Peter J. Tonge

Find an error

Name: Tonge, Peter
Organization: Stony Brook University , USA
Department: Department of Chemistry
Title: Professor(PhD)

TOPICS

Co-reporter:Agnieszka A. Gil, Sergey P. Laptenok, James N. Iuliano, Andras Lukacs, Anil Verma, Christopher R. Hall, Grace E. Yoon, Richard Brust, Gregory M. Greetham, Michael Towrie, Jarrod B. French, Stephen R. Meech, and Peter J. Tonge
Journal of the American Chemical Society October 18, 2017 Volume 139(Issue 41) pp:14638-14638
Publication Date(Web):September 6, 2017
DOI:10.1021/jacs.7b07849
The flavin chromophore in blue-light-using FAD (BLUF) photoreceptors is surrounded by a hydrogen bond network that senses and responds to changes in the electronic structure of the flavin on the ultrafast time scale. The hydrogen bond network includes a strictly conserved Tyr residue, and previously we explored the role of this residue, Y21, in the photoactivation mechanism of the BLUF protein AppABLUF by the introduction of fluorotyrosine (F-Tyr) analogues that modulated the pKa and reduction potential of Y21 by 3.5 pH units and 200 mV, respectively. Although little impact on the forward (dark- to light-adapted form) photoreaction was observed, the change in Y21 pKa led to a 4000-fold increase in the rate of dark-state recovery. In the present work we have extended these studies to the BLUF protein PixD, where, in contrast to AppABLUF, modulation in the Tyr (Y8) pKa has a profound impact on the forward photoreaction. In particular, a decrease in Y8 pKa by 2 or more pH units prevents formation of a stable light state, consistent with a photoactivation mechanism that involves proton transfer or proton-coupled electron transfer from Y8 to the electronically excited FAD. Conversely, the effect of pKa on the rate of dark recovery is markedly reduced in PixD. These observations highlight very significant differences between the photocycles of PixD and AppABLUF, despite their sharing highly conserved FAD binding architectures.
Co-reporter:Lauren A. Spagnuolo, Sandra Eltschkner, Weixuan Yu, Fereidoon Daryaee, Shabnam Davoodi, Susan E. Knudson, Eleanor K. H. Allen, Jonathan Merino, Annica Pschibul, Ben Moree, Neil Thivalapill, James J. Truglio, Joshua Salafsky, Richard A. Slayden, Caroline Kisker, and Peter J. Tonge
Journal of the American Chemical Society March 8, 2017 Volume 139(Issue 9) pp:3417-3417
Publication Date(Web):February 2, 2017
DOI:10.1021/jacs.6b11148
A critical goal of lead compound selection and optimization is to maximize target engagement while minimizing off-target binding. Since target engagement is a function of both the thermodynamics and kinetics of drug–target interactions, it follows that the structures of both the ground states and transition states on the binding reaction coordinate are needed to rationally modulate the lifetime of the drug–target complex. Previously, we predicted the structure of the rate-limiting transition state that controlled the time-dependent inhibition of the enoyl-ACP reductase InhA. This led to the discovery of a triazole-containing diphenyl ether with an increased residence time on InhA due to transition-state destabilization rather than ground-state stabilization. In the present work, we evaluate the inhibition of InhA by 14 triazole-based diphenyl ethers and use a combination of enzyme kinetics and X-ray crystallography to generate a structure–kinetic relationship for time-dependent binding. We show that the triazole motif slows the rate of formation for the final drug–target complex by up to 3 orders of magnitude. In addition, we identify a novel inhibitor with a residence time on InhA of 220 min, which is 3.5-fold longer than that of the INH-NAD adduct formed by the tuberculosis drug, isoniazid. This study provides a clear example in which the lifetime of the drug–target complex is controlled by interactions in the transition state for inhibitor binding rather than the ground state of the enzyme–inhibitor complex, and demonstrates the important role that on-rates can play in drug–target residence time.
Co-reporter:Carla Neckles, Sandra Eltschkner, Jason E. Cummings, Maria Hirschbeck, Fereidoon Daryaee, Gopal R. Bommineni, Zhuo Zhang, Lauren Spagnuolo, Weixuan Yu, Shabnam Davoodi, Richard A. Slayden, Caroline Kisker, and Peter J. Tonge
Biochemistry April 4, 2017 Volume 56(Issue 13) pp:1865-1865
Publication Date(Web):February 22, 2017
DOI:10.1021/acs.biochem.6b01048
There is growing awareness of the link between drug–target residence time and in vivo drug activity, and there are increasing efforts to determine the molecular factors that control the lifetime of a drug–target complex. Rational alterations in the drug–target residence time require knowledge of both the ground and transition states on the inhibition reaction coordinate, and we have determined the structure–kinetic relationship for 22 ethyl- or hexyl-substituted diphenyl ethers that are slow-binding inhibitors of bpFabI1, the enoyl-ACP reductase FabI1 from Burkholderia pseudomallei. Analysis of enzyme inhibition using a two-dimensional kinetic map demonstrates that the ethyl and hexyl diphenyl ethers fall into two distinct clusters. Modifications to the ethyl diphenyl ether B ring result in changes to both on and off rates, where residence times of up to ∼700 min (∼11 h) are achieved by either ground state stabilization (PT444) or transition state destabilization (slower on rate) (PT404). By contrast, modifications to the hexyl diphenyl ether B ring result in residence times of 300 min (∼5 h) through changes in only ground state stabilization (PT119). Structural analysis of nine enzyme:inhibitor complexes reveals that the variation in structure–kinetic relationships can be rationalized by structural rearrangements of bpFabI1 and subtle changes to the orientation of the inhibitor in the binding pocket. Finally, we demonstrate that three compounds with residence times on bpFabI1 from 118 min (∼2 h) to 670 min (∼11 h) have in vivo efficacy in an acute B. pseudomallei murine infection model using the virulent B. pseudomallei strain Bp400.
Co-reporter:Agnieszka A. Gil, Sergey P. Laptenok, Jarrod B. FrenchJames N. Iuliano, Andras Lukacs, Christopher R. Hall, Igor V. Sazanovich, Gregory M. Greetham, Adelbert Bacher, Boris Illarionov, Markus Fischer, Peter J. TongeStephen R. Meech
The Journal of Physical Chemistry B 2017 Volume 121(Issue 5) pp:
Publication Date(Web):January 9, 2017
DOI:10.1021/acs.jpcb.7b00088
The rational engineering of photosensor proteins underpins the field of optogenetics, in which light is used for spatiotemporal control of cell signaling. Optogenetic elements function by converting electronic excitation of an embedded chromophore into structural changes on the microseconds to seconds time scale, which then modulate the activity of output domains responsible for biological signaling. Using time-resolved vibrational spectroscopy coupled with isotope labeling, we have mapped the structural evolution of the LOV2 domain of the flavin binding phototropin Avena sativa (AsLOV2) over 10 decades of time, reporting structural dynamics between 100 fs and 1 ms after optical excitation. The transient vibrational spectra contain contributions from both the flavin chromophore and the surrounding protein matrix. These contributions are resolved and assigned through the study of four different isotopically labeled samples. High signal-to-noise data permit the detailed analysis of kinetics associated with the light activated structural evolution. A pathway for the photocycle consistent with the data is proposed. The earliest events occur in the flavin binding pocket, where a subpicosecond perturbation of the protein matrix occurs. In this perturbed environment, the previously characterized reaction between triplet state isoalloxazine and an adjacent cysteine leads to formation of the adduct state; this step is shown to exhibit dispersive kinetics. This reaction promotes coupling of the optical excitation to successive time-dependent structural changes, initially in the β-sheet and then α-helix regions of the AsLOV2 domain, which ultimately gives rise to Jα-helix unfolding, yielding the signaling state. This model is tested through point mutagenesis, elucidating in particular the key mediating role played by Q513.
Co-reporter:Fereidoon Daryaee;Zhuo Zhang;Kayla R. Gogarty;Yong Li;Jonathan Merino;Stewart L. Fisher
Chemical Science (2010-Present) 2017 vol. 8(Issue 5) pp:3434-3443
Publication Date(Web):2017/05/03
DOI:10.1039/C6SC03306G
Correlating target engagement with in vivo drug activity remains a central challenge in efforts to improve the efficiency of drug discovery. Previously we described a mechanistic pharmacokinetic–pharmacodynamic (PK/PD) model that used drug–target binding kinetics to successfully predict the in vivo efficacy of antibacterial compounds in models of Pseudomonas aeruginosa and Staphylococcus aureus infection. In the present work we extend this model to quantitatively correlate the engagement of Bruton's tyrosine kinase (Btk) by the covalent inhibitor CC-292 with the ability of this compound to reduce ankle swelling in an animal model of arthritis. The modeling studies include the rate of Btk turnover and reveal the vulnerability of Btk to engagement by CC-292.
Co-reporter:Fereidoon Daryaee, Andrew Chang, Johannes Schiebel, Yang Lu, Zhuo Zhang, Kanishk Kapilashrami, Stephen G. Walker, Caroline Kisker, Christoph A. Sotriffer, Stewart L. Fisher and Peter J. Tonge  
Chemical Science 2016 vol. 7(Issue 9) pp:5945-5954
Publication Date(Web):25 May 2016
DOI:10.1039/C6SC01000H
Drug–target kinetics enable time-dependent changes in target engagement to be quantified as a function of drug concentration. When coupled to drug pharmacokinetics (PK), drug–target kinetics can thus be used to predict in vivo pharmacodynamics (PD). Previously we described a mechanistic PK/PD model that successfully predicted the antibacterial activity of an LpxC inhibitor in a model of Pseudomonas aeruginosa infection. In the present work we demonstrate that the same approach can be used to predict the in vivo activity of an enoyl-ACP reductase (FabI) inhibitor in a model of methicillin-resistant Staphylococcus aureus (MRSA) infection. This is significant because the LpxC inhibitors are cidal, whereas the FabI inhibitors are static. In addition P. aeruginosa is a Gram-negative organism whereas MRSA is Gram-positive. Thus this study supports the general applicability of our modeling approach across antibacterial space.
Co-reporter:Christopher E. EvansJoe S. Matarlo, Peter J. Tonge, Derek S. Tan
Organic Letters 2016 Volume 18(Issue 24) pp:6384-6387
Publication Date(Web):December 1, 2016
DOI:10.1021/acs.orglett.6b03272
A stereoselective synthesis has been developed to provide all four side-chain stereoisomers of difluoroindanediol 2, the mixture of which was previously identified as an inhibitor of the o-succinylbenzoate-CoA synthetase MenE in bacterial menaquinone biosynthesis, having promising in vitro activity against methicillin-resistant Staphylococcus aureus and Mycobacterium tuberculosis. Only the (1R,3S)-diastereomer inhibited the biochemical activity of MenE, consistent with computational docking studies, and this diastereomer also exhibited in vitro antibacterial activity comparable to that of the mixture. However, mechanism-of-action studies suggest that this inhibitor and its diastereomers may act via other mechanisms beyond inhibition of menaquinone biosynthesis.
Co-reporter:Gopal R. Bommineni; Kanishk Kapilashrami; Jason E. Cummings; Yang Lu; Susan E. Knudson; Chendi Gu; Stephen G. Walker; Richard A. Slayden
Journal of Medicinal Chemistry 2016 Volume 59(Issue 11) pp:5377-5390
Publication Date(Web):May 17, 2016
DOI:10.1021/acs.jmedchem.6b00236
β-Ketoacyl-ACP synthases (KAS) are key enzymes involved in the type II bacterial fatty acid biosynthesis (FASII) pathway and are putative targets for antibacterial discovery. Several natural product KAS inhibitors have previously been reported, including thiolactomycin (TLM), which is produced by Nocardia spp. Here we describe the synthesis and characterization of optically pure 5R-thiolactomycin (TLM) analogues that show improved whole cell activity against bacterial strains including methicillin-resistant Staphylococcus aureus (MRSA) and priority pathogens such as Francisella tularensis and Burkholderia pseudomallei. In addition, we identify TLM analogues with in vivo efficacy against MRSA and Klebsiella pneumoniae in animal models of infection.
Co-reporter:Carla Neckles, Annica Pschibul, Cheng-Tsung Lai, Maria Hirschbeck, Jochen Kuper, Shabnam Davoodi, Junjie Zou, Nina Liu, Pan Pan, Sonam Shah, Fereidoon Daryaee, Gopal R. Bommineni, Cristina Lai, Carlos Simmerling, Caroline Kisker, and Peter J. Tonge
Biochemistry 2016 Volume 55(Issue 21) pp:2992-3006
Publication Date(Web):May 2, 2016
DOI:10.1021/acs.biochem.5b01301
The enoyl-ACP reductase (ENR) catalyzes the last reaction in the elongation cycle of the bacterial type II fatty acid biosynthesis (FAS-II) pathway. While the FabI ENR is a well-validated drug target in organisms such as Mycobacterium tuberculosis and Staphylococcus aureus, alternate ENR isoforms have been discovered in other pathogens, including the FabV enzyme that is the sole ENR in Yersinia pestis (ypFabV). Previously, we showed that the prototypical ENR inhibitor triclosan was a poor inhibitor of ypFabV and that inhibitors based on the 2-pyridone scaffold were more potent [Hirschbeck, M. (2012) Structure 20 (1), 89–100]. These studies were performed with the T276S FabV variant. In the work presented here, we describe a detailed examination of the mechanism and inhibition of wild-type ypFabV and the T276S variant. The T276S mutation significantly reduces the affinity of diphenyl ether inhibitors for ypFabV (20-fold → 100-fold). In addition, while T276S ypFabV generally displays an affinity for 2-pyridone inhibitors higher than that of the wild-type enzyme, the 4-pyridone scaffold yields compounds with similar affinity for both wild-type and T276S ypFabV. T276 is located at the N-terminus of the helical substrate-binding loop, and structural studies coupled with site-directed mutagenesis reveal that alterations in this residue modulate the size of the active site portal. Subsequently, we were able to probe the mechanism of time-dependent inhibition in this enzyme family by extending the inhibition studies to include P142W ypFabV, a mutation that results in a gain of slow-onset inhibition for the 4-pyridone PT156.
Co-reporter:Joe S. Matarlo, Yang Lu, Fereidoon Daryaee, Taraneh Daryaee, Bela Ruzsicska, Stephen G. Walker, and Peter J. Tonge
ACS Infectious Diseases 2016 Volume 2(Issue 5) pp:329
Publication Date(Web):March 7, 2016
DOI:10.1021/acsinfecdis.6b00023
4-Oxo-4-phenyl-but-2-enoates inhibit MenB, the 1,4-dihydroxyl-2-naphthoyl-CoA synthase in the bacterial menaquinone (MK) biosynthesis pathway, through the formation of an adduct with coenzyme A (CoA). Here, we show that the corresponding methyl butenoates have minimum inhibitory concentration (MIC) values as low as 0.35–0.75 μg/mL against drug-sensitive and -resistant strains of Staphylococcus aureus. Mode of action studies on the most potent compound, methyl 4-(4-chlorophenyl)-4-oxobut-2-enoate (1), reveal that 1 is converted into the corresponding CoA adduct in S. aureus cells and that this adduct binds to the S. aureus MenB (saMenB) with a Kd value of 2 μM. The antibacterial spectrum of 1 is limited to bacteria that utilize MK for respiration, and the activity of 1 can be complemented with exogenous MK or menadione. Finally, treatment of methicillin-resistant S. aureus (MRSA) with 1 results in the small colony variant phenotype, and thus 1 phenocopies knockout of the menB gene. Taken together, the data indicate that the antibacterial activity of 1 results from a specific effect on MK biosynthesis. We also evaluated the in vivo efficacy of 1 using two mouse models of MRSA infection. Notably, compound 1 increased survival in a systemic infection model and resulted in a dose-dependent decrease in bacterial load in a thigh infection model, validating MenB as a target for the development of new anti-MRSA candidates.Keywords: DHNA; electrophilic antibacterial compound; menadione; menaquinone biosynthesis; MenB; Staphylococcus aureus
Co-reporter:Agnieszka A. Gil; Allison Haigney; Sergey P. Laptenok; Richard Brust; Andras Lukacs; James N. Iuliano; Jessica Jeng; Eduard H. Melief; Rui-Kun Zhao; EunBin Yoon; Ian P. Clark; Michael Towrie; Gregory M. Greetham; Annabelle Ng; James J. Truglio; Jarrod B. French; Stephen R. Meech
Journal of the American Chemical Society 2015 Volume 138(Issue 3) pp:926-935
Publication Date(Web):December 26, 2015
DOI:10.1021/jacs.5b11115
The transcriptional antirepressor AppA is a blue light using flavin (BLUF) photoreceptor that releases the transcriptional repressor PpsR upon photoexcitation. Light activation of AppA involves changes in a hydrogen-bonding network that surrounds the flavin chromophore on the nanosecond time scale, while the dark state of AppA is then recovered in a light-independent reaction with a dramatically longer half-life of 15 min. Residue Y21, a component of the hydrogen-bonding network, is known to be essential for photoactivity. Here, we directly explore the effect of the Y21 pKa on dark state recovery by replacing Y21 with fluorotyrosine analogues that increase the acidity of Y21 by 3.5 pH units. Ultrafast transient infrared measurements confirm that the structure of AppA is unperturbed by fluorotyrosine substitution, and that there is a small (3-fold) change in the photokinetics of the forward reaction over the fluorotyrosine series. However, reduction of 3.5 pH units in the pKa of Y21 increases the rate of dark state recovery by 4000-fold with a Brønsted coefficient of ∼1, indicating that the Y21 proton is completely transferred in the transition state leading from light to dark adapted AppA. A large solvent isotope effect of ∼6–8 is also observed on the rate of dark state recovery. These data establish that the acidity of Y21 is a crucial factor for stabilizing the light activated form of the protein, and have been used to propose a model for dark state recovery that will ultimately prove useful for tuning the properties of BLUF photosensors for optogenetic applications.
Co-reporter:Joe S. Matarlo, Christopher E. Evans, Indrajeet Sharma, Lubens J. Lavaud, Stephen C. Ngo, Roger Shek, Kanagalaghatta R. Rajashankar, Jarrod B. French, Derek S. Tan, and Peter J. Tonge
Biochemistry 2015 Volume 54(Issue 42) pp:6514-6524
Publication Date(Web):September 22, 2015
DOI:10.1021/acs.biochem.5b00966
MenE is an o-succinylbenzoyl-CoA (OSB-CoA) synthetase in the bacterial menaquinone biosynthesis pathway and is a promising target for the development of novel antibacterial agents. The enzyme catalyzes CoA ligation via an acyl-adenylate intermediate, and we have previously reported tight-binding inhibitors of MenE based on stable acyl-sulfonyladenosine analogues of this intermediate, including OSB-AMS (1), which has an IC50 value of ≤25 nM for Escherichia coli MenE. Herein, we show that OSB-AMS reduces menaquinone levels in Staphylococcus aureus, consistent with its proposed mechanism of action, despite the observation that the antibacterial activity of OSB-AMS is ∼1000-fold lower than the IC50 for enzyme inhibition. To inform the synthesis of MenE inhibitors with improved antibacterial activity, we have undertaken a structure–activity relationship (SAR) study stimulated by the knowledge that OSB-AMS can adopt two isomeric forms in which the OSB side chain exists either as an open-chain keto acid or a cyclic lactol. These studies revealed that negatively charged analogues of the keto acid form bind, while neutral analogues do not, consistent with the hypothesis that the negatively charged keto acid form of OSB-AMS is the active isomer. X-ray crystallography and site-directed mutagenesis confirm the importance of a conserved arginine for binding the OSB carboxylate. Although most lactol isomers tested were inactive, a novel difluoroindanediol inhibitor (11) with improved antibacterial activity was discovered, providing a pathway toward the development of optimized MenE inhibitors in the future.
Co-reporter:Cheng-Tsung Lai, Huei-Jiun Li, Weixuan Yu, Sonam Shah, Gopal R. Bommineni, Victoria Perrone, Miguel Garcia-Diaz, Peter J. Tonge, and Carlos Simmerling
Biochemistry 2015 Volume 54(Issue 30) pp:4683-4691
Publication Date(Web):July 6, 2015
DOI:10.1021/acs.biochem.5b00284
Slow-onset enzyme inhibitors are the subject of considerable interest as an approach to increasing the potency of pharmaceutical compounds by extending the residence time of the inhibitor on the target (the lifetime of the drug–receptor complex). However, rational modulation of residence time presents significant challenges because it requires additional mechanistic insight, such as the nature of the transition state for postbinding isomerization. Our previous work, based on X-ray crystallography, enzyme kinetics, and molecular dynamics simulation, suggested that the slow step in inhibition of the Mycobacterium tuberculosis enoyl-ACP reductase InhA involves a change in the conformation of the substrate binding loop from an open state in the initial enzyme–inhibitor complex to a closed state in the final enzyme–inhibitor complex. Here, we use multidimensional free energy landscapes for loop isomerization to obtain a computational model for the transition state. The results suggest that slow-onset inhibitors crowd key side chains on helices that slide past each other during isomerization, resulting in a steric clash. The landscapes become significantly flatter when residues involved in the steric clash are replaced with alanine. Importantly, this lower barrier can be increased by rational inhibitor redesign to restore the steric clash. Crystallographic studies and enzyme kinetics confirm the predicted effects on loop structure and flexibility, as well as inhibitor residence time. These loss and regain of function studies validate our mechanistic hypothesis for interactions controlling substrate binding loop isomerization, providing a platform for the future design of inhibitors with longer residence times and better in vivo potency. Similar opportunities for slow-onset inhibition via the same mechanism are identified in other pathogens.
Co-reporter:Johannes Schiebel, Andrew Chang, Benjamin Merget, Gopal R. Bommineni, Weixuan Yu, Lauren A. Spagnuolo, Michael V. Baxter, Mona Tareilus, Peter J. Tonge, Caroline Kisker, and Christoph A. Sotriffer
Biochemistry 2015 Volume 54(Issue 10) pp:1943-1955
Publication Date(Web):February 23, 2015
DOI:10.1021/bi5014358
One third of all drugs in clinical use owe their pharmacological activity to the functional inhibition of enzymes, highlighting the importance of enzymatic targets for drug development. Because of the close relationship between inhibition and catalysis, understanding the recognition and turnover of enzymatic substrates is essential for rational drug design. Although the Staphylococcus aureus enoyl-acyl carrier protein reductase (saFabI) involved in bacterial fatty acid biosynthesis constitutes a very promising target for the development of novel, urgently needed anti-staphylococcal agents, the substrate binding mode and catalytic mechanism remained unclear for this enzyme. Using a combined crystallographic, kinetic, and computational approach, we have explored the chemical properties of the saFabI binding cavity, obtaining a consistent mechanistic model for substrate binding and turnover. We identified a water-molecule network linking the active site with a water basin inside the homo-tetrameric protein, which seems to be crucial for the closure of the flexible substrate binding loop as well as for an effective hydride and proton transfer during catalysis. On the basis of our results, we also derive a new model for the FabI-ACP complex that reveals how the ACP-bound acyl-substrate is injected into the FabI binding crevice. These findings support the future development of novel FabI inhibitors that target the FabI-ACP interface leading to the disruption of the interaction between these two proteins.
Co-reporter:Hui Wang, Li Liu, Yang Lu, Pan Pan, Jacob M. Hooker, Joanna S. Fowler, Peter J. Tonge
Bioorganic & Medicinal Chemistry Letters 2015 Volume 25(Issue 21) pp:4782-4786
Publication Date(Web):1 November 2015
DOI:10.1016/j.bmcl.2015.07.019
PT70 is a diaryl ether inhibitor of InhA, the enoyl-ACP reductase in the Mycobacterium tuberculosis fatty acid biosynthesis pathway. It has a residence time of 24 min on the target, and also shows antibacterial activity in a mouse model of tuberculosis infection. Due to the interest in studying target tissue pharmacokinetics of PT70, we developed a method to radiolabel PT70 with carbon-11 and have studied its pharmacokinetics in mice and baboons using positron emission tomography.
Co-reporter:Weixuan Yu, Carla Neckles, Andrew Chang, Gopal Reddy Bommineni, Lauren Spagnuolo, Zhuo Zhang, Nina Liu, Christina Lai, James Truglio, Peter J. Tonge
Analytical Biochemistry 2015 Volume 474() pp:40-49
Publication Date(Web):1 April 2015
DOI:10.1016/j.ab.2014.12.022

Abstract

The classical methods for quantifying drug–target residence time (tR) use loss or regain of enzyme activity in progress curve kinetic assays. However, such methods become imprecise at very long residence times, mitigating the use of alternative strategies. Using the NAD(P)H-dependent FabI enoyl-acyl carrier protein (enoyl-ACP) reductase as a model system, we developed a Penefsky column-based method for direct measurement of tR, where the off-rate of the drug was determined with radiolabeled [adenylate-32P]NAD(P+) cofactor. In total, 23 FabI inhibitors were analyzed, and a mathematical model was used to estimate limits to the tR values of each inhibitor based on percentage drug–target complex recovery following gel filtration. In general, this method showed good agreement with the classical steady-state kinetic methods for compounds with tR values of 10 to 100 min. In addition, we were able to identify seven long tR inhibitors (100–1500 min) and to accurately determine their tR values. The method was then used to measure tR as a function of temperature, an analysis not previously possible using the standard kinetic approach due to decreased NAD(P)H stability at elevated temperatures. In general, a 4-fold difference in tR was observed when the temperature was increased from 25 to 37 °C.

Co-reporter:Dr. Sergey P. Laptenok; Andras Lukacs;Agnieszka Gil;Dr. Richard Brust;Dr. Igor V. Sazanovich;Dr. Gregory M. Greetham; Peter J. Tonge; Stephen R. Meech
Angewandte Chemie 2015 Volume 127( Issue 32) pp:9435-9439
Publication Date(Web):
DOI:10.1002/ange.201503672

Abstract

Proton transfer is critical in many important biochemical reactions. The unique three-step excited-state proton transfer in avGFP allows observations of protein proton transport in real-time. In this work we exploit femtosecond to microsecond transient IR spectroscopy to record, in D2O, the complete proton transfer photocycle of avGFP, and two mutants (T203V and S205V) which modify the structure of the proton wire. Striking differences and similarities are observed among the three mutants yielding novel information on proton transfer mechanism, rates, isotope effects, H-bond strength and proton wire stability. These data provide a detailed picture of the dynamics of long-range proton transfer in a protein against which calculations may be compared.

Co-reporter:Dr. Sergey P. Laptenok; Andras Lukacs;Agnieszka Gil;Dr. Richard Brust;Dr. Igor V. Sazanovich;Dr. Gregory M. Greetham; Peter J. Tonge; Stephen R. Meech
Angewandte Chemie International Edition 2015 Volume 54( Issue 32) pp:9303-9307
Publication Date(Web):
DOI:10.1002/anie.201503672

Abstract

Proton transfer is critical in many important biochemical reactions. The unique three-step excited-state proton transfer in avGFP allows observations of protein proton transport in real-time. In this work we exploit femtosecond to microsecond transient IR spectroscopy to record, in D2O, the complete proton transfer photocycle of avGFP, and two mutants (T203V and S205V) which modify the structure of the proton wire. Striking differences and similarities are observed among the three mutants yielding novel information on proton transfer mechanism, rates, isotope effects, H-bond strength and proton wire stability. These data provide a detailed picture of the dynamics of long-range proton transfer in a protein against which calculations may be compared.

Co-reporter:Andras Lukacs ; Richard Brust ; Allison Haigney ; Sergey P. Laptenok ; Kiri Addison ; Agnieszka Gil ; Michael Towrie ; Gregory M. Greetham ; Peter J. Tonge ;Stephen R. Meech
Journal of the American Chemical Society 2014 Volume 136(Issue 12) pp:4605-4615
Publication Date(Web):March 2, 2014
DOI:10.1021/ja4121082
BLUF (blue light using flavin) domain proteins are an important family of blue light-sensing proteins which control a wide variety of functions in cells. The primary light-activated step in the BLUF domain is not yet established. A number of experimental and theoretical studies points to a role for photoinduced electron transfer (PET) between a highly conserved tyrosine and the flavin chromophore to form a radical intermediate state. Here we investigate the role of PET in three different BLUF proteins, using ultrafast broadband transient infrared spectroscopy. We characterize and identify infrared active marker modes for excited and ground state species and use them to record photochemical dynamics in the proteins. We also generate mutants which unambiguously show PET and, through isotope labeling of the protein and the chromophore, are able to assign modes characteristic of both flavin and protein radical states. We find that these radical intermediates are not observed in two of the three BLUF domains studied, casting doubt on the importance of the formation of a population of radical intermediates in the BLUF photocycle. Further, unnatural amino acid mutagenesis is used to replace the conserved tyrosine with fluorotyrosines, thus modifying the driving force for the proposed electron transfer reaction; the rate changes observed are also not consistent with a PET mechanism. Thus, while intermediates of PET reactions can be observed in BLUF proteins they are not correlated with photoactivity, suggesting that radical intermediates are not central to their operation. Alternative nonradical pathways including a keto–enol tautomerization induced by electronic excitation of the flavin ring are considered.
Co-reporter:Huei-Jiun Li, Cheng-Tsung Lai, Pan Pan, Weixuan Yu, Nina Liu, Gopal R. Bommineni, Miguel Garcia-Diaz, Carlos Simmerling, and Peter J. Tonge
ACS Chemical Biology 2014 Volume 9(Issue 4) pp:986
Publication Date(Web):February 14, 2014
DOI:10.1021/cb400896g
Slow-onset enzyme inhibitors are of great interest for drug discovery programs since the slow dissociation of the inhibitor from the drug–target complex results in sustained target occupancy leading to improved pharmacodynamics. However, the structural basis for slow-onset inhibition is often not fully understood, hindering the development of structure-kinetic relationships and the rational optimization of drug-target residence time. Previously we demonstrated that slow-onset inhibition of the Mycobacterium tuberculosis enoyl-ACP reductase InhA correlated with motions of a substrate-binding loop (SBL) near the active site. In the present work, X-ray crystallography and molecular dynamics simulations have been used to map the structural and energetic changes of the SBL that occur upon enzyme inhibition. Helix-6 within the SBL adopts an open conformation when the inhibitor structure or binding kinetics is substrate-like. In contrast, slow-onset inhibition results in large-scale local refolding in which helix-6 adopts a closed conformation not normally populated during substrate turnover. The open and closed conformations of helix-6 are hypothesized to represent the EI and EI* states on the two-step induced-fit reaction coordinate for enzyme inhibition. These two states were used as the end points for nudged elastic band molecular dynamics simulations resulting in two-dimensional potential energy profiles that reveal the barrier between EI and EI*, thus rationalizing the binding kinetics observed with different inhibitors. Our findings indicate that the structural basis for slow-onset kinetics can be understood once the structures of both EI and EI* have been identified, thus providing a starting point for the rational control of enzyme–inhibitor binding kinetics.
Co-reporter:Hui Wang, Yang Lu, Li Liu, Sung Won Kim, Jacob M. Hooker, Joanna S. Fowler, Peter J. Tonge
European Journal of Medicinal Chemistry 2014 Volume 88() pp:66-73
Publication Date(Web):17 December 2014
DOI:10.1016/j.ejmech.2014.09.008
•A radiolabelling method for a potent Staphylococcus aureus enoyl-ACP reductase inhibitor was developed.•The biodistribution of [11C]PT119 was determined in vivo.•PT119 demonstrated antibacterial activity in two different S. aureus infection models.The pharmacokinetics (PK) and pharmacodynamics (PD) of PT119, a potent Staphylococcus aureus enoyl-ACP reductase (saFabI) inhibitor with a Ki value of 0.01 nM and a residence time of 750 min on the enzyme target, has been evaluated in mice. PT119 was found to have promising antibacterial activity in two different S. aureus infection models: it caused a 3 log reduction in the CFU's in a mouse thigh muscle infection model and increased the survival rate from 0% to 50% in a mouse systemic infection model. PT119 was then radiolabeled with carbon-11 to evaluate its biodistribution and PK in both healthy and S. aureus infected mice using positron emission tomography (PET). The biodistribution of [11C]PT119 and/or its labeled metabolites did not differ significantly between the healthy group and the infected group, and PT119 was found to distribute equally between serum and tissue during the ∼1 h of analysis permitted by the carbon-11 half life. This approach provides important data for PK/PD modeling and is the first step in identifying radiotracers that can non-invasively image bacterial infection in vivo.
Co-reporter:Richard Brust, Allison Haigney, Andras Lukacs, Agnieszka Gil, Shahrier Hossain, Kiri Addison, Cheng-Tsung Lai, Michael Towrie, Gregory M. Greetham, Ian P. Clark, Boris Illarionov, Adelbert Bacher, Ryu-Ryun Kim, Markus Fischer, Carlos Simmerling, Stephen R. Meech, and Peter J. Tonge
The Journal of Physical Chemistry Letters 2014 Volume 5(Issue 1) pp:220-224
Publication Date(Web):December 6, 2013
DOI:10.1021/jz4023738
Acinetobacter baumannii is an important human pathogen that can form biofilms and persist under harsh environmental conditions. Biofilm formation and virulence are modulated by blue light, which is thought to be regulated by a BLUF protein, BlsA. To understand the molecular mechanism of light sensing, we have used steady-state and ultrafast vibrational spectroscopy to compare the photoactivation mechanism of BlsA to the BLUF photosensor AppA from Rhodobacter sphaeroides. Although similar photocycles are observed, vibrational data together with homology modeling identify significant differences in the β5 strand in BlsA caused by photoactivation, which are proposed to be directly linked to downstream signaling.Keywords: Acinetobacter baumannii; AppA; BlsA; blue light using FAD; BLUF domain; photosensor; ultrafast time-resolved infrared;
Co-reporter:Richard Brust ; Andras Lukacs ; Allison Haigney ; Kiri Addison ; Agnieszka Gil ; Michael Towrie ; Ian P. Clark ; Gregory M. Greetham ; Peter J. Tonge ;Stephen R. Meech
Journal of the American Chemical Society 2013 Volume 135(Issue 43) pp:16168-16174
Publication Date(Web):October 1, 2013
DOI:10.1021/ja407265p
Living systems are fundamentally dependent on the ability of proteins to respond to external stimuli. The mechanism, the underlying structural dynamics, and the time scales for regulation of this response are central questions in biochemistry. Here we probe the structural dynamics of the BLUF domain found in several photoactive flavoproteins, which is responsible for light activated functions as diverse as phototaxis and gene regulation. Measurements have been made over 10 decades of time (from 100 fs to 1 ms) using transient vibrational spectroscopy. Chromophore (flavin ring) localized dynamics occur on the pico- to nanosecond time scale, while subsequent protein structural reorganization is observed over microseconds. Multiple time scales are observed for the dynamics associated with different vibrations of the protein, suggesting an underlying hierarchical relaxation pathway. Structural evolution in residues directly H-bonded to the chromophore takes place more slowly than changes in more remote residues. However, a point mutation which suppresses biological function is shown to ‘short circuit’ this structural relaxation pathway, suppressing the changes which occur further away from the chromophore while accelerating dynamics close to it.
Co-reporter:Andrew Chang, Johannes Schiebel, Weixuan Yu, Gopal R. Bommineni, Pan Pan, Michael V. Baxter, Avinash Khanna, Christoph A. Sotriffer, Caroline Kisker, and Peter J. Tonge
Biochemistry 2013 Volume 52(Issue 24) pp:
Publication Date(Web):May 22, 2013
DOI:10.1021/bi400413c
Drug-target kinetics has recently emerged as an especially important facet of the drug discovery process. In particular, prolonged drug-target residence times may confer enhanced efficacy and selectivity in the open in vivo system. However, the lack of accurate kinetic and structural data for a series of congeneric compounds hinders the rational design of inhibitors with decreased off-rates. Therefore, we chose the Staphylococcus aureus enoyl-ACP reductase (saFabI) — an important target for the development of new anti-staphylococcal drugs — as a model system to rationalize and optimize the drug-target residence time on a structural basis. Using our new, efficient, and widely applicable mechanistically informed kinetic approach, we obtained a full characterization of saFabI inhibition by a series of 20 diphenyl ethers complemented by a collection of 9 saFabI–inhibitor crystal structures. We identified a strong correlation between the affinities of the investigated saFabI diphenyl ether inhibitors and their corresponding residence times, which can be rationalized on a structural basis. Because of its favorable interactions with the enzyme, the residence time of our most potent compound exceeds 10 h. In addition, we found that affinity and residence time in this system can be significantly enhanced by modifications predictable by a careful consideration of catalysis. Our study provides a blueprint for investigating and prolonging drug-target kinetics and may aid in the rational design of long-residence-time inhibitors targeting the essential saFabI enzyme.
Co-reporter:Andras Lukacs, Allison Haigney, Richard Brust, Kiri Addison, Michael Towrie, Gregory M. Greetham, Garth A. Jones, Atsushi Miyawaki, Peter J. Tonge, and Stephen R. Meech
The Journal of Physical Chemistry B 2013 Volume 117(Issue 40) pp:11954-11959
Publication Date(Web):September 13, 2013
DOI:10.1021/jp406142g
Photochromic proteins, such as Dronpa, are of particular importance in bioimaging and form the basis of ultraresolution fluorescence microscopy. The photochromic reaction involves switching between a weakly emissive neutral trans form of the chromophore (A) and its emissive cis anion (B). Controlling the rates of switching has the potential to significantly enhance the spatial and temporal resolution in microscopy. However, the mechanism of the switching reaction has yet to be established. Here we report a high signal-to-noise ultrafast transient infrared investigation of the photochromic reaction in the mutant Dronpa2, which exhibits facile switching behavior. In these measurements we excite both the A and B forms and observe the evolution in the IR difference spectra over hundreds of picoseconds. Electronic excitation leads to bleaching of the ground electronic state and instantaneous (subpicosecond) changes in the vibrational spectrum of the protein. The chromophore and protein modes evolve with different kinetics. The chromophore ground state recovers in a fast nonsingle-exponential relaxation, while in a competing reaction the protein undergoes a structural change. This results in formation of a metastable form of the protein in its ground electronic state (A′), which subsequently evolves on the time scale of hundreds of picoseconds. The changes in the vibrational spectrum that occur on the subnanosecond time scale do not show unambiguous evidence for either proton transfer or isomerization, suggesting that these low-yield processes occur from the metastable state on a longer time scale and are thus not the primary photoreaction. Formation of A′, and further relaxation of this state to the cis anion B, are relatively rare events, thus accounting for the overall low yield of the photochemical reaction.
Co-reporter:Fereidoon Daryaee, Zhuo Zhang, Kayla R. Gogarty, Yong Li, Jonathan Merino, Stewart L. Fisher and Peter J. Tonge
Chemical Science (2010-Present) 2017 - vol. 8(Issue 5) pp:NaN3443-3443
Publication Date(Web):2017/03/14
DOI:10.1039/C6SC03306G
Correlating target engagement with in vivo drug activity remains a central challenge in efforts to improve the efficiency of drug discovery. Previously we described a mechanistic pharmacokinetic–pharmacodynamic (PK/PD) model that used drug–target binding kinetics to successfully predict the in vivo efficacy of antibacterial compounds in models of Pseudomonas aeruginosa and Staphylococcus aureus infection. In the present work we extend this model to quantitatively correlate the engagement of Bruton's tyrosine kinase (Btk) by the covalent inhibitor CC-292 with the ability of this compound to reduce ankle swelling in an animal model of arthritis. The modeling studies include the rate of Btk turnover and reveal the vulnerability of Btk to engagement by CC-292.
Co-reporter:Fereidoon Daryaee, Andrew Chang, Johannes Schiebel, Yang Lu, Zhuo Zhang, Kanishk Kapilashrami, Stephen G. Walker, Caroline Kisker, Christoph A. Sotriffer, Stewart L. Fisher and Peter J. Tonge
Chemical Science (2010-Present) 2016 - vol. 7(Issue 9) pp:NaN5954-5954
Publication Date(Web):2016/05/25
DOI:10.1039/C6SC01000H
Drug–target kinetics enable time-dependent changes in target engagement to be quantified as a function of drug concentration. When coupled to drug pharmacokinetics (PK), drug–target kinetics can thus be used to predict in vivo pharmacodynamics (PD). Previously we described a mechanistic PK/PD model that successfully predicted the antibacterial activity of an LpxC inhibitor in a model of Pseudomonas aeruginosa infection. In the present work we demonstrate that the same approach can be used to predict the in vivo activity of an enoyl-ACP reductase (FabI) inhibitor in a model of methicillin-resistant Staphylococcus aureus (MRSA) infection. This is significant because the LpxC inhibitors are cidal, whereas the FabI inhibitors are static. In addition P. aeruginosa is a Gram-negative organism whereas MRSA is Gram-positive. Thus this study supports the general applicability of our modeling approach across antibacterial space.
Benzamide, 4-(4-ethyl-5-fluoro-2-hydroxyphenoxy)-3-fluoro-
1-(2-Fluoro-4-hydroxy-5-methoxyphenyl)ethanone
4-ethyl-5-fluoro-2-methoxyphenol
3,5-HEXADIENOIC ACID, 2-MERCAPTO-2,4-DIMETHYL-, ETHYL ESTER, (2R,3E)-
1,3-Oxathiolan-5-one, 2-(1,1-dimethylethyl)-4-methyl-, (2R,4S)-
3-FLUORO-2-(METHYLSULFANYL)PYRIDINE
2,3-difluoro-l-tyrosine