Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Commun Biol ; 5(1): 1344, 2022 12 07.
Article in English | MEDLINE | ID: mdl-36477694

ABSTRACT

Nicotinic acetylcholine receptor of α7 type (α7-nAChR) presented in the nervous and immune systems and epithelium is a promising therapeutic target for cognitive disfunctions and cancer treatment. Weak toxin from Naja kaouthia venom (WTX) is a non-conventional three-finger neurotoxin, targeting α7-nAChR with weak affinity. There are no data on interaction mode of non-conventional neurotoxins with nAChRs. Using α-bungarotoxin (classical three-finger neurotoxin with high affinity to α7-nAChR), we showed applicability of cryo-EM to study complexes of α7-nAChR extracellular ligand-binding domain (α7-ECD) with toxins. Using cryo-EM structure of the α7-ECD/WTX complex, together with NMR data on membrane active site in the WTX molecule and mutagenesis data, we reconstruct the structure of α7-nAChR/WTX complex in the membrane environment. WTX interacts at the entrance to the orthosteric site located at the receptor intersubunit interface and simultaneously forms the contacts with the membrane surface. WTX interaction mode with α7-nAChR significantly differs from α-bungarotoxin's one, which does not contact the membrane. Our study reveals the important role of the membrane for interaction of non-conventional neurotoxins with the nicotinic receptors.


Subject(s)
Receptors, Nicotinic , Receptors, Nicotinic/genetics , Three Finger Toxins , Bungarotoxins , Neurotoxins/toxicity
2.
Int J Mol Sci ; 23(16)2022 Aug 17.
Article in English | MEDLINE | ID: mdl-36012488

ABSTRACT

Understanding fusion mechanisms employed by SARS-CoV-2 spike protein entails realistic transmembrane domain (TMD) models, while no reliable approaches towards predicting the 3D structure of transmembrane (TM) trimers exist. Here, we propose a comprehensive computational framework to model the spike TMD only based on its primary structure. We performed amino acid sequence pattern matching and compared the molecular hydrophobicity potential (MHP) distribution on the helix surface against TM homotrimers with known 3D structures and selected an appropriate template for homology modeling. We then iteratively built a model of spike TMD, adjusting "dynamic MHP portraits" and residue variability motifs. The stability of this model, with and without palmitoyl modifications downstream of the TMD, and several alternative configurations (including a recent NMR structure), was tested in all-atom molecular dynamics simulations in a POPC bilayer mimicking the viral envelope. Our model demonstrated unique stability under the conditions applied and conforms to known basic principles of TM helix packing. The original computational framework looks promising and could potentially be employed in the construction of 3D models of TM trimers for a wide range of membrane proteins.


Subject(s)
SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Molecular Dynamics Simulation , Protein Domains , Spike Glycoprotein, Coronavirus/chemistry
3.
Commun Biol ; 5(1): 706, 2022 07 15.
Article in English | MEDLINE | ID: mdl-35840781

ABSTRACT

"Fluorescence-Activating and absorption-Shifting Tag" (FAST) is a well-studied fluorogen-activating protein with high brightness and low size, able to activate a wide range of fluorogens. This makes FAST a promising target for both protein and fluorogen optimization. Here, we describe the structure-based rational design of the enhanced FAST mutants, optimized for the N871b fluorogen. Using the spatial structure of the FAST/N871b complex, NMR relaxation analysis, and computer simulations, we identify the mobile regions in the complex and suggest mutations that could stabilize both the protein and the ligand. Two of our mutants appear brighter than the wild-type FAST, and these mutants provide up to 35% enhancement for several other fluorogens of similar structure, both in vitro and in vivo. Analysis of the mutants by NMR reveals that brighter mutants demonstrate the highest stability and lowest length of intermolecular H-bonds. Computer simulations provide the structural basis for such stabilization.


Subject(s)
Fluorescent Dyes , Proteins , Fluorescence , Fluorescent Dyes/chemistry
4.
PLoS One ; 15(8): e0231560, 2020.
Article in English | MEDLINE | ID: mdl-32822353

ABSTRACT

The dehydroshikimate dehydratase (DSD) from Corynebacterium glutamicum encoded by the qsuB gene is related to the previously described QuiC1 protein (39.9% identity) from Pseudomonas putida. Both QuiC1 and QsuB are two-domain bacterial DSDs. The N-terminal domain provides dehydratase activity, while the C-terminal domain has sequence identity with 4-hydroxyphenylpyruvate dioxygenase. Here, the QsuB protein and its N-terminal domain (N-QsuB) were expressed in the T7 system, purified and characterized. QsuB was present mainly in octameric form (60%), while N-QsuB had a predominantly monomeric structure (80%) in aqueous buffer. Both proteins possessed DSD activity with one of the following cofactors (listed in the order of decreasing activity): Co2+, Mg2+, Mn2+. The Km and kcat values for the QsuB enzyme (Km ~ 1 mM, kcat ~ 61 s-1) were two and three times higher than those for N-QsuB. 3,4-DHBA inhibited QsuB (Ki ~ 0.38 mM, Ki' ~ 0.96 mM) and N-QsuB (Ki ~ 0.69 mM) enzymes via mixed and noncompetitive inhibition mechanism, respectively. E. coli MG1655ΔaroEPlac‒qsuB strain produced three times more 3,4-DHBA from glucose in test tube fermentation than the MG1655ΔaroEPlac‒n-qsuB strain. The C-terminal domain activity towards 3,4-DHBA was not established in vitro. This domain was proposed to promote protein oligomerization for maintaining structural stability of the enzyme. The dimer formation of QsuB protein was more predictable (ΔG = ‒15.8 kcal/mol) than the dimerization of its truncated version N-QsuB (ΔG = ‒0.4 kcal/mol).


Subject(s)
Biotechnology , Corynebacterium glutamicum/enzymology , Hydro-Lyases/metabolism , Hydroxybenzoates/metabolism , Corynebacterium glutamicum/genetics , DNA, Recombinant/genetics , Escherichia coli/metabolism , Hydro-Lyases/chemistry , Hydro-Lyases/genetics , Hydrogen-Ion Concentration , Models, Molecular , Protein Domains , Protein Multimerization , Protein Structure, Quaternary
5.
Sci Rep ; 9(1): 413, 2019 01 23.
Article in English | MEDLINE | ID: mdl-30674891

ABSTRACT

How is a water-soluble globular protein able to spontaneously cross a cellular membrane? It is commonly accepted that it undergoes significant structural rearrangements on the lipid-water interface, thus acquiring membrane binding and penetration ability. In this study molecular dynamics (MD) simulations have been used to explore large-scale conformational changes of the globular viscumin A chain in a complex environment - comprising urea and chloroform/methanol (CHCl3/MeOH) mixture. Being well-packed in aqueous solution, viscumin A undergoes global structural rearrangements in both organic media. In urea, the protein is "swelling" and gradually loses its long-distance contacts, thus resembling the "molten globule" state. In CHCl3/MeOH, viscumin A is in effect turned "inside out". This is accompanied with strengthening of the secondary structure and surface exposure of hydrophobic epitopes originally buried inside the globule. Resulting solvent-adapted models were further subjected to Monte Carlo simulations with an implicit hydrophobic slab membrane. In contrast to only a few point surface contacts in water and two short regions with weak protein-lipid interactions in urea, MD-derived structures in CHCl3/MeOH reveal multiple determinants of membrane interaction. Consequently it is now possible to propose a specific pathway for the structural adaptation of viscumin A with respect to the cell membrane - a probable first step of its translocation into cytoplasmic targets.


Subject(s)
Cell Membrane/chemistry , Membranes, Artificial , Protein Refolding , Ribosome Inactivating Proteins, Type 2/chemistry , Toxins, Biological/chemistry , Protein Transport
6.
Sci Rep ; 6: 33112, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27612191

ABSTRACT

Heat-activated transient receptor potential channel TRPV1 is one of the most studied eukaryotic proteins involved in temperature sensation. Upon heating, it exhibits rapid reversible pore gating, which depolarizes neurons and generates action potentials. Underlying molecular details of such effects in the pore region of TRPV1 is of a crucial importance to control temperature responses of the organism. Despite the spatial structure of the channel in both open (O) and closed (C) states is known, microscopic nature of channel gating and mechanism of thermal sensitivity are still poorly understood. In this work, we used unrestrained atomistic molecular dynamics simulations of TRPV1 (without N- and C-terminal cytoplasmic domains) embedded into explicit lipid bilayer in its O- and C-states. We found that the pore domain with its neighboring loops undergoes large temperature-dependent conformational transitions in an asymmetric way, when fragments of only one monomer move with large amplitude, freeing the pore upon heating. Such an asymmetrical gating looks rather biologically relevant because it is faster and more reliable than traditionally proposed "iris-like" symmetric scheme of channel opening. Analysis of structural, dynamic, and hydrophobic organization of the pore domain revealed entropy growth upon TRPV1 gating, which is in line with current concepts of thermal sensitivity.


Subject(s)
Computer Simulation , Hot Temperature , Molecular Dynamics Simulation , TRPV Cation Channels/chemistry , Humans , Protein Domains
7.
Bioinformatics ; 30(6): 889-90, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24202542

ABSTRACT

SUMMARY: Here we present PREDDIMER, a web tool for prediction of dimer structure of transmembrane (TM) helices. PREDDIMER allows (i) reconstruction of a number of dimer structures for given sequence(s) of TM protein fragments, (ii) ranking and filtering of predicted structures according to respective values of a scoring function, (iii) visualization of predicted 3D dimer structures and (iv) visualization of surface hydrophobicity of TM helices and their contacting (interface) regions represented as 2D maps. RESULTS: We implemented online the original PREDDIMER algorithm and benchmarked the server on 11 TM sequences, whose 3D dimer conformations were obtained previously by nuclear magnetic resonance spectroscopy. In the most of tested cases backbone root-mean-square deviations of closest predicted conformations from the experimental reference are below 3 Å. A randomization test displays good anticorrelation (-0.82) between values of the scoring function and statistical significance of the prediction 'by chance'. Going beyond a single dimer conformation, our web tool predicts an ensemble of possible conformations, which may be useful for explanation of a functioning of bitopic membrane proteins, e.g. receptor tyrosine kinases. AVAILABILITY AND IMPLEMENTATION: PREDDIMER can be accessed for free on the web at http://model.nmr.ru/preddimer/ CONTACT: newant@gmail.com SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Membrane Proteins/chemistry , Protein Multimerization , Algorithms , Internet , Models, Molecular , Protein Structure, Secondary , Protein Structure, Tertiary , Software
8.
J Biomol Struct Dyn ; 31(1): 87-95, 2013.
Article in English | MEDLINE | ID: mdl-22804614

ABSTRACT

Delineation and analysis of lateral clustering of lipids in model bilayers is an important step toward understanding of the physical processes underlying formation of lipid domains and rafts in cell membranes. Computer modeling methods represent a powerful tool to address the problem since they can detect clusters of only few lipid molecules - this issue still resists easy characterization with modern experimental techniques. In this work, we propose a computational method to detect and analyze parts of membrane with different packing densities and hydrogen bonding patterns. A series of one- and two-component fluid systems containing lipids with the same polar heads and different acyl chains, dioleoylphosphatidylcholine (18:1) and dipalmitoylphosphatidylcholine (16:0), or with same acyl chains and different polar heads, dioleoylphosphatidylserine (18:1) and dioleoylphosphatidylcholine (18:1), were studied via molecular dynamics simulations. Four criteria of clustering were considered. It was shown that the water-lipid interface of biomembranes represents a highly dynamic and "mosaic" picture, whose parameters depend on the bilayer composition. Some systems (e.g. with 20-30% of the anionic lipid) demonstrate unusual clustering properties and demand further investigation at molecular level. Lateral microheterogeneities in fluid lipid bilayers seem to be among the most important factors determining the nature of the membrane-water interface in a cell.


Subject(s)
Lipid Bilayers/chemistry , Lipids/chemistry , 1,2-Dipalmitoylphosphatidylcholine/chemistry , Cell Membrane/chemistry , Hydrogen Bonding , Models, Molecular , Molecular Dynamics Simulation , Phosphatidylcholines/chemistry , Phosphatidylserines/chemistry , Water/chemistry
9.
Bioinformatics ; 25(9): 1201-2, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19244385

ABSTRACT

UNLABELLED: The PLATINUM (Protein-Ligand ATtractions Investigation NUMerically) web service is designed for analysis and visualization of hydrophobic/hydrophilic properties of biomolecules supplied as 3D-structures. Furthermore, PLATINUM provides a number of tools for quantitative characterization of the hydrophobic/hydrophilic match in biomolecular complexes e.g. in docking poses. These complement standard scoring functions. The calculations are based on the concept of empirical Molecular Hydrophobicity Potential (MHP). AVAILABILITY: The PLATINUM web tool as well as detailed documentation and tutorial are available free of charge for academic users at http://model.nmr.ru/platinum/. PLATINUM requires Java 5 or higher and Adobe Flash Player 9. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Proteins/chemistry , Software , Binding Sites , Databases, Protein , Hydrophobic and Hydrophilic Interactions , Internet , Ligands , Protein Conformation , Proteins/metabolism
10.
J Biomol Struct Dyn ; 25(2): 195-205, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17718599

ABSTRACT

The membrane interface-partitioning region preceding the transmembrane anchor of the human immunodeficiency virus type 1 (HIV-1) gp41 envelope protein is one of the sites responsible for virus binding to its host cell membrane and subsequent fusion events. Here, we used molecular modeling techniques to assess membrane interactions, structure, and hydrophobic properties of the fusion-active peptide representing this region, several of its homologs from different HIV-1 strains, as well as a peptide - defective gp41 phenotype - unable to mediate cell-cell fusion and virus entry. It is shown that the wild-type peptides bind to the water-membrane interface in alpha-helical conformation, while the mutant adopts partly destabilized helix-break-helix structure on the membrane surface. The wild-type peptides reveal specific "tilted oblique-oriented" pattern of hydrophobicity on their surfaces - the property specific for fusion regions of other viruses. Fusion peptides penetrate into the membrane with their N-termini and reveal "fine-tuning" interactions with membrane and water environments: the shift of this balance (e.g., due to point mutations) may dramatically change the mode of membrane binding, and therefore, may cause loss of fusion activity. The modeling results agree well with experimental data and provide a strategy to delineate fusogenic regions in amino acid sequences of viral proteins.


Subject(s)
Amino Acid Sequence , Cell Membrane/metabolism , HIV Envelope Protein gp41 , Virus Internalization , HIV Envelope Protein gp41/genetics , HIV Envelope Protein gp41/metabolism , Humans , Molecular Sequence Data , Peptides/chemistry , Peptides/genetics , Peptides/metabolism , Protein Conformation , Sequence Alignment
11.
Proteins ; 69(2): 309-25, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17600828

ABSTRACT

BNIP3 is a mitochondrial 19-kDa proapoptotic protein, a member of the Bcl-2 family. It has a single COOH-terminal transmembrane (TM) alpha-helical domain, which is required for membrane targeting, proapoptotic activity, hetero- and homo-dimerization in membrane. The role and the molecular details of association of TM helices of BNIP3 are yet to be established. Here, we present a molecular modeling study of helix interactions in its membrane domain. The approach combines Monte Carlo conformational search in an implicit hydrophobic slab followed by molecular dynamics simulations in a hydrated full-atom lipid bilayer. The former technique was used for exhaustive sampling of the peptides' conformational space and for generation of putative "native-like" structures of the dimer. The latter ones were taken as realistic starting points to assess stability and dynamic behavior of the complex in explicit lipid-water surrounding. As a result, several groups of tightly packed right-handed structures of the dimer were proposed. They have almost similar helix-helix interface, which includes the motif A(176)xxxG(180)xxxG(184) and agrees well with previous mutagenesis data and preliminary NMR analysis. Molecular dynamics simulations of these structures reveal perfect adaptation of most of them to heterogeneous membrane environment. A remarkable feature of the predicted dimeric structures is the occurrence of a cluster of H-bonded histidine 173 and serines 168 and 172 on the helix interface, near the N-terminus. Because of specific polar interactions between the monomers, this part of the dimer has no such dense packing as the C-terminal one, thus allowing penetration of water from the extramembrane side into the membrane interior. We propose that the ionization state of His(173) can mediate structural and dynamic properties of the dimer. This, in turn, may be related to pH-dependent proapoptotic activity of BNIP3, which is triggering on by acidosis appearing under hypoxic conditions.


Subject(s)
Membrane Proteins/chemistry , Mitochondrial Membranes/chemistry , Models, Molecular , Protein Multimerization , Protein Structure, Secondary , Proto-Oncogene Proteins/chemistry , Amino Acid Sequence , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/physiology , Cell Death/genetics , Computer Simulation , Gene Deletion , Gene Targeting , Histidine/chemistry , Histidine/genetics , Hydrogen-Ion Concentration , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/physiology , Mitochondrial Membranes/metabolism , Molecular Sequence Data , Protein Structure, Tertiary , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Signal Transduction/physiology
12.
J Chem Inf Model ; 47(3): 1150-62, 2007.
Article in English | MEDLINE | ID: mdl-17371005

ABSTRACT

Integral membrane proteins (MPs) are pharmaceutical targets of exceptional importance. Modern methods of three-dimensional protein structure determination often fail to supply the fast growing field of structure-based drug design with the requested MPs' structures. That is why computational modeling techniques gain a special importance for these objects. Among the principal difficulties limiting application of these methods is the low quality of the MPs' models built in silico. In this series of two papers we present a computational approach to the assessment of the packing "quality" of transmembrane (TM) alpha-helical domains in proteins. The method is based on the concept of protein environment classes, whereby each amino acid residue is described in terms of its environment polarity and accessibility to the membrane. In the first paper we analyze a nonredundant set of 26 TM alpha-helical domains and compute the residues' propensities to five predefined classes of membrane-protein environments. Here we evaluate the proposed approach only by various test sets, cross-validation protocols and ability of the method to delimit the crystal structure of visual rhodopsin, and a number of its erroneous theoretical models. More advanced validation of the method is given in the second article of this series. We assume that the developed "membrane score" method will be helpful in optimizing computer models of TM domains of MPs, especially G-protein coupled receptors.


Subject(s)
Cell Membrane/chemistry , Protein Structure, Secondary , Models, Molecular , Protein Folding , Rhodopsin/chemistry
13.
J Chem Inf Model ; 47(3): 1163-70, 2007.
Article in English | MEDLINE | ID: mdl-17371006

ABSTRACT

We describe a set of tests designed to check the ability of the new "membrane score" method (see the first paper of this series) to assess the packing quality of transmembrane (TM) alpha-helical domains in proteins. The following issues were addressed: (1) Whether there is a relation between the score (S(mem)) of a model and its closeness to the "nativelike" conformation? (2) Is it possible to recognize a correct model among misfolded and erroneous ones? (3) To what extent the score of a homology-built model is sensitive to errors in sequence alignment? To answer the first question, two test cases were considered: (i) Several models of bovine aquaporin-1 (target protein) were built on the structural templates provided by its homologs with known X-ray structure. (ii) Side chains in the spatial models of visual rhodopsin and cytochrome c oxidase were rebuilt based on the backbone scaffolds taken from their crystal structures, and the resulting models were iteratively fitted into the full-atom X-ray conformations. It was shown that the higher the S(mem) value of a model is, the lower its root-mean-square deviation is from the "correct" (crystal) structure of a target. Furthermore, the "membrane score" method successfully identifies the rhodopsin crystal structure in an ensemble of "rotamer-type" decoys, thus providing the way to optimize mutual orientations of alpha-helices in models of TM domains. Finally, being applied to a set of homology models of rhodopsin built on its crystal structure with systematically shifted alignment, the approach demonstrates a prominent ability to detect alignment errors. We therefore assume that the "membrane score" method will be helpful in optimization of in silico models of TM domains in proteins, especially those in GPCRs.


Subject(s)
Cell Membrane/chemistry , Protein Structure, Secondary , Animals , Aquaporin 1/chemistry , Cattle , Reproducibility of Results
14.
J Comput Aided Mol Des ; 20(1): 27-45, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16775778

ABSTRACT

Self-association of two hydrophobic alpha-helices is studied via unrestrained Monte Carlo (MC) simulations in a hydrophobic slab described by an effective potential. The system under study represents two transmembrane (TM) segments of human glycophorin A (GpA), which form homo-dimers in membranes. The influence of TM electrostatic potential, thickness and hydrophobicity degree of lipid bilayer is investigated. It is shown that the membrane environment stabilizes alpha-helical conformation of GpA monomers, induces their TM insertion and facilitates inter-helical contacts. Head-to-head orientation of the helices is promoted by the voltage difference across the membrane. Subsequent "fine-tuned" assembling of the dimer is mediated by van der Waals interactions. Only the models of dimer, calculated in a hydrophobic slab with applied voltage agree with experimental data, while simulations in vacuo or without TM voltage fail to give reasonable results. The moderate structural heterogeneity of GpA dimers (existence of several groups of states with close energies) is proposed to reflect their equilibrium dynamics in membrane-mimics. The calculations performed for GpA mutants G83A and G86L permit rationalization of mutagenesis data for them. The results of Monte Carlo simulations critically depend on the parameters of the membrane model: adequate description of helix association is achieved in the water-cyclohexane-water system with the membrane thickness 30-34 A, while in membranes with different hydrophobicities and thickness unrealistic conformations of the dimer are found. The computational approach permits efficient prediction of TM helical oligomers based solely on the sequences of interacting peptides.


Subject(s)
Membrane Proteins/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Computational Biology , Computer Simulation , Dimerization , Glycophorins/chemistry , Glycophorins/metabolism , Humans , Membrane Proteins/metabolism , Models, Molecular , Protein Folding
15.
J Phys Chem B ; 109(31): 15052-9, 2005 Aug 11.
Article in English | MEDLINE | ID: mdl-16852905

ABSTRACT

Anionic unsaturated lipid bilayers represent suitable model systems that mimic real cell membranes: they are fluid and possess a negative surface charge. Understanding of detailed molecular organization of water-lipid interfaces in such systems may provide an important insight into the mechanisms of proteins' binding to membranes. Molecular dynamics (MD) of full-atom hydrated lipid bilayers is one of the most powerful tools to address this problem in silico. Unfortunately, wide application of computational methods for such systems is limited by serious technical problems. They are mainly related to correct treatment of long-range electrostatic effects. In this study a physically reliable model of an anionic unsaturated bilayer of 1,2-dioleoyl-sn-glycero-3-phosphoserine (DOPS) was elaborated and subjected to long-term MD simulations. Electrostatic interactions were treated with two different algorithms: spherical cutoff function and particle-mesh Ewald summation (PME). To understand the role of lipid charge in the system behavior, similar calculations were also carried out for zwitterionic bilayer composed of 1,2-dioleoyl-sn-glycero-3-phosphocholine (DOPC). It was shown that, for the charged DOPS bilayer, the PME protocol performs much better than the cutoff scheme. In the last case a number of artifacts in the structural organization of the bilayer were observed. All of them were attributed to inadequate treatment of electrostatic interactions of lipid headgroups with counterions. Electrostatic properties, along with structural and dynamic parameters, of both lipid bilayers were investigated. Comparative analysis of the MD data reveals that the water-lipid interface of the DOPC bilayer is looser than that for DOPS. This makes possible deeper penetration of water molecules inside the zwitterionic (DOPC) bilayer, where they strongly interact with carbonyls of lipids. This can lead to thickening of the membrane interface in zwitterionic as compared to negatively charged bilayers.


Subject(s)
Computer Simulation , Lipid Bilayers/chemistry , Phosphatidylserines/chemistry , Water/chemistry , Electrons , Ions/chemistry , Models, Molecular , Molecular Structure , Sodium/chemistry , Static Electricity
16.
J Chem Theory Comput ; 1(6): 1252-64, 2005 Nov.
Article in English | MEDLINE | ID: mdl-26631669

ABSTRACT

We describe one of the first attempts at unrestrained modeling of self-association of α-helices in implicit heterogeneous membrane-mimic media. The computational approach is based on the Monte Carlo conformational search for peptides in dihedral angles space. The membrane is approximated by an effective potential. The method is tested in calculations of two hydrophobic segments of human glycophorin A (GpA), known to form membrane-spanning dimers in real lipid bilayers. Our main findings may be summarized as follows. Modeling in vacuo does not adequately describe the behavior of GpA helices, failing to reproduce experimental structural data. The membrane environment stabilizes α-helical conformation of GpA monomers, inducing their transmembrane insertion and facilitating interhelical contacts. The voltage difference across the membrane promotes "head-to-head" orientation of the helices. "Fine-tuning" of the monomers in a complex is shown to be regulated by van der Waals interactions. Detailed exploration of conformational space of the system starting from arbitrary locations of two noninteracting helices reveals only several groups of energetically favorable structures. All of them represent tightly packed transmembrane helical dimers. In overall, they agree reasonably well with mutagenesis data, some of them are close to NMR-derived structures. A possibility of left-handed dimers is discussed. We assume that the observed moderate structural heterogeneity (the existence of several groups of states with close energies) reflects a real equilibrium dynamics of the monomers [Formula: see text] at least in membrane mimics used in experimental studies of GpA. The elaborated computational approach is universal and may be employed in studies of a wide class of membrane peptides and proteins.

17.
Biochem J ; 382(Pt 1): 293-305, 2004 Aug 15.
Article in English | MEDLINE | ID: mdl-15147237

ABSTRACT

WNDP (Wilson's disease protein) is a copper-transporting ATPase that plays an essential role in human physiology. Mutations in WNDP result in copper accumulation in tissues and cause a severe hepato-neurological disorder known as Wilson's disease. Several mutations were surmised to affect the nucleotide binding and hydrolysis by WNDP; however, how the nucleotides bind to normal and mutated WNDP remains unknown. To aid such studies, we performed the molecular modelling of the spatial structure and dynamics of the ATP-binding domain of WNDP and its interactions with ATP. The three-dimensional models of this domain in two conformations were built using the X-ray structures of the Ca2+-ATPase in the E1 and E2 states. To study the functional aspects of the models, they were subjected to long-term molecular dynamics simulations in an explicit solvent; similar calculations were performed for the ATP-binding domain of Ca2+-ATPase. In both cases, we found large-scale motions that lead to significant changes of distances between several functionally important residues. The ATP docking revealed two possible modes of ATP binding: via adenosine buried in the cleft near residues H1069, R1151 and D1164, and via phosphate moiety 'anchored' by H-bonds with residues in the vicinity of catalytic D1027. Furthermore, interaction of ATP with both sites occurs if they are spatially close to each other. This may be achieved after relative domain motions of the 'closure' type observed in molecular dynamics simulations. The results provide a framework for analysis of disease mutations and for future mutagenesis studies.


Subject(s)
Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphate/metabolism , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Hepatolenticular Degeneration/genetics , Models, Molecular , Mutation/physiology , Nucleotides/metabolism , Peptides/chemistry , Peptides/metabolism , Adenosine Triphosphatases/metabolism , Adenosine Triphosphatases/physiology , Amino Acid Sequence/genetics , Binding Sites , Calcium-Transporting ATPases/metabolism , Cation Transport Proteins/metabolism , Cation Transport Proteins/physiology , Circular Dichroism/methods , Computer Simulation , Copper-Transporting ATPases , Hepatolenticular Degeneration/pathology , Models, Structural , Molecular Sequence Data , Mutagenesis/genetics , Mutagenesis/physiology , Mutation/genetics , Protein Binding , Protein Structure, Tertiary
18.
FEBS Lett ; 526(1-3): 97-100, 2002 Aug 28.
Article in English | MEDLINE | ID: mdl-12208512

ABSTRACT

Transmembrane potentials play important but poorly understood roles in many biological processes, including signal sequence-mediated protein translocation across bacterial membranes. In this study we applied Monte Carlo techniques to simulate the way the potential acts on a signal sequence. The simulations demonstrate that in the absence of a potential the signal sequence prefers insertion in both helical hairpin and transmembrane alpha-helical conformations. However, in the presence of a potential only the transmembrane alpha-helical conformation is the state of lowest energy for the signal sequence. From these results it is concluded that the membrane potential stabilizes the transmembrane orientation of a signal sequence, explaining the membrane potential dependence of preprotein translocation.


Subject(s)
Membrane Potentials/physiology , Proteins/metabolism , Amino Acid Sequence , Computer Simulation , Molecular Sequence Data , Monte Carlo Method , Peptides/chemistry , Peptides/metabolism , Protein Conformation , Protein Transport , Thermodynamics
19.
Biophys J ; 83(1): 144-53, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12080107

ABSTRACT

Incorporation of beta-sheet proteins into membrane is studied theoretically for the first time, and the results are validated by the direct experimental data. Using Monte Carlo simulations with implicit membrane, we explore spatial structure, energetics, polarity, and mode of insertion of two cardiotoxins with different membrane-destabilizing activity. Both proteins, classified as P- and S-type cardiotoxins, are found to retain the overall "three-finger" fold interacting with membrane core and lipid/water interface by the tips of the "fingers" (loops). The insertion critically depends upon the structure, hydrophobicity, and electrostatics of certain regions. The simulations reveal apparently distinct binding modes for S- and P-type cardiotoxins via the first loop or through all three loops, respectively. This rationalizes an earlier empirical classification of cardiotoxins into S- and P-type, and provides a basis for the analysis of experimental data on their membrane affinities. Accomplished with our previous simulations of membrane alpha-helices, the computational method may be used to study partitioning of proteins with diverse folds into lipid bilayers.


Subject(s)
Cell Membrane/metabolism , Cobra Cardiotoxin Proteins/chemistry , Amino Acid Sequence , Animals , Biophysics/methods , Cell Membrane/chemistry , Elapidae , Lipid Bilayers , Micelles , Models, Molecular , Molecular Sequence Data , Monte Carlo Method , Protein Binding , Protein Conformation , Protein Structure, Secondary , Sequence Homology, Amino Acid , Software
SELECTION OF CITATIONS
SEARCH DETAIL
...