Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Proc Natl Acad Sci U S A ; 116(33): 16378-16383, 2019 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-31346089

RESUMO

Triosephosphate isomerase (TIM) barrel proteins have not only a conserved architecture that supports a myriad of enzymatic functions, but also a conserved folding mechanism that involves on- and off-pathway intermediates. Although experiments have proven to be invaluable in defining the folding free-energy surface, they provide only a limited understanding of the structures of the partially folded states that appear during folding. Coarse-grained simulations employing native centric models are capable of sampling the entire energy landscape of TIM barrels and offer the possibility of a molecular-level understanding of the readout from sequence to structure. We have combined sequence-sensitive native centric simulations with small-angle X-ray scattering and time-resolved Förster resonance energy transfer to monitor the formation of structure in an intermediate in the Sulfolobus solfataricus indole-3-glycerol phosphate synthase TIM barrel that appears within 50 µs and must at least partially unfold to achieve productive folding. Simulations reveal the presence of a major and 2 minor folding channels not detected in experiments. Frustration in folding, i.e., backtracking in native contacts, is observed in the major channel at the initial stage of folding, as well as late in folding in a minor channel before the appearance of the native conformation. Similarities in global and pairwise dimensions of the early intermediate, the formation of structure in the central region that spreads progressively toward each terminus, and a similar rate-limiting step in the closing of the ß-barrel underscore the value of combining simulation and experiment to unravel complex folding mechanisms at the molecular level.


Assuntos
Indol-3-Glicerolfosfato Sintase/química , Conformação Proteica , Dobramento de Proteína , Triose-Fosfato Isomerase/química , Sequência de Aminoácidos , Transferência Ressonante de Energia de Fluorescência , Indol-3-Glicerolfosfato Sintase/genética , Modelos Moleculares , Estrutura Secundária de Proteína , Espalhamento a Baixo Ângulo , Sulfolobus solfataricus/enzimologia , Termodinâmica , Triose-Fosfato Isomerase/genética
2.
Proc Natl Acad Sci U S A ; 112(41): E5608-17, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26420867

RESUMO

The MHC class I peptide loading complex (PLC) facilitates the assembly of MHC class I molecules with peptides, but factors that regulate the stability and dynamics of the assembly complex are largely uncharacterized. Based on initial findings that ATP, in addition to MHC class I-specific peptide, is able to induce MHC class I dissociation from the PLC, we investigated the interaction of ATP with the chaperone calreticulin, an endoplasmic reticulum (ER) luminal, calcium-binding component of the PLC that is known to bind ATP. We combined computational and experimental measurements to identify residues within the globular domain of calreticulin, in proximity to the high-affinity calcium-binding site, that are important for high-affinity ATP binding and for ATPase activity. High-affinity calcium binding by calreticulin is required for optimal nucleotide binding, but both ATP and ADP destabilize enthalpy-driven high-affinity calcium binding to calreticulin. ATP also selectively destabilizes the interaction of calreticulin with cellular substrates, including MHC class I molecules. Calreticulin mutants that affect ATP or high-affinity calcium binding display prolonged associations with monoglucosylated forms of cellular MHC class I, delaying MHC class I dissociation from the PLC and their transit through the secretory pathway. These studies reveal central roles for ATP and calcium binding as regulators of calreticulin-substrate interactions and as key determinants of PLC dynamics.


Assuntos
Trifosfato de Adenosina/imunologia , Calbindina 2/imunologia , Retículo Endoplasmático/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Trifosfato de Adenosina/genética , Animais , Calbindina 2/genética , Cálcio/imunologia , Retículo Endoplasmático/genética , Antígenos de Histocompatibilidade Classe I/genética , Camundongos , Camundongos Knockout , Ligação Proteica/genética , Ligação Proteica/imunologia , Estrutura Terciária de Proteína
3.
Proc Natl Acad Sci U S A ; 111(29): 10562-7, 2014 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-25002512

RESUMO

Folding of globular proteins can be envisioned as the contraction of a random coil unfolded state toward the native state on an energy surface rough with local minima trapping frustrated species. These substructures impede productive folding and can serve as nucleation sites for aggregation reactions. However, little is known about the relationship between frustration and its underlying sequence determinants. Chemotaxis response regulator Y (CheY), a 129-amino acid bacterial protein, has been shown previously to populate an off-pathway kinetic trap in the microsecond time range. The frustration has been ascribed to premature docking of the N- and C-terminal subdomains or, alternatively, to the formation of an unproductive local-in-sequence cluster of branched aliphatic side chains, isoleucine, leucine, and valine (ILV). The roles of the subdomains and ILV clusters in frustration were tested by altering the sequence connectivity using circular permutations. Surprisingly, the stability and buried surface area of the intermediate could be increased or decreased depending on the location of the termini. Comparison with the results of small-angle X-ray-scattering experiments and simulations points to the accelerated formation of a more compact, on-pathway species for the more stable intermediate. The effect of chain connectivity in modulating the structures and stabilities of the early kinetic traps in CheY is better understood in terms of the ILV cluster model. However, the subdomain model captures the requirement for an intact N-terminal domain to access the native conformation. Chain entropy and aliphatic-rich sequences play crucial roles in biasing the early events leading to frustration in the folding of CheY.


Assuntos
Dobramento de Proteína , Análise de Sequência de Proteína , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Simulação por Computador , Cinética , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Proteínas Quimiotáticas Aceptoras de Metil , Modelos Moleculares , Estabilidade Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Espalhamento a Baixo Ângulo , Termodinâmica , Difração de Raios X
4.
J Am Chem Soc ; 136(8): 3097-107, 2014 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-24495192

RESUMO

Many viruses undergo large-scale conformational changes during their life cycles. Blocking the transition from one stage of the life cycle to the next is an attractive strategy for the development of antiviral compounds. In this work, we have constructed an icosahedrally symmetric, low-energy pathway for the maturation transition of bacteriophage HK97. By conducting constant-pH molecular dynamics simulations on this pathway, we identify which residues are contributing most significantly to shifting the stability between the states along the pathway under differing pH conditions. We further analyze these data to establish the connection between critical residues and important structural motifs which undergo reorganization during maturation. We go on to show how DNA packaging can induce spontaneous reorganization of the capsid during maturation.


Assuntos
Capsídeo/química , Siphoviridae/crescimento & desenvolvimento , Capsídeo/ultraestrutura , Cristalografia por Raios X , Concentração de Íons de Hidrogênio , Simulação de Dinâmica Molecular , Siphoviridae/ultraestrutura , Termodinâmica
5.
Biophys J ; 105(11): 2559-68, 2013 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-24314086

RESUMO

The modified base 8-oxo-7,8-dihydro-2'-deoxyguanosine (oxoG) is a common DNA adduct produced by the oxidation of DNA by reactive oxygen species. Kinetic data reveal that DNA polymerase X (pol X) from the African swine fever virus incorporates adenine (dATP) opposite to oxoG with higher efficiency than the non-damaged G:C basepair. To help interpret the kinetic data, we perform molecular dynamics simulations of pol X/DNA complexes, in which the template base opposite to the incoming dNTP (dCTP, dATP, dGTP) is oxoG. Our results suggest that pol X accommodates the oxoGsyn:A mispair by sampling closed active conformations that mirror those observed in traditional Watson-Crick complexes. Moreover, for both the oxoGsyn:A and oxoG:C ternary complexes, conformational sampling of the polymerase follows previously described large subdomain movements, local residue motions, and active site reorganization. Interestingly, the oxoGsyn:A system exhibits superior active site geometry in comparison to the oxoG:C system. Simulations for the other mismatch basepair complexes reveal large protein subdomain movement for all systems, except for oxoG:G, which samples conformations close to the open state. In addition, active site geometry and basepairing of the template base with the incoming nucleotide, reveal distortions and misalignments that range from moderate (i.e., oxoG:Asyn) to extreme (i.e., oxoGanti/syn:G). These results agree with the available kinetic data for pol X and provide structural insights regarding the mechanism by which this polymerase can accommodate incoming nucleotides opposite oxoG. Our simulations also support the notion that α-helix E is involved both in DNA binding and active site stabilization. Our proposed mechanism by which pol X can preferentially accommodate dATP opposite template oxoG further underscores the role that enzyme dynamics and conformational sampling operate in polymerase fidelity and function.


Assuntos
DNA Polimerase Dirigida por DNA/química , DNA/química , Nucleotídeos de Desoxiadenina/química , Guanina/análogos & derivados , Simulação de Dinâmica Molecular , Sequência de Aminoácidos , Sequência de Bases , DNA/metabolismo , DNA Polimerase Dirigida por DNA/metabolismo , Nucleotídeos de Desoxiadenina/metabolismo , Guanina/química , Guanina/metabolismo , Simulação de Acoplamento Molecular , Dados de Sequência Molecular , Ligação Proteica , Especificidade por Substrato
6.
J Am Chem Soc ; 135(30): 10906-9, 2013 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-23841875

RESUMO

Activation of class-A G-protein-coupled receptors (GPCRs) involves large-scale reorganization of the H3/H6 interhelical network. In rhodopsin (Rh), this process is coupled to a change in the protonation state of a key residue, E134, whose exact role in activation is not well understood. Capturing this millisecond pH-dependent process is a well-appreciated challenge. We have developed a scheme combining the harmonic Fourier beads (HFB) method and constant-pH molecular dynamics with pH-based replica exchange (pH-REX) to gain insight into the structural changes that occur along the activation pathway as a function of the protonation state of E134. Our results indicate that E134 is protonated as a consequence of tilting of H6 by ca. 4.0° with respect to its initial position and simultaneous rotation by ca. 23° along its principal axis. The movement of H6 is associated with breakage of the E247-R135 and R135-E134 salt bridges and concomitant release of the E134 side chain, which results in an increase in its pKa value above physiological pH. An increase in the hydrophobicity of the environment surrounding E134 leads to further tilting and rotation of H6 and upshift of the E134 pKa. Such atomic-level information, which is not accessible through experiments, refines the earlier proposed sequential model of Rh activation (see: Zaitseva, E.; et al. Sequential Rearrangement of Interhelical Networks Upon Rhodopsin Activation in Membranes: The Meta IIa Conformational Substate . J. Am. Chem. Soc. 2010, 132, 4815) and argues that the E134 protonation switch is both a cause and a consequence of the H6 motion.


Assuntos
Simulação de Dinâmica Molecular , Rodopsina/metabolismo , Conformação Proteica , Rodopsina/química , Fatores de Tempo
7.
Top Curr Chem ; 337: 165-87, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23420416

RESUMO

It has become increasingly clear that protein motions play an essential role in enzyme catalysis. However, exactly how these motions are related to an enzyme's chemical step is still intensely debated. This chapter examines the possible role of protein motions that display a hierarchy of timescales in enzyme catalysis. The linkage between protein motions and catalysis is investigated in the context of a model enzyme, E. coli dihydrofolate reductase (DHFR), that catalyzes the hydride transfer reaction in the conversion of dihydrofolate to tetrahydrofolate. The results of extensive computer simulations probing the protein motions that are manifest during different steps along the turnover cycle of DHFR are summarized. Evidence is presented that the protein motions modulate the catalytic efficacy of DHFR by generating a conformational ensemble conducive to the hydride transfer. The alteration of the equilibrium conformational ensemble rather than any protein dynamical effects is found to be sufficient to explain the rate-diminishing effects of mutation on the kinetics of the enzyme. These data support the view that the protein motions facilitate catalysis by establishing reaction competent conformations of the enzyme, but they do not directly couple to the chemical reaction itself. These findings have broad implications for our understanding of enzyme mechanisms and the design of novel protein catalysts.


Assuntos
Hidrogênio/química , Tetra-Hidrofolato Desidrogenase/química , Catálise , Mutação , Conformação Proteica , Relação Estrutura-Atividade , Tetra-Hidrofolato Desidrogenase/genética
8.
Theor Chem Acc ; 131: 1287, 2012 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23459563

RESUMO

In recent papers, there has been a lively exchange concerning theories for enzyme catalysis, especially the role of protein dynamics/pre-chemistry conformational changes in the catalytic cycle of enzymes. Of particular interest is the notion that substrate-induced conformational changes that assemble the polymerase active site prior to chemistry are required for DNA synthesis and impact fidelity (i.e., substrate specificity). High-resolution crystal structures of DNA polymerase ß representing intermediates of substrate complexes prior to the chemical step are available. These structures indicate that conformational adjustments in both the protein and substrates must occur to achieve the requisite geometry of the reactive participants for catalysis. We discuss computational and kinetic methods to examine possible conformational change pathways that lead from the observed crystal structure intermediates to the final structures poised for chemistry. The results, as well as kinetic data from site-directed mutagenesis studies, are consistent with models requiring pre-chemistry conformational adjustments in order to achieve high fidelity DNA synthesis. Thus, substrate-induced conformational changes that assemble the polymerase active site prior to chemistry contribute to DNA synthesis even when they do not represent actual rate-determining steps for chemistry.

9.
Biophys J ; 98(8): 1449-57, 2010 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-20409463

RESUMO

Hexameric helicases are molecular motor proteins that utilize energy obtained from ATP hydrolysis to translocate along and/or unwind nucleic acids. In this study, we investigate the dynamic behavior of the Simian Virus 40 hexameric helicase bound to DNA by performing molecular dynamics simulations employing a coarse-grained model. Our results elucidate the two most important molecular features of the helicase motion. First, the attractive interactions between the DNA-binding domain of the helicase and the DNA backbone are essential for the helicase to exhibit a unidirectional motion along the DNA strand. Second, the sequence of ATP binding at multiple binding pockets affects the helicase motion. Specifically, concerted ATP binding does not generate a unidirectional motion of the helicase. It is only when the binding of ATP occurs sequentially from one pocket to the next that the helicase moves unidirectionally along the DNA. Interestingly, in the reverse order of sequential ATP binding, the helicase also moves unidirectionally but in the opposite direction. These observations suggest that in nature ATP molecules must distinguish between different available ATP binding pockets of the hexameric helicase in order to function efficiently. To this end, simulations reveal that the binding of ATP in one pocket induces an opening of the next ATP-binding pocket and such an asymmetric deformation may coordinate the sequential ATP binding in a unidirectional manner. Overall, these findings may provide clues toward understanding the mechanism of substrate translocation in other motor proteins.


Assuntos
DNA Helicases/química , DNA Helicases/metabolismo , Vírus 40 dos Símios/enzimologia , Trifosfato de Adenosina/metabolismo , Sítios de Ligação , DNA/metabolismo , Modelos Moleculares , Simulação de Dinâmica Molecular , Conformação Proteica , Transporte Proteico , Especificidade por Substrato
10.
Proc Natl Acad Sci U S A ; 104(47): 18496-501, 2007 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-18000050

RESUMO

Large-scale conformational changes in proteins are often associated with the binding of a substrate. Because conformational changes may be related to the function of an enzyme, understanding the kinetics and energetics of these motions is very important. We have delineated the atomically detailed conformational transition pathway of the phosphotransferase enzyme adenylate kinase (AdK) in the absence and presence of an inhibitor. The computed free energy profiles associated with conformational transitions offer detailed mechanistic insights into, as well as kinetic information on, the ligand binding mechanism. Specifically, potential of mean force calculations reveal that in the ligand-free state, there is no significant barrier separating the open and closed conformations of AdK. The enzyme samples near closed conformations, even in the absence of its substrate. The ligand binding event occurs late, toward the closed state, and transforms the free energy landscape. In the ligand-bound state, the closed conformation is energetically most favored with a large barrier to opening. These results emphasize the underlying dynamic nature of the enzyme and indicate that the conformational transitions in AdK are more intricate than a mere two-state jump between the crystal-bound and -unbound states. Based on the existence of the multiple conformations of the enzyme in the open and closed states, a different viewpoint of ligand binding is presented. Our estimated activation energy barrier for the conformational transition is also in reasonable accord with the experimental findings.


Assuntos
Adenilato Quinase/química , Adenilato Quinase/metabolismo , Adenilato Quinase/antagonistas & inibidores , Regulação Alostérica , Simulação por Computador , Cristalografia por Raios X , Ativação Enzimática , Ligantes , Modelos Moleculares , Ligação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/metabolismo , Estrutura Terciária de Proteína
11.
Biophys J ; 96(10): 3887-96, 2009 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-19450462

RESUMO

The macroscopic ion-selective behavior of K(+) channels is mediated by a multitude of physiological factors. However, considering the carbonyl-lined binding site of a conductive K(+) channel as a canonical eightfold coordinated construct can be useful in understanding the principles that correlate the channel's structure with its function. We probe the effects of structure and chemical composition on the K(+)/Na(+) selectivity provided by a variety of simplified droplet-like ion binding site models. We find that when carbonyl- and water-based models capture the qualitative structural features of the K(+) channel binding site, a selective preference for K(+) emerges. Thus our findings suggest that the preference for K(+) over Na(+) exhibited by such models is principally built-in, and is not due to a unique K(+)-selective property of carbonyl functional groups. This suggestion is confirmed by a general thermodynamic assessment, which provides a basis for using simplified models to study the design principles underlying the molecular evolution of K(+) channels.


Assuntos
Modelos Moleculares , Canais de Potássio/química , Canais de Potássio/metabolismo , Potássio/metabolismo , Sódio/metabolismo , Sítios de Ligação , Ligantes , Conformação Proteica , Solventes/metabolismo , Especificidade por Substrato , Água/metabolismo
12.
J Am Chem Soc ; 131(15): 5642-7, 2009 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-19323547

RESUMO

Conformational changes in enzymes are well recognized to play an important role in the organization of the reactive groups for efficient catalysis. This study reveals atomic and energetic details of the conformational change process that precedes the catalytic reaction of the enzyme dihydrofolate reductase. The computed free energy profile provides insights into the ligand binding mechanism and a quantitative estimate of barrier heights separating different conformational states along the pathway. Studies show that the ternary complex comprised of NADPH cofactor and substrate dihydrofolate undergoes transitions between a closed state and an occluded state via an intermediate "open" conformation. During these transitions the largest conformational change occurs in the Met20 loop of DHFR and is accompanied by the motion of the cofactor into and out of the binding pocket. When the cofactor is out of the binding pocket, the enzyme frequently samples open and occluded conformations with a small (approximately 5 k(B)T) free energy barrier between the two states. However, when the cofactor is in the binding pocket, the closed conformation is thermodynamically most favored. The determination of a profile characterizing the position-dependent diffusion of the Met20 loop allowed us to apply reaction rate theory and deduce the kinetics of loop motions based on the computed free energy landscape.


Assuntos
Tetra-Hidrofolato Desidrogenase/química , Sítios de Ligação , Ácido Fólico/análogos & derivados , Ácido Fólico/metabolismo , Cinética , Ligantes , NADP/metabolismo , Ligação Proteica , Conformação Proteica , Tetra-Hidrofolato Desidrogenase/metabolismo , Termodinâmica
13.
Protein Sci ; 15(1): 135-51, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16322565

RESUMO

The possible conformational changes of DNA polymerase IV (Dpo4) before and after the nucleotidyl-transfer reaction are investigated at the atomic level by dynamics simulations to gain insight into the mechanism of low-fidelity polymerases and identify slow and possibly critical steps. The absence of significant conformational changes in Dpo4 before chemistry when the incoming nucleotide is removed supports the notion that the "induced-fit" mechanism employed to interpret fidelity in some replicative and repair DNA polymerases does not exist in Dpo4. However, significant correlated movements in the little finger and finger domains, as well as DNA sliding and subtle catalytic-residue rearrangements, occur after the chemical reaction when both active-site metal ions are released. Subsequently, Dpo4's little finger grips the DNA through two arginine residues and pushes it forward. These metal ion correlated movements may define subtle, and possibly characteristic, conformational adjustments that operate in some Y-family polymerase members in lieu of the prominent subdomain motions required for catalytic cycling in other DNA polymerases like polymerase beta. Such subtle changes do not easily provide a tight fit for correct incoming substrates as in higher-fidelity polymerases, but introduce in low-fidelity polymerases different fidelity checks as well as the variable conformational-mobility potential required to bypass different lesions.


Assuntos
Dano ao DNA , DNA Polimerase beta/química , Replicação do DNA , DNA Arqueal/biossíntese , Sulfolobus solfataricus/enzimologia , Termodinâmica , Arginina/química , Arginina/metabolismo , Simulação por Computador , Cristalografia por Raios X , DNA Polimerase beta/metabolismo , Reparo do DNA , DNA Arqueal/química , DNA Arqueal/metabolismo , Guanosina/análogos & derivados , Guanosina/química , Modelos Moleculares , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Sulfolobus solfataricus/genética
14.
Biochemistry ; 45(51): 15142-56, 2006 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-17176036

RESUMO

With an increasing number of structural, kinetic, and modeling studies of diverse DNA polymerases in various contexts, a complex dynamical view of how atomic motions might define molecular "gates" or checkpoints that contribute to polymerase specificity and efficiency is emerging. Such atomic-level information can offer insights into rate-limiting conformational and chemical steps to help piece together mechanistic views of polymerases in action. With recent advances, modeling and dynamics simulations, subject to the well-appreciated limitations, can access transition states and transient intermediates along a reaction pathway, both conformational and chemical, and such information can help bridge the gap between experimentally determined equilibrium structures and mechanistic enzymology data. Focusing on DNA polymerase beta (pol beta), we present an emerging view of the geometric, energetic, and dynamic selection criteria governing insertion rate and fidelity mechanisms of DNA polymerases, as gleaned from various computational studies and based on the large body of existing kinetic and structural data. The landscape of nucleotide insertion for pol beta includes conformational changes, prechemistry, and chemistry "avenues", each with a unique deterministic or stochastic pathway that includes checkpoints for selective control of nucleotide insertion efficiency. For both correct and incorrect incoming nucleotides, pol beta's conformational rearrangements before chemistry include a cascade of slow and subtle side chain rearrangements, followed by active site adjustments to overcome higher chemical barriers, which include critical ion-polymerase geometries; this latter notion of a prechemistry avenue fits well with recent structural and NMR data. The chemical step involves an associative mechanism with several possibilities for the initial proton transfer and for the interaction among the active site residues and bridging water molecules. The conformational and chemical events and associated barriers define checkpoints that control enzymatic efficiency and fidelity. Understanding the nature of such active site rearrangements can facilitate interpretation of existing data and stimulate new experiments that aim to probe enzyme features that contribute to fidelity discrimination across various polymerases via such geometric, dynamic, and energetic selection criteria.


Assuntos
DNA Polimerase beta/biossíntese , DNA Polimerase beta/química , Reparo do DNA , DNA Polimerase beta/genética , DNA Polimerase beta/metabolismo , Reparo do DNA/genética , Humanos , Modelos Moleculares , Nucleotídeos/química , Nucleotídeos/genética , Nucleotídeos/metabolismo , Especificidade por Substrato/genética
15.
J Phys Chem B ; 109(11): 5358-67, 2005 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-16863202

RESUMO

The closing conformational transition of wild-type polymerase beta bound to DNA template/primer before the chemical step (nucleotidyl transfer reaction) is simulated using the stochastic difference equation (in length version, "SDEL") algorithm that approximates long-time dynamics. The order of the events and the intermediate states during pol beta's closing pathway are identified and compared to a separate study of pol beta using transition path sampling (TPS) (Radhakrishnan, R.; Schlick, T. Proc. Natl. Acad. Sci. USA 2004, 101, 5970-5975). Results highlight the cooperative and subtle conformational changes in the pol beta active site upon binding the correct substrate that may help explain DNA replication and repair fidelity. These changes involve key residues that differentiate the open from the closed conformation (Asp192, Arg258, Phe272), as well as residues contacting the DNA template/primer strand near the active site (Tyr271, Arg283, Thr292, Tyr296) and residues contacting the beta and gamma phosphates of the incoming nucleotide (Ser180, Arg183, Gly189). This study compliments experimental observations by providing detailed atomistic views of the intermediates along the polymerase closing pathway and by suggesting additional key residues that regulate events prior to or during the chemical reaction. We also show general agreement between two sampling methods (the stochastic difference equation and transition path sampling) and identify methodological challenges involved in the former method relevant to large-scale biomolecular applications. Specifically, SDEL is very quick relative to TPS for obtaining an approximate path of medium resolution and providing qualitative information on the sequence of events; however, associated free energies are likely very costly to obtain because this will require both successful further refinement of the path segments close to the bottlenecks and large computational time.


Assuntos
DNA Polimerase beta/metabolismo , DNA/metabolismo , Conformação Molecular , Reparo do DNA , Replicação do DNA , Ligação Proteica
16.
J Phys Chem B ; 114(49): 16263-72, 2010 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-20936808

RESUMO

Dynamic motions of human purine nucleoside phosphorylase (hPNP) in complex with transition-state analogues and reactants were studied using 10 ns explicit solvent molecular dynamics simulations. hPNP is a homotrimer that catalyzes the phosphorolysis of purine 6-oxynucleosides. The ternary complex of hPNP includes the binding of a ligand and phosphate to the active site. Molecular dynamics simulations were performed on the ternary complex of six ligands including the picomolar transition-state analogues, Immucillin-H (K(d) = 56 pM), DADMe-Immucillin-H (K(d) = 8.5 pM), DATMe-Immucillin-H (K(d) = 8.6 pM), SerMe-Immucillin-H (K(d) = 5.2 pM), the substrate inosine, and a complex containing only phosphate. Protein-inhibitor complexes of the late transition-state inhibitors, DADMe-Imm-H and DATMe-Imm-H, are inflexible. Despite the structural similarity of SerMe-Imm-H and DATMe-Imm-H, the protein complex of SerMe-Imm-H is flexible, and the inhibitor is highly mobile within the active sites. All inhibitors exhibit an increased number of nonbonding interactions in the active site relative to the substrate inosine. Water density within the catalytic site is lower for DADMe-ImmH, DATMe-Imm-H, and SerMe-Imm-H than that for the substrate inosine. Tight binding of the picomolar inhibitors results from increased interactions within the active site and a reduction in the number of water molecules organized within the catalytic site relative to the substrate inosine.


Assuntos
Purina-Núcleosídeo Fosforilase/química , Domínio Catalítico , Inibidores Enzimáticos/farmacologia , Humanos , Conformação Molecular , Estrutura Molecular , Purina-Núcleosídeo Fosforilase/antagonistas & inibidores
17.
J Chem Theory Comput ; 5(1): 47-58, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20046947

RESUMO

Low-dimensional stochastic models can summarize dynamical information and make long time predictions associated with observables of complex atomistic systems. Maximum likelihood based techniques for estimating low-dimensional surrogate diffusion models from relatively short time series are presented. It is found that a heterogeneous population of slowly evolving conformational degrees of freedom modulates the dynamics. This underlying heterogeneity results in a collection of estimated low-dimensional diffusion models. Numerical techniques for exploiting this finding to approximate skewed histograms associated with the simulation are presented. In addition, statistical tests are also used to assess the validity of the models and determine physically relevant sampling information, e.g. the maximum sampling frequency at which one can discretely sample from an atomistic time series and have a surrogate diffusion model pass goodness-of-fit tests. The information extracted from such analyses can possibly be used to assist umbrella sampling computations as well as help in approximating effective diffusion coefficients. The techniques are demonstrated on simulations of Adenylate Kinase.

18.
J Mol Biol ; 384(5): 1086-97, 2008 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-18955064

RESUMO

DNA polymerase X (pol X) from the African swine fever virus is a 174-amino-acid repair polymerase that likely participates in a viral base excision repair mechanism, characterized by low fidelity. Surprisingly, pol X's insertion rate of the G*G mispair is comparable to that of the four Watson-Crick base pairs. This behavior is in contrast with another X-family polymerase, DNA polymerase beta (pol beta), which inserts G*G mismatches poorly, and has higher DNA repair fidelity. Using molecular dynamics simulations, we previously provided support for an induced-fit mechanism for pol X in the presence of the correct incoming nucleotide. Here, we perform molecular dynamics simulations of pol X/DNA complexes with different incoming incorrect nucleotides in various orientations [C*C, A*G, and G*G (anti) and A*G and G*G (syn)] and compare the results to available kinetic data and prior modeling. Intriguingly, the simulations reveal that the G*G mispair with the incoming nucleotide in the syn configuration undergoes large-scale conformational changes similar to that observed in the presence of correct base pair (G*C). The base pairing in the G*G mispair is achieved via Hoogsteen hydrogen bonding with an overall geometry that is well poised for catalysis. Simulations for other mismatched base pairs show that an intermediate closed state is achieved for the A*G and G*G mispair with the incoming dGTP in anti conformation, while the protein remains near the open conformation for the C*C and the A*G syn mismatches. In addition, catalytic site geometry and base pairing at the nascent template-incoming nucleotide interaction reveal distortions and misalignments that range from moderate for A*G anti to worst for the C*C complex. These results agree well with kinetic data for pol X and provide a structural/dynamic basis to explain, at atomic level, the fidelity of this polymerase compared with other members of the X family. In particular, the more open and pliant active site of pol X, compared to pol beta, allows pol X to accommodate bulkier mismatches such as guanine opposite guanine, while the more structured and organized pol beta active site imposes higher discrimination, which results in higher fidelity. The possibility of syn conformers resonates with other low-fidelity enzymes such as Dpo4 (from the Y family), which readily accommodate oxidative lesions.


Assuntos
Vírus da Febre Suína Africana/enzimologia , Pareamento Incorreto de Bases , Pareamento de Bases , Simulação por Computador , DNA Viral/química , DNA Polimerase Dirigida por DNA/química , Catálise , Domínio Catalítico , DNA Polimerase beta/química , Cinética , Magnésio/metabolismo , Modelos Moleculares , Estrutura Secundária de Proteína , Homologia de Sequência de Aminoácidos , Moldes Genéticos , Fatores de Tempo
19.
Biophys J ; 91(9): 3182-95, 2006 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-16920835

RESUMO

The nature of conformational transitions in DNA polymerase lambda (pol lambda), a low-fidelity DNA repair enzyme in the X-family that fills short nucleotide gaps, is investigated. Specifically, to determine whether pol lambda has an induced-fit mechanism and open-to-closed transition before chemistry, we analyze a series of molecular dynamics simulations from both the binary and ternary states before chemistry, with and without the incoming nucleotide, with and without the catalytic Mg(2+) ion in the active site, and with alterations in active site residues Ile(492) and Arg(517). Though flips occurred for several side-chain residues (Ile(492), Tyr(505), Phe(506)) in the active site toward the binary (inactive) conformation and partial DNA motion toward the binary position occurred without the incoming nucleotide, large-scale subdomain motions were not observed in any trajectory from the ternary complex regardless of the presence of the catalytic ion. Simulations from the binary state with incoming nucleotide exhibit more thumb subdomain motion, particularly in the loop containing beta-strand 8 in the thumb, but closing occurred only in the Ile(492)Ala mutant trajectory started from the binary state with incoming nucleotide and both ions. Further connections between active site residues and the DNA position are also revealed through our Ile(492)Ala and Arg(517)Ala mutant studies. Our combined studies suggest that while pol lambda does not demonstrate large-scale subdomain movements as DNA polymerase beta (pol beta), significant DNA motion exists, and there are sequential subtle side chain and other motions-associated with Arg(514), Arg(517), Ile(492), Phe(506), Tyr(505), the DNA, and again Arg(514) and Arg(517)-all coupled to active site divalent ions and the DNA motion. Collectively, these motions transform pol lambda to the chemistry-competent state. Significantly, analogs of these residues in pol beta (Lys(280), Arg(283), Arg(258), Phe(272), and Tyr(271), respectively) have demonstrated roles in determining enzyme efficiency and fidelity. As proposed for pol beta, motions of these residues may serve as gate-keepers by controlling the evolution of the reaction pathway before the chemical reaction.


Assuntos
DNA Polimerase beta/química , DNA Polimerase beta/ultraestrutura , Modelos Químicos , Modelos Moleculares , Simulação por Computador , Ativação Enzimática , Movimento (Física) , Transição de Fase , Conformação Proteica , Estrutura Terciária de Proteína
20.
Biophys J ; 90(1): 42-56, 2006 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16214865

RESUMO

The African swine fever virus DNA polymerase X (pol X), a member of the X family of DNA polymerases, is thought to be involved in base excision repair. Kinetics data indicate that pol X catalyzes DNA polymerization with low fidelity, suggesting a role in viral mutagenesis. Though pol X lacks the fingers domain that binds the DNA in other members of the X family, it binds DNA tightly. To help interpret details of this interaction, molecular dynamics simulations of free pol X at different salt concentrations and of pol X bound to gapped DNA, in the presence and in the absence of the incoming nucleotide, are performed. Anchors for the simulations are two NMR structures of pol X without DNA and a model of one NMR structure plus DNA and incoming nucleotide. Our results show that, in its free form, pol X can exist in two stable conformations that interconvert to one another depending on the salt concentration. When gapped double stranded DNA is introduced near the active site, pol X prefers an open conformation, regardless of the salt concentration. Finally, under physiological conditions, in the presence of both gapped DNA and correct incoming nucleotide, and two divalent ions, the thumb subdomain of pol X undergoes a large conformational change, closing upon the DNA. These results predict for pol X a substrate-induced conformational change triggered by the presence of DNA and the correct incoming nucleotide in the active site, as in DNA polymerase beta. The simulations also suggest specific experiments (e.g., for mutants Phe-102Ala, Val-120Gly, and Lys-85Val that may reveal crucial DNA binding and active-site organization roles) to further elucidate the fidelity mechanism of pol X.


Assuntos
Vírus da Febre Suína Africana/enzimologia , DNA Polimerase Dirigida por DNA/química , Vírus da Febre Suína Africana/genética , Sítios de Ligação , Biofísica/métodos , Simulação por Computador , DNA/química , DNA Polimerase beta/metabolismo , Lisina/química , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Conformação Molecular , Mutagênese , Mutação , Conformação de Ácido Nucleico , Ligação Proteica , Estrutura Terciária de Proteína , Sais/farmacologia , Fatores de Tempo , Valina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA