of theseresults, the prospect for future development of a new generation of M2 inhib-itors will be discussed. Moreover, we explore the possibility of expanding thisfield of research to incorporate ion channel proteins from other viruses.1. Gonzalez, M and Carrasco, L (2003). FEBS Lett. (2003) 552(1):28-34.2. Rosenberg, M and Casarotto, M (2010) PNAS 107(31):13866-71.

2087-PlatKinome-Wide Spectroscopic Study of Drug Binding Site ElectrostaticsNick Levinson, Steven G. Boxer.Stanford University, Stanford, CA, USA.Small molecule kinase inhibitors have recently demonstrated dramatic po-tential for treating cancers caused by dysregulated protein kinases. The ef-ficacy of these compounds is due to their ability to selectively targetparticular protein kinases, and this selectivity is remarkable given the factthat they bind to the ATP-binding site of the kinase domain, which is highlyconserved in sequence across this large protein family. The origin of thisselectivity is unknown, but must relate to differences in physical propertiesof the ATP-binding site among members of this protein family. The goal ofthis project is to assess how the evolutionary divergence of sequence andstructure in the human kinome translates into variation in ATP-bindingsite electrostatics, and how this variation can be exploited to design highlyselective inhibitors. I am using a clinically important class of kinase

Page 2: Kinome-Wide Spectroscopic Study of Drug Binding Site Electrostatics

Tuesday, February 28, 2012 411a

inhibitors, the 4-anilinoquinazolines/4-anilinoquinolines, to introduce vibra-tional probes of electric field, which measure electrostatics via the vibra-tional Stark effect, into the ATP-binding sites of different kinases. Theresults yield direct insight into how chemical changes within this inhibitorclass influence the selectivity for particular kinases, potentially yieldinga method for rationally designing selective inhibitors.

2088-PlatSpecificity, Structure and Dynamics of Tiam1 PDZ Domain Ligand-BoundComplexesErnesto J. Fuentes, Xu Liu, Tyson R. Shepherd, David C. Speckhard,Ann M. Murray.University of Iowa, Iowa City, IA, USA.PSD-95/DlgA/ZO-1 (PDZ) domains are among the most abundant protein-protein interaction domains in the human proteome and typically bind the4-10 C-terminal residues of its interaction partner with exquisite specificity.We used two homologous PDZ domains from the Tiam-family of guanine nu-cleotide exchange factors to investigate PDZ specificity. The Tiam1 and Tiam2PDZ domains have overlapping but distinct ligand binding specificity, and thisis exemplified by their unique preferences for C-terminal peptides derived fromthe syndecan1, Caspr4 and neurexin1 adhesion proteins. The Tiam1 PDZ do-main binds syndecan1 and Caspr4 but not neurexin1, while the Tiam2 PDZ do-main binds Caspr4 and neurexin1 but not syndecan1. Amino acid sequencecomparison of Tiam-family PDZ domains revealed that four residues criticalfor ligand specificity are not conserved. Remarkably, substitution of thesefour residues in the Tiam1 PDZ for those found in the Tiam2 PDZ domainswitched ligand specificity. To understand the structural and dynamic basisfor this change in specificity we used X-ray crystallography and solutionNMR methods, respectively. We determined the crystal structures of wildtype Tiam1 PDZ domain bound to syndecan1 and phosphorylated syndecan1peptides and the Tiam1 PDZ quadruple mutant (QM) bound to Caspr4 and neu-rexin1 peptides. Comparison of the crystal structures of the Tiam1 PDZ-syndecan1 and PDZ-phosphorylated syndecan1 showed that a distinct specific-ity pocket is used to accommodate the phosphoryl group. The crystal structureof the Tiam1 QM PDZ domain showed a unique side chain stacking interactionbetween aromatic residues in the PDZ domain and the Caspr4 ligand. Sidechain methyl relaxation experiments revealed distinct patterns of dynamicsin the Tiam1 PDZ-syndecan1 and PDZ-Caspr4 complexes. Collectively, thestructures and dynamics of physiologically-based PDZ domain complexesare contributing to understanding the origin of PDZ specificity and functionof Tiam-family PDZ domains.

Platform: Channel Regulation & Modulation

2089-PlatStructural Basis For Alcohol Modulation of Pentameric Ligand-Gated IonChannelsRebecca J. Howard1, Samuel Murail2, Kathryn E. Ondricek1,Pierre-Jean Corringer3, Erik Lindahl2, James R. Trudell4, R. Adron Harris1.1The University of Texas at Austin, Austin, TX, USA, 2Kungliga TekniskaHogskolan, Stockholm, Sweden, 3Institut Pasteur, Paris, France, 4StanfordUniversity School of Medicine, Stanford, CA, USA.Despite its long history of use and abuse in human culture, the molecular basisfor alcohol action in the brain is poorly understood. The recent determination ofthe atomic-scale structure of GLIC, a prokaryotic member of the pentamericligand-gated ion channel (pLGIC) family, provides a unique opportunity tocharacterize the structural basis for modulation of these channels, many ofwhich are alcohol targets in brain. We observed bimodal modulation ofGLIC by n-alcohols, similar to some eukaryotic pLGICs: methanol and ethanolweakly potentiated proton-activated currents in GLIC, whereas n-alcoholslarger than ethanol inhibited them. Mapping of residues important to alcoholmodulation of ionotropic receptors for glycine, GABA, and acetylcholineonto GLIC revealed their proximity to transmembrane cavities that may accom-modate one or more alcohol molecules. Site-directed mutations in the pore-liningM2 helix allowed the identification of four residues that influence alcoholpotentiation, with the direction of their effects reflecting helical structure. Atone of the potentiation-enhancing residues, decreased side chain volume con-verted GLIC into a highly ethanol-sensitive channel, comparable to its eukary-otic relatives. Covalent labeling of M2 positions with a methanethiosulfonatereagent further implicated residues at the extracellular end of the helix in alco-hol binding. Molecular dynamics simulations elucidated the structural conse-quences of a potentiation-enhancing mutation and suggested a structuralmechanism for alcohol potentiation via interaction with a transmembrane cav-ity previously termed the ‘‘linking tunnel.’’ These results provide a unique

structural model for independent potentiating and inhibitory interactions ofn-alcohols with a pLGIC family member.

2090-PlatCeramide Channel Regulation by Bcl-2 Family Proteins: MolecularInsightsMeenu N. Perera, Shang-Hsuan Lin, Kaiti Chang, Marco Colombini.University of Maryland, College Park, MD, USA.The sphingolipid, ceramide can self-assemble in phospholipid membranes toform large channels capable of translocating proteins through membranes.Electronmicroscopic visualization and electrophysiological measurements re-veal a range of pore sizes with the most frequent being 10 nm in diameter.The channel size and the propensity to form channels are controlled by theapoptosis-related Bcl-2 family proteins. Bcl-xL favors channel disassemblywhereas Bax favors channel growth. By using ceramide analogs we have iden-tified regions of the ceramide molecule that are recognized by these proteinsand through which the proteins exert their influence. The ability of Bax to en-hance ceramide channel formation was sensitive to the stereochemistry of theceramide head group and the hydrogen bonding ability of the amide nitrogenwhereas the ability of Bcl-xL to favor channel disassembly was highly sensitiveto the length of the fatty acyl chain length of ceramide. It is likely that the N-acyl chain is binding to the hydrophobic pocket on Bcl-xL because the action ofBcl-xL is blocked by 2-methoxyantimycin A3, ABT-737 and ABT-263, inhib-itors that specifically bind at this site. The results are consistent with the con-clusion that the highly specific binding of these proteins to the ceramidechannel results in structural changes that propagate throughout the channel inan allosteric manner resulting in a disturbance of the dynamic equilibrium be-tween ceramides in the channel and ceramides in other forms in the membrane.Depending on the influence on the dynamic equilibrium channel growth or dis-assembly may be favored. (Supported by NSF grant: MCB-1023008)

2091-PlatActivation of M-Type Potassium Channels by Different Membrane Phos-pholipids and AnalogsVsevolod Telezhkin, Alastair J. Gibb, David A. Brown.University College London, London, United Kingdom.M-type (Kv7.2/7.3) channels are activated by the membrane phospholipidphosphatidylinositol-4,5-bisphosphate (PI(4,5)P2) [Li et al., 2005: J.Neurosci.,25,9825], through interaction with a cluster of basic residues in the C-terminus[Hernandez et al, 2008: J.Gen.Physiol., 132,361]. However, little is known ofthe phospholipid specificity and requirements for this activation. We have ex-plored this using inside-out membrane patches from CHO cells stably-expressing Kv7.2 and Kv7.3 subunits and held at a constant voltage ca. �20mV. The dioctanoyl mono-, di- and tri-phosphatidylinositides DiC8-PI(4)P,DiC8-PI(4,5)P2 and DiC8-PI(3,4,5)P3 all produced biphasic Popen-concentra-tion curves, maximizing at Popen ~0.8. EC50s for the ‘high-affinity’ component1 (maximum Popen ~0.2) were similar at ~1 mM; ‘low-affinity’ EC50s were in-versely proportional to the number of phosphates (DiC8-PI(4)P ~100 mM,DiC8-PI(4,5)P2 ~50 mM, DiC8-PI(3,4,5)P3 ~35 mM). In contrast, the inositolphosphates I(1,4,5)P3 and I(4,5)P2 neither activated nor inhibited the channelsup to 300 mM, suggesting a crucial role for the lipophilic moiety. This wastested further using sphingosine-1-phosphate (S-1-P), fingolimod phosphate(FTY720-P) and 1-oleoyl lysophosphatidic acid (LPA). All three activatedthe Kv7.2/7.3 channels, to Popen values at 100 mM of ~0.8 (LPA), 0.15 (S-1-P) and 0.022 (FTY720-P). In each case ‘high’ and ‘low’ affinity componentsto the activation curves could be discerned, with EC50s of 1.5 and 40 mM(LPA), 3 and 160 mM (S-1-P) and 0.5 and 61 mM (FTY720-P). No channel ac-tivation was observed using membrane lipids devoid of phosphate groups(D-erythrosphingosine, fingolimod, phosphatidylglycerol and phosphatidyl-choline, all at 100 mM). Thus, M-channels can be activated in a rather similarmanner by a range of membrane lipids, the minimal requirements at concentra-tions tested being one or more terminal phosphates and a lipophilic domain.Supported by Wellcome Trust grant 085419.

2092-PlatBK Channel Modulation by Leucine-Rich Repeat Containing ProteinsJiusheng Yan, Richard W. Aldrich.University of Texas at Austin, Austin, TX, USA.Molecular diversity of ion channel structure and function underlies variabilityin electrical signaling in nerve, muscle, and non-excitable cells. Regulation byvariable auxiliary subunits is a major mechanism to generate tissue- or cell-specific diversity of ion channel function.Mammalian large-conductance, volt-age and calcium-activated potassium (BK, KCa1.1) channels are ubiquitouslyexpressed with diverse functions in different tissues or cell types, consistingof the pore-forming, voltage- and Ca2þ-sensing a-subunits (BKa), or together