Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Mol Cell ; 68(2): 350-360.e7, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-29053958

ABSTRACT

The proper location and timing of Dnmt1 activation are essential for DNA methylation maintenance. We demonstrate here that Dnmt1 utilizes two-mono-ubiquitylated histone H3 as a unique ubiquitin mark for its recruitment to and activation at DNA methylation sites. The crystal structure of the replication foci targeting sequence (RFTS) of Dnmt1 in complex with H3-K18Ub/23Ub reveals striking differences to the known ubiquitin-recognition structures. The two ubiquitins are simultaneously bound to the RFTS with a combination of canonical hydrophobic and atypical hydrophilic interactions. The C-lobe of RFTS, together with the K23Ub surface, also recognizes the N-terminal tail of H3. The binding of H3-K18Ub/23Ub results in spatial rearrangement of two lobes in the RFTS, suggesting the opening of its active site. Actually, incubation of Dnmt1 with H3-K18Ub/23Ub increases its catalytic activity in vitro. Our results therefore shed light on the essential role of a unique ubiquitin-binding module in DNA methylation maintenance.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA Methylation , Histones/chemistry , Ubiquitin/chemistry , Animals , Crystallography, X-Ray , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , Histones/genetics , Histones/metabolism , Humans , Protein Binding , Protein Structure, Quaternary , Ubiquitin/genetics , Ubiquitin/metabolism , Xenopus laevis
2.
Nano Lett ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38916205

ABSTRACT

Mechanical stress significantly affects the physiological functions of cells, including tissue homeostasis, cytoskeletal alterations, and intracellular transport. As a major cytoskeletal component, microtubules respond to mechanical stimulation by altering their alignment and polymerization dynamics. Previously, we reported that microtubules may modulate cargo transport by one of the microtubule-associated motor proteins, dynein, under compressive mechanical stress. Despite the critical role of tensile stress in many biological functions, how tensile stress on microtubules regulates cargo transport is yet to be unveiled. The present study demonstrates that the low-level tensile stress-induced microtubule deformation facilitates dynein-driven transport. We validate our experimental findings using all-atom molecular dynamics simulation. Our study may provide important implications for developing new therapies for diseases that involve impaired intracellular transport.

3.
J Chem Inf Model ; 64(9): 3884-3895, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38670929

ABSTRACT

Epidermal growth factor receptor (EGFR) activation is accompanied by dimerization. During the activation of the intracellular kinase domain, two EGFR kinases form an asymmetric dimer, and one side of the dimer (receiver) is activated. Using the string method and Markov state model (MSM), we performed a computational analysis of the structural changes in the activation of the EGFR dimer in this study. The string method reveals the minimum free-energy pathway (MFEP) from the inactive to active structure. The MSM was constructed from numerous trajectories of molecular dynamics simulations around the MFEP, which revealed the free-energy map of structural changes. In the activation of the receiver kinase, the unfolding of the activation loop (A-loop) is followed by the rearrangement of the C-helix, as observed in other kinases. However, unlike other kinases, the free-energy map of EGFR at the asymmetric dimer showed that the active state yielded the highest stability and revealed how interactions at the dimer interface induced receiver activation. As the H-helix of the activator approaches the C-helix of the receiver during activation, the A-loop unfolds. Subsequently, L782 of the receiver enters the pocket between the G- and H-helices of the activator, leading to a rearrangement of the hydrophobic residues around L782 of the receiver, which constitutes a structural rearrangement of the C-helix of the receiver from an outward to an inner position. The MSM analysis revealed long-time scale trajectories via kinetic Monte Carlo.


Subject(s)
ErbB Receptors , Markov Chains , Enzyme Activation , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Molecular Dynamics Simulation , Protein Conformation , Protein Multimerization , Thermodynamics
4.
J Chem Inf Model ; 63(24): 7860-7872, 2023 Dec 25.
Article in English | MEDLINE | ID: mdl-38069816

ABSTRACT

Interleukin-2-inducible T-cell kinase (ITK) regulates the response to T-cell receptor signaling and is a drug target for inflammatory and immunological diseases. Molecules that bind preferentially to the active form of ITK have low selectivity between kinases, whereas those that bind preferentially to the inactive form have high selectivity for ITK. Therefore, computational methods to predict the conformational selectivity of compounds are required to design highly selective ITK inhibitors. In this study, we performed absolute binding free-energy perturbation (ABFEP) simulations for 11 compounds on both active and inactive forms of ITK, and the calculated binding free energies were compared with experimental data. The conformational selectivity of 10 of the 11 compounds was correctly predicted using ABFEP. To investigate the mechanism underlying the stabilization of the active and inactive structures by the compounds, we performed extensive, conventional molecular dynamics simulations, which revealed that the compound-induced stabilization of the P-loop and linkage of conformational changes in L489, V419, F501, and M410 upon compound binding were critical factors. A guideline for designing inactive-form binders is proposed based on these key structural factors. The ABFEP and the created guidelines are expected to facilitate the discovery of highly selective ITK inhibitors.


Subject(s)
Molecular Dynamics Simulation , Protein Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Signal Transduction , Molecular Conformation
5.
J Chem Inf Model ; 63(1): 240-250, 2023 01 09.
Article in English | MEDLINE | ID: mdl-36539353

ABSTRACT

Based on many crystal structures of ligand complexes, much study has been devoted to understanding the molecular recognition of SARS-CoV-2 3C-like protease (3CLpro), a potent drug target for COVID-19. In this research, to extend this present static view, we examined the kinetic process of binding/unbinding of an eight-residue substrate peptide to/from 3CLpro by evaluating the path ensemble with the weighted ensemble simulation. The path ensemble showed the mechanism of how a highly flexible peptide folded into the bound form. At the early stage, the dominant motion was the diffusion on the protein surface showing a broad distribution, whose center was led into the cleft of the chymotrypsin fold. We observed a definite sequential formation of the hydrogen bonds at the later stage occurring in the cleft, initiated between Glu166 (3CLpro) and P3_Val (peptide), followed by binding to the oxyanion hole and completed by the sequence-specific recognition at P1_Gln.


Subject(s)
COVID-19 , Peptide Hydrolases , Humans , Peptide Hydrolases/metabolism , SARS-CoV-2/metabolism , Peptides/chemistry , Computer Simulation , Protease Inhibitors , Antiviral Agents , Molecular Docking Simulation
6.
Nucleic Acids Res ; 49(5): 2916-2930, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33211877

ABSTRACT

TFIIH is a crucial transcription and DNA repair factor consisting of the seven-subunit core. The core subunit p62 contains a pleckstrin homology domain (PH-D), which is essential for locating TFIIH at transcription initiation and DNA damage sites, and two BSD (BTF2-like transcription factors, synapse-associated proteins and DOS2-like proteins) domains. A recent cryo-electron microscopy (cryo-EM) structure of human TFIIH visualized most parts of core, except for the PH-D. Here, by nuclear magnetic resonance spectroscopy we have established the solution structure of human p62 PH-D connected to the BSD1 domain by a highly flexible linker, suggesting the flexibility of PH-D in TFIIH. Based on this dynamic character, the PH-D was modeled in the cryo-EM structure to obtain the whole human TFIIH core structure, which indicates that the PH-D moves around the surface of core with a specific but limited spatial distribution; these dynamic structures were refined by molecular dynamics (MD) simulations. Furthermore, we built models, also refined by MD simulations, of TFIIH in complex with five p62-binding partners, including transcription factors TFIIEα, p53 and DP1, and nucleotide excision repair factors XPC and UVSSA. The models explain why the PH-D is crucially targeted by these factors, which use their intrinsically disordered acidic regions for TFIIH recruitment.


Subject(s)
Transcription Factor TFIIH/chemistry , Cryoelectron Microscopy , Humans , Molecular Dynamics Simulation , Pleckstrin Homology Domains , Protein Domains
7.
Int J Mol Sci ; 24(20)2023 Oct 21.
Article in English | MEDLINE | ID: mdl-37895101

ABSTRACT

Tubulin has been recently reported to form a large family consisting of various gene isoforms; however, the differences in the molecular features of tubulin dimers composed of a combination of these isoforms remain unknown. Therefore, we attempted to elucidate the physical differences in the molecular motility of these tubulin dimers using the method of measurable pico-meter-scale molecular motility, diffracted X-ray tracking (DXT) analysis, regarding characteristic tubulin dimers, including neuronal TUBB3 and ubiquitous TUBB5. We first conducted a DXT analysis of neuronal (TUBB3-TUBA1A) and ubiquitous (TUBB5-TUBA1B) tubulin dimers and found that the molecular motility around the vertical axis of the neuronal tubulin dimer was lower than that of the ubiquitous tubulin dimer. The results of molecular dynamics (MD) simulation suggest that the difference in motility between the neuronal and ubiquitous tubulin dimers was probably caused by a change in the major contact of Gln245 in the T7 loop of TUBB from Glu11 in TUBA to Val353 in TUBB. The present study is the first report of a novel phenomenon in which the pico-meter-scale molecular motility between neuronal and ubiquitous tubulin dimers is different.


Subject(s)
Molecular Dynamics Simulation , Tubulin , Tubulin/genetics , Tubulin/metabolism , X-Rays , Protein Isoforms/genetics , Neurons/metabolism
8.
J Am Chem Soc ; 144(26): 11802-11809, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35727684

ABSTRACT

Inspired by mechanosensitive potassium channels found in nature, we developed a fluorinated amphiphilic cyclophane composed of fluorinated rigid aromatic units connected via flexible hydrophilic octa(ethylene glycol) chains. Microscopic and emission spectroscopic studies revealed that the cyclophane could be incorporated into the hydrophobic layer of the lipid bilayer membranes and self-assembled to form a supramolecular transmembrane ion channel. Current recording measurements using cyclophane-containing planer lipid bilayer membranes successfully demonstrated an efficient transmembrane ion transport. We also demonstrated that the ion transport property was sensitive to the mechanical forces applied to the membranes. In addition, ion transport assays using pH-sensitive fluorescence dye revealed that the supramolecular channel possesses potassium ion selectivity. We also performed all-atom hybrid quantum-mechanical/molecular mechanical simulations to assess the channel structures at atomic resolution and the mechanism of selective potassium ion transport. This research demonstrated the first example of a synthetic mechanosensitive potassium channel, which would open a new door to sensing and manipulating biologically important processes and purification of key materials in industries.


Subject(s)
Lipid Bilayers , Potassium Channels , Hydrophobic and Hydrophilic Interactions , Ion Channels/chemistry , Lipid Bilayers/chemistry , Potassium , Potassium Channels/chemistry
9.
J Chem Inf Model ; 62(18): 4460-4473, 2022 09 26.
Article in English | MEDLINE | ID: mdl-36068974

ABSTRACT

Among the factors affecting biological processes such as protein folding and ligand binding, hydration, which is represented by a three-dimensional water site distribution function around the protein, is crucial. The typical methods for computing the distribution functions, including molecular dynamics simulations and the three-dimensional reference interaction site model (3D-RISM) theory, require a long computation time ranging from hours to tens of hours. Here, we propose a deep learning (DL) model that rapidly estimates the distribution functions around proteins obtained using the 3D-RISM theory from the protein 3D structure. The distribution functions predicted using our DL model are in good agreement with those obtained using the 3D-RISM theory. Particularly, the coefficient of determination between the distribution function obtained by the DL model and that obtained using the 3D-RISM theory is approximately 0.98. Furthermore, using a graphics processing unit, the prediction by the DL model is completed in less than 1 min, more than 2 orders of magnitude faster than the calculation time of the 3D-RISM theory. The position of water molecules around the protein was estimated based on the distribution function obtained by our DL model, and the position of waters estimated by our DL model was in good agreement with that of water molecules estimated using the 3D-RISM theory and of crystallographic waters. Therefore, our DL model provides a practical and efficient way to calculate the three-dimensional water site distribution functions and to estimate the position of water molecules around the protein. The program called "gr Predictor" is available under the GNU General Public License from https://github.com/YoshidomeGroup-Hydration/gr-predictor.


Subject(s)
Deep Learning , Ligands , Molecular Dynamics Simulation , Proteins , Thermodynamics , Water/chemistry
10.
J Chem Inf Model ; 61(7): 3583-3592, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34228431

ABSTRACT

The mineralocorticoid receptor (MR) is a nuclear receptor whose endogenous ligands are mineralocorticoids, a type of steroid hormone. The activating S810L mutation is known to cause severe early-onset and pregnancy-related hypertension. Progesterone binds to the wild-type (WT) MR as a passive antagonist with fast dissociation; however, it binds to the S810L mutant as a full agonist with slow dissociation. The switch in the biological activity of progesterone is considered to be one of the causes of the disease. First, we used steered molecular dynamics simulations to analyze the dissociation process of progesterone for the WT and the S810L mutant. Progesterone in the WT dissociated from the ligand-binding pocket with a weak force in comparison with progesterone in the S810L mutant due to the large inflow of water molecules into the pocket. Therefore, we used conventional molecular dynamics simulations for the ligand-free structures of the WT and the S810L mutant to investigate the effect of the mutation on the inflow of water. In the WT, water molecules enter the ligand-binding pocket in two ways: in the vicinity of (i) Arg817 and (ii) Ser810. In contrast, few water molecules enter the pocket in the S810L mutant because of the large size and hydrophobic nature of the Leu810 side chain. Fast dissociation is a common feature among passive antagonists of MR; therefore, we inferred that the water inflow could be responsible for the dissociation kinetics of progesterone in the WT and the S810L mutant.


Subject(s)
Hypertension , Receptors, Mineralocorticoid , Water , Female , Humans , Ligands , Mutation , Pregnancy , Receptors, Mineralocorticoid/chemistry , Receptors, Mineralocorticoid/genetics
11.
J Chem Inf Model ; 61(7): 3625-3637, 2021 07 26.
Article in English | MEDLINE | ID: mdl-34189910

ABSTRACT

The vitamin D receptor ligand-binding domain (VDR-LBD) undergoes conformational changes upon ligand binding. In this nuclear receptor family, agonistic or antagonistic activities are controlled by the conformation of the helix (H)12. However, all crystal structures of VDR-LBD reported to date correspond to the active H12 conformation, regardless of agonist/antagonist binding. To understand the mechanism of VDR-LBD regulation structurally, conformational samplings of agonist- and antagonist-bound rat VDR-LBD were performed using the generalized replica exchange with solute tempering (gREST) method. The gREST simulations demonstrated different structural responses of rat VDR-LBD to agonist or antagonist binding, whereas in conventional molecular dynamics simulations, the conformation was the same as that of the crystal structures, regardless of agonist/antagonist binding. In the gREST simulations, a spontaneous conformational change of H12 was observed only for the antagonist complex. The different responses to agonist/antagonist binding were attributed to hydrophobic core formation at the ligand-binding pocket and cooperative rearrangements of H11. The gREST method can be applied to the examination of structure-activity relationships for multiple VDR-LBD ligands.


Subject(s)
Molecular Dynamics Simulation , Receptors, Calcitriol , Animals , Binding Sites , Ligands , Molecular Conformation , Protein Binding , Rats , Receptors, Calcitriol/metabolism
12.
Bioorg Med Chem ; 52: 116500, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34801826

ABSTRACT

The accumulation of epigenetic alterations is one of the major causes of tumorigenesis. Aberrant DNA methylation patterns cause genome instability and silencing of tumor suppressor genes in various types of tumors. Therefore, drugs that target DNA methylation-regulating factors have great potential for cancer therapy. Ubiquitin-like containing PHD and RING finger domain 1 (UHRF1) is an essential factor for DNA methylation maintenance. UHRF1 is overexpressed in various cancer cells and down-regulation of UHRF1 in these cells reactivates the expression of tumor suppressor genes, thus UHRF1 is a promising target for cancer therapy. We have previously shown that interaction between the tandem Tudor domain (TTD) of UHRF1 and DNA ligase 1 (LIG1) di/trimethylated on Lys126 plays a key role in the recruitment of UHRF1 to replication sites and replication-coupled DNA methylation maintenance. An arginine binding cavity (Arg-binding cavity) of the TTD is essential for LIG1 interaction, thus the development of inhibitors that target the Arg-binding cavity could potentially repress UHRF1 function in cancer cells. To develop such an inhibitor, we performed in silico screening using not only static but also dynamic metrics based on all-atom molecular dynamics simulations, resulting in efficient identification of 5-amino-2,4-dimethylpyridine (5A-DMP) as a novel TTD-binding compound. Crystal structure of the TTD in complex with 5A-DMP revealed that the compound stably bound to the Arg-binding cavity of the TTD. Furthermore, 5A-DMP inhibits the full-length UHRF1:LIG1 interaction in Xenopus egg extracts. Our study uncovers a UHRF1 inhibitor which can be the basis of future experiments for cancer therapy.


Subject(s)
CCAAT-Enhancer-Binding Proteins/antagonists & inhibitors , DNA Ligase ATP/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Molecular Dynamics Simulation , Pyridines/pharmacology , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line, Tumor , DNA Ligase ATP/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Enzyme Inhibitors/chemistry , Humans , Molecular Structure , Pyridines/chemistry , Structure-Activity Relationship , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Xenopus
13.
J Comput Chem ; 41(28): 2406-2419, 2020 10 30.
Article in English | MEDLINE | ID: mdl-32815201

ABSTRACT

Hydration is a critical factor in the ligand binding process. Herein, to examine the hydration states of ligand binding sites, the three-dimensional distribution function for the water oxygen site, gO (r), is computed for 3,706 ligand-free protein structures based on the corresponding small molecule-protein complexes using the 3D-RISM theory. For crystallographic waters (CWs) close to the ligand, gO (r) reveals that several CWs are stabilized by interaction networks formed between the ligand, CW, and protein. Based on the gO (r) for the crystallographic binding pose of the ligand, hydrogen bond interactions are dominant in the highly hydrated regions while weak interactions such as CH-O are dominant in the moderately hydrated regions. The polar heteroatoms of the ligand occupy the highly hydrated and moderately hydrated regions in the crystallographic (correct) and wrongly docked (incorrect) poses, respectively. Thus, the gO (r) of polar heteroatoms may be used to distinguish the correct binding poses.


Subject(s)
Proteins/chemistry , Amino Acid Sequence , Binding Sites , Crystallization , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Ligands , Molecular Dynamics Simulation , Protein Binding , Protein Conformation , Solvents/chemistry , Thermodynamics , Water/chemistry
14.
J Chem Inf Model ; 60(7): 3361-3368, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32496771

ABSTRACT

Here, we have constructed neural network-based models that predict atomic partial charges with high accuracy at low computational cost. The models were trained using high-quality data acquired from quantum mechanics calculations using the fragment molecular orbital method. We have succeeded in obtaining highly accurate atomic partial charges for three representative molecular systems of proteins, including one large biomolecule (approx. 2000 atoms). The novelty of our approach is the ability to take into account the electronic polarization in the system, which is a system-dependent phenomenon, being important in the field of drug design. Our high-precision models are useful for the prediction of atomic partial charges and expected to be widely applicable in structure-based drug designs such as structural optimization, high-speed and high-precision docking, and molecular dynamics calculations.


Subject(s)
Molecular Dynamics Simulation , Proteins , Drug Design , Machine Learning , Neural Networks, Computer
15.
J Chem Phys ; 152(6): 065103, 2020 Feb 14.
Article in English | MEDLINE | ID: mdl-32061219

ABSTRACT

Using experimentally determined structures of ubiquitin at 1 and 3000 bar, we generate sufficiently large ensembles of model structures in the native and pressure-induced (denatured) states by means of molecular dynamics simulations with explicit water. We calculate the values of a free-energy function (FEF), which comprises the hydration free energy (HFE) and the intramolecular (conformational) energy and entropy, for the two states at 1 and 3000 bar. The HFE and the conformational entropy, respectively, are calculated using our statistical-mechanical method, which has recently been shown to be accurate, and the Boltzmann-quasi-harmonic method. The HFE is decomposed into a variety of physically insightful components. We show that the FEF of the native state is lower than that of the denatured state at 1 bar, whereas the opposite is true at 3000 bar, thus being successful in reproducing the pressure denaturation. We argue that the following two quantities of hydration play essential roles in the denaturation: the WASA-dependent term in the water-entropy loss upon cavity creation for accommodating the protein (WASA is the water-accessible surface area of the cavity) and the protein-water Lennard-Jones interaction energy. At a high pressure, the mitigation of the serious water crowding in the system is the most important, and the WASA needs to be sufficiently enlarged with the increase in the excluded-volume being kept as small as possible. The denatured structure thus induced is characterized by the water penetration into the protein interior. The pressure denaturation is accompanied by a significantly large gain of water entropy.

16.
J Comput Chem ; 40(29): 2577-2585, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31343749

ABSTRACT

We propose a novel force-field-parametrization procedure that fits the parameters of potential functions in a manner that the pair distribution function (DF) of molecules derived from candidate parameters can reproduce the given target DF. Conventionally, approaches to minimize the difference between the candidate and target DFs employ radial DFs (RDF). RDF itself has been reported to be insufficient for uniquely identifying the parameters of a molecule. To overcome the weakness, we introduce energy DF (EDF) as a target DF, which describes the distribution of the pairwise energy of molecules. We found that the EDF responds more sensitively to a small perturbation in the pairwise potential parameters and provides better fitting accuracy compared to that of RDF. These findings provide valuable insights into a wide range of coarse graining methods, which determine parameters using information obtained from a higher-level calculation than that of the developed force field. © 2019 The Authors. Journal of Computational Chemistry published by Wiley Periodicals, Inc.

17.
J Chem Inf Model ; 59(8): 3533-3544, 2019 08 26.
Article in English | MEDLINE | ID: mdl-31282659

ABSTRACT

An oncoprotein MDM2 binds to the extreme N-terminal peptide region of a tumor suppressor protein p53 (p53NTD) and inhibits its anticancer activity. We recently discovered a peptide named MIP which exhibits much higher binding affinity for MDM2 than p53NTD. Experiments showed that the binding free energy (BFE) of MDM2-MIP is lower than that of MDM2-p53NTD by approximately -4 kcal/mol. Here, we develop a theoretical method which is successful in reproducing this quantitative difference and elucidating its physical origins. It enables us to decompose the BFE into a variety of energetic and entropic components, evaluate their relative magnitudes, and identify the physical factors driving or opposing the binding. It should be applicable also to the assessment of differences among ligands in the binding affinity for a particular receptor, which is a central issue in modern chemistry. In the MDM2 case, the higher affinity of MIP is ascribed to a larger gain of translational, configurational entropy of water upon binding. This result is useful to the design of a peptide possessing even higher affinity for MDM2 as a reliable drug against a cancer.


Subject(s)
Molecular Dynamics Simulation , Peptides/metabolism , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/metabolism , Binding Sites , Protein Binding , Protein Conformation , Proto-Oncogene Proteins c-mdm2/chemistry , Substrate Specificity , Thermodynamics , Tumor Suppressor Protein p53/chemistry
18.
J Chem Phys ; 150(17): 175101, 2019 May 07.
Article in English | MEDLINE | ID: mdl-31067910

ABSTRACT

A new method is developed for calculating hydration free energies (HFEs) of polyatomic solutes. The solute insertion is decomposed into the creation of a cavity in water matching the geometric characteristics of the solute at the atomic level (process 1) and the incorporation of solute-water van der Waals and electrostatic interactions (process 2). The angle-dependent integral equation theory combined with our morphometric approach and the three-dimensional interaction site model theory are applied to processes 1 and 2, respectively. Neither a stage of training nor parameterization is necessitated. For solutes with various sizes including proteins, the HFEs calculated by the new method are compared to those obtained using a molecular dynamics simulation based on solution theory in energy representation (the ER method developed by Matubayasi and co-workers), currently the most reliable tool. The agreement is very good especially for proteins. The new method is characterized by the following: The calculation can rapidly be finished; a solute possessing a significantly large total charge can be handled without difficulty; and since it yields not only the HFE but also its many physically insightful energetic and entropic components, it is best suited to the elucidation of mechanisms of diverse phenomena such as the receptor-ligand binding, different types of molecular recognition, and protein folding, denaturation, and association.


Subject(s)
Dipeptides/chemistry , Proteins/chemistry , Water/chemistry , Models, Chemical , Molecular Dynamics Simulation , Thermodynamics
19.
Proc Natl Acad Sci U S A ; 113(33): 9280-5, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27493216

ABSTRACT

The dystrophin glycoprotein complex, which connects the cell membrane to the basement membrane, is essential for a variety of biological events, including maintenance of muscle integrity. An O-mannose-type GalNAc-ß1,3-GlcNAc-ß1,4-(phosphate-6)-Man structure of α-dystroglycan (α-DG), a subunit of the complex that is anchored to the cell membrane, interacts directly with laminin in the basement membrane. Reduced glycosylation of α-DG is linked to some types of inherited muscular dystrophy; consistent with this relationship, many disease-related mutations have been detected in genes involved in O-mannosyl glycan synthesis. Defects in protein O-linked mannose ß1,2-N-acetylglucosaminyltransferase 1 (POMGnT1), a glycosyltransferase that participates in the formation of GlcNAc-ß1,2-Man glycan, are causally related to muscle-eye-brain disease (MEB), a congenital muscular dystrophy, although the role of POMGnT1 in postphosphoryl modification of GalNAc-ß1,3-GlcNAc-ß1,4-(phosphate-6)-Man glycan remains elusive. Our crystal structures of POMGnT1 agreed with our previous results showing that the catalytic domain recognizes substrate O-mannosylated proteins via hydrophobic interactions with little sequence specificity. Unexpectedly, we found that the stem domain recognizes the ß-linked GlcNAc of O-mannosyl glycan, an enzymatic product of POMGnT1. This interaction may recruit POMGnT1 to a specific site of α-DG to promote GlcNAc-ß1,2-Man clustering and also may recruit other enzymes that interact with POMGnT1, e.g., fukutin, which is required for further modification of the GalNAc-ß1,3-GlcNAc-ß1,4-(phosphate-6)-Man glycan. On the basis of our findings, we propose a mechanism for the deficiency in postphosphoryl modification of the glycan observed in POMGnT1-KO mice and MEB patients.


Subject(s)
Dystroglycans/chemistry , N-Acetylglucosaminyltransferases/chemistry , Binding Sites , Crystallization , Glycosylation , Humans , Mannose/chemistry
20.
Phys Chem Chem Phys ; 20(36): 23684-23693, 2018 Sep 19.
Article in English | MEDLINE | ID: mdl-30191211

ABSTRACT

We unravel the physical origins of the large difference between cellobiose and maltose, which consist of two ß-1,4 and α-1,4 linked d-glucose units, respectively, in terms of the solubility in water. We construct a thermodynamic theory where the chemical-potential difference between disaccharides in water and in vacuum is identified as the key free-energy function. Its energetic and entropic components are calculated for cellobiose and maltose by statistical-mechanical theories for solute hydration. The disaccharide structures are taken into account at the atomic level and a molecular model is adopted for water. Molecular dynamics simulations are used to account for the conformational fluctuation of a disaccharide molecule, which also enables us to estimate the conformational entropy. We show that the cellobiose/maltose solubility ratio calculated is in good agreement with the experimental value. The solubility becomes much lower for cellobiose due to conformational-entropy and water-entropy effects. The former effect is relevant to higher stability of the intramolecular hydrogen bond between oxygen atoms in the six-membered ring and in the neighboring hydroxyl group: the hydration alters the fluctuation of a molecular conformation to a larger or less regular one, but the degree of this alteration is smaller. The latter effect is attributed to more separation of two hydroxymethyl groups in a molecule, causing lower probability of the overlap of excluded volumes generated by the groups for water molecules. We suggest that physicochemical properties of disaccharides in water become variable depending on the stereoisomerism through hydration effects and the origins of the variety are entropic.

SELECTION OF CITATIONS
SEARCH DETAIL