Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
1.
J Comput Chem ; 44(25): 1976-1985, 2023 09 30.
Article in English | MEDLINE | ID: mdl-37352129

ABSTRACT

Understanding the molecular basis for protein stability requires a thermodynamic analysis of protein folding. Thermodynamic analysis is often performed by sampling many atomistic conformations using molecular simulations that employ either explicit or implicit water models. However, it remains unclear to what extent thermodynamic results from different solvation models are reliable at the molecular level. In this study, we quantify the influence of both solvation models on folding stability at the individual backbone and side chain resolutions. We assess the residue-specific folding free energy components of a ß-sheet protein and a helical protein using trajectories resulting from TIP3P explicit and generalized Born/surface area implicit solvent simulations of model proteins. We found that the thermodynamic discrepancy due to the implicit solvent mostly originates from charged side chains, followed by the under-stabilized hydrophobic ones. In contrast, the contributions of backbone residue in both proteins were comparable for explicit and implicit water models. Our study lays out the foundation for detailed thermodynamic assessment of solvation models in the context of protein simulation.


Subject(s)
Protein Folding , Proteins , Proteins/chemistry , Thermodynamics , Computer Simulation , Solvents/chemistry , Water/chemistry
2.
Proteins ; 91(5): 694-704, 2023 05.
Article in English | MEDLINE | ID: mdl-36564921

ABSTRACT

Understanding how protein-protein binding affinity is determined from molecular interactions at the interface is essential in developing protein therapeutics such as antibodies, but this has not yet been fully achieved. Among the major difficulties are the facts that it is generally difficult to decompose thermodynamic quantities into contributions from individual molecular interactions and that the solvent effect-dehydration penalty-must also be taken into consideration for every contact formation at the binding interface. Here, we present an atomic-level thermodynamics analysis that overcomes these difficulties and illustrate its utility through application to SARS-CoV-2 neutralizing antibodies. Our analysis is based on the direct interaction energy computed from simulated antibody-protein complex structures and on the decomposition of solvation free energy change upon complex formation. We find that the formation of a single contact such as a hydrogen bond at the interface barely contributes to binding free energy due to the dehydration penalty. On the other hand, the simultaneous formation of multiple contacts between two interface residues favorably contributes to binding affinity. This is because the dehydration penalty is significantly alleviated: the total penalty for multiple contacts is smaller than a sum of what would be expected for individual dehydrations of those contacts. Our results thus provide a new perspective for designing protein therapeutics of improved binding affinity.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/metabolism , Dehydration , Thermodynamics , Antibodies, Viral/metabolism , Protein Binding , Antibodies, Neutralizing/chemistry
3.
J Comput Chem ; 44(9): 1002-1009, 2023 04 05.
Article in English | MEDLINE | ID: mdl-36571461

ABSTRACT

The question of whether amino acids critical to protein folding kinetics are evolutionarily conserved has been investigated intensively in the past, but no consensus has yet been reached. Recently, we have demonstrated that the transition state, dictating folding kinetics, is characterized as the state of maximum dynamic cooperativity, i.e., the state of maximum correlations between amino acid contact formations. Here, we investigate the evolutionary conservation of those amino acids contributing significantly to the dynamic cooperativity. We find a strong indication of a new kind of relationship-necessary but not sufficient causality-between the evolutionary conservation and the dynamic cooperativity: larger contributions to the dynamic cooperativity arise from more conserved residues, but not vice versa. This holds for all the protein systems for which long folding simulation trajectories are available. To our knowledge, this is the first systematic demonstration of any kind of evolutionary conservation of amino acids relevant to folding kinetics.


Subject(s)
Amino Acids , Proteins , Amino Acids/chemistry , Proteins/chemistry , Protein Folding , Kinetics , Protein Conformation
4.
Biochim Biophys Acta Proteins Proteom ; 1870(3): 140746, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34942360

ABSTRACT

Mutations in the fasciclin 1 domain 4 (FAS1-4) of transforming growth factor ß-induced protein (TGFBIp) are associated with insoluble extracellular deposits and corneal dystrophies (CDs). The decrease in solubility upon mutation has been implicated in CD; however, the exact molecular mechanisms are not well understood. Here, we performed molecular dynamics simulations followed by solvation thermodynamic analyses of the FAS1-4 domain and its three mutants-R555W, R555Q, and A546T-linked to granular corneal dystrophy type 1, Thiel-Behnke corneal dystrophy and lattice corneal dystrophy, respectively. We found that both R555W and R555Q mutants have less affinity toward solvent water relative to the wild-type protein. In the R555W mutant, a remarkable increase in solvation free energy was observed because of the structural changes near the mutation site. The mutation site W555 is buried in other hydrophobic residues, and R557 simultaneously forms salt bridges with E554 and D561. In the R555Q mutant, the increase in solvation free energy is caused by structural rearrangements far from the mutation site. R558 separately forms salt bridges with D575, E576, and E598. Thus, we thus identified the relationship between the decrease in solubility and conformational changes caused by mutations, which may be useful in designing potential therapeutics and in blocking FAS1 aggregation related to CD.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Corneal Dystrophies, Hereditary/genetics , Extracellular Matrix Proteins/genetics , Mutation , Transforming Growth Factor beta/genetics , Amyloid/chemistry , Amyloid/metabolism , Cell Adhesion Molecules, Neuronal/chemistry , Corneal Dystrophies, Hereditary/metabolism , Extracellular Matrix Proteins/chemistry , Humans , Molecular Dynamics Simulation , Molecular Structure , Protein Aggregation, Pathological/metabolism , Solubility , Transforming Growth Factor beta/chemistry
5.
J Phys Chem B ; 125(26): 7108-7116, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34165991

ABSTRACT

The native structure of a protein is stabilized by a number of interactions such as main-chain hydrogen bonds and side-chain hydrophobic contacts. However, it has been challenging to determine how these interactions contribute to protein stability at single amino acid resolution. Here, we quantified site-specific thermodynamic stability at the molecular level to extend our understanding of the stabilizing forces in protein folding. We derived the free energy components of individual amino acid residues separately for the folding of the human Pin WW domain based on simulated structures. A further decomposition of the thermodynamic properties into contributions from backbone and side-chain groups enabled us to identify the critical residues in the secondary structure and hydrophobic core formation, without introducing physical modifications to the system as in site-directed mutagenesis methods. By relating the structural and thermodynamic changes upon folding for each residue, we find that the simultaneous formation of the backbone hydrogen bonds and side-chain contacts cooperatively stabilizes the folded structure. The identification of stabilizing interactions in a folding protein at atomic resolution will provide molecular insights into understanding the origin of the protein structure and into engineering a more stable protein.


Subject(s)
Protein Folding , Humans , Hydrogen Bonding , Models, Molecular , Protein Structure, Secondary , Thermodynamics , WW Domains
6.
Chem Sci ; 12(16): 5944-5951, 2021 Apr 28.
Article in English | MEDLINE | ID: mdl-35342544

ABSTRACT

Cooperativity is considered to be a key organizing principle behind biomolecular assembly, recognition and folding. However, it has remained very challenging to quantitatively characterize how cooperative processes occur on a concerted, multiple-interaction basis. Here, we address how and when the folding process is cooperative on a molecular scale. To this end, we analyze multipoint time-correlation functions probing time-dependent communication between multiple amino acids, which were computed from long folding simulation trajectories. We find that the simultaneous multiple amino-acid contact formation, which is absent in the unfolded state, starts to develop only upon entering the folding transition path. Interestingly, the transition state, whose presence is connected to the macrostate cooperative behavior known as the two-state folding, can be identified as the state in which the amino-acid cooperativity is maximal. Thus, our work not only provides a new mechanistic view on how protein folding proceeds on a multiple-interaction basis, but also offers a conceptually novel characterization of the folding transition state and the molecular origin of the phenomenological cooperative folding behavior. Moreover, the multipoint correlation function approach adopted here is general and can be used to expand the understanding of cooperative processes in complex chemical and biomolecular systems.

7.
Sci Rep ; 10(1): 21191, 2020 12 03.
Article in English | MEDLINE | ID: mdl-33273506

ABSTRACT

Protein aggregation is a major concern in biotherapeutic applications of monoclonal antibodies. Introducing charged mutations is among the promising strategies to improve aggregation resistance. However, the impact of such mutations on solubilizing activity depends largely on the inserting location, whose mechanism is still not well understood. Here, we address this issue from a solvation viewpoint, and this is done by analyzing how the change in solvation free energy upon charged mutation is composed of individual contributions from constituent residues. To this end, we perform molecular dynamics simulations for a number of antibody mutants and carry out the residue-wise decomposition of the solvation free energy. We find that, in addition to the previously identified "global" principle emphasizing the key role played by the protein total net charge, a local net charge within [Formula: see text]15 Å from the mutation site exerts significant effects. For example, when the net charge of an antibody is positive, the global principle states that introducing a positively charged mutation will lead to more favorable solvation. Our finding further adds that an even more optimal mutation can be done at the site around which more positively charged residues and fewer negatively charged residues are present. Such a "local" design principle accounts for the location dependence of charged mutations, and will be useful in producing aggregation-resistant antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Mutation , Protein Aggregates , Antibodies, Monoclonal/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Molecular Dynamics Simulation , Solubility
8.
PLoS One ; 15(4): e0232613, 2020.
Article in English | MEDLINE | ID: mdl-32353067

ABSTRACT

Inactivation of the tumor suppressor p53 resulting from the binding with a negative regulator HDM2 is among the predominant defects in human cancers. p53-mimicking peptides whose conformational and proteolytic stability is enhanced by an all-hydrocarbon staple are being recognized as promising anticancer agents for disrupting the p53-HDM2 binding and reactivating p53. Herein, we conduct a computational modeling and thermodynamic characterization of stapled p53/HDM2 complex via molecular docking, simulations, and binding free energy analysis. The binding thermodynamics analysis is done based on the end-point calculation of the effective binding energy-a sum of the direct peptide-protein interaction energy and the dehydration penalty-and on its decomposition into contributions from specific groups constituting the complex. This allows us to investigate how individual amino acids in the stapled p53 and HDM2 contribute to the binding affinity. We find that not only the epitope residues (F19, W23 and L26), but also the hydrocarbon linker of the stapled p53 impart significant contributions. Our computational approach will be useful in designing new stapled peptides in which the staple location is also optimized to improve the binding affinity.


Subject(s)
Peptides/chemistry , Proto-Oncogene Proteins c-mdm2/chemistry , Tumor Suppressor Protein p53/chemistry , Amino Acids/chemistry , Molecular Docking Simulation , Peptides/metabolism , Protein Binding , Proto-Oncogene Proteins c-mdm2/metabolism , Thermodynamics , Tumor Suppressor Protein p53/metabolism
9.
Chem Commun (Camb) ; 55(88): 13311-13314, 2019 Oct 31.
Article in English | MEDLINE | ID: mdl-31631199

ABSTRACT

Herein, we report a strategy for generating conformationally restricted α-helix mimetic small molecules by introducing covalent bridges that limit rotation about the central axis of α-helix mimetics. We demonstrate that the bridged α-helix mimetics have enhanced binding affinity and specificity to the target protein due to the restricted conformation as well as extra interaction of the bridge with the protein surface.


Subject(s)
Heterocyclic Compounds, Bridged-Ring/chemistry , Myeloid Cell Leukemia Sequence 1 Protein/chemistry , Small Molecule Libraries/chemistry , Heterocyclic Compounds, Bridged-Ring/pharmacology , Humans , Jurkat Cells , Models, Molecular , Molecular Conformation , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Small Molecule Libraries/pharmacology
10.
Sci Rep ; 9(1): 14927, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31624293

ABSTRACT

Folding funnel is the essential concept of the free energy landscape for ordered proteins. How does this concept apply to intrinsically disordered proteins (IDPs)? Here, we address this fundamental question through the explicit characterization of the free energy landscapes of the representative α-helical (HP-35) and ß-sheet (WW domain) proteins and of an IDP (pKID) that folds upon binding to its partner (KIX). We demonstrate that HP-35 and WW domain indeed exhibit the steep folding funnel: the landscape slope for these proteins is ca. -50 kcal/mol, meaning that the free energy decreases by ~5 kcal/mol upon the formation of 10% native contacts. On the other hand, the landscape of pKID is funneled but considerably shallower (slope of -24 kcal/mol), which explains why pKID is disordered in free environments. Upon binding to KIX, the landscape of pKID now becomes significantly steep (slope of -54 kcal/mol), which enables otherwise disordered pKID to fold. We also show that it is the pKID-KIX intermolecular interactions originating from hydrophobic residues that mainly confer the steep folding funnel. The present work not only provides the quantitative characterization of the protein folding free energy landscape, but also establishes the usefulness of the folding funnel concept to IDPs.


Subject(s)
Intrinsically Disordered Proteins/metabolism , Models, Molecular , Protein Folding , Entropy , Feasibility Studies , Hydrophobic and Hydrophilic Interactions , Intrinsically Disordered Proteins/chemistry , Kinetics , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand
11.
ACS Cent Sci ; 5(8): 1342-1351, 2019 Aug 28.
Article in English | MEDLINE | ID: mdl-31482116

ABSTRACT

The most fundamental aspect of the free energy landscape of proteins is that it is globally funneled such that protein folding is energetically biased. Then, what are the distinctive characteristics of the landscape of intrinsically disordered proteins, apparently lacking such energetic bias, that nevertheless fold upon binding? Here, we address this fundamental issue through the explicit characterization of the free energy landscape of the paradigmatic pKID-KIX system (pKID, phosphorylated kinase-inducible domain; KIX, kinase interacting domain). This is done based on unguided, fully atomistic, explicit-water molecular dynamics simulations with an aggregated simulation time of >30 µs and on the computation of the free energy that defines the landscape. We find that, while the landscape of pKID before binding is considerably shallower than the one for a protein that autonomously folds, it becomes progressively more funneled as the binding of pKID with KIX proceeds. This explains why pKID is disordered in a free state, and the binding of pKID with KIX is a prerequisite for pKID's folding. In addition, we observe that the key event in completing the pKID-KIX coupled folding and binding is the directed self-assembly where pKID is docked upon the KIX surface to maximize the surface electrostatic complementarity, which, in turn, require pKID to adopt the correct folded structure. This key process shows up as the free energy barrier in the pKID landscape separating the intermediate nonspecific complex state and the specific complex state. The present work not only provides a detailed molecular picture of the coupled folding and binding of pKID but also expands the funneled landscape perspective to intrinsically disordered proteins.

12.
ACS Nano ; 13(8): 8766-8783, 2019 08 27.
Article in English | MEDLINE | ID: mdl-31310506

ABSTRACT

Complex amyloid aggregation of amyloid-ß (1-40) (Aß1-40) in terms of monomer structures has not been fully understood. Herein, we report the microscopic mechanism and pathways of Aß1-40 aggregation with macroscopic viewpoints through tuning its initial structure and solubility. Partial helical structures of Aß1-40 induced by low solvent polarity accelerated cytotoxic Aß1-40 amyloid fibrillation, while predominantly helical folds did not aggregate. Changes in the solvent polarity caused a rapid formation of ß-structure-rich protofibrils or oligomers via aggregation-prone helical structures. Modulation of the pH and salt concentration transformed oligomers to protofibrils, which proceeded to amyloid formation. We reveal diverse molecular mechanisms underlying Aß1-40 aggregation with conceptual energy diagrams and propose that aggregation-prone partial helical structures are key to inducing amyloidogenesis. We demonstrate that context-dependent protein aggregation is comprehensively understood using the macroscopic phase diagram, which provides general insights into differentiation of amyloid formation and phase separation from unfolded and folded structures.


Subject(s)
Alzheimer Disease/genetics , Amyloid beta-Peptides/ultrastructure , Peptide Fragments/ultrastructure , Protein Aggregation, Pathological/genetics , Protein Conformation, alpha-Helical/genetics , Alzheimer Disease/pathology , Amyloid/chemistry , Amyloid/genetics , Amyloid beta-Peptides/chemistry , Humans , Peptide Fragments/chemistry , Protein Conformation, beta-Strand/genetics , Protein Folding/drug effects , Protein Stability/drug effects , Signal Transduction/drug effects , Solubility
13.
Biochem Biophys Res Commun ; 510(3): 442-448, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30722990

ABSTRACT

The self-assembly of amyloid-beta (Aß) proteins in aqueous extracellular environments is implicated in Alzheimer's disease. Among several alloforms of Aß proteins differing in sequence length, the 42- and 40-residue forms (Aß42 and Aß40) are the most abundant ones in the human body. Although the only difference is the additional I41A42 residues in the C-terminus, Aß42 exhibits more aggregation tendency and stronger neurotoxicity than Aß40. Here, we investigate the molecular factors that confer more aggregation potential to Aß42 than to Aß40 based on molecular dynamics simulations combined with solvation thermodynamic analyses. It is observed that the most salient structural feature of Aß42 relative to Aß40 is the more enhanced ß-sheet forming tendency, in particular in the C-terminal region. While such a structural characteristic of Aß42 will certainly serve to facilitate the formation of aggregate species rich in ß-sheet structure, we also detect its interesting thermodynamic consequence. Indeed, we find from the decomposition analysis that the C-terminal region substantially increases the solvation free energy (i.e., overall "hydrophobicity") of Aß42, which is caused by the dehydration of the backbone moieties showing the enhanced tendency of forming the ß-structure. Together with the two additional hydrophobic residues (I41A42), this leads to the higher solvation free energy of Aß42, implying the larger water-mediated attraction toward the self-assembly. Thus, our computational results provide structural and thermodynamic grounds on why Aß42 has more aggregation propensity than Aß40 in aqueous environments.


Subject(s)
Amyloid beta-Peptides/chemistry , Peptide Fragments/chemistry , Molecular Dynamics Simulation , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Structure, Secondary , Thermodynamics
14.
Sci Rep ; 8(1): 8269, 2018 May 24.
Article in English | MEDLINE | ID: mdl-29799017

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

15.
Sci Rep ; 8(1): 7148, 2018 05 08.
Article in English | MEDLINE | ID: mdl-29740018

ABSTRACT

Dimensionality reduction with a suitable choice of order parameters or reaction coordinates is commonly used for analyzing high-dimensional time-series data generated by atomistic biomolecular simulations. So far, geometric order parameters, such as the root mean square deviation, fraction of native amino acid contacts, and collective coordinates that best characterize rare or large conformational transitions, have been prevailing in protein folding studies. Here, we show that the solvent-averaged effective energy, which is a thermodynamic quantity but unambiguously defined for individual protein conformations, serves as a good order parameter of protein folding. This is illustrated through the application to the folding-unfolding simulation trajectory of villin headpiece subdomain. We rationalize the suitability of the effective energy as an order parameter by the funneledness of the underlying protein free energy landscape. We also demonstrate that an improved conformational space discretization is achieved by incorporating the effective energy. The most distinctive feature of this thermodynamic order parameter is that it works in pointing to near-native folded structures even when the knowledge of the native structure is lacking, and the use of the effective energy will also find applications in combination with methods of protein structure prediction.


Subject(s)
Protein Conformation , Protein Folding , Proteins/chemistry , Thermodynamics , Entropy , Models, Molecular , Proteins/metabolism , Solvents/chemistry
16.
Sci Rep ; 7(1): 8744, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28821854

ABSTRACT

Interfacial waters are considered to play a crucial role in protein-protein interactions, but in what sense and why are they important? Here, using molecular dynamics simulations and statistical thermodynamic analyses, we demonstrate distinctive dynamic characteristics of the interfacial water and investigate their implications for the binding thermodynamics. We identify the presence of extraordinarily slow (~1,000 times slower than in bulk water) hydrogen-bond rearrangements in interfacial water. We rationalize the slow rearrangements by introducing the "trapping" free energies, characterizing how strongly individual hydration waters are captured by the biomolecular surface, whose magnitude is then traced back to the number of water-protein hydrogen bonds and the strong electrostatic field produced at the binding interface. We also discuss the impact of the slow interfacial waters on the binding thermodynamics. We find that, as expected from their slow dynamics, the conventional approach to the water-mediated interaction, which assumes rapid equilibration of the waters' degrees of freedom, is inadequate. We show instead that an explicit treatment of the extremely slow interfacial waters is critical. Our results shed new light on the role of water in protein-protein interactions, highlighting the need to consider its dynamics to improve our understanding of biomolecular bindings.


Subject(s)
Molecular Dynamics Simulation , Multiprotein Complexes/chemistry , Proteins/chemistry , Thermodynamics , Water/chemistry , Algorithms , Hydrogen Bonding , Protein Binding , Protein Conformation , Structure-Activity Relationship
17.
Mol Cell ; 67(2): 334-347.e5, 2017 Jul 20.
Article in English | MEDLINE | ID: mdl-28689660

ABSTRACT

Multi-subunit SMC complexes control chromosome superstructure and promote chromosome disjunction, conceivably by actively translocating along DNA double helices. SMC subunits comprise an ABC ATPase "head" and a "hinge" dimerization domain connected by a 49 nm coiled-coil "arm." The heads undergo ATP-dependent engagement and disengagement to drive SMC action on the chromosome. Here, we elucidate the architecture of prokaryotic Smc dimers by high-throughput cysteine cross-linking and crystallography. Co-alignment of the Smc arms tightly closes the interarm space and misaligns the Smc head domains at the end of the rod by close apposition of their ABC signature motifs. Sandwiching of ATP molecules between Smc heads requires them to substantially tilt and translate relative to each other, thereby opening up the Smc arms. We show that this mechanochemical gating reaction regulates chromosome targeting and propose a mechanism for DNA translocation based on the merging of DNA loops upon closure of Smc arms.


Subject(s)
Bacillus subtilis/metabolism , Bacterial Proteins/metabolism , Cell Cycle Proteins/metabolism , Chromosome Segregation , Chromosomes, Bacterial , Adenosine Triphosphate/metabolism , Bacillus subtilis/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Binding Sites , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Crystallography, X-Ray , Cysteine , High-Throughput Screening Assays , Models, Molecular , Mutation , Nucleic Acid Conformation , Protein Conformation , Protein Multimerization , Protein Stability , Structure-Activity Relationship
18.
Annu Rev Phys Chem ; 68: 117-134, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28226222

ABSTRACT

The investigation of intrinsically disordered proteins (IDPs) is a new frontier in structural and molecular biology that requires a new paradigm to connect structural disorder to function. Molecular dynamics simulations and statistical thermodynamics potentially offer ideal tools for atomic-level characterizations and thermodynamic descriptions of this fascinating class of proteins that will complement experimental studies. However, IDPs display sensitivity to inaccuracies in the underlying molecular mechanics force fields. Thus, achieving an accurate structural characterization of IDPs via simulations is a challenge. It is also daunting to perform a configuration-space integration over heterogeneous structural ensembles sampled by IDPs to extract, in particular, protein configurational entropy. In this review, we summarize recent efforts devoted to the development of force fields and the critical evaluations of their performance when applied to IDPs. We also survey recent advances in computational methods for protein configurational entropy that aim to provide a thermodynamic link between structural disorder and protein activity.


Subject(s)
Intrinsically Disordered Proteins/chemistry , Animals , Entropy , Humans , Molecular Dynamics Simulation , Protein Conformation , Thermodynamics , Water/chemistry
19.
J Phys Chem Lett ; 7(19): 3967-3972, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27660882

ABSTRACT

Confined water often exhibits anomalous properties not observable in the bulk phase. Although water in hydrophobic confinement has been the focus of intense investigation, the behavior of water confined between hydrophilic surfaces, which are more frequently found in biological systems, has not been fully explored. Here, we investigate using molecular dynamics simulations dynamical properties of the water confined in hydrophilic protein-protein and protein-DNA interfaces. We find that the interfacial water exhibits glassy slow relaxations even at 300 K. In particular, the rotational dynamics show a logarithmic decay that was observed in glass-forming liquids at deeply supercooled states. We argue that such slow water dynamics are indeed induced by the hydrophilic binding surfaces, which is in opposition to the picture that the hydration water slaves protein motions. Our results will significantly impact the view on the role of water in biomolecular interactions.


Subject(s)
DNA/chemistry , Proteins/chemistry , Water/chemistry , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , DNA/metabolism , Hydrophobic and Hydrophilic Interactions , Lac Repressors/chemistry , Lac Repressors/metabolism , Molecular Dynamics Simulation , Nucleic Acid Conformation , Protein Structure, Tertiary , Proteins/metabolism , Ribonucleases/chemistry , Ribonucleases/metabolism
20.
Angew Chem Int Ed Engl ; 55(36): 10612-5, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27467415

ABSTRACT

The design of inhibitors of intracellular protein-protein interactions (PPIs) remains a challenge in chemical biology and drug discovery. We propose a cyclized helix-loop-helix (cHLH) peptide as a scaffold for generating cell-permeable PPI inhibitors through bifunctional grafting: epitope grafting to provide binding activity, and arginine grafting to endow cell-permeability. To inhibit p53-HDM2 interactions, the p53 epitope was grafted onto the C-terminal helix and six Arg residues were grafted onto another helix. The designed peptide cHLHp53-R showed high inhibitory activity for this interaction, and computational analysis suggested a binding mode for HDM2. Confocal microscopy of cells treated with fluorescently labeled cHLHp53-R revealed cell membrane penetration and cytosolic localization. The peptide inhibited the growth of HCT116 and LnCap cancer cells. This strategy of bifunctional grafting onto a well-structured peptide scaffold could facilitate the generation of inhibitors for intracellular PPIs.


Subject(s)
Arginine/analogs & derivatives , Arginine/pharmacology , Drug Design , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Protein Interaction Maps/drug effects , Amino Acid Sequence , Cell Line, Tumor , Humans , Molecular Docking Simulation , Protein Conformation, alpha-Helical , Protein Interaction Mapping , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Proto-Oncogene Proteins c-mdm2/chemistry , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/chemistry , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...