Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 14(1): 2929, 2023 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-37217466

RESUMO

Cytotoxic-T-lymphocyte (CTL) mediated control of HIV-1 is enhanced by targeting highly networked epitopes in complex with human-leukocyte-antigen-class-I (HLA-I). However, the extent to which the presenting HLA allele contributes to this process is unknown. Here we examine the CTL response to QW9, a highly networked epitope presented by the disease-protective HLA-B57 and disease-neutral HLA-B53. Despite robust targeting of QW9 in persons expressing either allele, T cell receptor (TCR) cross-recognition of the naturally occurring variant QW9_S3T is consistently reduced when presented by HLA-B53 but not by HLA-B57. Crystal structures show substantial conformational changes from QW9-HLA to QW9_S3T-HLA by both alleles. The TCR-QW9-B53 ternary complex structure manifests how the QW9-B53 can elicit effective CTLs and suggests sterically hindered cross-recognition by QW9_S3T-B53. We observe populations of cross-reactive TCRs for B57, but not B53 and also find greater peptide-HLA stability for B57 in comparison to B53. These data demonstrate differential impacts of HLAs on TCR cross-recognition and antigen presentation of a naturally arising variant, with important implications for vaccine design.


Assuntos
Infecções por HIV , Humanos , Antígenos HLA-B/genética , Linfócitos T Citotóxicos , Peptídeos , Epitopos de Linfócito T , Receptores de Antígenos de Linfócitos T
2.
Brief Bioinform ; 25(1)2023 11 22.
Artigo em Inglês | MEDLINE | ID: mdl-38233090

RESUMO

Immunologic recognition of peptide antigens bound to class I major histocompatibility complex (MHC) molecules is essential to both novel immunotherapeutic development and human health at large. Current methods for predicting antigen peptide immunogenicity rely primarily on simple sequence representations, which allow for some understanding of immunogenic features but provide inadequate consideration of the full scale of molecular mechanisms tied to peptide recognition. We here characterize contributions that unsupervised and supervised artificial intelligence (AI) methods can make toward understanding and predicting MHC(HLA-A2)-peptide complex immunogenicity when applied to large ensembles of molecular dynamics simulations. We first show that an unsupervised AI method allows us to identify subtle features that drive immunogenicity differences between a cancer neoantigen and its wild-type peptide counterpart. Next, we demonstrate that a supervised AI method for class I MHC(HLA-A2)-peptide complex classification significantly outperforms a sequence model on small datasets corrected for trivial sequence correlations. Furthermore, we show that both unsupervised and supervised approaches reveal determinants of immunogenicity based on time-dependent molecular fluctuations and anchor position dynamics outside the MHC binding groove. We discuss implications of these structural and dynamic immunogenicity correlates for the induction of T cell responses and therapeutic T cell receptor design.


Assuntos
Antígeno HLA-A2 , Simulação de Dinâmica Molecular , Humanos , Antígeno HLA-A2/metabolismo , Inteligência Artificial , Peptídeos/química , Antígenos de Histocompatibilidade Classe I/metabolismo , Ligação Proteica
3.
J Chem Inf Model ; 62(4): 801-816, 2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-35130440

RESUMO

The application of deep learning to generative molecule design has shown early promise for accelerating lead series development. However, questions remain concerning how factors like training, data set, and seed bias impact the technology's utility to medicinal and computational chemists. In this work, we analyze the impact of seed and training bias on the output of an activity-conditioned graph-based variational autoencoder (VAE). Leveraging a massive, labeled data set corresponding to the dopamine D2 receptor, our graph-based generative model is shown to excel in producing desired conditioned activities and favorable unconditioned physical properties in generated molecules. We implement an activity-swapping method that allows for the activation, deactivation, or retention of activity of molecular seeds, and we apply independent deep learning classifiers to verify the generative results. Overall, we uncover relationships between noise, molecular seeds, and training set selection across a range of latent-space sampling procedures, providing important insights for practical AI-driven molecule generation.


Assuntos
Inteligência Artificial , Modelos Moleculares , Receptores de Dopamina D2 , Receptores de Dopamina D2/química
4.
J Comput Aided Mol Des ; 36(5): 391-404, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34817762

RESUMO

We here present a streamlined, explainable graph convolutional neural network (gCNN) architecture for small molecule activity prediction. We first conduct a hyperparameter optimization across nearly 800 protein targets that produces a simplified gCNN QSAR architecture, and we observe that such a model can yield performance improvements over both standard gCNN and RF methods on difficult-to-classify test sets. Additionally, we discuss how reductions in convolutional layer dimensions potentially speak to the "anatomical" needs of gCNNs with respect to radial coarse graining of molecular substructure. We augment this simplified architecture with saliency map technology that highlights molecular substructures relevant to activity, and we perform saliency analysis on nearly 100 data-rich protein targets. We show that resultant substructural clusters are useful visualization tools for understanding substructure-activity relationships. We go on to highlight connections between our models' saliency predictions and observations made in the medicinal chemistry literature, focusing on four case studies of past lead finding and lead optimization campaigns.


Assuntos
Redes Neurais de Computação , Proteínas
5.
AIDS ; 34(12): 1713-1723, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32501836

RESUMO

OBJECTIVE: Viral infections influence intracellular peptide repertoires available for presentation by HLA-I. Alterations in HLA-I/peptide complexes can modulate binding of killer immunoglobuline-like receptors (KIRs) and thereby the function of natural killer (NK) cells. Although multiple studies have provided evidence that HLA-I/KIR interactions play a role in HIV-1 disease progression, the consequence of HIV-1 infection for HLA-I/KIR interactions remain largely unknown. DESIGN: We determined changes in HLA-I presented peptides resulting from HIV-1-infection of primary human CD4 T cells and assessed the impact of changes in peptide repertoires on HLA-I/KIR interactions. METHODS: Liquid chromatography-coupled tandem mass spectrometry to identify HLA-I presented peptides, cell-based in-vitro assays to evaluate functional consequences of alterations in immunopeptidome and atomistic molecular dynamics simulations to confirm experimental data. RESULTS: A total of 583 peptides exclusively presented on HIV-1-infected cells were identified, of which only 0.2% represented HIV-1 derived peptides. Focusing on HLA-C*03 : 04/KIR2DL3 interactions, we observed that HLA-C*03 : 04-presented peptides derived from noninfected CD4 T cells mediated stronger binding of inhibitory KIR2DL3 than peptides derived from HIV-1-infected cells. Furthermore, the most abundant peptide presented by HLA-C*03 : 04 on noninfected CD4 T cells (VIYPARISL) mediated the strongest KIR2DL3-binding, while the most abundant peptide presented on HIV-1-infected cells (YAIQATETL) did not mediate KIR2DL3-binding. Molecular dynamics simulations of HLA-C*03 : 04/KIR2DL3 interactions in the context of these two peptides revealed that VIYPARISL significantly enhanced the HLA-C*03 : 04/peptide contact area to KIR2DL3 compared with YAIQATETL. CONCLUSION: These data demonstrate that HIV-1 infection-induced changes in HLA-I-presented peptides can reduce engagement of inhibitory KIRs, providing a mechanism for enhanced activation of NK cells by virus-infected cells.


Assuntos
Infecções por HIV , HIV-1 , Antígenos HLA-C , Humanos , Peptídeos , Receptores KIR , Receptores de Células Matadoras Naturais
6.
Proc Natl Acad Sci U S A ; 117(15): 8486-8493, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32234785

RESUMO

Nucleic acid aptamers hold great promise for therapeutic applications due to their favorable intrinsic properties, as well as high-throughput experimental selection techniques. Despite the utility of the systematic evolution of ligands by the exponential enrichment (SELEX) method for aptamer determination, complementary in silico aptamer design is highly sought after to facilitate virtual screening and increased understanding of important nucleic acid-protein interactions. Here, with a combined experimental and theoretical approach, we have developed two optimal epithelial cellular adhesion molecule (EpCAM) aptamers. Our structure-based in silico method first predicts their binding modes and then optimizes them for EpCAM with molecular dynamics simulations, docking, and free energy calculations. Our isothermal titration calorimetry experiments further confirm that the EpCAM aptamers indeed exhibit enhanced affinity over a previously patented nanomolar aptamer, EP23. Moreover, our study suggests that EP23 and the de novo designed aptamers primarily bind to EpCAM dimers (and not monomers, as hypothesized in previous published works), suggesting a paradigm for developing EpCAM-targeted therapies.


Assuntos
Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/metabolismo , Molécula de Adesão da Célula Epitelial/química , Molécula de Adesão da Célula Epitelial/metabolismo , Magnésio/metabolismo , Calorimetria , Cristalografia por Raios X , Humanos , Ligantes , Modelos Moleculares , Simulação de Dinâmica Molecular , Conformação de Ácido Nucleico , Conformação Proteica , Multimerização Proteica , Técnica de Seleção de Aptâmeros
7.
J Chem Inf Model ; 60(9): 4170-4179, 2020 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-32077698

RESUMO

We present a simple, modular graph-based convolutional neural network that takes structural information from protein-ligand complexes as input to generate models for activity and binding mode prediction. Complex structures are generated by a standard docking procedure and fed into a dual-graph architecture that includes separate subnetworks for the ligand bonded topology and the ligand-protein contact map. Recent work has indicated that data set bias drives many past promising results derived from combining deep learning and docking. Our dual-graph network allows contributions from ligand identity that give rise to such biases to be distinguished from effects of protein-ligand interactions on classification. We show that our neural network is capable of learning from protein structural information when, as in the case of binding mode prediction, an unbiased data set is constructed. We next develop a deep learning model for binding mode prediction that uses docking ranking as input in combination with docking structures. This strategy mirrors past consensus models and outperforms a baseline docking program (AutoDock Vina) in a variety of tests, including on cross-docking data sets that mimic real-world docking use cases. Furthermore, the magnitudes of network predictions serve as reliable measures of model confidence.


Assuntos
Aprendizado Profundo , Ligantes , Simulação de Acoplamento Molecular , Ligação Proteica , Proteínas/metabolismo
8.
Biophys J ; 114(4): 812-821, 2018 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-29490243

RESUMO

Protein unfolding dynamics are bound by their degree of entropy production, a quantity that relates the amount of heat dissipated by a nonequilibrium process to a system's forward and time-reversed trajectories. We here explore the statistics of heat dissipation that emerge in protein molecules subjected to a chemical denaturant. Coupling large molecular dynamics datasets and Markov state models with the theory of entropy production, we demonstrate that dissipative processes can be rigorously characterized over the course of the urea-induced unfolding of the protein chymotrypsin inhibitor 2. By enumerating full entropy production probability distributions as a function of time, we first illustrate that distinct passive and dissipative regimes are present in the denaturation dynamics. Within the dissipative dynamical region, we next find that chymotrypsin inhibitor 2 is strongly driven into unfolded states in which the protein's hydrophobic core has been penetrated by urea molecules and disintegrated. Detailed analyses reveal that urea's interruption of key hydrophobic contacts between core residues causes many of the protein's native structural features to dissolve.


Assuntos
Modelos Teóricos , Peptídeos/química , Peptídeos/metabolismo , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Desnaturação Proteica , Dobramento de Proteína , Desdobramento de Proteína , Entropia , Temperatura Alta , Interações Hidrofóbicas e Hidrofílicas , Cadeias de Markov , Simulação de Dinâmica Molecular , Conformação Proteica , Domínios Proteicos , Ureia
9.
Proc Natl Acad Sci U S A ; 115(8): 1877-1882, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29437954

RESUMO

HIV controllers (HCs) are individuals who can naturally control HIV infection, partially due to potent HIV-specific CD8+ T cell responses. Here, we examined the hypothesis that superior function of CD8+ T cells from HCs is encoded by their T cell receptors (TCRs). We compared the functional properties of immunodominant HIV-specific TCRs obtained from HLA-B*2705 HCs and chronic progressors (CPs) following expression in primary T cells. T cells transduced with TCRs from HCs and CPs showed equivalent induction of epitope-specific cytotoxicity, cytokine secretion, and antigen-binding properties. Transduced T cells comparably, albeit modestly, also suppressed HIV infection in vitro and in humanized mice. We also performed extensive molecular dynamics simulations that provided a structural basis for similarities in cytotoxicity and epitope cross-reactivity. These results demonstrate that the differential abilities of HIV-specific CD8+ T cells from HCs and CPs are not genetically encoded in the TCRs alone and must depend on additional factors.


Assuntos
Linfócitos T CD8-Positivos/fisiologia , Epitopos de Linfócito T/genética , Infecções por HIV/imunologia , HIV-1/imunologia , Receptores de Antígenos de Linfócitos T/genética , Clonagem Molecular , Regulação da Expressão Gênica/imunologia , Células HEK293 , Antígeno HLA-B27 , Humanos , Células Jurkat
10.
Biomaterials ; 152: 24-36, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29080421

RESUMO

Gadolinium-containing fullerenol Gd@C82(OH)22 has demonstrated low-toxicity and highly therapeutic efficacy in inhibiting tumor growth and metastasis through new strategy of encaging cancer, however, little is known about the mechanisms how this nanoparticle regulates fibroblast cells to prison (instead of poison) cancer cells. Here, we report that Gd@C82(OH)22 promote the binding activity of tumor necrosis factor (TNFα) to tumor necrosis factor receptors 2 (TNFR2), activate TNFR2/p38 MAPK signaling pathway to increase cellular collagen expression in fibrosarcoma cells and human primary lung cancer associated fibroblasts isolated from patients. We also employ molecular dynamics simulations to study the atomic-scale mechanisms that dictate how Gd@C82(OH)22 mediates interactions between TNFα and TNFRs. Our data suggest that Gd@C82(OH)22 might enhance the association between TNFα and TNFR2 through a "bridge-like" mode of interaction; by contrast, the fullerenol appears to inhibit TNFα-TNFR1 association by binding to two of the receptor's cysteine-rich domains. In concert, our results uncover a sequential, systemic process by which Gd@C82(OH)22 acts to prison tumor cells, providing new insights into principles of designs of cancer therapeutics.


Assuntos
Antineoplásicos/química , Colágeno/metabolismo , Fulerenos/química , Gadolínio/química , Nanopartículas Metálicas/química , Animais , Linhagem Celular Tumoral , Fibrossarcoma/patologia , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Simulação de Dinâmica Molecular , Tamanho da Partícula , Receptores Tipo II do Fator de Necrose Tumoral/química , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Propriedades de Superfície , Células Tumorais Cultivadas , Fator de Necrose Tumoral alfa/química , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Science ; 359(6375): 582-587, 2018 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-29217585

RESUMO

CD8+ T cell-dependent killing of cancer cells requires efficient presentation of tumor antigens by human leukocyte antigen class I (HLA-I) molecules. However, the extent to which patient-specific HLA-I genotype influences response to anti-programmed cell death protein 1 or anti-cytotoxic T lymphocyte-associated protein 4 is currently unknown. We determined the HLA-I genotype of 1535 advanced cancer patients treated with immune checkpoint blockade (ICB). Maximal heterozygosity at HLA-I loci ("A," "B," and "C") improved overall survival after ICB compared with patients who were homozygous for at least one HLA locus. In two independent melanoma cohorts, patients with the HLA-B44 supertype had extended survival, whereas the HLA-B62 supertype (including HLA-B*15:01) or somatic loss of heterozygosity at HLA-I was associated with poor outcome. Molecular dynamics simulations of HLA-B*15:01 revealed different elements that may impair CD8+ T cell recognition of neoantigens. Our results have important implications for predicting response to ICB and for the design of neoantigen-based therapeutic vaccines.


Assuntos
Antígeno CTLA-4/antagonistas & inibidores , Antígenos de Histocompatibilidade Classe I/genética , Imunoterapia/métodos , Melanoma/imunologia , Melanoma/terapia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Neoplasias Cutâneas/imunologia , Neoplasias Cutâneas/terapia , Adulto , Idoso , Apresentação de Antígeno , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Estudos de Coortes , Triagem de Portadores Genéticos , Antígenos de Histocompatibilidade Classe I/química , Humanos , Melanoma/genética , Melanoma/mortalidade , Pessoa de Meia-Idade , Conformação Proteica , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/mortalidade , Resultado do Tratamento , Adulto Jovem
12.
Sci Rep ; 7(1): 13579, 2017 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-29051586

RESUMO

In the face of chronic cancers and protracted viral infections, human immune cells are known to adopt an exhausted state in which their effector functions are lost. In recent years, a number of inhibitory receptors have been connected to the immune cell exhaustion phenotype; furthermore, ligands capable of activating these receptors have been discovered. The molecular mechanisms by which these ligands affect the exhausted states of immune cells, however, are largely unknown. Here, we present the results of molecular dynamics simulations of one potential exhaustion-associated system: the complex of human inhibitory receptor TIM3 (hTIM3) and its ligand phosphatidylserine (PSF). We find that PSF fundamentally alters the electrostatic environment within hTIM3's Ca2+ binding site, facilitating the formation of a salt bridge and freeing a tyrosine-containing strand. This liberated tyrosine then collapses into a nearby hydrophobic pocket, anchoring a modified conformational ensemble typified by a ß-strand rearrangement. The "electrostatic switching/hydrophobic anchoring" mechanism of conformational modulation reported here suggests a new type of process by which TIM3 activation might be achieved. This work also highlights strategies by which PSF-mediated conformational change could be controlled, either through administration of small molecules, execution of mutations, or modification of receptor phosphorylation states.


Assuntos
Receptor Celular 2 do Vírus da Hepatite A/química , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Fosfatidilserinas/química , Sítios de Ligação , Domínios de Imunoglobulina , Modelos Moleculares , Simulação de Dinâmica Molecular , Fosfatidilserinas/metabolismo , Fosforilação , Conformação Proteica
13.
Sci Adv ; 3(5): e1601684, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28560321

RESUMO

The molecular motor exploited by bacteriophage φ29 to pack DNA into its capsid is regarded as one of the most powerful mechanical devices present in viral, bacterial, and eukaryotic systems alike. Acting as a linker element, a prohead RNA (pRNA) effectively joins the connector and ATPase (adenosine triphosphatase) components of the φ29 motor. During DNA packing, this pRNA needs to withstand enormous strain along the capsid's portal axis-how this remarkable stability is achieved remains to be elucidated. We investigate the mechanical properties of the φ29 motor's three-way junction (3WJ)-pRNA using a combined steered molecular dynamics and atomic force spectroscopy approach. The 3WJ exhibits strong resistance to stretching along its coaxial helices, demonstrating its super structural robustness. This resistance disappears, however, when external forces are applied to the transverse directions. From a molecular standpoint, we demonstrate that this direction-dependent stability can be attributed to two Mg clamps that cooperate and generate mechanical resistance in the pRNA's coaxial direction. Our results suggest that the asymmetric nature of the 3WJ's mechanical stability is entwined with its biological function: Enhanced rigidity along the portal axis is likely essential to withstand the strain caused by DNA condensation, and flexibility in other directions should aid in the assembly of the pRNA and its association with other motor components.


Assuntos
Adenosina Trifosfatases/química , Fagos Bacilares/química , Bacillus subtilis/virologia , Podoviridae/química , RNA Viral/química , Proteínas Virais/química , Adenosina Trifosfatases/metabolismo , Fagos Bacilares/fisiologia , Capsídeo/química , Capsídeo/metabolismo , DNA Viral/química , DNA Viral/metabolismo , Podoviridae/fisiologia , RNA Viral/metabolismo , Proteínas Virais/metabolismo , Montagem de Vírus/fisiologia
14.
Sci Rep ; 7: 42767, 2017 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-28218295

RESUMO

Examining interactions between nanomaterials and cell membranes can expose underlying mechanisms of nanomaterial cytotoxicity and guide the design of safer nanomedical technologies. Recently, graphene has been shown to exhibit potential toxicity to cells; however, the molecular processes driving its lethal properties have yet to be fully characterized. We here demonstrate that graphene nanosheets (both pristine and oxidized) can produce holes (pores) in the membranes of A549 and Raw264.7 cells, substantially reducing cell viability. Electron micrographs offer clear evidence of pores created on cell membranes. Our molecular dynamics simulations reveal that multiple graphene nanosheets can cooperate to extract large numbers of phospholipids from the membrane bilayer. Strong dispersion interactions between graphene and lipid-tail carbons result in greatly depleted lipid density within confined regions of the membrane, ultimately leading to the formation of water-permeable pores. This cooperative lipid extraction mechanism for membrane perforation represents another distinct process that contributes to the molecular basis of graphene cytotoxicity.


Assuntos
Membrana Celular/efeitos dos fármacos , Grafite/toxicidade , Fosfolipídeos/isolamento & purificação , Células A549 , Animais , Membrana Celular/ultraestrutura , Sobrevivência Celular/efeitos dos fármacos , Humanos , Bicamadas Lipídicas/química , Camundongos , Simulação de Dinâmica Molecular , Nanoestruturas/toxicidade , Fosfolipídeos/química , Células RAW 264.7
15.
Nat Commun ; 8: 14537, 2017 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-28233871

RESUMO

Engineered nanomaterials promise to transform medicine at the bio-nano interface. However, it is important to elucidate how synthetic nanomaterials interact with critical biological systems before such products can be safely utilized in humans. Past evidence suggests that polyethylene glycol-functionalized (PEGylated) nanomaterials are largely biocompatible and elicit less dramatic immune responses than their pristine counterparts. We here report results that contradict these findings. We find that PEGylated graphene oxide nanosheets (nGO-PEGs) stimulate potent cytokine responses in peritoneal macrophages, despite not being internalized. Atomistic molecular dynamics simulations support a mechanism by which nGO-PEGs preferentially adsorb onto and/or partially insert into cell membranes, thereby amplifying interactions with stimulatory surface receptors. Further experiments demonstrate that nGO-PEG indeed provokes cytokine secretion by enhancing integrin ß8-related signalling pathways. The present results inform that surface passivation does not always prevent immunological reactions to 2D nanomaterials but also suggest applications for PEGylated nanomaterials wherein immune stimulation is desired.


Assuntos
Materiais Revestidos Biocompatíveis/farmacologia , Grafite/farmacologia , Imunidade/efeitos dos fármacos , Nanoestruturas/química , Polietilenoglicóis/farmacologia , Adsorção , Animais , Engenharia Biomédica/métodos , Linhagem Celular , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Materiais Revestidos Biocompatíveis/química , Citocinas/imunologia , Citocinas/metabolismo , Grafite/química , Cadeias beta de Integrinas/imunologia , Cadeias beta de Integrinas/metabolismo , Macrófagos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Simulação de Dinâmica Molecular , Polietilenoglicóis/química , Cultura Primária de Células , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia , Propriedades de Superfície
16.
Sci Rep ; 6: 25138, 2016 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-27113844

RESUMO

Though the problem of sequence-reversed protein folding is largely unexplored, one might speculate that reversed native protein sequences should be significantly more foldable than purely random heteropolymer sequences. In this article, we investigate how the reverse-sequences of native proteins might fold by examining a series of small proteins of increasing structural complexity (α-helix, ß-hairpin, α-helix bundle, and α/ß-protein). Employing a tandem protein structure prediction algorithmic and molecular dynamics simulation approach, we find that the ability of reverse sequences to adopt native-like folds is strongly influenced by protein size and the flexibility of the native hydrophobic core. For ß-hairpins with reverse-sequences that fail to fold, we employ a simple mutational strategy for guiding stable hairpin formation that involves the insertion of amino acids into the ß-turn region. This systematic look at reverse sequence duality sheds new light on the problem of protein sequence-structure mapping and may serve to inspire new protein design and protein structure prediction protocols.


Assuntos
Dobramento de Proteína , Proteínas/química , Proteínas/metabolismo , Simulação de Dinâmica Molecular , Conformação Proteica , Estabilidade Proteica
17.
Nanoscale ; 8(24): 12143-51, 2016 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-26899409

RESUMO

An assortment of biological processes, like protein degradation and the transport of proteins across membranes, depend on protein unfolding events mediated by nanopore interfaces. In this work, we exploit fully atomistic simulations of an artificial, CNT-based nanopore to investigate the nature of ubiquitin unfolding. With one end of the protein subjected to an external force, we observe non-canonical unfolding behaviour as ubiquitin is pulled through the pore opening. Secondary structural elements are sequentially detached from the protein and threaded into the nanotube, interestingly, the remaining part maintains native-like characteristics. The constraints of the nanopore interface thus facilitate the formation of stable "unfoldon" motifs above the nanotube aperture that can exist in the absence of specific native contacts with the other secondary structure. Destruction of these unfoldons gives rise to distinct force peaks in our simulations, providing us with a sensitive probe for studying the kinetics of serial unfolding events. Our detailed analysis of nanopore-mediated protein unfolding events not only provides insight into how related processes might proceed in the cell, but also serves to deepen our understanding of structural arrangements which form the basis for protein conformational stability.


Assuntos
Nanoporos , Nanotubos de Carbono , Desdobramento de Proteína , Proteínas/química , Modelos Moleculares , Conformação Proteica , Dobramento de Proteína
18.
Sci Rep ; 6: 21316, 2016 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-26899743

RESUMO

We report a detailed computational and experimental study of the interaction of single-walled carbon nanotubes (SWCNTs) with the drug-metabolizing cytochrome P450 enzyme, CYP3A4. Dose-dependent inhibition of CYP3A4-mediated conversion of the model compound, testosterone, to its major metabolite, 6ß-hydroxy testosterone was noted. Evidence for a direct interaction between SWCNTs and CYP3A4 was also provided. The inhibition of enzyme activity was alleviated when SWCNTs were pre-coated with bovine serum albumin. Furthermore, covalent functionalization of SWCNTs with polyethylene glycol (PEG) chains mitigated the inhibition of CYP3A4 enzymatic activity. Molecular dynamics simulations suggested that inhibition of the catalytic activity of CYP3A4 is mainly due to blocking of the exit channel for substrates/products through a complex binding mechanism. This work suggests that SWCNTs could interfere with metabolism of drugs and other xenobiotics and provides a molecular mechanism for this toxicity. Our study also suggests means to reduce this toxicity, eg., by surface modification.


Assuntos
Inibidores do Citocromo P-450 CYP3A/química , Citocromo P-450 CYP3A/metabolismo , Inativação Metabólica/efeitos dos fármacos , Nanotubos de Carbono/química , Animais , Bovinos , Inibidores do Citocromo P-450 CYP3A/efeitos adversos , Humanos , Ligação de Hidrogênio/efeitos dos fármacos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Simulação de Dinâmica Molecular , Nanotubos de Carbono/efeitos adversos , Conformação Proteica/efeitos dos fármacos , Testosterona/metabolismo
19.
ACS Nano ; 10(2): 1829-35, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26800095

RESUMO

The deteriorating state of global fresh water resources represents one of the most serious challenges that scientists and policymakers currently face. Desalination technologies, which are designed to extract potable water from the planet's bountiful stores of seawater, could serve to alleviate much of the stress that presently plagues fresh water supplies. In recent decades, desalination methods have improved via water-filtering architectures based on nanoporous graphene filters and artificial membranes integrated with biological water channels. Here, we report the auspicious performance (in simulations) of an alternative nanoporous desalination filter constructed from a MoS2 nanosheet. In striking contrast to graphene-based filters, we find that the "open" and "closed" states of the MoS2 filter can be regulated by the introduction of mechanical strain, yielding a highly tunable nanopore interface. By applying lateral strain to the MoS2 filter in our simulations, we see that the transition point between "open" and "closed" states occurs under tension that induces about 6% cross-sectional expansion in the membrane (6% strain); the open state of the MoS2 filter demonstrates high water transparency and a strong salt filtering capability even under 12% strain. Our results thus demonstrate the promise of a controllable nanoporous MoS2 desalination filter, wherein the morphology and size of the central nanopore can be precisely regulated by tensile strain. These findings support the design and proliferation of tunable nanodevices for filtration and other applications.

20.
J Chem Theory Comput ; 11(6): 2412-20, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-26575541

RESUMO

As simulators attempt to replicate the dynamics of large cellular components in silico, problems related to sampling slow, glassy degrees of freedom in molecular systems will be amplified manyfold. It is tempting to augment simulation techniques with external biases to overcome such barriers with ease; biased simulations, however, offer little utility unless equilibrium properties of interest (both kinetic and thermodynamic) can be recovered from the data generated. In this Article, we present a general scheme that harnesses the power of Markov state models (MSMs) to extract equilibrium kinetic properties from molecular dynamics trajectories collected on biased potential energy surfaces. We first validate our reweighting protocol on a simple two-well potential, and we proceed to test our method on potential-biased simulations of the Trp-cage miniprotein. In both cases, we find that equilibrium populations, time scales, and dynamical processes are reliably reproduced as compared to gold standard, unbiased data sets. We go on to discuss the limitations of our dynamical reweighting approach, and we suggest auspicious target systems for further application.


Assuntos
Cadeias de Markov , Simulação de Dinâmica Molecular , Proteínas/química , Cinética , Peso Molecular , Proteínas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA