Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 97
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Med Res Rev ; 43(2): 293-318, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36104980

RESUMO

The centrosome in animal cells is instrumental in spindle pole formation, nucleation, proper alignment of microtubules during cell division, and distribution of chromosomes in each daughter cell. Centrosome amplification involving structural and numerical abnormalities in the centrosome can cause chromosomal instability and dysregulation of the cell cycle, leading to cancer development and metastasis. However, disturbances caused by centrosome amplification can also limit cancer cell survival by activating mitotic checkpoints and promoting mitotic catastrophe. As a smart escape, cancer cells cluster their surplus of centrosomes into pseudo-bipolar spindles and progress through the cell cycle. This phenomenon, known as centrosome clustering (CC), involves many proteins and has garnered considerable attention as a specific cancer cell-targeting weapon. The kinesin-14 motor protein KIFC1 is a minus end-directed motor protein that is involved in CC. Because KIFC1 is upregulated in various cancers and modulates oncogenic signaling cascades, it has emerged as a potential chemotherapeutic target. Many molecules have been identified as KIFC1 inhibitors because of their centrosome declustering activity in cancer cells. Despite the ever-increasing literature in this field, there have been few efforts to review the progress. The current review aims to collate and present an in-depth analysis of known KIFC1 inhibitors and their biological activities. Additionally, we present computational docking data of putative KIFC1 inhibitors with their binding sites and binding affinities. This first-of-kind comparative analysis involving experimental biology, chemistry, and computational docking of different KIFC1 inhibitors may help guide decision-making in the selection and design of potent inhibitors.


Assuntos
Benchmarking , Neoplasias , Animais , Neoplasias/patologia , Centrossomo/metabolismo , Sítios de Ligação , Microtúbulos
2.
Biochemistry ; 62(5): 1070-1081, 2023 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-36795942

RESUMO

Numerous studies demonstrate that enzymes undergo multiple conformational changes during catalysis. The malleability of enzymes forms the basis for allosteric regulation: residues located far from the active site can exert long-range dynamical effects on the active site residues to modulate catalysis. The structure of Pseudomonas aeruginosa d-arginine dehydrogenase (PaDADH) shows four loops (L1, L2, L3, and L4) that span the substrate and the FAD-binding domains. Loop L4 comprises residues 329-336, spanning over the flavin cofactor. The I335 residue on loop L4 is ∼10 Šaway from the active site and ∼3.8 Šfrom N(1)-C(2)═O atoms of the flavin. In this study, we used molecular dynamics and biochemical techniques to investigate the effect of the mutation of I335 to histidine on the catalytic function of PaDADH. Molecular dynamics showed that the conformational dynamics of PaDADH are shifted to a more closed conformation in the I335H variant. In agreement with an enzyme that samples more in a closed conformation, the kinetic data of the I335H variant showed a 40-fold decrease in the rate constant of substrate association (k1), a 340-fold reduction in the rate constant of substrate dissociation from the enzyme-substrate complex (k2), and a 24-fold decrease in the rate constant of product release (k5), compared to that of the wild-type. Surprisingly, the kinetic data are consistent with the mutation having a negligible effect on the reactivity of the flavin. Altogether, the data indicate that the residue at position 335 has a long-range dynamical effect on the catalytic function in PaDADH.


Assuntos
Aminoácido Oxirredutases , Simulação de Dinâmica Molecular , Aminoácido Oxirredutases/química , Domínio Catalítico , Catálise , Flavinas/metabolismo , Cinética , Especificidade por Substrato , Sítios de Ligação , Conformação Proteica
3.
J Chem Inf Model ; 63(12): 3892-3902, 2023 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-37285207

RESUMO

Drug resistance in antiviral treatments is a serious public health problem. Viral proteins mutate very fast, giving them a way to escape drugs by lowering drug binding affinity but with compromised function. Human immunodeficiency virus type I (HIV-1) protease, a critical antiretroviral therapeutic target, represents a model for such viral regulation under inhibition. Drug inhibitors of HIV-1 protease lose effectiveness as the protein evolves through several variants to become more resistant. However, the detailed mechanism of drug resistance in HIV-1 protease is still unclear. Here, we test the hypothesis that mutations throughout the protease alter the protein conformational ensemble to weaken protein-inhibitor binding, resulting in an inefficient protease but still viable virus. Comparing conformational ensembles between variants and the wild type helps detect these function-related dynamical changes. All analyses of over 30 µs simulations converge to the conclusion that conformational dynamics of more drug-resistant variants are more different from that of the wild type. Distinct roles of mutations during viral evolution are discussed, including a mutation predominantly contributing to the increase of drug resistance and a mutation that is responsible (synergistically) for restoring catalytic efficiency. Drug resistance is mainly due to altered flap dynamics that hinder the access to the active site. The mutant variant showing the highest drug resistance has the most ″collapsed″ active-site pocket and hence the largest magnitude of hindrance of drug binding. An enhanced difference contact network community analysis is applied to understand allosteric communications. The method summarizes multiple conformational ensembles in one community network and can be used in future studies to detect function-related dynamics in proteins.


Assuntos
Inibidores da Protease de HIV , Humanos , Inibidores da Protease de HIV/química , Sítios de Ligação , Farmacorresistência Viral/genética , Domínio Catalítico , Mutação , Protease de HIV/metabolismo
4.
Biochemistry ; 60(8): 597-606, 2021 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-33591178

RESUMO

The multifunctional protein p53 is the central molecular sensor of cellular stresses. The canonical function of p53 is to transcriptionally activate target genes in response to, for example, DNA damage that may trigger apoptosis. Recently, p53 was also found to play a role in the regulation of necrosis, another type of cell death featured by the mitochondrial permeability transition (mPT). In this process, p53 directly interacts with the mPT regulator cyclophilin D, the detailed mechanism of which however remains poorly understood. Here, we report a comprehensive computational investigation of the p53-cyclophilin D interaction using molecular dynamics simulations and associated analyses. We have identified the specific cyclophilin D binding site on p53 that is located at proline 151 in the DNA binding domain. As a peptidyl-prolyl isomerase, cyclophilin D binds p53 and catalyzes the cis-trans isomerization of the peptide bond preceding proline 151. We have also characterized the effect of such an isomerization and found that the p53 domain in the cis state is overall more rigid than the trans state except for the local region around proline 151. Dynamical changes upon isomerization occur in both local and distal regions, indicating an allosteric effect elicited by the isomerization. We present potential allosteric communication pathways between proline 151 and distal sites, including the DNA binding surface. Our work provides, for the first time, a model for how cyclophilin D binds p53 and regulates its activity by switching the configuration of a specific site.


Assuntos
Ciclofilinas/metabolismo , DNA/metabolismo , Simulação de Dinâmica Molecular , Prolina/química , Proteína Supressora de Tumor p53/metabolismo , Sítios de Ligação , Catálise , Ciclofilinas/química , Ciclofilinas/genética , DNA/química , Humanos , Prolina/metabolismo , Domínios Proteicos , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética
5.
Biochemistry ; 60(9): 711-724, 2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33630571

RESUMO

Proteins are inherently dynamic, and proper enzyme function relies on conformational flexibility. In this study, we demonstrated how an active site residue changes an enzyme's reactivity by modulating fluctuations between conformational states. Replacement of tyrosine 249 (Y249) with phenylalanine in the active site of the flavin-dependent d-arginine dehydrogenase yielded an enzyme with both an active yellow FAD (Y249F-y) and an inactive chemically modified green FAD, identified as 6-OH-FAD (Y249F-g) through various spectroscopic techniques. Structural investigation of Y249F-g and Y249F-y variants by comparison to the wild-type enzyme showed no differences in the overall protein structure and fold. A closer observation of the active site of the Y249F-y enzyme revealed an alternative conformation for some active site residues and the flavin cofactor. Molecular dynamics simulations probed the alternate conformations observed in the Y249F-y enzyme structure and showed that the enzyme variant with FAD samples a metastable conformational state, not available to the wild-type enzyme. Hybrid quantum/molecular mechanical calculations identified differences in flavin electronics between the wild type and the alternate conformation of the Y249F-y enzyme. The computational studies further indicated that the alternate conformation in the Y249F-y enzyme is responsible for the higher spin density at the C6 atom of flavin, which is consistent with the formation of 6-OH-FAD in the variant enzyme. The observations in this study are consistent with an alternate conformational space that results in fine-tuning the microenvironment around a versatile cofactor playing a critical role in enzyme function.


Assuntos
Aminoácido Oxirredutases/química , Aminoácido Oxirredutases/metabolismo , Flavinas/metabolismo , Fenilalanina/química , Mutação Puntual , Pseudomonas aeruginosa/enzimologia , Tirosina/química , Aminoácido Oxirredutases/genética , Sítios de Ligação , Catálise , Domínio Catalítico , Cinética , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Fenilalanina/genética , Fenilalanina/metabolismo , Conformação Proteica , Tirosina/genética , Tirosina/metabolismo
6.
Org Biomol Chem ; 19(8): 1835-1846, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33565564

RESUMO

Donor-π-acceptor (D-π-A) fluorophores consisting of a donor unit, a π linker, and an acceptor moiety have attracted attention in the last decade. In this study, we report the synthesis, characterization, optical properties, TD-DFT, and cytotoxicity studies of 17 near infrared (NIR) D-π-A analogs which have not been reported so far to the best of our knowledge. These fluorophores have chloroacrylic acid as the acceptor unit and various donor units such as indole, benzothiazole, benzo[e]indole, and quinoline. The fluorophores showed strong absorption in the NIR (700-970 nm) region due to their enhanced intramolecular charge transfer (ICT) between chloroacrylic acid and the donor moieties connected with the Vilsmeier-Haack linker. The emission wavelength maxima of the fluorophores were in between 798 and 870 nm. Compound 20 with a 4-quinoline donor moiety showed an emission wavelength above 1000 nm in the NIR II window. The synthesized fluorophores were characterized by 1H NMR and 13C NMR, and their optical properties were studied. Time dependent density functional theory (TD-DFT) calculations showed that the charge transfer occurs from the donor groups (indole, benzothiazole, benzo[e]indole, and quinoline) to the acceptor chloroacrylic acid moiety. Fluorophores with [HOMO] to [LUMO+1] transitions were shown to possess a charge separation character. The cytotoxicity of selected fluorophores, 4, 7, 10 and 12 was investigated against breast cancer cell lines and they showed better activity than the anti-cancer agent docetaxel.


Assuntos
Acrilatos/farmacologia , Antineoplásicos/farmacologia , Corantes Fluorescentes/farmacologia , Compostos Heterocíclicos com 2 Anéis/farmacologia , Acrilatos/síntese química , Acrilatos/efeitos da radiação , Antineoplásicos/síntese química , Antineoplásicos/efeitos da radiação , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Teoria da Densidade Funcional , Ensaios de Seleção de Medicamentos Antitumorais , Estabilidade de Medicamentos , Corantes Fluorescentes/síntese química , Corantes Fluorescentes/efeitos da radiação , Compostos Heterocíclicos com 2 Anéis/síntese química , Compostos Heterocíclicos com 2 Anéis/efeitos da radiação , Humanos , Luz , Modelos Químicos , Fenômenos Ópticos
7.
Angew Chem Int Ed Engl ; 60(43): 23289-23298, 2021 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-34436811

RESUMO

Multi-scale calcium (Ca2+ ) dynamics, exhibiting wide-ranging temporal kinetics, constitutes a ubiquitous mode of signal transduction. We report a novel endoplasmic-reticulum (ER)-targeted Ca2+ indicator, R-CatchER, which showed superior kinetics in vitro (koff ≥2×103  s-1 , kon ≥7×106  M-1 s-1 ) and in multiple cell types. R-CatchER captured spatiotemporal ER Ca2+ dynamics in neurons and hotspots at dendritic branchpoints, enabled the first report of ER Ca2+ oscillations mediated by calcium sensing receptors (CaSRs), and revealed ER Ca2+ -based functional cooperativity of CaSR. We elucidate the mechanism of R-CatchER and propose a principle to rationally design genetically encoded Ca2+ indicators with a single Ca2+ -binding site and fast kinetics by tuning rapid fluorescent-protein dynamics and the electrostatic potential around the chromophore. The design principle is supported by the development of G-CatchER2, an upgrade of our previous (G-)CatchER with improved dynamic range. Our work may facilitate protein design, visualizing Ca2+ dynamics, and drug discovery.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Cálcio/análise , Retículo Endoplasmático/metabolismo , Proteínas Luminescentes/metabolismo , Animais , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Proteínas de Ligação ao Cálcio/química , Células HEK293 , Células HeLa , Humanos , Proteínas Luminescentes/química , Camundongos , Simulação de Dinâmica Molecular , Ligação Proteica , Engenharia de Proteínas , Espectrometria de Fluorescência
8.
J Biol Chem ; 294(48): 18451-18464, 2019 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-31645439

RESUMO

Soluble guanylyl cyclase (sGC) is the main receptor for nitric oxide (NO) and a central component of the NO-cGMP pathway, critical to cardiovascular function. NO binding to the N-terminal sensor domain in sGC enhances the cyclase activity of the C-terminal catalytic domain. Our understanding of the structural elements regulating this signaling cascade is limited, hindering structure-based drug design efforts that target sGC to improve the management of cardiovascular diseases. Conformational changes are thought to propagate the NO-binding signal throughout the entire sGC heterodimer, via its coiled-coil domain, to reorient the catalytic domain into an active conformation. To identify the structural elements involved in this signal transduction cascade, here we optimized a cGMP-based luciferase assay that reports on heterologous sGC activity in Escherichia coli and identified several mutations that activate sGC. These mutations resided in the dorsal flaps, dimer interface, and GTP-binding regions of the catalytic domain. Combinations of mutations from these different elements synergized, resulting in even greater activity and indicating a complex cross-talk among these regions. Molecular dynamics simulations further revealed conformational changes underlying the functional impact of these mutations. We propose that the interfacial residues play a central role in the sGC activation mechanism by coupling the coiled-coil domain to the active site via a series of hot spots. Our results provide new mechanistic insights not only into the molecular pathway for sGC activation but also for other members of the larger nucleotidyl cyclase family.


Assuntos
GMP Cíclico/metabolismo , Simulação de Dinâmica Molecular , Mutação , Óxido Nítrico/metabolismo , Guanilil Ciclase Solúvel/genética , Sequência de Aminoácidos , Animais , Domínio Catalítico , GMP Cíclico/química , Ativação Enzimática/genética , Humanos , Cinética , Óxido Nítrico/química , Ligação Proteica , Conformação Proteica , Multimerização Proteica , Subunidades Proteicas/química , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Homologia de Sequência de Aminoácidos , Transdução de Sinais , Guanilil Ciclase Solúvel/química , Guanilil Ciclase Solúvel/metabolismo
9.
Acc Chem Res ; 52(12): 3455-3464, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31793290

RESUMO

Recent advances have made all-atom molecular dynamics (MD) a powerful tool to sample the conformational energy landscape. There are still however three major challenges in the application of MD to biological systems: accuracy of force field, time scale, and the analysis of simulation trajectories. Significant progress in addressing the first two challenges has been made and extensively reviewed previously. This Account focuses on strategies of analyzing simulation data of biomolecules that also covers ways to properly design simulations and validate simulation results. In particular, we examine an approach named comparative perturbed-ensembles analysis, which we developed to efficiently detect dynamics in protein MD simulations that can be linked to biological functions. In our recent studies, we implemented this approach to understand allosteric regulations in several disease-associated human proteins. The central task of a comparative perturbed-ensembles analysis is to compare two or more conformational ensembles of a system generated by MD simulations under distinct perturbation conditions. Perturbations can be different sequence variations, ligand-binding conditions, and other physical/chemical modifications of the system. Each simulation is long enough (e.g., microsecond-long) to ensure sufficient sampling of the local substate. Then, sophisticated bioinformatic and statistical tools are applied to extract function-related information from the simulation data, including principal component analysis, residue-residue contact analysis, difference contact network analysis (dCNA) based on the graph theory, and statistical analysis of side-chain conformations. Computational findings are further validated with experimental data. By comparing distinct conformational ensembles, functional micro- to millisecond dynamics can be inferred. In contrast, such a time scale is difficult to reach in a single simulation; even when reached for a single condition of a system, it is elusive as to what dynamical motions are related to functions without, for example, comparing free and substrate-bound proteins at the minimum. We illustrate our approach with three examples. First, we discuss using the approach to identify allosteric pathways in cyclophilin A (CypA), a member of a ubiquitous class of peptidyl-prolyl cis-trans isomerase enzymes. By comparing side-chain torsion-angle distributions of CypA in wild-type and mutant forms, we identified three pathways: two are consistent with recent nuclear magnetic resonance experiments, whereas the third is a novel pathway. Second, we show how the approach enables a dynamical-evolution analysis of the human cyclophilin family. In the analysis, both conserved and divergent conformational dynamics across three cyclophilin isoforms (CypA, CypD, and CypE) were summarized. The conserved dynamics led to the discovery of allosteric networks resembling those found in CypA. A residue wise determinant underlying the unique dynamics in CypD was also detected and validated with additional mutational MD simulations. In the third example, we applied the approach to elucidate a peptide sequence-dependent allosteric mechanism in human Pin 1, a phosphorylation-dependent peptidyl-prolyl isomerase. We finally present our outlook of future directions. Especially, we envisage how the approach could help open a new avenue in drug discovery.


Assuntos
Proteínas/química , Proteínas/metabolismo , Regulação Alostérica , Sequência de Aminoácidos , Animais , Humanos , Simulação de Dinâmica Molecular , Domínios Proteicos
10.
Bioconjug Chem ; 30(10): 2647-2663, 2019 10 16.
Artigo em Inglês | MEDLINE | ID: mdl-31518105

RESUMO

G-Quadruplex DNA has been recognized as a highly appealing target for the development of new selective chemotherapeutics, which could result in markedly reduced toxicity toward normal cells. In particular, the cyanine dyes that bind selectively to G-quadruplex structures without targeting duplex DNA have attracted attention due to their high amenability to structural modifications that allows fine-tuning of their biomolecular interactions. We have previously reported pentamethine and symmetric trimethine cyanines designed to effectively bind G-quadruplexes through end stacking interactions. Herein, we are reporting a second generation of drug candidates, the asymmetric trimethine cyanines. These have been synthesized and evaluated for their quadruplex binding properties. Incorporating a benz[c,d]indolenine heterocyclic unit increased overall quadruplex binding, and elongating the alkyl length increases the quadruplex-to-duplex binding specificity.


Assuntos
Alcinos/química , Alcinos/farmacologia , Quadruplex G/efeitos dos fármacos , Sequência de Bases , DNA/química , DNA/genética , Desenho de Fármacos , Indóis/química , Modelos Moleculares
11.
J Chem Inf Model ; 59(7): 3222-3228, 2019 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-31268315

RESUMO

Detailed understanding of interactions between amino acid residues is critical in using promising difference network analysis approaches to map allosteric communication pathways. Using experimental data as benchmarks, we scan values of two essential residue-residue contact parameters: the distance cutoff (dc) and the cutoff of residue separation in sequence (nc). The optimal dc = 4.5 Å is revealed, which defines the upper bound of the first shell of residue-residue packing in proteins, whereas nc is found to have little effects on performance. We also develop a new energy-based contact method for network analyses and find an equivalency between the energy network using the optimal energy cutoff ec = 1.0 kBT and the structure network using dc = 4.5 Å. The simple 4.5-Å contact method is further shown to have comparable prediction accuracy to a contact method using amino acid type-specific distance cutoffs and chemical shift prediction-based methods. This study provides necessary tools in mapping dynamics to functions.


Assuntos
Proteínas/química , Sequência de Aminoácidos , Biologia Computacional/métodos , Bicamadas Lipídicas/química , Modelos Moleculares , Simulação de Dinâmica Molecular , Conformação Proteica , Análise de Sequência de Proteína , Termodinâmica
12.
Proc Natl Acad Sci U S A ; 113(17): 4735-40, 2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27071107

RESUMO

Detailed understanding of how conformational dynamics orchestrates function in allosteric regulation of recognition and catalysis remains ambiguous. Here, we simulate CypA using multiple-microsecond-long atomistic molecular dynamics in explicit solvent and carry out NMR experiments. We analyze a large amount of time-dependent multidimensional data with a coarse-grained approach and map key dynamical features within individual macrostates by defining dynamics in terms of residue-residue contacts. The effects of substrate binding are observed to be largely sensed at a location over 15 Å from the active site, implying its importance in allostery. Using NMR experiments, we confirm that a dynamic cluster of residues in this distal region is directly coupled to the active site. Furthermore, the dynamical network of interresidue contacts is found to be coupled and temporally dispersed, ranging over 4 to 5 orders of magnitude. Finally, using network centrality measures we demonstrate the changes in the communication network, connectivity, and influence of CypA residues upon substrate binding, mutation, and during catalysis. We identify key residues that potentially act as a bottleneck in the communication flow through the distinct regions in CypA and, therefore, as targets for future mutational studies. Mapping these dynamical features and the coupling of dynamics to function has crucial ramifications in understanding allosteric regulation in enzymes and proteins, in general.


Assuntos
Aminoácidos/química , Ciclofilina A/química , Ciclofilina A/ultraestrutura , Espectroscopia de Ressonância Magnética/métodos , Modelos Químicos , Simulação de Dinâmica Molecular , Aminoácidos/genética , Sítios de Ligação , Catálise , Ciclofilina A/genética , Mutação , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
13.
J Chem Inf Model ; 58(7): 1325-1330, 2018 07 23.
Artigo em Inglês | MEDLINE | ID: mdl-29956925

RESUMO

A difference contact network analysis (dCNA) method is developed for delineating allosteric mechanisms in proteins. The new method addresses limitations of conventional network analysis methods and is particularly suitable for allosteric systems undergoing large-amplitude conformational changes during function. Tests show that dCNA works well for proteins of varying sizes and functions. The design of dCNA is general enough to facilitate analyses of diverse dynamic data generated by molecular dynamics, crystallography, or nuclear magnetic resonance.


Assuntos
Simulação de Dinâmica Molecular , Proteínas/química , Algoritmos , Regulação Alostérica , DNA/química , NF-kappa B/química , Ligação Proteica , Conformação Proteica , Termodinâmica
14.
Biochemistry ; 56(37): 5002-5010, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28825294

RESUMO

The molecular basis of gene regulation by Nuclear Factor-κB (NF-κB) transcription factors and their coregulators is not well understood. This family of transcription factors controls a number of essential subcellular processes. Human Pirin, a nonheme iron (Fe) binding protein, has been shown to modulate the binding affinity between p65 homodimeric NF-κB and κB DNA. However, the allosteric effect of the active Fe(III) form of Pirin on the DNA has not been established. Here, we use multiple microsecond-long molecular dynamics simulations to explore the conformational dynamics of the free DNA, the p65-DNA complex, and the Pirin-p65-DNA supramolecular complex. We show that only the Fe(III) form of Pirin enhances the affinity between p65 and the DNA in the Pirin-p65-DNA supramolecular complex, in agreement with experiments. Additionally, the results provide atomistic details of the effect of the active Fe(III) form of Pirin on the DNA upon binding to the p65-DNA complex. In general, unlike the Fe(II) form of Pirin, binding of the Fe(III) form of Pirin to the p65-DNA complex significantly alters both the conformational dynamics of the DNA and the interactions between p65 and the DNA. The results provide atomic level understanding of the modulation of the DNA as a result of a redox-specific Fe(II)/Fe(III) coregulation of NF-κB by Pirin, knowledge that is necessary to fully understand normal and aberrant subcellular processes and the role of a subtle single electron redox process in gene regulation.


Assuntos
DNA de Forma B/metabolismo , Dioxigenases/metabolismo , Modelos Moleculares , Fator de Transcrição RelA/metabolismo , Regulação Alostérica , Substituição de Aminoácidos , Cristalografia por Raios X , DNA de Forma B/química , Bases de Dados de Proteínas , Dimerização , Dioxigenases/química , Dioxigenases/genética , Transferência de Energia , Regulação da Expressão Gênica , Cinética , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Conformação de Ácido Nucleico , Oxirredução , Mutação Puntual , Análise de Componente Principal , Conformação Proteica , Multimerização Proteica , Fator de Transcrição RelA/química
15.
Biochemistry ; 56(19): 2477-2487, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28445031

RESUMO

Mobile loops located at the active site entrance in enzymes often participate in conformational changes required to shield the reaction from bulk solvent, to control the access of the substrate to the active site, and to position residues for substrate binding and catalysis. In d-arginine dehydrogenase from Pseudomonas aeruginosa (PaDADH), previous crystallographic data suggested that residues 45-47 in the FAD-binding domain and residues 50-56 in the substrate-binding domain in loop L1 could adopt two distinct conformations. In this study, we have used molecular dynamics, kinetics, and fluorescence spectroscopy on the S45A and A46G enzyme variants of PaDADH to investigate the impact of mutations in loop L1 on the catalytic function of the enzyme. Molecular dynamics showed that the mutant enzymes have probabilities of being in open conformations that are higher than that of wild-type PaDADH of loop L1, yielding an increased level of solvent exposure of the active site. In agreement, the flavin fluorescence intensity was ∼2-fold higher in the S45A and A46G enzymes than in wild-type PaDADH, with a 9 nm bathochromic shift of the emission band. In the variant enzymes, the kcat/Km values with d-arginine were ∼13-fold lower than in wild-type PaDADH. Moreover, the pH profiles for the kcat value with d-arginine showed a hollow, consistent with restricted proton movements in catalysis, and no saturation was achieved with the alternate substrate d-leucine in the reductive half-reaction of the variant enzymes. Taken together, the computational and experimental data are consistent with the dynamics of loop L1 being important for substrate capture and catalysis in PaDADH.


Assuntos
Aminoácido Oxirredutases/metabolismo , Proteínas de Bactérias/metabolismo , Modelos Moleculares , Pseudomonas aeruginosa/enzimologia , Algoritmos , Aminoácido Oxirredutases/química , Aminoácido Oxirredutases/genética , Arginina/química , Arginina/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Biocatálise , Domínio Catalítico , Bases de Dados de Proteínas , Concentração de Íons de Hidrogênio , Leucina/química , Leucina/metabolismo , Ligantes , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Mutação , Oxirredução , Conformação Proteica , Dobramento de Proteína , Desdobramento de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Estereoisomerismo , Especificidade por Substrato
16.
Biochemistry ; 56(29): 3800-3807, 2017 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-28640638

RESUMO

Flavins, including flavin adenine dinucleotide (FAD), are fundamental catalytic cofactors that are responsible for the redox functionality of a diverse set of proteins. Alternatively, modified flavin analogues are rarely found in nature as their incorporation typically results in inactivation of flavoproteins, thus leading to the disruption of important cellular pathways. Here, we report that the fungal flavoenzyme formate oxidase (FOX) catalyzes the slow conversion of noncovalently bound FAD to 8-formyl FAD and that this conversion results in a nearly 10-fold increase in formate oxidase activity. Although the presence of an enzyme-bound 8-formyl FMN has been reported previously as a result of site-directed mutagenesis studies of lactate oxidase, FOX is the first reported case of 8-formyl FAD in a wild-type enzyme. Therefore, the formation of the 8-formyl FAD cofactor in formate oxidase was investigated using steady-state kinetics, site-directed mutagenesis, ultraviolet-visible, circular dichroism, and fluorescence spectroscopy, liquid chromatography with mass spectrometry, and computational analysis. Surprisingly, the results from these studies indicate not only that 8-formyl FAD forms spontaneously and results in the active form of FOX but also that its autocatalytic formation is dependent on a nearby arginine residue, R87. Thus, this work describes a new enzyme cofactor and provides insight into the little-understood mechanism of enzyme-mediated 8α-flavin modifications.


Assuntos
Aspergillus oryzae/enzimologia , Coenzimas/química , Flavina-Adenina Dinucleotídeo/química , Proteínas Fúngicas/química , Oxirredutases Atuantes sobre Doadores de Grupos Aldeído ou Oxo/química , Substituição de Aminoácidos , Aspergillus oryzae/genética , Dicroísmo Circular , Coenzimas/metabolismo , Mononucleotídeo de Flavina/química , Mononucleotídeo de Flavina/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Cinética , Mutagênese Sítio-Dirigida , Mutação de Sentido Incorreto , Oxirredutases Atuantes sobre Doadores de Grupos Aldeído ou Oxo/genética , Oxirredutases Atuantes sobre Doadores de Grupos Aldeído ou Oxo/metabolismo
17.
Biochemistry ; 55(10): 1473-84, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26907558

RESUMO

Choline oxidase from Arthrobacter globiformis catalyzes the oxidation of choline to glycine betaine by using oxygen as an electron acceptor. A partially rate limiting isomerization of the reduced wild-type enzyme during the reaction with oxygen was previously detected using solvent viscosity effects. In this study, we hypothesized that the side chains of M62 and F357, located at the entrance to the active site of choline oxidase, may be related to the slow isomerization detected. We engineered a double-variant enzyme M62A/F357A. The kinetic characterization of the double-variant enzyme showed a lack of the isomerization detected in wild-type choline oxidase, and a lack of saturation with an oxygen concentration as high as 1 mM, while most other kinetic parameters were similar to those of wild-type choline oxidase. The kinetic characterization of the single-variant enzymes established that only the side chain of F357 plays a role in the isomerization of choline oxidase in the oxidative half-reaction. Molecular dynamics studies suggest that the slow isomerization related to F357 is possibly due to the participation of the phenyl ring in a newly proposed gating mechanism for a narrow tunnel, assumed to regulate the access of oxygen to the reduced cofactor.


Assuntos
Oxirredutases do Álcool/metabolismo , Arthrobacter , Proteínas de Bactérias/metabolismo , Oxigênio/metabolismo , Oxirredutases do Álcool/química , Oxirredutases do Álcool/genética , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Variação Genética/genética , Oxirredução , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
18.
J Biol Chem ; 290(25): 15621-15634, 2015 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-25918158

RESUMO

The rubredoxin motif is present in over 74,000 protein sequences and 2,000 structures, but few have known functions. A secondary, non-catalytic, rubredoxin-like iron site is conserved in 3-hydroxyanthranilate 3,4-dioxygenase (HAO), from single cellular sources but not multicellular sources. Through the population of the two metal binding sites with various metals in bacterial HAO, the structural and functional relationship of the rubredoxin-like site was investigated using kinetic, spectroscopic, crystallographic, and computational approaches. It is shown that the first metal presented preferentially binds to the catalytic site rather than the rubredoxin-like site, which selectively binds iron when the catalytic site is occupied. Furthermore, an iron ion bound to the rubredoxin-like site is readily delivered to an empty catalytic site of metal-free HAO via an intermolecular transfer mechanism. Through the use of metal analysis and catalytic activity measurements, we show that a downstream metabolic intermediate can selectively remove the catalytic iron. As the prokaryotic HAO is often crucial for cell survival, there is a need for ensuring its activity. These results suggest that the rubredoxin-like site is a possible auxiliary iron source to the catalytic center when it is lost during catalysis in a pathway with metabolic intermediates of metal-chelating properties. A spare tire concept is proposed based on this biochemical study, and this concept opens up a potentially new functional paradigm for iron-sulfur centers in iron-dependent enzymes as transient iron binding and shuttling sites to ensure full metal loading of the catalytic site.


Assuntos
3-Hidroxiantranilato 3,4-Dioxigenase/química , Proteínas de Bactérias/química , Burkholderiaceae/enzimologia , Ferro/química , Motivos de Aminoácidos , Catálise , Domínio Catalítico , Humanos , Saccharomyces cerevisiae/enzimologia , Relação Estrutura-Atividade
19.
Biochim Biophys Acta ; 1850(5): 878-888, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25153688

RESUMO

BACKGROUND: Accelerated molecular dynamics (aMD) has been proven to be a powerful biasing method for enhanced sampling of biomolecular conformations on general-purpose computational platforms. Biologically important long timescale events that are beyond the reach of standard molecular dynamics can be accessed without losing the detailed atomistic description of the system in aMD. Over other biasing methods, aMD offers the advantages of tuning the level of acceleration to access the desired timescale without any advance knowledge of the reaction coordinate. SCOPE OF REVIEW: Recent advances in the implementation of aMD and its applications to small peptides and biological macromolecules are reviewed here along with a brief account of all the aMD variants introduced in the last decade. MAJOR CONCLUSIONS: In comparison to the original implementation of aMD, the recent variant in which all the rotatable dihedral angles are accelerated (RaMD) exhibits faster convergence rates and significant improvement in statistical accuracy of retrieved thermodynamic properties. RaMD in conjunction with accelerating diffusive degrees of freedom, i.e. dual boosting, has been rigorously tested for the most difficult conformational sampling problem, protein folding. It has been shown that RaMD with dual boosting is capable of efficiently sampling multiple folding and unfolding events in small fast folding proteins. GENERAL SIGNIFICANCE: RaMD with the dual boost approach opens exciting possibilities for sampling multiple timescales in biomolecules. While equilibrium properties can be recovered satisfactorily from aMD-based methods, directly obtaining dynamics and kinetic rates for larger systems presents a future challenge. This article is part of a Special Issue entitled Recent developments of molecular dynamics.


Assuntos
Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Preparações Farmacêuticas/química , Proteínas/química , Cinética , Conformação Proteica , Dobramento de Proteína , Desdobramento de Proteína , Relação Estrutura-Atividade , Termodinâmica
20.
Biochim Biophys Acta ; 1850(10): 1994-2004, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25585011

RESUMO

BACKGROUND: Peptidyl prolyl cis-trans isomerization of the protein backbone is involved in the regulation of many biological processes. Cis-trans isomerization is notoriously slow and is catalyzed by a family of cis-trans peptidyl prolyl isomerases (PPIases) that have been implicated in many diseases. A general consensus on how these enzymes speed up prolyl isomerization has not been reached after decades of both experimental and computational studies. SCOPE OF REVIEW: Computational studies carried out to understand the catalytic mechanism of the prototypical FK506 binding protein 12, Cyclophilin A and peptidyl-prolyl cis-trans isomerase NIMA-interacting 1 (Pin1) are reviewed. A summary and an evaluation of the implications of the proposed mechanisms from computational studies are presented. MAJOR CONCLUSIONS: The analysis of computational studies and evaluation of the proposed mechanisms provide a general consensus and a better understanding of PPIase catalysis. The speedup of the rate of peptidyl-prolyl isomerization by PPIases can be best described by a catalytic mechanism in which the substrate in transition state configuration is stabilized. The enzymes preferentially bind the transition state configuration of the substrate relative to the cis conformation, which in most cases is bound better than the trans conformation of the substrate. Stabilization of the transition state configuration of the substrate leads to a lower free energy barrier and a faster rate of isomerization when compared to the uncatalyzed isomerization reaction. GENERAL SIGNIFICANCE: Fully understanding the catalytic mechanism of PPIases has broad implications for drug design, elucidation of the molecular basis of many diseases, protein engineering, and enzyme catalysis in general. This article is part of a Special Issue entitled Proline-directed Foldases: Cell Signaling Catalysts and Drug Targets.


Assuntos
Simulação por Computador , Modelos Moleculares , Peptidilprolil Isomerase/química , Animais , Catálise , Humanos , Peptidilprolil Isomerase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA