Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
1.
Cell ; 172(5): 966-978.e12, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29474922

ABSTRACT

Ebola virus nucleoprotein (eNP) assembles into higher-ordered structures that form the viral nucleocapsid (NC) and serve as the scaffold for viral RNA synthesis. However, molecular insights into the NC assembly process are lacking. Using a hybrid approach, we characterized the NC-like assembly of eNP, identified novel regulatory elements, and described how these elements impact function. We generated a three-dimensional structure of the eNP NC-like assembly at 5.8 Å using electron cryo-microscopy and identified a new regulatory role for eNP helices α22-α23. Biochemical, biophysical, and mutational analyses revealed that inter-eNP contacts within α22-α23 are critical for viral NC assembly and regulate viral RNA synthesis. These observations suggest that the N terminus and α22-α23 of eNP function as context-dependent regulatory modules (CDRMs). Our current study provides a framework for a structural mechanism for NC-like assembly and a new therapeutic target.


Subject(s)
Cryoelectron Microscopy , Ebolavirus/physiology , Ebolavirus/ultrastructure , Nucleocapsid/ultrastructure , Nucleoproteins/ultrastructure , Virus Assembly , Models, Biological , Mutant Proteins/chemistry , Mutation/genetics , Nucleoproteins/chemistry , Protein Multimerization , Protein Structure, Secondary , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA, Viral/biosynthesis , RNA, Viral/chemistry , RNA, Viral/metabolism
2.
Proc Natl Acad Sci U S A ; 120(7): e2215371120, 2023 02 14.
Article in English | MEDLINE | ID: mdl-36749730

ABSTRACT

The ε4-allele variant of apolipoprotein E (ApoE4) is the strongest genetic risk factor for Alzheimer's disease, although it only differs from its neutral counterpart ApoE3 by a single amino acid substitution. While ApoE4 influences the formation of plaques and neurofibrillary tangles, the structural determinants of pathogenicity remain undetermined due to limited structural information. Previous studies have led to conflicting models of the C-terminal region positioning with respect to the N-terminal domain across isoforms largely because the data are potentially confounded by the presence of heterogeneous oligomers. Here, we apply a combination of single-molecule spectroscopy and molecular dynamics simulations to construct an atomically detailed model of monomeric ApoE4 and probe the effect of lipid association. Importantly, our approach overcomes previous limitations by allowing us to work at picomolar concentrations where only the monomer is present. Our data reveal that ApoE4 is far more disordered and extended than previously thought and retains significant conformational heterogeneity after binding lipids. Comparing the proximity of the N- and C-terminal domains across the three major isoforms (ApoE4, ApoE3, and ApoE2) suggests that all maintain heterogeneous conformations in their monomeric form, with ApoE2 adopting a slightly more compact ensemble. Overall, these data provide a foundation for understanding how ApoE4 differs from nonpathogenic and protective variants of the protein.


Subject(s)
Apolipoprotein E4 , Apolipoproteins E , Apolipoprotein E4/genetics , Apolipoprotein E3/chemistry , Apolipoprotein E2 , Protein Conformation , Protein Isoforms/metabolism
3.
Proc Natl Acad Sci U S A ; 120(4): e2212694120, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36652481

ABSTRACT

Multidrug-resistant Acinetobacter baumannii infections are an urgent clinical problem and can cause difficult-to-treat nosocomial infections. During such infections, like catheter-associated urinary tract infections (CAUTI), A. baumannii rely on adhesive, extracellular fibers, called chaperone-usher pathway (CUP) pili for critical binding interactions. The A. baumannii uropathogenic strain, UPAB1, and the pan-European subclone II isolate, ACICU, use the CUP pili Abp1 and Abp2 (previously termed Cup and Prp, respectively) in tandem to establish CAUTIs, specifically to facilitate bacterial adherence and biofilm formation on the implanted catheter. Abp1 and Abp2 pili are tipped with two domain tip adhesins, Abp1D and Abp2D, respectively. We discovered that both adhesins bind fibrinogen, a critical host wound response protein that is released into the bladder upon catheterization and is subsequently deposited on the catheter. The crystal structures of the Abp1D and Abp2D receptor-binding domains were determined and revealed that they both contain a large, distally oriented pocket, which mediates binding to fibrinogen and other glycoproteins. Genetic, biochemical, and biophysical studies revealed that interactions with host proteins are governed by several critical residues in and along the edge of the binding pocket, one of which regulates the structural stability of an anterior loop motif. K34, located outside of the pocket but interacting with the anterior loop, also regulates the binding affinity of the protein. This study illuminates the mechanistic basis of the critical fibrinogen-coated catheter colonization step in A. baumannii CAUTI pathogenesis.


Subject(s)
Acinetobacter baumannii , Urinary Tract Infections , Humans , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Urinary Tract Infections/microbiology , Catheters , Acinetobacter baumannii/genetics , Fibrinogen/metabolism
4.
Nat Immunol ; 13(12): 1187-95, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23104097

ABSTRACT

Interleukin 15 (IL-15) and IL-2 have distinct immunological functions even though both signal through the receptor subunit IL-2Rß and the common γ-chain (γ(c)). Here we found that in the structure of the IL-15-IL-15Rα-IL-2Rß-γ(c) quaternary complex, IL-15 binds to IL-2Rß and γ(c) in a heterodimer nearly indistinguishable from that of the IL-2-IL-2Rα-IL-2Rß-γ(c) complex, despite their different receptor-binding chemistries. IL-15Rα substantially increased the affinity of IL-15 for IL-2Rß, and this allostery was required for IL-15 trans signaling. Consistent with their identical IL-2Rß-γ(c) dimer geometries, IL-2 and IL-15 showed similar signaling properties in lymphocytes, with any differences resulting from disparate receptor affinities. Thus, IL-15 and IL-2 induced similar signals, and the cytokine specificity of IL-2Rα versus IL-15Rα determined cellular responsiveness. Our results provide new insights for the development of specific immunotherapeutics based on IL-15 or IL-2.


Subject(s)
Interleukin-15/immunology , Interleukin-2/immunology , Animals , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Humans , Interleukin-15/chemistry , Interleukin-15/metabolism , Interleukin-2/chemistry , Interleukin-2/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-2 Receptor beta Subunit/metabolism , Ligands , Lymphocytes/immunology , Lymphocytes/metabolism , Mice , Models, Molecular , Molecular Dynamics Simulation , Protein Binding , Protein Multimerization , Protein Structure, Quaternary , Signal Transduction
5.
Annu Rev Phys Chem ; 74: 1-27, 2023 04 24.
Article in English | MEDLINE | ID: mdl-36719975

ABSTRACT

Phillip L. Geissler made important contributions to the statistical mechanics of biological polymers, heterogeneous materials, and chemical dynamics in aqueous environments. He devised analytical and computational methods that revealed the underlying organization of complex systems at the frontiers of biology, chemistry, and materials science. In this retrospective we celebrate his work at these frontiers.


Subject(s)
Physics , Male , Humans , Retrospective Studies , Chemistry, Physical
6.
Proc Natl Acad Sci U S A ; 118(47)2021 11 23.
Article in English | MEDLINE | ID: mdl-34799442

ABSTRACT

Understanding the functional role of protein-excited states has important implications in protein design and drug discovery. However, because these states are difficult to find and study, it is still unclear if excited states simply result from thermal fluctuations and generally detract from function or if these states can actually enhance protein function. To investigate this question, we consider excited states in ß-lactamases and particularly a subset of states containing a cryptic pocket which forms under the Ω-loop. Given the known importance of the Ω-loop and the presence of this pocket in at least two homologs, we hypothesized that these excited states enhance enzyme activity. Using thiol-labeling assays to probe Ω-loop pocket dynamics and kinetic assays to probe activity, we find that while this pocket is not completely conserved across ß-lactamase homologs, those with the Ω-loop pocket have a higher activity against the substrate benzylpenicillin. We also find that this is true for TEM ß-lactamase variants with greater open Ω-loop pocket populations. We further investigate the open population using a combination of NMR chemical exchange saturation transfer experiments and molecular dynamics simulations. To test our understanding of the Ω-loop pocket's functional role, we designed mutations to enhance/suppress pocket opening and observed that benzylpenicillin activity is proportional to the probability of pocket opening in our designed variants. The work described here suggests that excited states containing cryptic pockets can be advantageous for function and may be favored by natural selection, increasing the potential utility of such cryptic pockets as drug targets.


Subject(s)
Penicillinase/chemistry , Penicillinase/drug effects , beta-Lactamases/chemistry , beta-Lactamases/pharmacology , Binding Sites , Escherichia coli , Escherichia coli Proteins , Molecular Dynamics Simulation , Mutation , Penicillin G/chemistry , Penicillin G/metabolism , Penicillinase/metabolism , Protein Conformation , Proteins/chemistry , Proteins/genetics , Proteins/metabolism , beta-Lactamases/genetics
7.
Trends Biochem Sci ; 44(4): 351-364, 2019 04.
Article in English | MEDLINE | ID: mdl-30555007

ABSTRACT

A protein is a dynamic shape-shifter whose function is determined by the set of structures it adopts. Unfortunately, atomically detailed structures are only available for a few conformations of any given protein, and these structures have limited explanatory and predictive power. Here, we provide a brief historical perspective on protein dynamics and introduce recent advances in computational and experimental methods that are providing unprecedented access to protein shape-shifting. Next, we focus on how these tools are revealing the mechanism of allosteric communication and features like cryptic pockets; both of which present new therapeutic opportunities. A major theme is the importance of considering the relative probabilities of different structures and the control one can exert over protein function by modulating this balance.


Subject(s)
Computational Biology , Proteins/chemistry , Humans , Protein Conformation , Proteins/metabolism
8.
Biophys J ; 122(14): 2852-2863, 2023 07 25.
Article in English | MEDLINE | ID: mdl-36945779

ABSTRACT

Simulations of biomolecules have enormous potential to inform our understanding of biology but require extremely demanding calculations. For over 20 years, the Folding@home distributed computing project has pioneered a massively parallel approach to biomolecular simulation, harnessing the resources of citizen scientists across the globe. Here, we summarize the scientific and technical advances this perspective has enabled. As the project's name implies, the early years of Folding@home focused on driving advances in our understanding of protein folding by developing statistical methods for capturing long-timescale processes and facilitating insight into complex dynamical processes. Success laid a foundation for broadening the scope of Folding@home to address other functionally relevant conformational changes, such as receptor signaling, enzyme dynamics, and ligand binding. Continued algorithmic advances, hardware developments such as graphics processing unit (GPU)-based computing, and the growing scale of Folding@home have enabled the project to focus on new areas where massively parallel sampling can be impactful. While previous work sought to expand toward larger proteins with slower conformational changes, new work focuses on large-scale comparative studies of different protein sequences and chemical compounds to better understand biology and inform the development of small-molecule drugs. Progress on these fronts enabled the community to pivot quickly in response to the COVID-19 pandemic, expanding to become the world's first exascale computer and deploying this massive resource to provide insight into the inner workings of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus and aid the development of new antivirals. This success provides a glimpse of what is to come as exascale supercomputers come online and as Folding@home continues its work.


Subject(s)
COVID-19 , Citizen Science , Humans , Pandemics , COVID-19/epidemiology , SARS-CoV-2 , Computer Simulation
9.
J Biol Chem ; 298(9): 102355, 2022 09.
Article in English | MEDLINE | ID: mdl-35952758

ABSTRACT

Plasmepsin V (PM V) is a pepsin-like aspartic protease essential for growth of the malarial parasite Plasmodium falciparum. Previous work has shown PM V to be an endoplasmic reticulum-resident protease that processes parasite proteins destined for export into the host cell. Depletion or inhibition of the enzyme is lethal during asexual replication within red blood cells as well as during the formation of sexual stage gametocytes. The structure of the Plasmodium vivax PM V has been characterized by X-ray crystallography, revealing a canonical pepsin fold punctuated by structural features uncommon to secretory aspartic proteases; however, the function of this unique structure is unclear. Here, we used parasite genetics to probe these structural features by attempting to rescue lethal PM V depletion with various mutant enzymes. We found an unusual nepenthesin 1-type insert in the PM V gene to be essential for parasite growth and PM V activity. Mutagenesis of the nepenthesin insert suggests that both its amino acid sequence and one of the two disulfide bonds that undergird its structure are required for the insert's role in PM V function. Furthermore, molecular dynamics simulations paired with Markov state modeling suggest that mutations to the nepenthesin insert may allosterically affect PM V catalysis through multiple mechanisms. Taken together, these data provide further insights into the structure of the P. falciparum PM V protease.


Subject(s)
Malaria, Falciparum , Plasmodium falciparum , Aspartic Acid Endopeptidases/metabolism , Disulfides/metabolism , Humans , Pepsin A/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism
10.
Biophys J ; 120(14): 2880-2889, 2021 07 20.
Article in English | MEDLINE | ID: mdl-33794150

ABSTRACT

Coronaviruses have caused multiple epidemics in the past two decades, in addition to the current COVID-19 pandemic that is severely damaging global health and the economy. Coronaviruses employ between 20 and 30 proteins to carry out their viral replication cycle, including infection, immune evasion, and replication. Among these, nonstructural protein 16 (Nsp16), a 2'-O-methyltransferase, plays an essential role in immune evasion. Nsp16 achieves this by mimicking its human homolog, CMTr1, which methylates mRNA to enhance translation efficiency and distinguish self from other. Unlike human CMTr1, Nsp16 requires a binding partner, Nsp10, to activate its enzymatic activity. The requirement of this binding partner presents two questions that we investigate in this manuscript. First, how does Nsp10 activate Nsp16? Although experimentally derived structures of the active Nsp16/Nsp10 complex exist, structures of inactive, monomeric Nsp16 have yet to be solved. Therefore, it is unclear how Nsp10 activates Nsp16. Using over 1 ms of molecular dynamics simulations of both Nsp16 and its complex with Nsp10, we investigate how the presence of Nsp10 shifts Nsp16's conformational ensemble to activate it. Second, guided by this activation mechanism and Markov state models, we investigate whether Nsp16 adopts inactive structures with cryptic pockets that, if targeted with a small molecule, could inhibit Nsp16 by stabilizing its inactive state. After identifying such a pocket in SARS-CoV2 Nsp16, we show that this cryptic pocket also opens in SARS-CoV1 and MERS but not in human CMTr1. Therefore, it may be possible to develop pan-coronavirus antivirals that target this cryptic pocket.

11.
J Biol Chem ; 295(21): 7376-7390, 2020 05 22.
Article in English | MEDLINE | ID: mdl-32299911

ABSTRACT

CTX-M ß-lactamases are widespread in Gram-negative bacterial pathogens and provide resistance to the cephalosporin cefotaxime but not to the related antibiotic ceftazidime. Nevertheless, variants have emerged that confer resistance to ceftazidime. Two natural mutations, causing P167S and D240G substitutions in the CTX-M enzyme, result in 10-fold increased hydrolysis of ceftazidime. Although the combination of these mutations would be predicted to increase ceftazidime hydrolysis further, the P167S/D240G combination has not been observed in a naturally occurring CTX-M variant. Here, using recombinantly expressed enzymes, minimum inhibitory concentration measurements, steady-state enzyme kinetics, and X-ray crystallography, we show that the P167S/D240G double mutant enzyme exhibits decreased ceftazidime hydrolysis, lower thermostability, and decreased protein expression levels compared with each of the single mutants, indicating negative epistasis. X-ray structures of mutant enzymes with covalently trapped ceftazidime suggested that a change of an active-site Ω-loop to an open conformation accommodates ceftazidime leading to enhanced catalysis. 10-µs molecular dynamics simulations further correlated Ω-loop opening with catalytic activity. We observed that the WT and P167S/D240G variant with acylated ceftazidime both favor a closed conformation not conducive for catalysis. In contrast, the single substitutions dramatically increased the probability of open conformations. We conclude that the antagonism is due to restricting the conformation of the Ω-loop. These results reveal the importance of conformational heterogeneity of active-site loops in controlling catalytic activity and directing evolutionary trajectories.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Evolution, Molecular , Mutation, Missense , beta-Lactam Resistance , beta-Lactamases/chemistry , Amino Acid Substitution , Catalysis , Ceftazidime/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , beta-Lactamases/genetics , beta-Lactamases/metabolism
12.
Acc Chem Res ; 53(3): 654-661, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32134250

ABSTRACT

This Account highlights recent advances and discusses major challenges in investigations of cryptic (hidden) binding sites by molecular simulations. Cryptic binding sites are not visible in protein targets crystallized without a ligand and only become visible crystallographically upon binding events. These sites have been shown to be druggable and might provide a rare opportunity to target difficult proteins. However, due to their hidden nature, they are difficult to find through experimental screening. Computational methods based on atomistic molecular simulations remain one of the best approaches to identify and characterize cryptic binding sites. However, not all methods are equally efficient. Some are more apt at quickly probing protein dynamics but do not provide thermodynamic or druggability information, while others that are able to provide such data are demanding in terms of time and resources. Here, we review the recent contributions of mixed-solvent simulations, metadynamics, Markov state models, and other enhanced sampling methods to the field of cryptic site identification and characterization. We discuss how these methods were able to provide precious information on the nature of the site opening mechanisms, to predict previously unknown sites which were used to design new ligands, and to compute the free energy landscapes and kinetics associated with the opening of the sites and the binding of the ligands. We highlight the potential and the importance of such predictions in drug discovery, especially for difficult ("undruggable") targets. We also discuss the major challenges in the field and their possible solutions.


Subject(s)
Molecular Dynamics Simulation , Binding Sites , Drug Discovery , Ligands , Markov Chains , Solvents/chemistry
13.
Biophys J ; 116(5): 818-830, 2019 03 05.
Article in English | MEDLINE | ID: mdl-30744991

ABSTRACT

Proteins are dynamic molecules that undergo conformational changes to a broad spectrum of different excited states. Unfortunately, the small populations of these states make it difficult to determine their structures or functional implications. Computer simulations are an increasingly powerful means to identify and characterize functionally relevant excited states. However, this advance has uncovered a further challenge: it can be extremely difficult to identify the most salient features of large simulation data sets. We reasoned that many functionally relevant conformational changes are likely to involve large, cooperative changes to the surfaces that are available to interact with potential binding partners. To examine this hypothesis, we introduce a method that returns a prioritized list of potentially functional conformational changes by segmenting protein structures into clusters of residues that undergo cooperative changes in their solvent exposure, along with the hierarchy of interactions between these groups. We term these groups exposons to distinguish them from other types of clusters that arise in this analysis and others. We demonstrate, using three different model systems, that this method identifies experimentally validated and functionally relevant conformational changes, including conformational switches, allosteric coupling, and cryptic pockets. Our results suggest that key functional sites are hubs in the network of exposons. As a further test of the predictive power of this approach, we apply it to discover cryptic allosteric sites in two different ß-lactamase enzymes that are widespread sources of antibiotic resistance. Experimental tests confirm our predictions for both systems. Importantly, we provide the first evidence, to our knowledge, for a cryptic allosteric site in CTX-M-9 ß-lactamase. Experimentally testing this prediction did not require any mutations and revealed that this site exerts the most potent allosteric control over activity of any pockets found in ß-lactamases to date. Discovery of a similar pocket that was previously overlooked in the well-studied TEM-1 ß-lactamase demonstrates the utility of exposons.


Subject(s)
Allosteric Site , Models, Molecular , Proteins/chemistry , Solvents/chemistry , Cyclic AMP Receptor Protein/chemistry , Escherichia coli Proteins/chemistry , Protein Conformation , beta-Lactamases/chemistry
14.
Biochemistry ; 57(1): 117-135, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29039929

ABSTRACT

Tabtoxinine-ß-lactam (TßL), also known as wildfire toxin, is a time- and ATP-dependent inhibitor of glutamine synthetase produced by plant pathogenic strains of Pseudomonas syringae. Here we demonstrate that recombinant glutamine synthetase from Escherichia coli phosphorylates the C3-hydroxyl group of the TßL 3-(S)-hydroxy-ß-lactam (3-HßL) warhead. Phosphorylation of TßL generates a stable, noncovalent enzyme-ADP-inhibitor complex that resembles the glutamine synthetase tetrahedral transition state. The TßL ß-lactam ring remains intact during enzyme inhibition, making TßL mechanistically distinct from traditional ß-lactam antibiotics such as penicillin. Our findings could enable the design of new 3-HßL transition state inhibitors targeting enzymes in the ATP-dependent carboxylate-amine ligase superfamily with broad therapeutic potential in many disease areas.


Subject(s)
Adenosine Triphosphate/metabolism , Azetidines/pharmacology , Bacterial Toxins/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Escherichia coli/enzymology , Glutamate-Ammonia Ligase/antagonists & inhibitors , Azetidines/isolation & purification , Azetidines/metabolism , Bacterial Toxins/biosynthesis , Bacterial Toxins/isolation & purification , Catalysis , Chromatography, Liquid , Escherichia coli/drug effects , Escherichia coli/growth & development , Mass Spectrometry , Microbial Sensitivity Tests , Nuclear Magnetic Resonance, Biomolecular , Phosphorylation , Pseudomonas syringae/metabolism
15.
Nature ; 484(7395): 529-33, 2012 Mar 25.
Article in English | MEDLINE | ID: mdl-22446627

ABSTRACT

The immunostimulatory cytokine interleukin-2 (IL-2) is a growth factor for a wide range of leukocytes, including T cells and natural killer (NK) cells. Considerable effort has been invested in using IL-2 as a therapeutic agent for a variety of immune disorders ranging from AIDS to cancer. However, adverse effects have limited its use in the clinic. On activated T cells, IL-2 signals through a quaternary 'high affinity' receptor complex consisting of IL-2, IL-2Rα (termed CD25), IL-2Rß and IL-2Rγ. Naive T cells express only a low density of IL-2Rß and IL-2Rγ, and are therefore relatively insensitive to IL-2, but acquire sensitivity after CD25 expression, which captures the cytokine and presents it to IL-2Rß and IL-2Rγ. Here, using in vitro evolution, we eliminated the functional requirement of IL-2 for CD25 expression by engineering an IL-2 'superkine' (also called super-2) with increased binding affinity for IL-2Rß. Crystal structures of the IL-2 superkine in free and receptor-bound forms showed that the evolved mutations are principally in the core of the cytokine, and molecular dynamics simulations indicated that the evolved mutations stabilized IL-2, reducing the flexibility of a helix in the IL-2Rß binding site, into an optimized receptor-binding conformation resembling that when bound to CD25. The evolved mutations in the IL-2 superkine recapitulated the functional role of CD25 by eliciting potent phosphorylation of STAT5 and vigorous proliferation of T cells irrespective of CD25 expression. Compared to IL-2, the IL-2 superkine induced superior expansion of cytotoxic T cells, leading to improved antitumour responses in vivo, and elicited proportionally less expansion of T regulatory cells and reduced pulmonary oedema. Collectively, we show that in vitro evolution has mimicked the functional role of CD25 in enhancing IL-2 potency and regulating target cell specificity, which has implications for immunotherapy.


Subject(s)
Directed Molecular Evolution , Interleukin-2/chemistry , Interleukin-2/immunology , Mutant Proteins/chemistry , Mutant Proteins/immunology , Protein Engineering , Animals , Binding Sites , Cell Line , Cell Proliferation , Crystallography, X-Ray , Humans , Immunotherapy , Interleukin-2/genetics , Interleukin-2/pharmacology , Interleukin-2 Receptor alpha Subunit/chemistry , Interleukin-2 Receptor alpha Subunit/deficiency , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-2 Receptor beta Subunit/chemistry , Interleukin-2 Receptor beta Subunit/metabolism , Killer Cells, Natural/immunology , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Dynamics Simulation , Mutant Proteins/genetics , Mutant Proteins/pharmacology , Mutation , Neoplasm Transplantation , Neoplasms/drug therapy , Neoplasms/immunology , Phosphorylation , Protein Conformation , STAT5 Transcription Factor/metabolism , Surface Plasmon Resonance , T-Lymphocytes/cytology , T-Lymphocytes/immunology
16.
Proc Natl Acad Sci U S A ; 112(9): 2734-9, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25730859

ABSTRACT

The discovery of drug-like molecules that bind pockets in proteins that are not present in crystallographic structures yet exert allosteric control over activity has generated great interest in designing pharmaceuticals that exploit allosteric effects. However, there have only been a small number of successes, so the therapeutic potential of these pockets--called hidden allosteric sites--remains unclear. One challenge for assessing their utility is that rational drug design approaches require foreknowledge of the target site, but most hidden allosteric sites are only discovered when a small molecule is found to stabilize them. We present a means of decoupling the identification of hidden allosteric sites from the discovery of drugs that bind them by drawing on new developments in Markov state modeling that provide unprecedented access to microsecond- to millisecond-timescale fluctuations of a protein's structure. Visualizing these fluctuations allows us to identify potential hidden allosteric sites, which we then test via thiol labeling experiments. Application of these methods reveals multiple hidden allosteric sites in an important antibiotic target--TEM-1 ß-lactamase. This result supports the hypothesis that there are many as yet undiscovered hidden allosteric sites and suggests our methodology can identify such sites, providing a starting point for future drug design efforts. More generally, our results demonstrate the power of using Markov state models to guide experiments.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli/enzymology , Models, Chemical , Models, Molecular , beta-Lactamases/chemistry , Allosteric Site , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Markov Chains , beta-Lactamases/genetics
17.
Chemistry ; 23(19): 4615-4624, 2017 Apr 03.
Article in English | MEDLINE | ID: mdl-28182309

ABSTRACT

Allosteric sodium in the helix bundle of a G protein-coupled receptor (GPCR) can modulate the receptor activation on the intracellular side. This phenomenon has confounded the GPCR community for decades. In this work, we present a theoretical model that reveals the mechanism of the allosteric modulation induced by sodium in the δ-opioid receptor. We found that the allosteric sodium ion exploits a distinct conformation of the key residue Trp2746.48 to propagate the modulation to helices 5 and 6, which further transmits along the helices and regulates their positions on the intracellular side. This mechanism is supported by subsequent functional assays. Remarkably, our results highlight the contrast between the allosteric effects towards two GPCR partners, the G protein and ß-arrestin, as indicated by the fact that the allosteric modulation initiated by the sodium ion significantly affects the ß-arrestin recruitment, while it alters the G protein signaling only moderately. We believe that the mechanism revealed in this work can be used to explain allosteric effects initiated by sodium in other GPCRs since the allosteric sodium is highly conserved across GPCRs.


Subject(s)
Receptors, Opioid, delta/metabolism , Sodium/metabolism , Allosteric Regulation , Allosteric Site , Humans , Molecular Dynamics Simulation , Protein Structure, Tertiary , Receptors, Opioid, delta/chemistry , Sodium/chemistry , Thermodynamics
18.
J Comput Chem ; 37(6): 558-66, 2016 Mar 05.
Article in English | MEDLINE | ID: mdl-26077712

ABSTRACT

Advances in hardware and algorithms have greatly extended the timescales accessible to molecular simulation. This article assesses whether such long timescale simulations improve our ability to calculate order parameters that describe conformational heterogeneity on ps-ns timescales or if force fields are now a limiting factor. Order parameters from experiment are compared with order parameters calculated in three different ways from simulations ranging from 10 ns to 100 µs in length. Importantly, bootstrapping is employed to assess the variability in results for each simulation length. The results of 10-100 ns timescale simulations are highly variable, possibly explaining the variation in levels of agreement between simulation and experiment in published works examining different proteins. Fortunately, microsecond timescale simulations improve both the accuracy and precision of calculated order parameters, at least so long as the full exponential fit or truncated average approximation is used instead of the common long-time limit approximation. The improved precision of these long timescale simulations allows a statistically sound comparison of a number of modern force fields, such as Amber03, Amber99sb-ILDN, and Charmm27. While there is some variation between these force fields, they generally give very similar results for sufficiently long simulations. The fact that so much simulation is required to precisely capture ps-ns timescale processes suggests that extremely extensive simulations are required for slower processes. Advanced sampling techniques could aid greatly, however, such methods will need to maintain accurate kinetics if they are to be of value for calculating dynamical properties like order parameters. © 2015 Wiley Periodicals, Inc.


Subject(s)
Molecular Dynamics Simulation , Proteins/chemistry , Algorithms , Escherichia coli/chemistry , Escherichia coli/enzymology , Humans , Protein Conformation , Ribonucleases/chemistry , Time Factors , Ubiquitin/chemistry
19.
Acc Chem Res ; 48(4): 1098-105, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25688669

ABSTRACT

Folded protein structures are both stable and dynamic. Historically, our clearest window into these structures came from X-ray crystallography, which generally provided a static image of each protein's singular "folded state", highlighting its stability. Deviations away from that crystallographic structure were difficult to quantify, and as a result, their potential functional consequences were often neglected. However, several dynamical and statistical studies now highlight the structural variability that is present within the protein's folded state. Here we review mounting evidence of the importance of these structural rearrangements; both experiment and computation indicate that folded proteins undergo substantial fluctuations that can greatly influence their function. Crucially, recent studies have shown that structural elements of proteins, especially their side-chain degrees of freedom, fluctuate in ways that generate significant conformational heterogeneity. The entropy associated with these motions contributes to the folded structure's thermodynamic stability. In addition, since these fluctuations can shift in response to perturbations such as ligand binding, they may play an important role in the protein's capacity to respond to environmental cues. In one compelling example, the entropy associated with side-chain fluctuations contributes significantly to regulating the binding of calmodulin to a set of peptide ligands. The neglect of fluctuations within proteins' native states was often justified by the dense packing within folded proteins, which has inspired comparisons with crystalline solids. Many liquids, however, can achieve similarly dense packing yet fluidity is maintained through correlated molecular motions. Indeed, the studies we discuss favor comparison of folded proteins not with solids but instead with dense liquids, where the internal side chain fluidity is facilitated by collective motions that are correlated over long distances. These correlated rearrangements can enable allosteric communication between different parts of a protein, through subtle and varied channels. Such long-range correlations appear to be an innate feature of proteins in general, manifest even in molecules lacking known allosteric regulators and arising robustly from the physical nature of their internal environment. Given their ubiquity, it is only to be expected that, over time, nature has refined some subset of these correlated motions and put them to use. Native state fluctuations increasingly appear to be vital for proteins' natural functions. Understanding the diversity, origin, and range of these rearrangements may provide novel routes for rationally manipulating biomolecular activity.


Subject(s)
Proteins/chemistry , Proteins/metabolism , Thermodynamics , Allosteric Regulation , Crystallography, X-Ray , Models, Molecular , Protein Conformation , Protein Folding
20.
Proc Natl Acad Sci U S A ; 109(29): 11681-6, 2012 Jul 17.
Article in English | MEDLINE | ID: mdl-22753506

ABSTRACT

Cryptic allosteric sites--transient pockets in a folded protein that are invisible to conventional experiments but can alter enzymatic activity via allosteric communication with the active site--are a promising opportunity for facilitating drug design by greatly expanding the repertoire of available drug targets. Unfortunately, identifying these sites is difficult, typically requiring resource-intensive screening of large libraries of small molecules. Here, we demonstrate that Markov state models built from extensive computer simulations (totaling hundreds of microseconds of dynamics) can identify prospective cryptic sites from the equilibrium fluctuations of three medically relevant proteins--ß-lactamase, interleukin-2, and RNase H--even in the absence of any ligand. As in previous studies, our methods reveal a surprising variety of conformations--including bound-like configurations--that implies a role for conformational selection in ligand binding. Moreover, our analyses lead to a number of unique insights. First, direct comparison of simulations with and without the ligand reveals that there is still an important role for an induced fit during ligand binding to cryptic sites and suggests new conformations for docking. Second, correlations between amino acid sidechains can convey allosteric signals even in the absence of substantial backbone motions. Most importantly, our extensive sampling reveals a multitude of potential cryptic sites--consisting of transient pockets coupled to the active site--even in a single protein. Based on these observations, we propose that cryptic allosteric sites may be even more ubiquitous than previously thought and that our methods should be a valuable means of guiding the search for such sites.


Subject(s)
Allosteric Site/genetics , Interleukin-2/chemistry , Models, Chemical , Ribonuclease H/chemistry , beta-Lactamases/chemistry , Molecular Dynamics Simulation , Protein Binding , Protein Folding
SELECTION OF CITATIONS
SEARCH DETAIL