Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
J Chem Inf Model ; 63(20): 6177-6182, 2023 10 23.
Artículo en Inglés | MEDLINE | ID: mdl-37782001

RESUMEN

The industry's transition from three-dimensional (3D) glasses to virtual reality (VR) headsets has left modelers stranded without hardware supply, since walking around and waving arms in a virtual world is a great experience but also very tiring when doing time-intensive modeling work. We present a novel software implementation that uses a VR headset while sitting at a desk in front of the normal screen, which is beamed into the virtual reality together with keyboard, mouse, and chair using the headset's cameras and an extra tracker attached to the seat-back. Compared to 3D glasses, this yields a comparably relaxing but much more immersive workplace and provides additional possibilities such as taking molecules into one's hands, standing up, and walking or teleporting through the models. This VR functionality has been combined with a molecular graphics engine based on Vulkan, a next-generation cross-platform application programming interface (API) for GPUs and the successor of the widely used Open Graphics Library (OpenGL). It is built into the YASARA Model program, which includes many features like small and large molecule builders, electron densities, partial surfaces, contact analysis, coordinate manipulation, and animations. Interactive tutorials are provided to guide modelers into VR and familiarize them with the molecular modeling features. YASARA Model is available for Linux, Windows, Android, and MacOS (the latter without VR) with an introductory video at www.YASARA.org/vr.


Asunto(s)
Interfaz Usuario-Computador , Realidad Virtual , Humanos , Programas Informáticos , Computadores , Modelos Moleculares
2.
J Chem Inf Model ; 61(10): 5293-5303, 2021 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-34528431

RESUMEN

Building and displaying all-atom models of biomolecular structures with millions or billions of atoms, like virus particles or cells, remain a challenge due to the sheer size of the data, the required levels of automated building, and the visualization limits of today's graphics hardware. Based on concepts introduced with the CellPack program, we report new algorithms to create such large-scale models using an intermediate coarse-grained "pet representation" of biomolecules with 1/10th the normal size. Pet atoms are placed such that they optimally trace the surface of the original molecule with just ∼1/50th the original atom number and are joined with covalent bonds. Molecular dynamics simulations of pet molecules allow for efficient packing optimization, as well as the generation of realistic DNA/RNA conformations. This pet world can be expanded back to the all-atom representation to be explored and visualized with full details. Essential for the efficient interactive visualization of gigastructures is the use of multiple levels of detail (LODs), where distant molecules are drawn with a heavily reduced polygon count. We present a grid-based algorithm to create such LODs for all common molecular graphics styles (including ball-and-sticks, ribbons, and cartoons) that do not require monochrome molecules to hide LOD transitions. As a practical application, we built all-atom models of SARS-CoV-2, HIV, and an entire presynaptic bouton with 1 µm diameter and 3.6 billion atoms, using modular building blocks to significantly reduce GPU memory requirements through instancing. We employ the Vulkan graphics API to maximize performance on consumer grade hardware and describe how to use the mmCIF format to efficiently store such giant models. An implementation is available as part of the YASARA molecular modeling and simulation program from www.YASARA.org. The free YASARA View program can be used to explore the presented models, which can be downloaded from www.YASARA.org/petworld, a Creative Commons platform for sharing giant biomolecular structures.


Asunto(s)
COVID-19 , Gráficos por Computador , Algoritmos , Humanos , Simulación de Dinámica Molecular , SARS-CoV-2
3.
Protein Sci ; 29(1): 330-344, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31724231

RESUMEN

We describe a series of databases and tools that directly or indirectly support biomedical research on macromolecules, with focus on their applicability in protein structure bioinformatics research. DSSP, that determines secondary structures of proteins, has been updated to work well with extremely large structures in multiple formats. The PDBREPORT database that lists anomalies in protein structures has been remade to remove many small problems. These reports are now available as PDF-formatted files with a computer-readable summary. The VASE software has been added to analyze and visualize HSSP multiple sequence alignments for protein structures. The Lists collection of databases has been extended with a series of databases, most noticeably with a database that gives each protein structure a grade for usefulness in protein structure bioinformatics projects. The PDB-REDO collection of reanalyzed and re-refined protein structures that were solved by X-ray crystallography has been improved by dealing better with sugar residues and with hydrogen bonds, and adding many missing surface loops. All academic software underlying these protein structure bioinformatics applications and databases are now publicly accessible, either directly from the authors or from the GitHub software repository.


Asunto(s)
Biología Computacional/métodos , Recolección de Datos/métodos , Proteínas/química , Bases de Datos de Proteínas , Modelos Moleculares , Estructura Secundaria de Proteína , Programas Informáticos
4.
Biochim Biophys Acta Gen Subj ; 1863(3): 528-533, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30586626

RESUMEN

BACKGROUND: Binding of chemokines to glycosaminoglycans (GAGs) is a crucial step in leukocyte recruitment to inflamed tissues. METHODS: A disaccharide compositional analysis of the HS dp6 fraction in combination with MS analysis of the CCL2-depleted dp6 fraction was the basis for target GAG ligand structure suggestions. Four experimentally-derived heparan sulfate hexasaccharides, two potentially chemokine-specific and two unspecific, have been docked to CCL2. Subsequent 300 ns molecular dynamics simulations were used to improve the docked complexes. RESULTS: Hexasaccharides with four sulfations and no acetylations are suggested for selective and high affinity chemokine binding. Using the Antithromin-III/heparin complex as positive control for docking, we were able to recover the correct complex structure only if the previously liganded ATIII structure was used as input. Since the liganded structure is not known for a CCL2-GAG complex, we investigated if molecular dynamics simulations could improve initial docking results. We found that all four GAG oligosaccharides ended up in close contact with the known binding residues after about 100 ns simulation time. CONCLUSIONS: A discrimination of specific vs. unspecific CCL2 GAG ligands is not possible by this approach. Long-time molecular dynamics simulations are, however, well suited to capture the delicate enthalpy/entropy balance of GAG binding and improve results obtained from docking. GENERAL SIGNIFICANCE: With the comparison of two methods, MS-based ligand identification and molecular modelling, we have shown the current limitations of our molecular understanding of complex ligand binding which is could be due to the numerical inaccessibility of ligand-induced protein conformational changes.


Asunto(s)
Quimiocina CCL2/química , Quimiocina CCL2/metabolismo , Heparitina Sulfato/metabolismo , Simulación de Dinámica Molecular , Oligosacáridos/metabolismo , Secuencia de Carbohidratos , Fraccionamiento Químico/métodos , Cromatografía Líquida de Alta Presión/métodos , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Heparitina Sulfato/química , Heparitina Sulfato/aislamiento & purificación , Oligosacáridos/química , Oligosacáridos/aislamiento & purificación , Unión Proteica
5.
Sci Rep ; 7: 44751, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-28303975

RESUMEN

AAA-ATPases fulfil essential roles in different cellular pathways and often act in form of hexameric complexes. Interaction with pathway-specific substrate and adaptor proteins recruits them to their targets and modulates their catalytic activity. This substrate dependent regulation of ATP hydrolysis in the AAA-domains is mediated by a non-catalytic N-terminal domain. The exact mechanisms that transmit the signal from the N-domain and coordinate the individual AAA-domains in the hexameric complex are still the topic of intensive research. Here, we present the characterization of a novel mutant variant of the eukaryotic AAA-ATPase Drg1 that shows dysregulation of ATPase activity and altered interaction with Rlp24, its substrate in ribosome biogenesis. This defective regulation is the consequence of amino acid exchanges at the interface between the regulatory N-domain and the adjacent D1 AAA-domain. The effects caused by these mutations strongly resemble those of pathological mutations of the AAA-ATPase p97 which cause the hereditary proteinopathy IBMPFD (inclusion body myopathy associated with Paget's disease of the bone and frontotemporal dementia). Our results therefore suggest well conserved mechanisms of regulation between structurally, but not functionally related members of the AAA-family.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Adenosina Trifosfatasas/química , Alelos , Secuencia Conservada , Modelos Moleculares , Mutación/genética , Fenotipo , Dominios Proteicos , Relación Estructura-Actividad , Especificidad por Sustrato , Supresión Genética , Temperatura
6.
J Comput Chem ; 36(13): 996-1007, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25824339

RESUMEN

We describe a set of algorithms that allow to simulate dihydrofolate reductase (DHFR, a common benchmark) with the AMBER all-atom force field at 160 nanoseconds/day on a single Intel Core i7 5960X CPU (no graphics processing unit (GPU), 23,786 atoms, particle mesh Ewald (PME), 8.0 Å cutoff, correct atom masses, reproducible trajectory, CPU with 3.6 GHz, no turbo boost, 8 AVX registers). The new features include a mixed multiple time-step algorithm (reaching 5 fs), a tuned version of LINCS to constrain bond angles, the fusion of pair list creation and force calculation, pressure coupling with a "densostat," and exploitation of new CPU instruction sets like AVX2. The impact of Intel's new transactional memory, atomic instructions, and sloppy pair lists is also analyzed. The algorithms map well to GPUs and can automatically handle most Protein Data Bank (PDB) files including ligands. An implementation is available as part of the YASARA molecular modeling and simulation program from www.YASARA.org.


Asunto(s)
Algoritmos , Modelos Químicos , Simulación de Dinámica Molecular , Secuencia de Aminoácidos
7.
Bioinformatics ; 30(20): 2981-2, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24996895

RESUMEN

SUMMARY: Today's graphics processing units (GPUs) compose the scene from individual triangles. As about 320 triangles are needed to approximate a single sphere-an atom-in a convincing way, visualizing larger proteins with atomic details requires tens of millions of triangles, far too many for smooth interactive frame rates. We describe a new approach to solve this 'molecular graphics problem', which shares the work between GPU and multiple CPU cores, generates high-quality results with perfectly round spheres, shadows and ambient lighting and requires only OpenGL 1.0 functionality, without any pixel shader Z-buffer access (a feature which is missing in most mobile devices). AVAILABILITY AND IMPLEMENTATION: YASARA View, a molecular modeling program built around the visualization algorithm described here, is freely available (including commercial use) for Linux, MacOS, Windows and Android (Intel) from www.YASARA.org. CONTACT: elmar@yasara.org SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Teléfono Celular , Gráficos por Computador , Computadores , Modelos Moleculares , Algoritmos , Proteínas/química
8.
J Biol Chem ; 289(7): 3913-22, 2014 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-24371142

RESUMEN

The drug diazaborine is the only known inhibitor of ribosome biogenesis and specifically blocks large subunit formation in eukaryotic cells. However, the target of this drug and the mechanism of inhibition were unknown. Here we identify the AAA-ATPase Drg1 as a target of diazaborine. Inhibitor binding into the second AAA domain of Drg1 requires ATP loading and results in inhibition of ATP hydrolysis in this site. As a consequence the physiological activity of Drg1, i.e. the release of Rlp24 from pre-60S particles, is blocked, and further progression of cytoplasmic preribosome maturation is prevented. Our results identify the first target of an inhibitor of ribosome biogenesis and provide the mechanism of inhibition of a key step in large ribosomal subunit formation.


Asunto(s)
Adenosina Trifosfatasas/antagonistas & inhibidores , Compuestos de Boro/farmacología , Inhibidores Enzimáticos/farmacología , Proteínas de Saccharomyces cerevisiae/antagonistas & inhibidores , Saccharomyces cerevisiae/enzimología , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Sitios de Unión , Compuestos de Boro/química , Citoplasma/enzimología , Citoplasma/genética , Inhibidores Enzimáticos/química , Proteínas Ribosómicas/biosíntesis , Proteínas Ribosómicas/genética , Subunidades Ribosómicas Grandes de Eucariotas/genética , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
9.
J Biomol NMR ; 54(3): 267-83, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22986687

RESUMEN

We present a suite of programs, named CING for Common Interface for NMR Structure Generation that provides for a residue-based, integrated validation of the structural NMR ensemble in conjunction with the experimental restraints and other input data. External validation programs and new internal validation routines compare the NMR-derived models with empirical data, measured chemical shifts, distance- and dihedral restraints and the results are visualized in a dynamic Web 2.0 report. A red-orange-green score is used for residues and restraints to direct the user to those critiques that warrant further investigation. Overall green scores below ~20 % accompanied by red scores over ~50 % are strongly indicative of poorly modelled structures. The publically accessible, secure iCing webserver ( https://nmr.le.ac.uk ) allows individual users to upload the NMR data and run a CING validation analysis.


Asunto(s)
Resonancia Magnética Nuclear Biomolecular/métodos , Proteínas/química , Programas Informáticos , Modelos Moleculares , Conformación Proteica , Reproducibilidad de los Resultados , Interfaz Usuario-Computador
10.
Biochem Pharmacol ; 84(3): 366-73, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22542979

RESUMEN

Multidrug resistance-associated protein 4 (MRP4) is a membrane transporter that mediates the cellular efflux of a wide range of anionic drugs and endogenous molecules. MRP4 transport can influence the pharmacokinetics of drugs and their metabolites, therefore more knowledge about the molecular determinants important for its transport function would be of relevance. Here, we substituted amino acids Phe(368), Trp(995), and Arg(998) with conservative or non-conservative residues, and determined the effect on transport of the model substrates estradiol 17-ß-d-glucuronide (E(2)17ßG), cyclic guanosine monophosphate (cGMP), methotrexate (MTX), and folic acid into membrane vesicles isolated from baculovirus transduced HEK293 cells overexpressing the mutant MRP4 proteins. This revealed that all Arg(998) mutations appeared to be deleterious, whereas the effect of a Phe(368) or Trp(995) replacement was dependent on the amino acid introduced and the substrate studied. Substitution of Phe(368) with Trp (F368W) induced a gain-of-function of E(2)17ßG transport and a loss-of-function of MTX transport, which could not be attributed to an altered substrate binding. Moreover, we did not observe any modification in ATP or ADP handling for F368W. These results, in combination with docking of substrates in a homology model of MRP4 in the inward- and outward-facing conformation, suggest that Phe(368) and Trp(995) do not play an important role in the initial binding of substrates. They, however, might interact with the substrates during rearrangement of helixes for substrate translocation, funneling the substrates to the exit site in the outward-facing conformation.


Asunto(s)
Proteínas Asociadas a Resistencia a Múltiples Medicamentos/química , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/fisiología , Fenilalanina/fisiología , Secuencia de Aminoácidos , Sustitución de Aminoácidos/genética , Regulación hacia Abajo/genética , Células HEK293 , Humanos , Datos de Secuencia Molecular , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Fenilalanina/química , Fenilalanina/genética , Estructura Secundaria de Proteína/genética , Transporte de Proteínas/genética , Especificidad por Sustrato/genética
11.
Methods Mol Biol ; 819: 405-21, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22183550

RESUMEN

Among the many applications of molecular modeling, drug design is probably the one with the highest demands on the accuracy of the underlying structures. During lead optimization, the position of every atom in the binding site should ideally be known with high precision to identify those chemical modifications that are most likely to increase drug affinity. Unfortunately, X-ray crystallography at common resolution yields an electron density map that is too coarse, since the chemical elements and their protonation states cannot be fully resolved.This chapter describes the steps required to fill in the missing knowledge, by devising an algorithm that can detect and resolve the ambiguities. First, the pK (a) values of acidic and basic groups are predicted. Second, their potential protonation states are determined, including all permutations (considering for example protons that can jump between the oxygens of a phosphate group). Third, those groups of atoms are identified that can adopt alternative but indistinguishable conformations with essentially the same electron density. Fourth, potential hydrogen bond donors and acceptors are located. Finally, all these data are combined in a single "configuration energy function," whose global minimum is found with the SCWRL algorithm, which employs dead-end elimination and graph theory. As a result, one obtains a complete model of the protein and its bound ligand, with ambiguous groups rotated to the best orientation and with protonation states assigned considering the current pH and the H-bonding network. An implementation of the algorithm has been available since 2008 as part of the YASARA modeling & simulation program.


Asunto(s)
Biología Computacional/métodos , Proteínas/química , Protones , Algoritmos , Enlace de Hidrógeno , Ligandos , Modelos Moleculares , Termodinámica
12.
Nucleic Acids Res ; 39(Database issue): D411-9, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21071423

RESUMEN

The Protein Data Bank (PDB) is the world-wide repository of macromolecular structure information. We present a series of databases that run parallel to the PDB. Each database holds one entry, if possible, for each PDB entry. DSSP holds the secondary structure of the proteins. PDBREPORT holds reports on the structure quality and lists errors. HSSP holds a multiple sequence alignment for all proteins. The PDBFINDER holds easy to parse summaries of the PDB file content, augmented with essentials from the other systems. PDB_REDO holds re-refined, and often improved, copies of all structures solved by X-ray. WHY_NOT summarizes why certain files could not be produced. All these systems are updated weekly. The data sets can be used for the analysis of properties of protein structures in areas ranging from structural genomics, to cancer biology and protein design.


Asunto(s)
Bases de Datos de Proteínas , Proteínas/química , Conformación Proteica , Estructura Secundaria de Proteína , Alineación de Secuencia , Análisis de Secuencia de Proteína , Integración de Sistemas , Interfaz Usuario-Computador
13.
Hepatology ; 51(1): 286-96, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19918981

RESUMEN

UNLABELLED: Deficiency in P-type ATP8B1 is a severe and clinically highly variable hereditary disorder that is primarily characterized by intrahepatic cholestasis. It presents either as a progressive (progressive familial intrahepatic cholestasis type 1 [PFIC1]) or intermittent (benign recurrent intrahepatic cholestasis type 1 [BRIC1]) disease. ATP8B1 deficiency is caused by autosomal recessive mutations in the gene encoding ATP8B1, a putative aminophospholipid-translocating P-type adenosine triphosphatase. The exact pathogenesis of the disease is elusive, and no effective pharmacological therapy is currently available. Here, the molecular consequences of six distinct ATP8B1 missense mutations (p.L127P, p.G308V, p.D454G, p.D554N, p.I661T, and p.G1040R) and one nonsense mutation (p.R1164X) associated with PFIC1 and/or BRIC1 were systematically characterized. Except for the p.L127P mutation, all mutations resulted in markedly reduced ATP8B1 protein expression, whereas messenger RNA expression was unaffected. Five of seven mutations resulted in (partial) retention of ATP8B1 in the endoplasmic reticulum. Reduced protein expression was partially restored by culturing the cells at 30 degrees C and by treatment with proteasomal inhibitors, indicating protein misfolding and subsequent proteosomal degradation. Protein misfolding was corroborated by predicting the consequences of most mutations onto a homology model of ATP8B1. Treatment with 4-phenylbutyrate, a clinically approved pharmacological chaperone, partially restored defects in expression and localization of ATP8B1 substitutions G308V, D454G, D554N, and in particular I661T, which is the most frequently identified mutation in BRIC1. CONCLUSION: A surprisingly large proportion of ATP8B1 mutations resulted in aberrant folding and decreased expression at the plasma membrane. These effects were partially restored by treatment with 4-phenylbutyrate. We propose that treatment with pharmacological chaperones may represent an effective therapeutic strategy to ameliorate the recurrent attacks of cholestasis in patients with intermittent (BRIC1) disease.


Asunto(s)
Adenosina Trifosfatasas/genética , Colestasis/genética , Fenilbutiratos/farmacología , Pliegue de Proteína/efectos de los fármacos , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/efectos de los fármacos , Células Cultivadas , Proteínas de la Membrana/metabolismo , Mutación Missense , Complejo de la Endopetidasa Proteasomal/metabolismo
14.
Eur Biophys J ; 39(4): 551-63, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19718498

RESUMEN

Homology modelling is normally the technique of choice when experimental structure data are not available but three-dimensional coordinates are needed, for example, to aid with detailed interpretation of results of spectroscopic studies. Herein, the state of the art of homology modelling will be described in the light of a series of recent developments, and an overview will be given of the problems and opportunities encountered in this field. The major topic, the accuracy and precision of homology models, will be discussed extensively due to its influence on the reliability of conclusions drawn from the combination of homology models and spectroscopic data. Three real-world examples will illustrate how both homology modelling and spectroscopy can be beneficial for (bio)medical research.


Asunto(s)
Modelos Moleculares , Homología de Secuencia , Análisis Espectral/métodos , Secuencia de Aminoácidos , Humanos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Proteínas/química , Proteínas/metabolismo , Marcadores de Spin
15.
Hepatology ; 50(6): 1783-95, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19937698

RESUMEN

UNLABELLED: Wilson disease (WD) is an autosomal recessive copper overload disorder of the liver and basal ganglia. WD is caused by mutations in the gene encoding ATP7B, a protein localized to the trans-Golgi network that primarily facilitates hepatic copper excretion. Current treatment comprises reduction of circulating copper by zinc supplementation or copper chelation. Despite treatment, a significant number of patients have neurological deterioration. The aim of this study was to investigate the possibility that defects arising from some WD mutations are ameliorated by drug treatment aimed at improvement of protein folding and restoration of protein function. This necessitated systematic characterization of the molecular consequences of distinct ATP7B missense mutations associated with WD. With the exception of p.S1363F, all mutations tested (p.G85V, p.R778L, p.H1069Q, p.C1104F, p.V1262F, p.G1343V, and p.S1363F) resulted in reduced ATP7B protein expression, whereas messenger RNA abundance was unaffected. Retention of mutant ATP7B in the endoplasmic reticulum, increased protein expression, and normalization of localization after culturing cells at 30 degrees C, and homology modeling suggested that these proteins were misfolded. Four distinct mutations exhibited residual copper export capacity, whereas other mutations resulted in complete disruption of copper export by ATP7B. Treatment with pharmacological chaperones 4-phenylbutyrate (4-PBA) and curcumin, a clinically approved compound, partially restored protein expression of most ATP7B mutants. CONCLUSION: These findings might enable novel treatment strategies in WD by directly enhancing the protein expression of mutant ATP7B with residual copper export activity. 1795.).


Asunto(s)
Adenosina Trifosfatasas/genética , Proteínas de Transporte de Catión/genética , Curcumina/farmacología , Degeneración Hepatolenticular/tratamiento farmacológico , Mutación , Fenilbutiratos/farmacología , Pliegue de Proteína/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales , Adenosina Trifosfatasas/química , Proteínas Portadoras/química , Proteínas de Transporte de Catión/química , Línea Celular Tumoral , Cobre/metabolismo , Proteínas Transportadoras de Cobre , ATPasas Transportadoras de Cobre , Degeneración Hepatolenticular/genética , Humanos , Metalochaperonas , Chaperonas Moleculares/química , Conformación Proteica
16.
Proteins ; 77 Suppl 9: 114-22, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19768677

RESUMEN

A correct alignment is an essential requirement in homology modeling. Yet in order to bridge the structural gap between template and target, which may not only involve loop rearrangements, but also shifts of secondary structure elements and repacking of core residues, high-resolution refinement methods with full atomic details are needed. Here, we describe four approaches that address this "last mile of the protein folding problem" and have performed well during CASP8, yielding physically realistic models: YASARA, which runs molecular dynamics simulations of models in explicit solvent, using a new partly knowledge-based all atom force field derived from Amber, whose parameters have been optimized to minimize the damage done to protein crystal structures. The LEE-SERVER, which makes extensive use of conformational space annealing to create alignments, to help Modeller build physically realistic models while satisfying input restraints from templates and CHARMM stereochemistry, and to remodel the side-chains. ROSETTA, whose high resolution refinement protocol combines a physically realistic all atom force field with Monte Carlo minimization to allow the large conformational space to be sampled quickly. And finally UNDERTAKER, which creates a pool of candidate models from various templates and then optimizes them with an adaptive genetic algorithm, using a primarily empirical cost function that does not include bond angle, bond length, or other physics-like terms.


Asunto(s)
Biología Computacional/métodos , Modelos Moleculares , Proteínas/química , Alineación de Secuencia/métodos , Algoritmos , Conformación Proteica , Programas Informáticos
17.
Mol Pharmacol ; 74(4): 964-71, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18612080

RESUMEN

Multidrug resistance protein (MRP) 4 transports a variety of endogenous and xenobiotic organic anions. MRP4 is widely expressed in the body and specifically localized to the renal apical proximal tubule cell membrane, where it mediates the excretion of these compounds into urine. To characterize the MRP4 substrate-binding site, the amino acids Phe368, Phe369, Glu374, Arg375, and Glu378 of transmembrane helix 6, and Arg998 of helix 12, localized in the intracellular half of the central pore, were mutated into the corresponding amino acids of MRP1 and MRP2. Membrane vesicles isolated from human embryonic kidney 293 cells overexpressing these mutants showed significantly reduced methotrexate (MTX) and cGMP transport activity compared with vesicles that expressed wild-type MRP4. The only exception was substitution of Arg375 with serine, which had no effect on cGMP transport but significantly decreased the affinity of MTX. Substitution of the same amino acid with a positively charged lysine returned the MTX affinity to that of the wild type. Furthermore, MTX inhibition of MRP4-mediated cGMP transport was noncompetitive, and the inhibition constant was increased by introduction of the R375S mutation. A homology model of MRP4 showed that Arg375 and Arg998 face right into the central aqueous pore of MRP4. We conclude that positively charged amino acids in transmembrane helices 6 and 12 contribute to the MRP4 substrate-binding pocket.


Asunto(s)
Arginina/metabolismo , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/química , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Alanina/metabolismo , Sustitución de Aminoácidos , Baculoviridae/genética , Sitios de Unión , Transporte Biológico/efectos de los fármacos , Línea Celular , Membrana Celular/química , Estructuras Celulares/metabolismo , ADN Complementario/genética , Relación Dosis-Respuesta a Droga , Técnica del Anticuerpo Fluorescente Indirecta , Guanosina Monofosfato/metabolismo , Humanos , Riñón/citología , Cinética , Metotrexato/farmacología , Modelos Moleculares , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Unión Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
18.
Am J Med Genet A ; 143A(20): 2382-9, 2007 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17853461

RESUMEN

Myosin XVA is an unconventional myosin which has been implicated in autosomal recessive nonsyndromic hearing impairment (ARNSHI) in humans. In Myo15A mouse models, vestibular dysfunction accompanies the autosomal recessive hearing loss. Genomewide homozygosity mapping and subsequent fine mapping in two Turkish families with ARNSHI revealed significant linkage to a critical interval harboring a known deafness gene MYO15A on chromosome 17p13.1-17q11.2. Subsequent sequencing of the MYO15A gene led to the identification of a novel missense mutation, c.5492G-->T (p.Gly1831Val) and a novel splice site mutation, c.8968-1G-->C. These mutations were not detected in additional 64 unrelated ARNSHI index patients and in 230 Turkish control chromosomes. Gly1831 is a conserved residue located in the motor domains of the different classes of myosins of different species. Molecular modeling of the motor head domain of the human myosin XVa protein suggests that the Gly1831Val mutation inhibits the powerstroke by reducing backbone flexibility and weakening the hydrophobic interactions necessary for signal transmission to the converter domain.


Asunto(s)
Pérdida Auditiva/genética , Mutación , Miosinas/química , Miosinas/genética , Secuencia de Bases , Análisis Mutacional de ADN , Haplotipos , Homocigoto , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Datos de Secuencia Molecular , Linaje , Estructura Terciaria de Proteína , Turquía , Valina/genética , Valina/metabolismo
19.
Hum Mol Genet ; 16(16): 1993-2003, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17584769

RESUMEN

The highly ordered distribution of neurons is an essential feature of a functional mammalian retina. Disruptions in the apico-basal polarity complexes at the outer limiting membrane (OLM) of the retina are associated with retinal patterning defects in vertebrates. We have analyzed the binding repertoire of MPP5/Pals1, a key member of the apico-basal Crumbs polarity complex, that has functionally conserved counterparts in zebrafish (nagie oko) and Drosophila (Stardust). We show that MPP5 interacts with its MAGUK family member MPP1/p55 at the OLM. Mechanistically, this interaction involves heterodimerization of both MAGUK modules in a directional fashion. MPP1 expression in the retina throughout development resembles the expression of whirlin, a multi-PDZ scaffold protein and an important organizer in the Usher protein network. We demonstrate that both proteins interact strongly by both a classical PDZ domain-to-PDZ binding motif (PBM) mechanism, and a mechanism involving internal epitopes. MPP1 and whirlin colocalize in the retina at the OLM, at the outer synaptic layer and at the basal bodies and the ciliary axoneme. In view of the known roles of the Crumbs and Usher protein networks, our findings suggest a novel link of the core developmental processes of actin polymerization and establishment/maintenance of apico-basal cell polarity through MPP1. These processes, essential in neural development and patterning of the retina, may be disrupted in eye disorders that are associated with defects in these protein networks.


Asunto(s)
Proteínas Sanguíneas/metabolismo , Proteínas del Ojo/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Nucleósido-Fosfato Quinasa/metabolismo , Retina/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Proteínas Sanguíneas/genética , Membrana Celular/metabolismo , Embrión de Mamíferos/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Proteínas del Ojo/genética , Humanos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Modelos Biológicos , Modelos Genéticos , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/genética , Nucleósido-Fosfato Quinasa/química , Nucleósido-Fosfato Quinasa/genética , Estructura Terciaria de Proteína , Ratas , Ratas Wistar , Técnicas del Sistema de Dos Híbridos
20.
Biochem J ; 398(3): 399-409, 2006 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16737443

RESUMEN

The retromer complex is involved in the retrograde transport of the CI-M6PR (cation-independent mannose 6-phosphate receptor) from endosomes to the Golgi. It is a hetero-trimeric complex composed of Vps26 (vacuolar sorting protein 26), Vps29 and Vps35 proteins, which are conserved in eukaryote evolution. Recently, elucidation of the crystal structure of Vps29 revealed that Vps29 contains a metallo-phosphoesterase fold [Wang, Guo, Liang, Fan, Zhu, Zang, Zhu, Li, Teng, Niu et al. (2005) J. Biol. Chem. 280, 22962-22967; Collins, Skinner, Watson, Seaman and Owen (2005) Nat. Struct. Mol. Biol. 12, 594-602]. We demonstrate that recombinant hVps29 (human Vps29) displays in vitro phosphatase activity towards a serine-phosphorylated peptide, containing the acidic-cluster dileucine motif of the cytoplasmatic tail of the CI-M6PR. Efficient dephosphorylation required the additional presence of recombinant hVps26 and hVps35 proteins, which interact with hVps29. Phosphatase activity of hVps29 was greatly decreased by alanine substitutions of active-site residues that are predicted to co-ordinate metal ions. Using inductively coupled plasma MS, we demonstrate that recombinant hVps29 binds zinc. Moreover, hVps29-dependent phosphatase activity is greatly reduced by non-specific and zinc-specific metal ion chelators, which can be completely restored by addition of excess ZnCl2. The binuclear Zn2+ centre and phosphate group were modelled into the hVps29 catalytic site and pKa calculations provided further insight into the molecular mechanisms of Vps29 phosphatase activity. We conclude that the retromer complex displays Vps29-dependent in vitro phosphatase activity towards a serinephosphorylated acidic-cluster dileucine motif that is involved in endosomal trafficking of the CI-M6PR. The potential significance of these findings with respect to regulation of transport of cycling trans-Golgi network proteins is discussed.


Asunto(s)
Proteínas Portadoras/metabolismo , Receptor IGF Tipo 2/metabolismo , Secuencias de Aminoácidos , Animales , Sitios de Unión , Cationes , Quelantes , Escherichia coli/metabolismo , Humanos , Ratones , Modelos Moleculares , Células 3T3 NIH , Conformación Proteica , Pliegue de Proteína , Especificidad por Sustrato , Proteínas de Transporte Vesicular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA