Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Syst ; 15(3): 246-263.e7, 2024 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-38366601

RESUMEN

Autoinhibition is a prevalent allosteric regulatory mechanism in signaling proteins. Reduced autoinhibition underlies the tumorigenic effect of some known cancer drivers, but whether autoinhibition is altered generally in cancer remains elusive. Here, we demonstrate that cancer-associated missense mutations, in-frame insertions/deletions, and fusion breakpoints are enriched within inhibitory allosteric switches (IASs) across all cancer types. Selection for IASs that are recurrently mutated in cancers identifies established and unknown cancer drivers. Recurrent missense mutations in IASs of these drivers are associated with distinct, cancer-specific changes in molecular signaling. For the specific case of PPP3CA, the catalytic subunit of calcineurin, we provide insights into the molecular mechanisms of altered autoinhibition by cancer mutations using biomolecular simulations, and demonstrate that such mutations are associated with transcriptome changes consistent with increased calcineurin signaling. Our integrative study shows that autoinhibition-modulating genetic alterations are positively selected for by cancer cells.


Asunto(s)
Calcineurina , Neoplasias , Humanos , Calcineurina/genética , Neoplasias/genética , Mutación/genética , Carcinogénesis , Mutación Missense/genética
2.
Mol Syst Biol ; 19(12): e11801, 2023 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-37984409

RESUMEN

The accumulation of misfolded and aggregated proteins is a hallmark of neurodegenerative proteinopathies. Although multiple genetic loci have been associated with specific neurodegenerative diseases (NDs), molecular mechanisms that may have a broader relevance for most or all proteinopathies remain poorly resolved. In this study, we developed a multi-layered network expansion (MLnet) model to predict protein modifiers that are common to a group of diseases and, therefore, may have broader pathophysiological relevance for that group. When applied to the four NDs Alzheimer's disease (AD), Huntington's disease, and spinocerebellar ataxia types 1 and 3, we predicted multiple members of the insulin pathway, including PDK1, Akt1, InR, and sgg (GSK-3ß), as common modifiers. We validated these modifiers with the help of four Drosophila ND models. Further evaluation of Akt1 in human cell-based ND models revealed that activation of Akt1 signaling by the small molecule SC79 increased cell viability in all models. Moreover, treatment of AD model mice with SC79 enhanced their long-term memory and ameliorated dysregulated anxiety levels, which are commonly affected in AD patients. These findings validate MLnet as a valuable tool to uncover molecular pathways and proteins involved in the pathophysiology of entire disease groups and identify potential therapeutic targets that have relevance across disease boundaries. MLnet can be used for any group of diseases and is available as a web tool at http://ssbio.cau.ac.kr/software/mlnet.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Huntington , Deficiencias en la Proteostasis , Animales , Humanos , Ratones , Enfermedad de Alzheimer/genética , Glucógeno Sintasa Quinasa 3 beta , Enfermedad de Huntington/genética , Transducción de Señal
3.
Biomolecules ; 13(3)2023 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-36979462

RESUMEN

Research in the field of biochemistry and cellular biology has entered a new phase due to the discovery of phase separation driving the formation of biomolecular condensates, or membraneless organelles, in cells. The implications of this novel principle of cellular organization are vast and can be applied at multiple scales, spawning exciting research questions in numerous directions. Of fundamental importance are the molecular mechanisms that underly biomolecular condensate formation within cells and whether insights gained into these mechanisms provide a gateway for accurate predictions of protein phase behavior. Within the last six years, a significant number of predictors for protein phase separation and condensate localization have emerged. Herein, we compare a collection of state-of-the-art predictors on different tasks related to protein phase behavior. We show that the tested methods achieve high AUCs in the identification of biomolecular condensate drivers and scaffolds, as well as in the identification of proteins able to phase separate in vitro. However, our benchmark tests reveal that their performance is poorer when used to predict protein segments that are involved in phase separation or to classify amino acid substitutions as phase-separation-promoting or -inhibiting mutations. Our results suggest that the phenomenological approach used by most predictors is insufficient to fully grasp the complexity of the phenomenon within biological contexts and make reliable predictions related to protein phase behavior at the residue level.


Asunto(s)
Condensados Biomoleculares , Proteínas , Proteínas/análisis , Orgánulos/química , Citoplasma , Sustitución de Aminoácidos
4.
Nucleic Acids Res ; 51(1): 99-116, 2023 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-36535377

RESUMEN

Numerous cancers, including prostate cancer (PCa), are addicted to transcription programs driven by specific genomic regions known as super-enhancers (SEs). The robust transcription of genes at such SEs is enabled by the formation of phase-separated condensates by transcription factors and coactivators with intrinsically disordered regions. The androgen receptor (AR), the main oncogenic driver in PCa, contains large disordered regions and is co-recruited with the transcriptional coactivator mediator complex subunit 1 (MED1) to SEs in androgen-dependent PCa cells, thereby promoting oncogenic transcriptional programs. In this work, we reveal that full-length AR forms foci with liquid-like properties in different PCa models. We demonstrate that foci formation correlates with AR transcriptional activity, as this activity can be modulated by changing cellular foci content chemically or by silencing MED1. AR ability to phase separate was also validated in vitro by using recombinant full-length AR protein. We also demonstrate that AR antagonists, which suppress transcriptional activity by targeting key regions for homotypic or heterotypic interactions of this receptor, hinder foci formation in PCa cells and phase separation in vitro. Our results suggest that enhanced compartmentalization of AR and coactivators may play an important role in the activation of oncogenic transcription programs in androgen-dependent PCa.


Asunto(s)
Neoplasias de la Próstata , Receptores Androgénicos , Masculino , Humanos , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Andrógenos , Factores de Transcripción/metabolismo , Regulación de la Expresión Génica , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/metabolismo , Expresión Génica , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica
5.
Biomolecules ; 10(8)2020 07 24.
Artículo en Inglés | MEDLINE | ID: mdl-32722039

RESUMEN

Because proteins are fundamental to most biological processes, many genetic diseases can be traced back to single nucleotide variants (SNVs) that cause changes in protein sequences. However, not all SNVs that result in amino acid substitutions cause disease as each residue is under different structural and functional constraints. Influential studies have shown that protein-protein interaction interfaces are enriched in disease-associated SNVs and depleted in SNVs that are common in the general population. These studies focus primarily on folded (globular) protein domains and overlook the prevalent class of protein interactions mediated by intrinsically disordered regions (IDRs). Therefore, we investigated the enrichment patterns of missense mutation-causing SNVs that are associated with disease and cancer, as well as those present in the healthy population, in structures of IDR-mediated interactions with comparisons to classical globular interactions. When comparing the different categories of interaction interfaces, division of the interface regions into solvent-exposed rim residues and buried core residues reveal distinctive enrichment patterns for the various types of missense mutations. Most notably, we demonstrate a strong enrichment at the interface core of interacting IDRs in disease mutations and its depletion in neutral ones, which supports the view that the disruption of IDR interactions is a mechanism underlying many diseases. Intriguingly, we also found an asymmetry across the IDR interaction interface in the enrichment of certain missense mutation types, which may hint at an increased variant tolerance and urges further investigations of IDR interactions.


Asunto(s)
Bases de Datos de Proteínas , Proteínas Intrínsecamente Desordenadas/genética , Mutación Missense , Polimorfismo de Nucleótido Simple , Algoritmos , Humanos , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Modelos Moleculares , Unión Proteica , Dominios Proteicos
6.
Structure ; 28(8): 922-932.e5, 2020 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-32492370

RESUMEN

STAC3 is a soluble protein essential for skeletal muscle excitation-contraction (EC) coupling. Through its tandem SH3 domains, it interacts with the cytosolic II-III loop of the skeletal muscle voltage-gated calcium channel. STAC3 is the target for a mutation (W284S) that causes Native American myopathy, but multiple other sequence variants have been reported. Here, we report a crystal structure of the human STAC3 tandem SH3 domains. We analyzed the effect of five disease-associated variants, spread over both SH3 domains, on their ability to bind to the CaV1.1 II-III loop and on muscle EC coupling. In addition to W284S, we find the F295L and K329N variants to affect both binding and EC coupling. The ability of the K329N variant, located in the second SH3 domain, to affect the interaction highlights the importance of both SH3 domains in association with CaV1.1. Our results suggest that multiple STAC3 variants may cause myopathy.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Canales de Calcio Tipo L/metabolismo , Fisura del Paladar/genética , Acoplamiento Excitación-Contracción , Hipertermia Maligna/genética , Miotonía Congénita/genética , Potenciales de Acción , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Sitios de Unión , Canales de Calcio Tipo L/química , Línea Celular , Humanos , Simulación de Dinámica Molecular , Mutación Missense , Unión Proteica , Conformación Proteica en Lámina beta , Dominios Homologos src
7.
Proc Natl Acad Sci U S A ; 116(33): 16326-16331, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31366629

RESUMEN

Phase separation drives numerous cellular processes, ranging from the formation of membrane-less organelles to the cooperative assembly of signaling proteins. Features such as multivalency and intrinsic disorder that enable condensate formation are found not only in cytosolic and nuclear proteins, but also in membrane-associated proteins. The ABC transporter Rv1747, which is important for Mycobacterium tuberculosis (Mtb) growth in infected hosts, has a cytoplasmic regulatory module consisting of 2 phosphothreonine-binding Forkhead-associated domains joined by an intrinsically disordered linker with multiple phospho-acceptor threonines. Here we demonstrate that the regulatory modules of Rv1747 and its homolog in Mycobacterium smegmatis form liquid-like condensates as a function of concentration and phosphorylation. The serine/threonine kinases and sole phosphatase of Mtb tune phosphorylation-enhanced phase separation and differentially colocalize with the resulting condensates. The Rv1747 regulatory module also phase-separates on supported lipid bilayers and forms dynamic foci when expressed heterologously in live yeast and M. smegmatis cells. Consistent with these observations, single-molecule localization microscopy reveals that the endogenous Mtb transporter forms higher-order clusters within the Mycobacterium membrane. Collectively, these data suggest a key role for phase separation in the function of these mycobacterial ABC transporters and their regulation via intracellular signaling.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Proteínas de la Membrana/genética , Mycobacterium tuberculosis/genética , Tuberculosis/genética , Transportadoras de Casetes de Unión a ATP/química , Citosol/metabolismo , Regulación Bacteriana de la Expresión Génica/genética , Humanos , Membrana Dobles de Lípidos/metabolismo , Proteínas de la Membrana/ultraestructura , Mycobacterium smegmatis/genética , Mycobacterium smegmatis/patogenicidad , Mycobacterium tuberculosis/patogenicidad , Mycobacterium tuberculosis/ultraestructura , Proteínas Nucleares/genética , Fosforilación/genética , Transducción de Señal/genética , Imagen Individual de Molécula , Tuberculosis/microbiología
8.
J Phys Chem Lett ; 9(17): 4901-4909, 2018 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-30102541

RESUMEN

Mapping the nanomechanical properties of amyloids can provide valuable insights into structure and assembly mechanisms of protein aggregates that underlie the development of various human diseases. Although it is well-known that amyloids exhibit an intrinsic stiffness comparable to that of silk (1-10 GPa), a detailed understanding of the directional dependence (anisotropy) of the stiffness of amyloids and how it relates to structural features in these protein aggregates is missing. Here we used steered molecular dynamics (SMD) simulations and amplitude modulation-frequency modulation (AM-FM) atomic force microscopy to measure the directional variation in stiffness of GNNQQNY amyloid crystals. We reveal that individual crystals display significant mechanical anisotropy and relate this anisotropy to subtle but mechanically important differences in interactions between interfaces that define the crystal architecture. Our results provide detailed insights into the structure-mechanics relationship of amyloid that may help in designing amyloid-based nanomaterials with tailored mechanical properties.

9.
Structure ; 26(7): 972-986.e6, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29861345

RESUMEN

The Mycobacterium tuberculosis ATP-binding cassette transporter Rv1747 is a putative exporter of cell wall biosynthesis intermediates. Rv1747 has a cytoplasmic regulatory module consisting of two pThr-interacting Forkhead-associated (FHA) domains connected by a conformationally disordered linker with two phospho-acceptor threonines (pThr). The structures of FHA-1 and FHA-2 were determined by X-ray crystallography and nuclear magnetic resonance (NMR) spectroscopy, respectively. Relative to the canonical 11-strand ß-sandwich FHA domain fold of FHA-1, FHA-2 is circularly permuted and lacking one ß-strand. Nevertheless, the two share a conserved pThr-binding cleft. FHA-2 is less stable and more dynamic than FHA-1, yet binds model pThr peptides with moderately higher affinity (∼50 µM versus 500 µM equilibrium dissociation constants). Based on NMR relaxation and chemical shift perturbation measurements, when joined within a polypeptide chain, either FHA domain can bind either linker pThr to form intra- and intermolecular complexes. We hypothesize that this enables tunable phosphorylation-dependent multimerization to regulate Rv1747 transporter activity.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/metabolismo , Mycobacterium tuberculosis/metabolismo , Sitios de Unión , Cristalografía por Rayos X , Citoplasma/metabolismo , Modelos Moleculares , Mycobacterium tuberculosis/química , Resonancia Magnética Nuclear Biomolecular , Fosfotreonina/metabolismo , Unión Proteica , Estructura Secundaria de Proteína
10.
Biophys J ; 112(4): 584-594, 2017 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-28256219

RESUMEN

Amyloids are fibrillar nanostructures of proteins that are assembled in several physiological processes in human cells (e.g., hormone storage) but also during the course of infectious (prion) and noninfectious (nonprion) diseases such as Creutzfeldt-Jakob and Alzheimer's diseases, respectively. How the amyloid state, a state accessible to all proteins and peptides, can be exploited for functional purposes but also have detrimental effects remains to be determined. Here, we measure the nanomechanical properties of different amyloids and link them to features found in their structure models. Specifically, we use shape fluctuation analysis and sonication-induced scission in combination with full-atom molecular dynamics simulations to reveal that the amyloid fibrils of the mammalian prion protein PrP are mechanically unstable, most likely due to a very low hydrogen bond density in the fibril structure. Interestingly, amyloid fibrils formed by HET-s, a fungal protein that can confer functional prion behavior, have a much higher Young's modulus and tensile strength than those of PrP, i.e., they are much stiffer and stronger due to a tighter packing in the fibril structure. By contrast, amyloids of the proteins RIP1/RIP3 that have been shown to be of functional use in human cells are significantly stiffer than PrP fibrils but have comparable tensile strength. Our study demonstrates that amyloids are biomaterials with a broad range of nanomechanical properties, and we provide further support for the strong link between nanomechanics and ß-sheet characteristics in the amyloid core.


Asunto(s)
Amiloide/química , Fenómenos Mecánicos , Multimerización de Proteína , Fenómenos Biomecánicos , Humanos , Enlace de Hidrógeno , Insulina/química , Simulación de Dinámica Molecular , Estructura Secundaria de Proteína
11.
J Comput Chem ; 37(7): 629-40, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26558440

RESUMEN

Implicit solvent models for biomolecular simulations have been developed to use in place of more expensive explicit models; however, these models make many assumptions and approximations that are likely to affect accuracy. Here, the changes in free energies of solvation upon folding ΔΔGsolv of several fast folding proteins are calculated from previously run µs-ms simulations with a number of implicit solvent models and compared to the values needed to be consistent with the explicit solvent model used in the simulations. In the majority of cases, there is a significant and substantial difference between the ΔΔGsolv values calculated from the two approaches that is robust to the details of the calculations. These differences could only be remedied by selecting values for the model parameters-the internal dielectric constant for the polar term and the surface tension coefficient for the nonpolar term-that were system-specific or physically unrealistic. We discuss the potential implications of our findings for both implicit and explicit solvent simulations. © 2015 Wiley Periodicals, Inc.


Asunto(s)
Simulación por Computador , Modelos Moleculares , Péptidos/química , Oligopéptidos/química , Conformación Proteica , Pliegue de Proteína , Solventes/química , Termodinámica
12.
Chembiochem ; 15(11): 1607-13, 2014 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-25044481

RESUMEN

Protein arginine N-methyltransferases (PRMTs) catalyze methyl-group transfer from S-adenosyl-L-methionine onto arginine residues in proteins. In this study, modifications were introduced at the guanidine moiety of a peptidyl arginine residue to investigate how changes to the PRMT substrate can modulate enzyme activity. We found that peptides bearing Nη-hydroxy or Nη-amino substituted arginine showed higher apparent kcat values than for the monomethylated substrate when using PRMT1, whereas this catalytic preference was not observed for PRMT4 and PRMT6. Methylation by compromised PRMT1 variants E153Q and D51N further supports the finding that the N-hydroxy substitution facilitates methyl transfer by tuning the reactivity of the guanidine moiety. In contrast, Nη-nitro and Nη-canavanine substituted substrates inhibit PRMT activity. These findings demonstrate that methylation of these PRMT substrates is dependent on the nature of the modification at the guanidine moiety.


Asunto(s)
Arginina/metabolismo , Péptidos/química , Péptidos/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Estructura Molecular , Proteína-Arginina N-Metiltransferasas/química , Especificidad por Sustrato
13.
Structure ; 22(8): 1196-1203, 2014 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-25017730

RESUMEN

Functions of many proteins are affected by posttranslational modifications of intrinsically disordered (ID) regions, yet little is known about the underlying molecular mechanisms. By combining molecular dynamics simulations and protein docking, we demonstrate that the addition of phosphates to an ID segment adjacent to the PNT domain of Ets1 directs conformational sampling toward substates that are most compatible with high-affinity binding of the TAZ1 domain of its coactivator CBP. The phosphate charges disrupt salt bridges and thereby open a hydrophobic cleft and expose hydrophobic residues at the ID N terminus. The structure of the PNT-TAZ1 complex that we determined shows that PNT binds to TAZ1 via these hydrophobic regions in a similar manner to how it interacts with other partners. Our calculations reveal a dual effect of phosphorylation in that it changes the dynamics of PNT so that it becomes more compatible for TAZ1 binding and increases complementarity with this binding partner.


Asunto(s)
Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo , Modelos Moleculares , Proteína Proto-Oncogénica c-ets-1/química , Proteína Proto-Oncogénica c-ets-1/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Simulación de Dinámica Molecular , Fragmentos de Péptidos/metabolismo , Fosforilación , Unión Proteica , Conformación Proteica , Sialoglicoproteínas/metabolismo
14.
FEBS Lett ; 584(6): 1235-9, 2010 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-20171214

RESUMEN

The giant protein titin, which comprises immunoglobulin (Ig) domains, acts as a bidirectional spring in muscle. The unfolding of Ig domains has been extensively studied, but their dynamics under native states have not been well-characterized. We performed molecular dynamics simulation on a single titin Ig domain and multi-domains. Mobile regions displaying concerted motions were identified. The dynamics of Ig domains are constrained by evolutionary pressures, in such a way that global dominant motion is conserved, yet different flexibilities within Ig domains and in linkers connecting neighbouring domains were observed. We explain these heterogeneous conserved dynamics in relation to sequence conservation across species and the sequence diversity among neighbouring Ig domains.


Asunto(s)
Secuencia Conservada , Evolución Molecular , Inmunoglobulinas/química , Proteínas Musculares/química , Proteínas Quinasas/química , Secuencia de Aminoácidos , Conectina , Entropía , Humanos , Modelos Moleculares , Simulación de Dinámica Molecular , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
15.
Biophys J ; 97(9): 2513-20, 2009 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-19883594

RESUMEN

The functions of proteins depend on the dynamical behavior of their native states on a wide range of timescales. To investigate these dynamics in the case of the small protein Gbeta1, we analyzed molecular dynamics simulations with the model-free approach of nuclear magnetic relaxation. We found amplitudes of fast timescale motions (sub-tau(c), where tau(c) is the rotational correlation time) consistent with S(2) obtained from spin relaxation measurements as well as amplitudes of slow timescale motions (supra-tau(c)) in quantitative agreement with S(2) order parameters derived from residual dipolar coupling measurements. The slow timescale motions are associated with the large variations of the (3)J couplings that follow transitions between different conformational substates. These results provide further characterization of the large structural fluctuations in the native states of proteins that occur on timescales longer than the rotational correlation time.


Asunto(s)
Proteínas Bacterianas/química , Streptococcus/metabolismo , Proteínas Bacterianas/metabolismo , Biofisica/métodos , Simulación por Computador , Concentración de Iones de Hidrógeno , Espectroscopía de Resonancia Magnética , Magnetismo , Modelos Estadísticos , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Electricidad Estática , Factores de Tiempo
17.
Structure ; 16(5): 736-46, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18462678

RESUMEN

We used nuclear magnetic resonance data to determine ensembles of conformations representing the structure and dynamics of calmodulin (CaM) in the calcium-bound state (Ca(2+)-CaM) and in the state bound to myosin light chain kinase (CaM-MLCK). These ensembles reveal that the Ca(2+)-CaM state includes a range of structures similar to those present when CaM is bound to MLCK. Detailed analysis of the ensembles demonstrates that correlated motions within the Ca(2+)-CaM state direct the structural fluctuations toward complex-like substates. This phenomenon enables initial ligation of MLCK at the C-terminal domain of CaM and induces a population shift among the substates accessible to the N-terminal domain, thus giving rise to the cooperativity associated with binding. Based on these results and the combination of modern free energy landscape theory with classical allostery models, we suggest that a coupled equilibrium shift mechanism controls the efficient binding of CaM to a wide range of ligands.


Asunto(s)
Calmodulina/metabolismo , Transducción de Señal , Calcio/metabolismo , Calmodulina/química , Ligandos , Modelos Moleculares , Movimiento (Física) , Quinasa de Cadena Ligera de Miosina/metabolismo , Resonancia Magnética Nuclear Biomolecular , Conformación Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
18.
Chem Biol Interact ; 175(1-3): 303-4, 2008 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-18452905

RESUMEN

Proteins have been metaphorically described--due to the introduction and extraordinary advances in biomolecular dynamics and computational biophysics over the past decades--as "kicking and screaming" molecules [G. Weber, Adv. Protein Chem. 29 (1975) 1-83]. In fact, dynamic fluctuations in protein structural conformation have been known to play an important role in protein function. However, fundamental mechanisms by which protein fluctuations couple with catalytic function of particular enzymes remain poorly understood. To understand the dynamical properties of acetylcholinesterase (AChE) in rapid termination of cationic neurotransmitter, acetylcholine at neurosynaptic junctions, multiple molecular dynamics (MD) trajectories of AChE in the presence and absence of its inhibitors [J.M. Bui, J.A. McCammon, Proc. Natl. Acad. Sci. U.S.A. 103 (2006) 15451-15456; J.M. Bui, Z. Radic, P. Taylor, J.A. McCammon, Biophys. J. 90 (2006) 3280-3287; J.M. Bui, K. Tai, J.A. McCammon, J. Am. Chem. Soc. 126 (2004) 7198-7205; J.M. Bui, R.H. Henchman, J.A. McCammon, Biophys. J. 85 (2003) 2267-2272] have been conducted and correlated with its inhibitory mechanisms. The intrinsic flexibilities of AChE, particularly of the long omega loop, are important in facilitating the ligand's inhibition of the enzyme.


Asunto(s)
Acetilcolinesterasa/química , Acetilcolinesterasa/metabolismo , Biocatálisis , Modelos Moleculares , Conformación Proteica
19.
Proc Natl Acad Sci U S A ; 103(42): 15451-6, 2006 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-17021015

RESUMEN

Specific, rapid association of protein complexes is essential for all forms of cellular existence. The initial association of two molecules in diffusion-controlled reactions is often influenced by the electrostatic potential. Yet, the detailed binding mechanisms of proteins highly depend on the particular system. A complete protein complex formation pathway has been delineated by using structural information sampled over the course of the transformation reaction. The pathway begins at an encounter complex that is formed by one of the apo forms of neurotoxin fasciculin-2 (FAS2) and its high-affinity binding protein, acetylcholinesterase (AChE), followed by rapid conformational rearrangements into an intermediate complex that subsequently converts to the final complex as observed in crystal structures. Formation of the intermediate complex has also been independently captured in a separate 20-ns molecular dynamics simulation of the encounter complex. Conformational transitions between the apo and liganded states of FAS2 in the presence and absence of AChE are described in terms of their relative free energy profiles that link these two states. The transitions of FAS2 after binding to AChE are significantly faster than in the absence of AChE; the energy barrier between the two conformational states is reduced by half. Conformational rearrangements of FAS2 to the final liganded form not only bring the FAS2/AChE complex to lower energy states, but by controlling transient motions that lead to opening or closing one of the alternative passages to the active site of the enzyme also maximize the ligand's inhibition of the enzyme.


Asunto(s)
Acetilcolinesterasa/química , Venenos Elapídicos/química , Conformación Proteica , Acetilcolinesterasa/metabolismo , Regulación Alostérica , Venenos Elapídicos/metabolismo , Modelos Moleculares , Complejos Multiproteicos , Unión Proteica , Solventes/química , Termodinámica
20.
Biophys J ; 90(9): 3280-7, 2006 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-16473897

RESUMEN

The neurotoxin fasciculin-2 (FAS2) is a picomolar inhibitor of synaptic acetylcholinesterase (AChE). The dynamics of binding between FAS2 and AChE is influenced by conformational fluctuations both before and after protein encounter. Submicrosecond molecular dynamics trajectories of apo forms of fasciculin, corresponding to different conformational substates, are reported here with reference to the conformational changes of loop I of this three-fingered toxin. This highly flexible loop exhibits an ensemble of conformations within each substate corresponding to its functions. The high energy barrier found between the two major substates leads to transitions that are slow on the timescale of the diffusional encounter of noninteracting FAS2 and AChE. The more stable of the two apo substates may not be the one observed in the complex with AChE. It seems likely that the more stable apo form binds rapidly to AChE and conformational readjustments then occur in the resulting encounter complex.


Asunto(s)
Acetilcolinesterasa/metabolismo , Inhibidores de la Colinesterasa/metabolismo , Venenos Elapídicos/metabolismo , Acetilcolinesterasa/química , Animales , Simulación por Computador , Venenos Elapídicos/química , Cinética , Modelos Biológicos , Unión Proteica/fisiología , Conformación Proteica , Mapeo de Interacción de Proteínas , Estructura Terciaria de Proteína , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA