Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
1.
Nature ; 604(7905): 330-336, 2022 04.
Article in English | MEDLINE | ID: mdl-35172323

ABSTRACT

The animal reservoir of SARS-CoV-2 is unknown despite reports of SARS-CoV-2-related viruses in Asian Rhinolophus bats1-4, including the closest virus from R. affinis, RaTG13 (refs. 5,6), and pangolins7-9. SARS-CoV-2 has a mosaic genome, to which different progenitors contribute. The spike sequence determines the binding affinity and accessibility of its receptor-binding domain to the cellular angiotensin-converting enzyme 2 (ACE2) receptor and is responsible for host range10-12. SARS-CoV-2 progenitor bat viruses genetically close to SARS-CoV-2 and able to enter human cells through a human ACE2 (hACE2) pathway have not yet been identified, although they would be key in understanding the origin of the epidemic. Here we show that such viruses circulate in cave bats living in the limestone karstic terrain in northern Laos, in the Indochinese peninsula. We found that the receptor-binding domains of these viruses differ from that of SARS-CoV-2 by only one or two residues at the interface with ACE2, bind more efficiently to the hACE2 protein than that of the SARS-CoV-2 strain isolated in Wuhan from early human cases, and mediate hACE2-dependent entry and replication in human cells, which is inhibited by antibodies that neutralize SARS-CoV-2. None of these bat viruses contains a furin cleavage site in the spike protein. Our findings therefore indicate that bat-borne SARS-CoV-2-like viruses that are potentially infectious for humans circulate in Rhinolophus spp. in the Indochinese peninsula.


Subject(s)
COVID-19 , Chiroptera , Angiotensin-Converting Enzyme 2 , Animals , Asia , Caves , Chiroptera/virology , Disease Reservoirs , Humans , Protein Binding , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/chemistry
2.
PLoS Comput Biol ; 20(7): e1012180, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39008528

ABSTRACT

Converting cryo-electron microscopy (cryo-EM) data into high-quality structural models is a challenging problem of outstanding importance. Current refinement methods often generate unbalanced models in which physico-chemical quality is sacrificed for excellent fit to the data. Furthermore, these techniques struggle to represent the conformational heterogeneity averaged out in low-resolution regions of density maps. Here we introduce EMMIVox, a Bayesian inference approach to determine single-structure models as well as structural ensembles from cryo-EM maps. EMMIVox automatically balances experimental information with accurate physico-chemical models of the system and the surrounding environment, including waters, lipids, and ions. Explicit treatment of data correlation and noise as well as inference of accurate B-factors enable determination of structural models and ensembles with both excellent fit to the data and high stereochemical quality, thus outperforming state-of-the-art refinement techniques. EMMIVox represents a flexible approach to determine high-quality structural models that will contribute to advancing our understanding of the molecular mechanisms underlying biological functions.


Subject(s)
Bayes Theorem , Cryoelectron Microscopy , Models, Molecular , Cryoelectron Microscopy/methods , Computational Biology/methods , Protein Conformation , Algorithms , Proteins/chemistry , Proteins/ultrastructure
3.
EMBO Rep ; 24(4): e56055, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36876574

ABSTRACT

Bat sarbecovirus BANAL-236 is highly related to SARS-CoV-2 and infects human cells, albeit lacking the furin cleavage site in its spike protein. BANAL-236 replicates efficiently and pauci-symptomatically in humanized mice and in macaques, where its tropism is enteric, strongly differing from that of SARS-CoV-2. BANAL-236 infection leads to protection against superinfection by a virulent strain. We find no evidence of antibodies recognizing bat sarbecoviruses in populations in close contact with bats in which the virus was identified, indicating that such spillover infections, if they occur, are rare. Six passages in humanized mice or in human intestinal cells, mimicking putative early spillover events, select adaptive mutations without appearance of a furin cleavage site and no change in virulence. Therefore, acquisition of a furin site in the spike protein is likely a pre-spillover event that did not occur upon replication of a SARS-CoV-2-like bat virus in humans or other animals. Other hypotheses regarding the origin of the SARS-CoV-2 should therefore be evaluated, including the presence of sarbecoviruses carrying a spike with a furin cleavage site in bats.


Subject(s)
COVID-19 , Humans , Animals , Mice , SARS-CoV-2 , Furin/genetics , Furin/metabolism , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism , Mutation
4.
Proc Natl Acad Sci U S A ; 118(37)2021 09 14.
Article in English | MEDLINE | ID: mdl-34507995

ABSTRACT

ASCT2 (SLC1A5) is a sodium-dependent neutral amino acid transporter that controls amino acid homeostasis in peripheral tissues. In cancer, ASCT2 is up-regulated where it modulates intracellular glutamine levels, fueling cell proliferation. Nutrient deprivation via ASCT2 inhibition provides a potential strategy for cancer therapy. Here, we rationally designed stereospecific inhibitors exploiting specific subpockets in the substrate binding site using computational modeling and cryo-electron microscopy (cryo-EM). The final structures combined with molecular dynamics simulations reveal multiple pharmacologically relevant conformations in the ASCT2 binding site as well as a previously unknown mechanism of stereospecific inhibition. Furthermore, this integrated analysis guided the design of a series of unique ASCT2 inhibitors. Our results provide a framework for future development of cancer therapeutics targeting nutrient transport via ASCT2, as well as demonstrate the utility of combining computational modeling and cryo-EM for solute carrier ligand discovery.


Subject(s)
Amino Acid Transport System ASC/antagonists & inhibitors , Binding, Competitive , Computational Chemistry , Cryoelectron Microscopy/methods , Glutamine/metabolism , Pharmaceutical Preparations/administration & dosage , Amino Acid Transport System ASC/metabolism , Binding Sites , Drug Design , Humans , Minor Histocompatibility Antigens/metabolism , Molecular Docking Simulation , Pharmaceutical Preparations/chemistry , Protein Binding , Protein Domains , Protein Structure, Tertiary , Structure-Activity Relationship
5.
Biochemistry ; 62(16): 2407-2416, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37477459

ABSTRACT

In recent years, major advances in cryo-electron microscopy (cryo-EM) have enabled the routine determination of complex biomolecular structures at atomistic resolution. An open challenge for this approach, however, concerns large systems that exhibit continuous dynamics. To address this problem, we developed the metadynamic electron microscopy metainference (MEMMI) method, which incorporates metadynamics, an enhanced conformational sampling approach, into the metainference method of integrative structural biology. MEMMI enables the simultaneous determination of the structure and dynamics of large heterogeneous systems by combining cryo-EM density maps with prior information through molecular dynamics, while at the same time modeling the different sources of error. To illustrate the method, we apply it to elucidate the dynamics of an amyloid fibril of the islet amyloid polypeptide (IAPP). The resulting conformational ensemble provides an accurate description of the structural variability of the disordered region of the amyloid fibril, known as fuzzy coat. The conformational ensemble also reveals that in nearly half of the structural core of this amyloid fibril, the side chains exhibit liquid-like dynamics despite the presence of the highly ordered network backbone of hydrogen bonds characteristic of the cross-ß structure of amyloid fibrils.


Subject(s)
Amyloid , Islet Amyloid Polypeptide , Cryoelectron Microscopy , Islet Amyloid Polypeptide/chemistry , Amyloid/chemistry , Molecular Dynamics Simulation , Microscopy, Electron
7.
Biophys J ; 121(23): 4476-4491, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36369754

ABSTRACT

The human L-type amino acid transporter 1 (LAT1; SLC7A5) is a membrane transporter of amino acids, thyroid hormones, and drugs such as the Parkinson's disease drug levodopa (L-Dopa). LAT1 is found in the blood-brain barrier, testis, bone marrow, and placenta, and its dysregulation has been associated with various neurological diseases, such as autism and epilepsy, as well as cancer. In this study, we combine metainference molecular dynamics simulations, molecular docking, and experimental testing, to characterize LAT1-inhibitor interactions. We first conducted a series of molecular docking experiments to identify the most relevant interactions between LAT1's substrate-binding site and ligands, including both inhibitors and substrates. We then performed metainference molecular dynamics simulations using cryoelectron microscopy structures in different conformations of LAT1 with the electron density map as a spatial restraint, to explore the inherent heterogeneity in the structures. We analyzed the LAT1 substrate-binding site to map important LAT1-ligand interactions as well as newly described druggable pockets. Finally, this analysis guided the discovery of previously unknown LAT1 ligands using virtual screening and cellular uptake experiments. Our results improve our understanding of LAT1-inhibitor recognition, providing a framework for rational design of future lead compounds targeting this key drug target.


Subject(s)
Amino Acid Transport Systems , Humans , Molecular Docking Simulation , Cryoelectron Microscopy
8.
Proc Natl Acad Sci U S A ; 116(21): 10366-10371, 2019 05 21.
Article in English | MEDLINE | ID: mdl-31072936

ABSTRACT

Acetylation of K40 in α-tubulin is the sole posttranslational modification to mark the luminal surface of microtubules. It is still controversial whether its relationship with microtubule stabilization is correlative or causative. We have obtained high-resolution cryo-electron microscopy (cryo-EM) reconstructions of pure samples of αTAT1-acetylated and SIRT2-deacetylated microtubules to visualize the structural consequences of this modification and reveal its potential for influencing the larger assembly properties of microtubules. We modeled the conformational ensembles of the unmodified and acetylated states by using the experimental cryo-EM density as a structural restraint in molecular dynamics simulations. We found that acetylation alters the conformational landscape of the flexible loop that contains αK40. Modification of αK40 reduces the disorder of the loop and restricts the states that it samples. We propose that the change in conformational sampling that we describe, at a location very close to the lateral contacts site, is likely to affect microtubule stability and function.


Subject(s)
Microtubules/metabolism , Tubulin/metabolism , Acetylation , Animals , Cryoelectron Microscopy/methods , Protein Processing, Post-Translational/physiology , Swine
9.
J Chem Phys ; 154(17): 174108, 2021 May 07.
Article in English | MEDLINE | ID: mdl-34241080

ABSTRACT

In recent years, π-conjugated polymers are attracting considerable interest in view of their light-dependent torsional reorganization around the π-conjugated backbone, which determines peculiar light-emitting properties. Motivated by the interest in designing conjugated polymers with tunable photoswitchable pathways, we devised a computational framework to enhance the sampling of the torsional conformational space and, at the same time, estimate ground- to excited-state free-energy differences. This scheme is based on a combination of Hamiltonian Replica Exchange Method (REM), parallel bias metadynamics, and free-energy perturbation theory. In our scheme, each REM samples an intermediate unphysical state between the ground and the first two excited states, which are characterized by time-dependent density functional theory simulations at the B3LYP/6-31G* level of theory. We applied the method to a 5-mer of 9,9-dioctylfluorene and found that upon irradiation, this system can undergo a dihedral inversion from -155° to 155°, crossing a barrier that decreases from 0.1 eV in the ground state (S0) to 0.05 eV and 0.04 eV in the first (S1) and second (S2) excited states. Furthermore, S1 and even more S2 were predicted to stabilize coplanar dihedrals, with a local free-energy minimum located at ±44°. The presence of a free-energy barrier of 0.08 eV for the S1 state and 0.12 eV for the S2 state can trap this conformation in a basin far from the global free-energy minimum located at 155°. The simulation results were compared with the experimental emission spectrum, showing a quantitative agreement with the predictions provided by our framework.

10.
Proc Natl Acad Sci U S A ; 115(28): E6447-E6456, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29941580

ABSTRACT

Protein homeostasis is critically important for cell viability. Key to this process is the refolding of misfolded or aggregated proteins by molecular chaperones or, alternatively, their degradation by proteases. In most prokaryotes and in chloroplasts and mitochondria, protein degradation is performed by the caseinolytic protease ClpP, a tetradecamer barrel-like proteolytic complex. Dysregulating ClpP function has shown promise in fighting antibiotic resistance and as a potential therapy for acute myeloid leukemia. Here we use methyl-transverse relaxation-optimized spectroscopy (TROSY)-based NMR, cryo-EM, biochemical assays, and molecular dynamics simulations to characterize the structural dynamics of ClpP from Staphylococcus aureus (SaClpP) in wild-type and mutant forms in an effort to discover conformational hotspots that regulate its function. Wild-type SaClpP was found exclusively in the active extended form, with the N-terminal domains of its component protomers in predominantly ß-hairpin conformations that are less well-defined than other regions of the protein. A hydrophobic site was identified that, upon mutation, leads to unfolding of the N-terminal domains, loss of SaClpP activity, and formation of a previously unobserved split-ring conformation with a pair of 20-Å-wide pores in the side of the complex. The extended form of the structure and partial activity can be restored via binding of ADEP small-molecule activators. The observed structural plasticity of the N-terminal gates is shown to be a conserved feature through studies of Escherichia coli and Neisseria meningitidis ClpP, suggesting a potential avenue for the development of molecules to allosterically modulate the function of ClpP.


Subject(s)
Bacterial Proteins/chemistry , Endopeptidase Clp/chemistry , Molecular Dynamics Simulation , Staphylococcus aureus/enzymology , Hydrophobic and Hydrophilic Interactions , Protein Domains
11.
J Am Chem Soc ; 141(5): 1998-2011, 2019 02 06.
Article in English | MEDLINE | ID: mdl-30618247

ABSTRACT

Elucidation of the structure and interactions of proteins at native mineral interfaces is key to understanding how biological systems regulate the formation of hard tissue structures. In addition, understanding how these same proteins interact with non-native mineral surfaces has important implications for the design of medical and dental implants, chromatographic supports, diagnostic tools, and a host of other applications. Here, we combine solid-state NMR spectroscopy, isotherm measurements, and molecular dynamics simulations to study how SNa15, a peptide derived from the hydroxyapatite (HAP) recognition domain of the biomineralization protein statherin, interacts with HAP, silica (SiO2), and titania (TiO2) mineral surfaces. Adsorption isotherms are used to characterize the binding affinity of SNa15 to HAP, SiO2, and TiO2. We also apply 1D 13C CP MAS, 1D 15N CP MAS, and 2D 13C-13C DARR experiments to SNa15 samples with uniformly 13C- and 15N-enriched residues to determine backbone and side-chain chemical shifts. Different computational tools, namely TALOS-N and molecular dynamics simulations, are used to deduce secondary structure from backbone and side-chain chemical shift data. Our results show that SNa15 adopts an α-helical conformation when adsorbed to HAP and TiO2, but the helix largely unravels upon adsorption to SiO2. Interactions with HAP are mediated in general by acidic and some basic amino acids, although the specific amino acids involved in direct surface interaction vary with surface. The integrated experimental and computational approach used in this study is able to provide high-resolution insights into adsorption of proteins on interfaces.


Subject(s)
Durapatite/chemistry , Molecular Dynamics Simulation , Nuclear Magnetic Resonance, Biomolecular , Salivary Proteins and Peptides/chemistry , Silicon Dioxide/chemistry , Titanium/chemistry , Humans , Mutation , Protein Conformation , Salivary Proteins and Peptides/genetics
12.
Nature ; 499(7456): 107-10, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23685453

ABSTRACT

Eukaryotic Ca(2+) regulation involves sequestration into intracellular organelles, and expeditious Ca(2+) release into the cytosol is a hallmark of key signalling transduction pathways. Bulk removal of Ca(2+) after such signalling events is accomplished by members of the Ca(2+):cation (CaCA) superfamily. The CaCA superfamily includes the Na(+)/Ca(2+) (NCX) and Ca(2+)/H(+) (CAX) antiporters, and in mammals the NCX and related proteins constitute families SLC8 and SLC24, and are responsible for the re-establishment of Ca(2+) resting potential in muscle cells, neuronal signalling and Ca(2+) reabsorption in the kidney. The CAX family members maintain cytosolic Ca(2+) homeostasis in plants and fungi during steep rises in intracellular Ca(2+) due to environmental changes, or following signal transduction caused by events such as hyperosmotic shock, hormone response and response to mating pheromones. The cytosol-facing conformations within the CaCA superfamily are unknown, and the transport mechanism remains speculative. Here we determine a crystal structure of the Saccharomyces cerevisiae vacuolar Ca(2+)/H(+) exchanger (Vcx1) at 2.3 Å resolution in a cytosol-facing, substrate-bound conformation. Vcx1 is the first structure, to our knowledge, within the CAX family, and it describes the key cytosol-facing conformation of the CaCA superfamily, providing the structural basis for a novel alternating access mechanism by which the CaCA superfamily performs high-throughput Ca(2+) transport across membranes.


Subject(s)
Antiporters/chemistry , Antiporters/metabolism , Calcium/metabolism , Cytosol/metabolism , Protons , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/chemistry , Amino Acid Sequence , Binding Sites , Crystallography, X-Ray , Ion Transport , Methanococcus/chemistry , Models, Molecular , Molecular Sequence Data , Protein Conformation , Protein Folding , Structure-Activity Relationship
13.
Nature ; 496(7446): 533-6, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23542591

ABSTRACT

Phosphate is crucial for structural and metabolic needs, including nucleotide and lipid synthesis, signalling and chemical energy storage. Proton-coupled transporters of the major facilitator superfamily (MFS) are essential for phosphate uptake in plants and fungi, and also have a function in sensing external phosphate levels as transceptors. Here we report the 2.9 Å structure of a fungal (Piriformospora indica) high-affinity phosphate transporter, PiPT, in an inward-facing occluded state, with bound phosphate visible in the membrane-buried binding site. The structure indicates both proton and phosphate exit pathways and suggests a modified asymmetrical 'rocker-switch' mechanism of phosphate transport. PiPT is related to several human transporter families, most notably the organic cation and anion transporters of the solute carrier family (SLC22), which are implicated in cancer-drug resistance. We modelled representative cation and anion SLC22 transporters based on the PiPT structure to surmise the structural basis for substrate binding and charge selectivity in this important family. The PiPT structure demonstrates and expands on principles of substrate transport by the MFS transporters and illuminates principles of phosphate uptake in particular.


Subject(s)
Basidiomycota/chemistry , Eukaryotic Cells/chemistry , Phosphate Transport Proteins/chemistry , Binding Sites , Crystallography, X-Ray , Humans , Models, Biological , Models, Molecular , Phosphate Transport Proteins/metabolism , Phosphates/metabolism , Protein Conformation , Protons , Structure-Activity Relationship
14.
Biophys J ; 114(7): 1604-1613, 2018 04 10.
Article in English | MEDLINE | ID: mdl-29642030

ABSTRACT

Cryo-electron microscopy is rapidly emerging as a powerful technique to determine the structures of complex macromolecular systems elusive to other techniques. Because many of these systems are highly dynamical, characterizing their movements is also a crucial step to unravel their biological functions. To achieve this goal, we report an integrative modeling approach to simultaneously determine structure and dynamics of macromolecular systems from cryo-electron microscopy density maps. By quantifying the level of noise in the data and dealing with their ensemble-averaged nature, this approach enables the integration of multiple sources of information to model ensembles of structures and infer their populations. We illustrate the method by characterizing structure and dynamics of the integral membrane receptor STRA6, thus providing insights into the mechanisms by which it interacts with retinol binding protein and translocates retinol across the membrane.


Subject(s)
Cryoelectron Microscopy , Proteins/chemistry , Proteins/metabolism , Cell Membrane/metabolism , Molecular Dynamics Simulation , Protein Conformation , Thermodynamics , Time Factors
15.
Bioinformatics ; 33(24): 3999-4000, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-28961689

ABSTRACT

SUMMARY: Accurate structural models of biological systems can be obtained by properly combining experimental data with a priori physico-chemical knowledge. Here we present PLUMED-ISDB, an open-source, freely-available module of the popular PLUMED library, which enables the simultaneous determination of structure and dynamics of conformationally heterogeneous systems by integrating experimental data with a priori information. This integration is achieved using metainference, a general Bayesian framework that accounts for both noise in the data and their ensemble-averaged nature. PLUMED-ISDB implements different types of experimental data, such as several NMR observables, FRET, SAXS and cryo-electron microscopy data, and enables modelling structure and dynamics of individual proteins, protein complexes, membrane proteins, RNA and DNA, using a variety of enhanced sampling methods and resolutions of the system. AVAILABILITY AND IMPLEMENTATION: PLUMED-ISDB is freely available at www.plumed.org. CONTACT: mb2006@cam.ac.uk or carlo.camilloni@unimi.it. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Models, Molecular , Software , Bayes Theorem , Molecular Conformation
16.
PLoS Comput Biol ; 11(10): e1004477, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26444490

ABSTRACT

The Alanine-Serine-Cysteine transporter ASCT2 (SLC1A5) is a membrane protein that transports neutral amino acids into cells in exchange for outward movement of intracellular amino acids. ASCT2 is highly expressed in peripheral tissues such as the lung and intestines where it contributes to the homeostasis of intracellular concentrations of neutral amino acids. ASCT2 also plays an important role in the development of a variety of cancers such as melanoma by transporting amino acid nutrients such as glutamine into the proliferating tumors. Therefore, ASCT2 is a key drug target with potentially great pharmacological importance. Here, we identify seven ASCT2 ligands by computational modeling and experimental testing. In particular, we construct homology models based on crystallographic structures of the aspartate transporter GltPh in two different conformations. Optimization of the models' binding sites for protein-ligand complementarity reveals new putative pockets that can be targeted via structure-based drug design. Virtual screening of drugs, metabolites, fragments-like, and lead-like molecules from the ZINC database, followed by experimental testing of 14 top hits with functional measurements using electrophysiological methods reveals seven ligands, including five activators and two inhibitors. For example, aminooxetane-3-carboxylate is a more efficient activator than any other known ASCT2 natural or unnatural substrate. Furthermore, two of the hits inhibited ASCT2 mediated glutamine uptake and proliferation of a melanoma cancer cell line. Our results improve our understanding of how substrate specificity is determined in amino acid transporters, as well as provide novel scaffolds for developing chemical tools targeting ASCT2, an emerging therapeutic target for cancer and neurological disorders.


Subject(s)
Amino Acid Transport System ASC/chemistry , Amino Acid Transport System ASC/ultrastructure , Drug Evaluation, Preclinical/methods , Models, Chemical , Molecular Docking Simulation , Protein Interaction Mapping/methods , Algorithms , Amino Acid Sequence , Binding Sites , Minor Histocompatibility Antigens , Molecular Sequence Data , Protein Binding , Sequence Analysis, Protein/methods , Sequence Homology, Amino Acid
17.
Phys Chem Chem Phys ; 18(13): 9036-41, 2016 Apr 07.
Article in English | MEDLINE | ID: mdl-26967312

ABSTRACT

Exploring the free energy landscapes of metal cations on phospholipid membrane surfaces is important for the understanding of chemical and biological processes in cellular environments. Using metadynamics simulations we have performed systematic free energy calculations of sodium cations bound to DMPC phospholipid membranes with cholesterol concentration varying between 0% (cholesterol-free) and 50% (cholesterol-rich) at infinite dilution. The resulting free energy landscapes reveal the competition between binding of sodium to water and to lipid head groups. Moreover, the binding competitiveness of lipid head groups is diminished by cholesterol contents. As cholesterol concentration increases, the ionic affinity to membranes decreases. When cholesterol concentration is greater than 30%, the ionic binding is significantly reduced, which coincides with the phase transition point of DMPC-cholesterol membranes from a liquid-disordered phase to a liquid-ordered phase. We have also evaluated the contributions of different lipid head groups to the binding free energy separately. The DMPC's carbonyl group is the most favorable binding site for sodium, followed by DMPC's phosphate group and then the hydroxyl group of cholesterol.


Subject(s)
Cholesterol/chemistry , Dimyristoylphosphatidylcholine/chemistry , Sodium/chemistry
18.
Mol Cell Proteomics ; 13(11): 2812-23, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25139910

ABSTRACT

The use of in vivo Förster resonance energy transfer (FRET) data to determine the molecular architecture of a protein complex in living cells is challenging due to data sparseness, sample heterogeneity, signal contributions from multiple donors and acceptors, unequal fluorophore brightness, photobleaching, flexibility of the linker connecting the fluorophore to the tagged protein, and spectral cross-talk. We addressed these challenges by using a Bayesian approach that produces the posterior probability of a model, given the input data. The posterior probability is defined as a function of the dependence of our FRET metric FRETR on a structure (forward model), a model of noise in the data, as well as prior information about the structure, relative populations of distinct states in the sample, forward model parameters, and data noise. The forward model was validated against kinetic Monte Carlo simulations and in vivo experimental data collected on nine systems of known structure. In addition, our Bayesian approach was validated by a benchmark of 16 protein complexes of known structure. Given the structures of each subunit of the complexes, models were computed from synthetic FRETR data with a distance root-mean-squared deviation error of 14 to 17 Å. The approach is implemented in the open-source Integrative Modeling Platform, allowing us to determine macromolecular structures through a combination of in vivo FRETR data and data from other sources, such as electron microscopy and chemical cross-linking.


Subject(s)
Bacterial Proteins/metabolism , Fluorescence Resonance Energy Transfer , Luminescent Proteins/metabolism , Saccharomyces cerevisiae Proteins/analysis , Saccharomyces cerevisiae Proteins/metabolism , Algorithms , Bayes Theorem , Computer Simulation , Molecular Structure , Monte Carlo Method , Protein Interaction Mapping , Protein Structure, Quaternary , Saccharomyces cerevisiae
19.
Proc Natl Acad Sci U S A ; 110(16): 6358-63, 2013 Apr 16.
Article in English | MEDLINE | ID: mdl-23553839

ABSTRACT

A detailed description of the events ruling ligand/protein interaction and an accurate estimation of the drug affinity to its target is of great help in speeding drug discovery strategies. We have developed a metadynamics-based approach, named funnel metadynamics, that allows the ligand to enhance the sampling of the target binding sites and its solvated states. This method leads to an efficient characterization of the binding free-energy surface and an accurate calculation of the absolute protein-ligand binding free energy. We illustrate our protocol in two systems, benzamidine/trypsin and SC-558/cyclooxygenase 2. In both cases, the X-ray conformation has been found as the lowest free-energy pose, and the computed protein-ligand binding free energy in good agreement with experiments. Furthermore, funnel metadynamics unveils important information about the binding process, such as the presence of alternative binding modes and the role of waters. The results achieved at an affordable computational cost make funnel metadynamics a valuable method for drug discovery and for dealing with a variety of problems in chemistry, physics, and material science.


Subject(s)
Drug Delivery Systems/methods , Drug Discovery/methods , Models, Chemical , Models, Molecular , Pharmaceutical Preparations/metabolism , Benzamidines/metabolism , Cyclooxygenase 2/metabolism , Ligands , Protein Binding , Pyrazoles/metabolism , Trypsin/metabolism
20.
Proc Natl Acad Sci U S A ; 110(49): E4708-13, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24248370

ABSTRACT

In the realm of protein-protein interactions, the assembly process of homooligomers plays a fundamental role because the majority of proteins fall into this category. A comprehensive understanding of this multistep process requires the characterization of the driving molecular interactions and the transient intermediate species. The latter are often short-lived and thus remain elusive to most experimental investigations. Molecular simulations provide a unique tool to shed light onto these complex processes complementing experimental data. Here we combine advanced sampling techniques, such as metadynamics and parallel tempering, to characterize the oligomerization landscape of fibritin foldon domain. This system is an evolutionarily optimized trimerization motif that represents an ideal model for experimental and computational mechanistic studies. Our results are fully consistent with previous experimental nuclear magnetic resonance and kinetic data, but they provide a unique insight into fibritin foldon assembly. In particular, our simulations unveil the role of nonspecific interactions and suggest that an interplay between thermodynamic bias toward native structure and residual conformational disorder may provide a kinetic advantage.


Subject(s)
Models, Molecular , Polymerization , Protein Conformation , Proteins/chemistry , Biophysics , Evolution, Molecular , Molecular Dynamics Simulation , Protein Folding
SELECTION OF CITATIONS
SEARCH DETAIL