Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Brief Bioinform ; 25(3)2024 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-38581420

RESUMO

Protein-ligand interaction prediction presents a significant challenge in drug design. Numerous machine learning and deep learning (DL) models have been developed to accurately identify docking poses of ligands and active compounds against specific targets. However, current models often suffer from inadequate accuracy or lack practical physical significance in their scoring systems. In this research paper, we introduce IGModel, a novel approach that utilizes the geometric information of protein-ligand complexes as input for predicting the root mean square deviation of docking poses and the binding strength (pKd, the negative value of the logarithm of binding affinity) within the same prediction framework. This ensures that the output scores carry intuitive meaning. We extensively evaluate the performance of IGModel on various docking power test sets, including the CASF-2016 benchmark, PDBbind-CrossDocked-Core and DISCO set, consistently achieving state-of-the-art accuracies. Furthermore, we assess IGModel's generalizability and robustness by evaluating it on unbiased test sets and sets containing target structures generated by AlphaFold2. The exceptional performance of IGModel on these sets demonstrates its efficacy. Additionally, we visualize the latent space of protein-ligand interactions encoded by IGModel and conduct interpretability analysis, providing valuable insights. This study presents a novel framework for DL-based prediction of protein-ligand interactions, contributing to the advancement of this field. The IGModel is available at GitHub repository https://github.com/zchwang/IGModel.


Assuntos
Aprendizado Profundo , Proteínas , Proteínas/química , Ligação Proteica , Ligantes , Desenho de Fármacos
2.
Brief Bioinform ; 24(1)2023 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-36502369

RESUMO

The recently reported machine learning- or deep learning-based scoring functions (SFs) have shown exciting performance in predicting protein-ligand binding affinities with fruitful application prospects. However, the differentiation between highly similar ligand conformations, including the native binding pose (the global energy minimum state), remains challenging that could greatly enhance the docking. In this work, we propose a fully differentiable, end-to-end framework for ligand pose optimization based on a hybrid SF called DeepRMSD+Vina combined with a multi-layer perceptron (DeepRMSD) and the traditional AutoDock Vina SF. The DeepRMSD+Vina, which combines (1) the root mean square deviation (RMSD) of the docking pose with respect to the native pose and (2) the AutoDock Vina score, is fully differentiable; thus is capable of optimizing the ligand binding pose to the energy-lowest conformation. Evaluated by the CASF-2016 docking power dataset, the DeepRMSD+Vina reaches a success rate of 94.4%, which outperforms most reported SFs to date. We evaluated the ligand conformation optimization framework in practical molecular docking scenarios (redocking and cross-docking tasks), revealing the high potentialities of this framework in drug design and discovery. Structural analysis shows that this framework has the ability to identify key physical interactions in protein-ligand binding, such as hydrogen-bonding. Our work provides a paradigm for optimizing ligand conformations based on deep learning algorithms. The DeepRMSD+Vina model and the optimization framework are available at GitHub repository https://github.com/zchwang/DeepRMSD-Vina_Optimization.


Assuntos
Aprendizado Profundo , Ligantes , Simulação de Acoplamento Molecular , Proteínas/química , Desenho de Fármacos , Ligação Proteica
3.
Brief Bioinform ; 23(3)2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35289359

RESUMO

Scoring functions are important components in molecular docking for structure-based drug discovery. Traditional scoring functions, generally empirical- or force field-based, are robust and have proven to be useful for identifying hits and lead optimizations. Although multiple highly accurate deep learning- or machine learning-based scoring functions have been developed, their direct applications for docking and screening are limited. We describe a novel strategy to develop a reliable protein-ligand scoring function by augmenting the traditional scoring function Vina score using a correction term (OnionNet-SFCT). The correction term is developed based on an AdaBoost random forest model, utilizing multiple layers of contacts formed between protein residues and ligand atoms. In addition to the Vina score, the model considerably enhances the AutoDock Vina prediction abilities for docking and screening tasks based on different benchmarks (such as cross-docking dataset, CASF-2016, DUD-E and DUD-AD). Furthermore, our model could be combined with multiple docking applications to increase pose selection accuracies and screening abilities, indicating its wide usage for structure-based drug discoveries. Furthermore, in a reverse practice, the combined scoring strategy successfully identified multiple known receptors of a plant hormone. To summarize, the results show that the combination of data-driven model (OnionNet-SFCT) and empirical scoring function (Vina score) is a good scoring strategy that could be useful for structure-based drug discoveries and potentially target fishing in future.


Assuntos
Descoberta de Drogas , Proteínas , Descoberta de Drogas/métodos , Ligantes , Aprendizado de Máquina , Simulação de Acoplamento Molecular , Ligação Proteica , Proteínas/química
4.
Proteins ; 91(12): 1837-1849, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37606194

RESUMO

We introduce a deep learning-based ligand pose scoring model called zPoseScore for predicting protein-ligand complexes in the 15th Critical Assessment of Protein Structure Prediction (CASP15). Our contributions are threefold: first, we generate six training and evaluation data sets by employing advanced data augmentation and sampling methods. Second, we redesign the "zFormer" module, inspired by AlphaFold2's Evoformer, to efficiently describe protein-ligand interactions. This module enables the extraction of protein-ligand paired features that lead to accurate predictions. Finally, we develop the zPoseScore framework with zFormer for scoring and ranking ligand poses, allowing for atomic-level protein-ligand feature encoding and fusion to output refined ligand poses and ligand per-atom deviations. Our results demonstrate excellent performance on various testing data sets, achieving Pearson's correlation R = 0.783 and 0.659 for ranking docking decoys generated based on experimental and predicted protein structures of CASF-2016 protein-ligand complexes. Additionally, we obtain an averaged local distance difference test (lDDT pli = 0.558) of AIchemy LIG2 in CASP15 for de novo protein-ligand complex structure predictions. Detailed analysis shows that accurate ligand binding site prediction and side-chain orientation are crucial for achieving better prediction performance. Our proposed model is one of the most accurate protein-ligand pose prediction models and could serve as a valuable tool in small molecule drug discovery.


Assuntos
Proteínas , Ligantes , Ligação Proteica , Proteínas/química , Sítios de Ligação , Simulação de Acoplamento Molecular
5.
Phys Chem Chem Phys ; 24(7): 4324-4333, 2022 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-35107451

RESUMO

The COVID-19 pandemic caused by SARS-CoV-2 has been declared a global health crisis. The development of anti-SARS-CoV-2 drugs heavily depends on the systematic study of the critical biological processes of key proteins of coronavirus among which the main proteinase (Mpro) dimerization is a key step for virus maturation. Because inhibiting the Mpro dimerization can efficiently suppress virus maturation, the key residues that mediate dimerization can be treated as targets of drug and antibody developments. In this work, the structure and energy features of the Mpro dimer of SARS-CoV-2 and SARS-CoV were studied using molecular dynamics (MD) simulations. The free energy calculations using the Generalized Born (GB) model showed that the dimerization free energy of the SARS-CoV-2 Mpro dimer (-107.5 ± 10.89 kcal mol-1) is larger than that of the SARS-CoV Mpro dimer (-92.83 ± 9.81 kcal mol-1), indicating a more stable and possibly a quicker formation of the Mpro dimer of SARS-CoV-2. In addition, the energy decomposition of each residue revealed 11 key attractive residues. Furthermore, Thr285Ala weakens the steric hindrance between the two protomers of SARS-CoV-2 that can form more intimate interactions. It is interesting to find 11 repulsive residues which effectively inhibit the dimerization process. At the interface of the Mpro dimer, we detected three regions that are rich in interfacial water which stabilize the SARS-CoV-2 Mpro dimer by forming hydrogen bonds with two protomers. The key residues and rich water regions provide important targets for the future design of anti-SARS-CoV-2 drugs through inhibiting Mpro dimerization.


Assuntos
Proteases 3C de Coronavírus/química , SARS-CoV-2/enzimologia , COVID-19 , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Pandemias , Multimerização Proteica
6.
Proteins ; 89(12): 1901-1910, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34473376

RESUMO

In this paper, we report our tFold framework's performance on the inter-residue contact prediction task in the 14th Critical Assessment of protein Structure Prediction (CASP14). Our tFold framework seamlessly combines both homologous sequences and structural decoys under an ultra-deep network architecture. Squeeze-excitation and axial attention mechanisms are employed to effectively capture inter-residue interactions. In CASP14, our best predictor achieves 41.78% in the averaged top-L precision for long-range contacts for all the 22 free-modeling (FM) targets, and ranked 1st among all the 60 participating teams. The tFold web server is now freely available at: https://drug.ai.tencent.com/console/en/tfold.


Assuntos
Redes Neurais de Computação , Dobramento de Proteína , Proteínas , Software , Homologia Estrutural de Proteína , Biologia Computacional , Modelos Moleculares , Proteínas/química , Proteínas/metabolismo , Reprodutibilidade dos Testes , Análise de Sequência de Proteína
7.
Biochemistry ; 58(36): 3777-3788, 2019 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-31424191

RESUMO

Recognition of RNAs under physiological conditions is important for the development of chemical probes and therapeutic ligands. Nucleobase-modified dsRNA-binding PNAs (dbPNAs) are promising for the recognition of dsRNAs in a sequence and structure specific manner under near-physiological conditions. Guanidinium is often present in proteins and small molecules for the recognition of G bases in nucleic acids, in cell-penetrating carriers, and in bioactive drug molecules, which might be due to the fact that guanidinium is amphiphilic and has unique hydrogen bonding and stacking properties. We hypothesized that a simple guanidinium moiety can be directly incorporated into PNAs to facilitate enhanced molecular recognition of G-C pairs in dsRNAs and improved bioactivity. We grafted a guanidinium moiety directly into a PNA monomer (designated as R) using a two-carbon linker as guided by computational modeling studies. The synthetic scheme of the PNA R monomer is relatively simple compared to that of the previously reported L monomer. We incorporated the R residue into various dbPNAs for binding studies. dbPNAs incorporated with R residues are excellent in sequence specifically recognizing G-C pairs in dsRNAs over dsDNA and ssRNAs. We demonstrated that the R residue is compatible with unmodified T and C and previously developed modified L and Q residues in dbPNAs for targeting model dsRNAs, the influenza A viral panhandle duplex structure, and the HIV-1 frameshift site RNA hairpin. Furthermore, R residues enhance the cellular uptake of PNAs.


Assuntos
DNA/metabolismo , Guanidinas/química , Ácidos Nucleicos Peptídicos/metabolismo , RNA de Cadeia Dupla/metabolismo , Animais , Pareamento de Bases , Transporte Biológico , DNA/genética , HIV-1/química , Ligação de Hidrogênio , Concentração de Íons de Hidrogênio , Modelos Moleculares , Conformação de Ácido Nucleico , Orthomyxoviridae/química , Ácidos Nucleicos Peptídicos/química , Ácidos Nucleicos Peptídicos/genética , RNA de Cadeia Dupla/genética , RNA Viral/genética , RNA Viral/metabolismo , Spodoptera/química
8.
Phys Chem Chem Phys ; 20(43): 27439-27448, 2018 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-30357163

RESUMO

CRISPR-Cas9, a powerful genome editing tool, has widely been applied in biological fields. Since the discovery of CRISPR-Cas9 as an adaptive immune system, it has been gradually modified to perform precise genome editing in eukaryotic cells by creating double-strand breaks. Although it is robust and efficient, the current CRISPR-Cas9 system faces a major flaw: off-target effects, which are not well understood. Several Cas9 mutants show significant improvement, with very low off-target effects; however, they also show relatively lower cleavage efficiency for on-target sequences. In this study, the dynamics of wild-type Cas9 from Streptococcus pyogenes and a high fidelity Cas9 mutant have been explored using molecular dynamics simulations. It was found that the mutations cause decreased electrostatic interactions between Cas9 and the R-loop. Consequently, the flexibility of the tDNA/sgRNA heteroduplex is decreased, which may explain the lower tolerance of mismatches in the heteroduplex region. The mutations also affect the protein dynamics and the correlation networks among Cas9 domains. In mutant Cas9, weakened communications between two catalytic domains as well as a slight opening of the conformation induced by the mutations account for the lower on-target cleavage efficiency and probably the lower off-target efficiency as well. These findings will facilitate more precise Cas9 engineering in future.


Assuntos
Proteínas de Bactérias/metabolismo , Endonucleases/metabolismo , Mutação , Proteínas de Bactérias/genética , Proteína 9 Associada à CRISPR , Sistemas CRISPR-Cas , Endonucleases/genética , Ligação Proteica/genética , Domínios Proteicos , Eletricidade Estática
9.
J Hazard Mater ; 475: 134828, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-38876015

RESUMO

The prediction of ecological toxicity plays an increasingly important role in modern society. However, the existing models often suffer from poor performance and limited predictive capabilities. In this study, we propose a novel approach for ecological toxicity assessment based on pre-trained models. By leveraging pre-training techniques and graph neural network models, we establish a highperformance predictive model. Furthermore, we incorporate a variational autoencoder to optimize the model, enabling simultaneous discrimination of toxicity to bees and molecular degradability. Additionally, despite the low similarity between the endogenous hormones in bees and the compounds in our dataset, our model confidently predicts that these hormones are non-toxic to bees, which further strengthens the credibility and accuracy of our model. We also discovered the negative correlation between the degradation and bee toxicity of compounds. In summary, this study presents an ecological toxicity assessment model with outstanding performance. The proposed model accurately predicts the toxicity of chemicals to bees and their degradability capabilities, offering valuable technical support to relevant fields.


Assuntos
Redes Neurais de Computação , Abelhas/efeitos dos fármacos , Animais , Ecotoxicologia , Testes de Toxicidade
10.
Commun Biol ; 7(1): 586, 2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38755285

RESUMO

Bats serve as reservoirs for numerous zoonotic viruses, yet they typically remain asymptomatic owing to their unique immune system. Of particular significance is the MHC-I in bats, which plays crucial role in anti-viral response and exhibits polymorphic amino acid (AA) insertions. This study demonstrated that both 5AA and 3AA insertions enhance the thermal stability of the bat MHC-I complex and enrich the diversity of bound peptides in terms of quantity and length distribution, by stabilizing the 310 helix, a region prone to conformational changes during peptide loading. However, the mismatched insertion could diminish the stability of bat pMHC-I. We proposed that a suitable insertion may help bat MHC-I adapt to high body temperatures during flight while enhancing antiviral responses. Moreover, this site-specific insertions may represent a strategy of evolutionary adaptation of MHC-I molecules to fluctuations in body temperature, as similar insertions have been found in other lower vertebrates.


Assuntos
Quirópteros , Antígenos de Histocompatibilidade Classe I , Animais , Antígenos de Histocompatibilidade Classe I/metabolismo , Antígenos de Histocompatibilidade Classe I/química , Antígenos de Histocompatibilidade Classe I/genética , Estabilidade Proteica , Peptídeos/química , Peptídeos/metabolismo , Aminoácidos/química , Aminoácidos/metabolismo , Apresentação de Antígeno , Mutagênese Insercional
11.
Gut Microbes ; 15(1): 2227434, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37349961

RESUMO

A demonstration of cellulose degrading bacterium from human gut changed our view that human cannot degrade the cellulose. However, investigation of cellulose degradation by human gut microbiota on molecular level has not been completed so far. We showed here, using cellobiose as a model that promoted the growth of human gut key members, such as Bacteroides ovatus (BO), to clarify the molecular mechanism. Our results showed that a new polysaccharide utilization locus (PUL) from BO was involved in the cellobiose capturing and degradation. Further, two new cellulases BACOVA_02626GH5 and BACOVA_02630GH5 on the cell surface performed the degradation of cellobiose into glucose were determined. The predicted structures of BACOVA_02626GH5 and BACOVA_02630GH5 were highly homologous with the cellulase from soil bacteria, and the catalytic residues were highly conservative with two glutamate residues. In murine experiment, we observed cellobiose reshaped the composition of gut microbiota and probably modified the metabolic function of bacteria. Taken together, our findings further highlight the evidence of cellulose can be degraded by human gut microbes and provide new insight in the field of investigation on cellulose.


Assuntos
Celobiose , Microbioma Gastrointestinal , Humanos , Animais , Camundongos , Celobiose/metabolismo , Celulose/metabolismo , Polissacarídeos/metabolismo
12.
Sci Data ; 9(1): 71, 2022 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-35241693

RESUMO

Intrinsic solubility is a critical property in pharmaceutical industry that impacts in-vivo bioavailability of small molecule drugs. However, solubility prediction with Artificial Intelligence(AI) are facing insufficient data, poor data quality, and no unified measurements for AI and physics-based approaches. We collect 7 aqueous solubility datasets, and present a dataset curation workflow. Evaluating the curated data with two expanded deep learning methods, improved RMSE scores on all curated thermodynamic datasets are observed. We also compare expanded Chemprop enhanced with curated data and state-of-art physics-based approach using pearson and spearman correlation coefficients. A similar performance on pearson with 0.930 and spearman with 0.947 from expanded Chemprop is achieved. A steadily improved pearson and spearman values with increasing data points are also illustrated. Besides that, the computation advantage of AI models enables quick evaluation of a large set of molecules during the hit identification or lead optimization stages, which helps further decision making within the time cycle at drug discovery stage.

13.
Front Chem ; 9: 753002, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34778208

RESUMO

One key task in virtual screening is to accurately predict the binding affinity (△G) of protein-ligand complexes. Recently, deep learning (DL) has significantly increased the predicting accuracy of scoring functions due to the extraordinary ability of DL to extract useful features from raw data. Nevertheless, more efforts still need to be paid in many aspects, for the aim of increasing prediction accuracy and decreasing computational cost. In this study, we proposed a simple scoring function (called OnionNet-2) based on convolutional neural network to predict △G. The protein-ligand interactions are characterized by the number of contacts between protein residues and ligand atoms in multiple distance shells. Compared to published models, the efficacy of OnionNet-2 is demonstrated to be the best for two widely used datasets CASF-2016 and CASF-2013 benchmarks. The OnionNet-2 model was further verified by non-experimental decoy structures from docking program and the CSAR NRC-HiQ data set (a high-quality data set provided by CSAR), which showed great success. Thus, our study provides a simple but efficient scoring function for predicting protein-ligand binding free energy.

14.
ACS Omega ; 4(14): 15956-15965, 2019 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-31592466

RESUMO

Computational drug discovery provides an efficient tool for helping large-scale lead molecule screening. One of the major tasks of lead discovery is identifying molecules with promising binding affinities toward a target, a protein in general. The accuracies of current scoring functions that are used to predict the binding affinity are not satisfactory enough. Thus, machine learning or deep learning based methods have been developed recently to improve the scoring functions. In this study, a deep convolutional neural network model (called OnionNet) is introduced; its features are based on rotation-free element-pair-specific contacts between ligands and protein atoms, and the contacts are further grouped into different distance ranges to cover both the local and nonlocal interaction information between the ligand and the protein. The prediction power of the model is evaluated and compared with other scoring functions using the comparative assessment of scoring functions (CASF-2013) benchmark and the v2016 core set of the PDBbind database. The robustness of the model is further explored by predicting the binding affinities of the complexes generated from docking simulations instead of experimentally determined PDB structures.

15.
Front Chem ; 7: 315, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31134186

RESUMO

Progesterone receptor (PR) is a member of the nuclear receptor (NR) superfamily and plays a vital role in the female reproductive system. The malfunction of it would lead to several types of cancers. The understanding of conformational changes in its ligand binding domain (LBD) is valuable for both biological function studies and therapeutically intervenes. A key unsolved question is how the binding of a ligand (agonist, antagonist, or a selective modulator) induces conformational changes of PR LBD, especially its helix 12. We applied molecular dynamics (MD) simulations to explore the conformational adaptations of PR LBD with or without a ligand or the co-repressor peptides binding. From the simulations, both the agonist progesterone (P4) and the selective PR modulator (SPRM) asoprisnil induces agonistic-like helix 12 conformations (the "closed" states) in PR LBD and the complex of LBD-SPRM is less stable, comparing to the agonist-liganded PR LBD. The results, therefore, explain the partial agonism of the SPRM, which could induce weak agonistic effects in PR. We also found that co-repressor peptides could be stably associated with the LBD and stabilize the LBD in a "semi-open" state for helix 12. These findings would enhance our understanding of PR structural and functional relationships and would also be useful for future structure and knowledge-based drug discovery.

16.
Trends Pharmacol Sci ; 40(11): 897-910, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31662208

RESUMO

Recent clinical and therapeutic success with RAF and MEK1/2 inhibitors has revolutionized the existing treatment schemes for previously incurable cancers like melanomas. However, the overall therapeutic efficacies are still largely compromised by the dose-limiting side effects and emerging drug resistance mechanisms. Accumulating evidence has revealed the intricate nature of the RAS-RAF-MEK1/2-ERK1/2 pathway, such as activation mechanisms, kinase-substrate relationships, crosstalk with parallel signaling pathways, feedback regulations, and intimate interplay with immune responses. Limited strategies are currently available to exploit the benefits of combining RAF-MEK1/2-ERK1/2 pathway inhibitors with other targeted therapies or immunotherapies. Here, we compiled the kinase-substrate relationships and analyzed the intricate signaling networks of the renowned pathway, providing an integrated and simplified visualization, to reveal the potentials of RAS-RAF-MEK1/2-ERK1/2-based combination therapies.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Fatores Imunológicos/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Humanos , Fatores Imunológicos/administração & dosagem , Fatores Imunológicos/uso terapêutico , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/imunologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/imunologia , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/administração & dosagem , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/imunologia , Neoplasias/enzimologia , Neoplasias/imunologia , Inibidores de Proteínas Quinases/uso terapêutico , Quinases raf/antagonistas & inibidores , Quinases raf/imunologia , Quinases raf/metabolismo , Proteínas ras/antagonistas & inibidores , Proteínas ras/imunologia , Proteínas ras/metabolismo
17.
FEBS J ; 285(15): 2762-2784, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29722136

RESUMO

Actin filament assembly contributes to the endocytic pathway pleiotropically, with active roles in clathrin-dependent and clathrin-independent endocytosis as well as subsequent endosomal trafficking. Endocytosis comprises a series of dynamic events, including the initiation of membrane curvature, bud invagination, vesicle abscission and subsequent vesicular transport. The ultimate success of endocytosis requires the coordinated activities of proteins that trigger actin polymerization, recruit actin-binding proteins (ABPs) and organize endocytic proteins (EPs) that promote membrane curvature through molecular crowding or scaffolding mechanisms. A particularly interesting phenomenon is that multiple EPs and ABPs contain a substantial percentage of intrinsically disordered regions (IDRs), which can contribute to protein coacervation and phase separation. In addition, intrinsically disordered proteins (IDPs) frequently contain sites for post-translational modifications (PTMs) such as phosphorylation, and these modifications exhibit a high preference for IDR residues [Groban ES et al. (2006) PLoS Comput Biol 2, e32]. PTMs are implicated in regulating protein function by modulating the protein conformation, protein-protein interactions and the transition between order and disorder states of IDPs. The molecular mechanisms by which IDRs of ABPs and EPs fine-tune actin assembly and endocytosis remain mostly unexplored and elusive. In this review, we analyze protein sequences of budding yeast EPs and ABPs, and discuss the potential underlying mechanisms for regulating endocytosis and actin assembly through the emerging concept of IDR-mediated protein multivalency, coacervation, and phase transition, with an emphasis on the phospho-regulation of IDRs. Finally, we summarize the current understanding of how these mechanisms coordinate actin cytoskeleton assembly and membrane curvature formation during endocytosis in budding yeast.


Assuntos
Actinas/metabolismo , Endocitose/fisiologia , Proteínas Intrinsicamente Desordenadas/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Endossomos/metabolismo , Proteínas Fúngicas/metabolismo , Humanos , Proteínas dos Microfilamentos/metabolismo , Miosinas/metabolismo , Fosforilação , Proteínas de Plantas/metabolismo , Transporte Proteico , Saccharomycetales/metabolismo
18.
Sci Rep ; 7(1): 17795, 2017 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-29259246

RESUMO

Host defense cationic Antimicrobial Peptides (AMPs) can kill microorganisms including bacteria, viruses and fungi using various modes of action. The negatively charged bacterial membranes serve as a key target for many AMPs. Bacterial cell death by membrane permeabilization has been well perceived. A number of cationic AMPs kill bacteria by cell agglutination which is a distinctly different mode of action compared to membrane pore formation. However, mechanism of cell agglutinating AMPs is poorly understood. The outer membrane lipopolysaccharide (LPS) or the cell-wall peptidoglycans are targeted by AMPs as a key step in agglutination process. Here, we report the first atomic-resolution structure of thanatin, a cell agglutinating AMP, in complex with LPS micelle by solution NMR. The structure of thanatin in complex with LPS, revealed four stranded antiparallel ß-sheet in a 'head-tail' dimeric topology. By contrast, thanatin in free solution assumed an antiparallel ß-hairpin conformation. Dimeric structure of thanatin displayed higher hydrophobicity and cationicity with sites of LPS interactions. MD simulations and biophysical interactions analyses provided mode of LPS recognition and perturbation of LPS micelle structures. Mechanistic insights of bacterial cell agglutination obtained in this study can be utilized to develop antibiotics of alternative mode of action.


Assuntos
Aglutinação/efeitos dos fármacos , Peptídeos Catiônicos Antimicrobianos/farmacologia , Bactérias/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Antibacterianos/farmacologia , Membrana Celular/efeitos dos fármacos , Parede Celular/efeitos dos fármacos , Interações Hidrofóbicas e Hidrofílicas/efeitos dos fármacos , Micelas
19.
PLoS One ; 11(11): e0165824, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27824891

RESUMO

Progesterone receptor (PR), a member of nuclear receptor (NR) superfamily, plays a vital role for female reproductive tissue development, differentiation and maintenance. PR ligand, such as progesterone, induces conformation changes in PR ligand binding domain (LBD), thus mediates subsequent gene regulation cascades. PR LBD may adopt different conformations upon an agonist or an antagonist binding. These different conformations would trigger distinct transcription events. Therefore, the dynamics of PR LBD would be of general interest to biologists for a deep understanding of its structure-function relationship. However, no apo-form (non-ligand bound) of PR LBD model has been proposed either by experiments or computational methods so far. In this study, we explored the structural dynamics of PR LBD using molecular dynamics simulations and advanced sampling tools in both ligand-bound and the apo-forms. Resolved by the simulation study, helix 11, helix 12 and loop 895-908 (the loop between these two helices) are quite flexible in antagonistic conformation. Several residues, such as Arg899 and Glu723, could form salt-bridging interaction between helix 11 and helix 3, and are important for the PR LBD dynamics. And we also propose that helix 12 in apo-form PR LBD, not like other NR LBDs, such as human estrogen receptor α (ERα) LBD, may not adopt a totally extended conformation. With the aid of umbrella sampling and metadynamics simulations, several stable conformations of apo-form PR LBD have been sampled, which may work as critical structural models for further large scale virtual screening study to discover novel PR ligands for therapeutic application.


Assuntos
Receptores de Progesterona/metabolismo , Sítios de Ligação , Feminino , Humanos , Ligantes , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Receptores de Progesterona/fisiologia , Receptores de Progesterona/ultraestrutura , Relação Estrutura-Atividade
20.
Nat Commun ; 7: 11265, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-27068241

RESUMO

Actin cables, composed of actin filament bundles nucleated by formins, mediate intracellular transport for cell polarity establishment and maintenance. We previously observed that metaphase cells preferentially promote actin cable assembly through cyclin-dependent kinase 1 (Cdk1) activity. However, the relevant metaphase Cdk1 targets were not known. Here we show that the highly conserved actin filament crosslinking protein fimbrin is a critical Cdk1 target for actin cable assembly regulation in budding yeast. Fimbrin is specifically phosphorylated on threonine 103 by the metaphase cyclin-Cdk1 complex, in vivo and in vitro. On the basis of conformational simulations, we suggest that this phosphorylation stabilizes fimbrin's N-terminal domain, and modulates actin filament binding to regulate actin cable assembly and stability in cells. Overall, this work identifies fimbrin as a key target for cell cycle regulation of actin cable assembly in budding yeast, and suggests an underlying mechanism.


Assuntos
Actinas/metabolismo , Proteína Quinase CDC2/metabolismo , Glicoproteínas de Membrana/metabolismo , Metáfase , Proteínas dos Microfilamentos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomycetales/citologia , Saccharomycetales/enzimologia , Proteína Quinase CDC2/química , Simulação por Computador , Ciclinas/metabolismo , Mutação , Fosforilação , Fosfotreonina/metabolismo , Ligação Proteica , Estrutura Secundária de Proteína , Proteínas de Saccharomyces cerevisiae/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA