Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-38895306

RESUMO

How can a single protein domain encode a conformational landscape with multiple stably-folded states, and how do those states interconvert? Here, we use real-time and relaxation-dispersion NMR to characterize the conformational landscape of the circadian rhythm protein KaiB from Rhodobacter sphaeroides. Unique among known natural metamorphic proteins, this KaiB variant spontaneously interconverts between two monomeric states: the "Ground" and "Fold-switched" (FS) state. KaiB in its FS state interacts with multiple binding partners, including the central KaiC protein, to regulate circadian rhythms. We find that KaiB itself takes hours to interconvert between the Ground and FS state, underscoring the ability of a single sequence to encode the slow process needed for function. We reveal the rate-limiting step between the Ground and FS state is the cis-trans isomerization of three prolines in the fold-switching region by demonstrating interconversion acceleration by the prolyl isomerase CypA. The interconversion proceeds through a "partially disordered" (PD) state, where the C-terminal half becomes disordered while the N-terminal half remains stably folded. We discovered two additional properties of KaiB's landscape. Firstly, the Ground state experiences cold denaturation: at 4°C, the PD state becomes the majorly populated state. Secondly, the Ground state exchanges with a fourth state, the "Enigma" state, on the millisecond timescale. We combine AlphaFold2-based predictions and NMR chemical shift predictions to predict this "Enigma" state is a beta-strand register shift that eases buried charged residues, and support this structure experimentally. These results provide mechanistic insight in how evolution can design a single sequence that achieves specific timing needed for its function.

2.
Nature ; 625(7996): 832-839, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37956700

RESUMO

AlphaFold2 (ref. 1) has revolutionized structural biology by accurately predicting single structures of proteins. However, a protein's biological function often depends on multiple conformational substates2, and disease-causing point mutations often cause population changes within these substates3,4. We demonstrate that clustering a multiple-sequence alignment by sequence similarity enables AlphaFold2 to sample alternative states of known metamorphic proteins with high confidence. Using this method, named AF-Cluster, we investigated the evolutionary distribution of predicted structures for the metamorphic protein KaiB5 and found that predictions of both conformations were distributed in clusters across the KaiB family. We used nuclear magnetic resonance spectroscopy to confirm an AF-Cluster prediction: a cyanobacteria KaiB variant is stabilized in the opposite state compared with the more widely studied variant. To test AF-Cluster's sensitivity to point mutations, we designed and experimentally verified a set of three mutations predicted to flip KaiB from Rhodobacter sphaeroides from the ground to the fold-switched state. Finally, screening for alternative states in protein families without known fold switching identified a putative alternative state for the oxidoreductase Mpt53 in Mycobacterium tuberculosis. Further development of such bioinformatic methods in tandem with experiments will probably have a considerable impact on predicting protein energy landscapes, essential for illuminating biological function.


Assuntos
Análise por Conglomerados , Aprendizado de Máquina , Conformação Proteica , Dobramento de Proteína , Proteínas , Alinhamento de Sequência , Mutação , Proteínas/química , Proteínas/genética , Proteínas/metabolismo , Rhodobacter sphaeroides , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo
3.
Proc Natl Acad Sci U S A ; 120(34): e2304611120, 2023 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-37590418

RESUMO

Selective orthosteric inhibition of kinases has been challenging due to the conserved active site architecture of kinases and emergence of resistance mutants. Simultaneous inhibition of distant orthosteric and allosteric sites, which we refer to as "double-drugging", has recently been shown to be effective in overcoming drug resistance. However, detailed biophysical characterization of the cooperative nature between orthosteric and allosteric modulators has not been undertaken. Here, we provide a quantitative framework for double-drugging of kinases employing isothermal titration calorimetry, Förster resonance energy transfer, coupled-enzyme assays, and X-ray crystallography. We discern positive and negative cooperativity for Aurora A kinase (AurA) and Abelson kinase (Abl) with different combinations of orthosteric and allosteric modulators. We find that a conformational equilibrium shift is the main principle governing cooperativity. Notably, for both kinases, we find a synergistic decrease of the required orthosteric and allosteric drug dosages when used in combination to inhibit kinase activities to clinically relevant inhibition levels. X-ray crystal structures of the double-drugged kinase complexes reveal the molecular principles underlying the cooperative nature of double-drugging AurA and Abl with orthosteric and allosteric inhibitors. Finally, we observe a fully closed conformation of Abl when bound to a pair of positively cooperative orthosteric and allosteric modulators, shedding light on the puzzling abnormality of previously solved closed Abl structures. Collectively, our data provide mechanistic and structural insights into rational design and evaluation of double-drugging strategies.


Assuntos
Aurora Quinase A , Mesilato de Imatinib , Niacinamida , Inibidores de Proteínas Quinases , Proteínas Proto-Oncogênicas c-abl , Humanos , Cristalografia por Raios X , Mesilato de Imatinib/química , Mesilato de Imatinib/farmacologia , Niacinamida/química , Niacinamida/farmacologia , Proteínas Proto-Oncogênicas c-abl/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-abl/química , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/química , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia
4.
Nature ; 616(7955): 183-189, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36949197

RESUMO

Circadian rhythms play an essential part in many biological processes, and only three prokaryotic proteins are required to constitute a true post-translational circadian oscillator1. The evolutionary history of the three Kai proteins indicates that KaiC is the oldest member and a central component of the clock2. Subsequent additions of KaiB and KaiA regulate the phosphorylation state of KaiC for time synchronization. The canonical KaiABC system in cyanobacteria is well understood3-6, but little is known about more ancient systems that only possess KaiBC. However, there are reports that they might exhibit a basic, hourglass-like timekeeping mechanism7-9. Here we investigate the primordial circadian clock in Rhodobacter sphaeroides, which contains only KaiBC, to elucidate its inner workings despite missing KaiA. Using a combination of X-ray crystallography and cryogenic electron microscopy, we find a new dodecameric fold for KaiC, in which two hexamers are held together by a coiled-coil bundle of 12 helices. This interaction is formed by the carboxy-terminal extension of KaiC and serves as an ancient regulatory moiety that is later superseded by KaiA. A coiled-coil register shift between daytime and night-time conformations is connected to phosphorylation sites through a long-range allosteric network that spans over 140 Å. Our kinetic data identify the difference in the ATP-to-ADP ratio between day and night as the environmental cue that drives the clock. They also unravel mechanistic details that shed light on the evolution of self-sustained oscillators.


Assuntos
Proteínas de Bactérias , Relógios Circadianos , Ritmo Circadiano , Rhodobacter sphaeroides , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Proteínas de Bactérias/ultraestrutura , Fosforilação , Rhodobacter sphaeroides/química , Rhodobacter sphaeroides/metabolismo , Cristalografia por Raios X , Microscopia Crioeletrônica , Trifosfato de Adenosina/metabolismo , Difosfato de Adenosina/metabolismo , Cinética , Dobramento de Proteína , Conformação Proteica , Regulação Alostérica
5.
Nature ; 603(7901): 528-535, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35236984

RESUMO

Macromolecular function frequently requires that proteins change conformation into high-energy states1-4. However, methods for solving the structures of these functionally essential, lowly populated states are lacking. Here we develop a method for high-resolution structure determination of minorly populated states by coupling NMR spectroscopy-derived pseudocontact shifts5 (PCSs) with Carr-Purcell-Meiboom-Gill (CPMG) relaxation dispersion6 (PCS-CPMG). Our approach additionally defines the corresponding kinetics and thermodynamics of high-energy excursions, thereby characterizing the entire free-energy landscape. Using a large set of simulated data for adenylate kinase (Adk), calmodulin and Src kinase, we find that high-energy PCSs accurately determine high-energy structures (with a root mean squared deviation of less than 3.5 angström). Applying our methodology to Adk during catalysis, we find that the high-energy excursion involves surprisingly small openings of the AMP and ATP lids. This previously unresolved high-energy structure solves a longstanding controversy about conformational interconversions that are rate-limiting for catalysis. Primed for either substrate binding or product release, the high-energy structure of Adk suggests a two-step mechanism combining conformational selection to this state, followed by an induced-fit step into a fully closed state for catalysis of the phosphoryl-transfer reaction. Unlike other methods for resolving high-energy states, such as cryo-electron microscopy and X-ray crystallography, our solution PCS-CPMG approach excels in cases involving domain rearrangements of smaller systems (less than 60 kDa) and populations as low as 0.5%, and enables the simultaneous determination of protein structure, kinetics and thermodynamics while proteins perform their function.


Assuntos
Adenilato Quinase , Adenilato Quinase/metabolismo , Microscopia Crioeletrônica , Cristalografia por Raios X , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica , Termodinâmica
6.
7.
Science ; 370(6519)2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-33214247

RESUMO

Park et al question one out of seven findings from Hadzipasic et al: whether TPX2 allosterically regulates the oldest Aurora. We had already addressed the two concerns raised-sparse sequence sampling and not forcing the gene to the species tree-before publication. Moreover, we believe their ancestral sequence reconstruction would be consistent with a nonallosteric common ancestor, and we show large sequence differences caused by species tree-enforced gene trees.


Assuntos
Proteínas de Ciclo Celular , Proteínas Associadas aos Microtúbulos , Proteínas Serina-Treonina Quinases/genética
8.
Science ; 370(6523): 1442-1446, 2020 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-33214289

RESUMO

The advent of biocatalysts designed computationally and optimized by laboratory evolution provides an opportunity to explore molecular strategies for augmenting catalytic function. Applying a suite of nuclear magnetic resonance, crystallography, and stopped-flow techniques to an enzyme designed for an elementary proton transfer reaction, we show how directed evolution gradually altered the conformational ensemble of the protein scaffold to populate a narrow, highly active conformational ensemble and accelerate this transformation by nearly nine orders of magnitude. Mutations acquired during optimization enabled global conformational changes, including high-energy backbone rearrangements, that cooperatively organized the catalytic base and oxyanion stabilizer, thus perfecting transition-state stabilization. The development of protein catalysts for many chemical transformations could be facilitated by explicitly sampling conformational substates during design and specifically stabilizing productive substates over all unproductive conformations.


Assuntos
Biocatálise , Desenho Assistido por Computador , Evolução Molecular Direcionada , Enzimas/química , Enzimas/genética , Proteínas/química , Proteínas/genética , Domínio Catalítico , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica
9.
Proc Natl Acad Sci U S A ; 117(32): 19221-19227, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32719139

RESUMO

Despite the outstanding success of the cancer drug imatinib, one obstacle in prolonged treatment is the emergence of resistance mutations within the kinase domain of its target, Abl. We noticed that many patient-resistance mutations occur in the dynamic hot spots recently identified to be responsible for imatinib's high selectivity toward Abl. In this study, we provide an experimental analysis of the mechanism underlying drug resistance for three major resistance mutations (G250E, Y253F, and F317L). Our data settle controversies, revealing unexpected resistance mechanisms. The mutations alter the energy landscape of Abl in complex ways: increased kinase activity, altered affinity, and cooperativity for the substrates, and, surprisingly, only a modestly decreased imatinib affinity. Only under cellular adenosine triphosphate (ATP) concentrations, these changes cumulate in an order of magnitude increase in imatinib's half-maximal inhibitory concentration (IC50). These results highlight the importance of characterizing energy landscapes of targets and its changes by drug binding and by resistance mutations developed by patients.


Assuntos
Antineoplásicos/farmacologia , Mesilato de Imatinib/farmacologia , Neoplasias/enzimologia , Proteínas Oncogênicas v-abl/genética , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Resistencia a Medicamentos Antineoplásicos , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Proteínas Oncogênicas v-abl/química , Proteínas Oncogênicas v-abl/metabolismo
10.
Science ; 367(6480): 912-917, 2020 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-32079772

RESUMO

A myriad of cellular events are regulated by allostery; therefore, evolution of this process is of fundamental interest. Here, we use ancestral sequence reconstruction to resurrect ancestors of two colocalizing proteins, Aurora A kinase and its allosteric activator TPX2 (targeting protein for Xklp2), to experimentally characterize the evolutionary path of allosteric activation. Autophosphorylation of the activation loop is the most ancient activation mechanism; it is fully developed in the oldest kinase ancestor and has remained stable over 1 billion years of evolution. As the microtubule-associated protein TPX2 appeared, efficient kinase binding to TPX2 evolved, likely owing to increased fitness by virtue of colocalization. Subsequently, TPX2-mediated allosteric kinase regulation gradually evolved. Surprisingly, evolution of this regulation is encoded in the kinase and did not arise by a dominating mechanism of coevolution.


Assuntos
Aurora Quinase A/classificação , Aurora Quinase A/metabolismo , Evolução Molecular , Regulação Alostérica , Animais , Aurora Quinase A/química , Proteínas de Ciclo Celular/metabolismo , Ativação Enzimática , Humanos , Proteínas Associadas aos Microtúbulos/metabolismo , Filogenia
11.
Proc Natl Acad Sci U S A ; 116(28): 13937-13942, 2019 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-31239342

RESUMO

Despite being the subject of intense effort and scrutiny, kinases have proven to be consistently challenging targets in inhibitor drug design. A key obstacle has been promiscuity and consequent adverse effects of drugs targeting the ATP binding site. Here we introduce an approach to controlling kinase activity by using monobodies that bind to the highly specific regulatory allosteric pocket of the oncoprotein Aurora A (AurA) kinase, thereby offering the potential for more specific kinase modulators. Strikingly, we identify a series of highly specific monobodies acting either as strong kinase inhibitors or activators via differential recognition of structural motifs in the allosteric pocket. X-ray crystal structures comparing AurA bound to activating vs inhibiting monobodies reveal the atomistic mechanism underlying allosteric modulation. The results reveal 3 major advantages of targeting allosteric vs orthosteric sites: extreme selectivity, ability to inhibit as well as activate, and avoidance of competing with ATP that is present at high concentrations in the cells. We envision that exploiting allosteric networks for inhibition or activation will provide a general, powerful pathway toward rational drug design.


Assuntos
Aurora Quinase A/química , Aurora Quinase B/química , Inibidores de Proteínas Quinases/química , Proteínas Quinases/química , Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Regulação Alostérica/genética , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/genética , Aurora Quinase B/antagonistas & inibidores , Aurora Quinase B/genética , Sítios de Ligação/genética , Proteínas de Transporte/química , Proteínas de Transporte/genética , Cristalografia por Raios X , Desenho de Fármacos , Domínio de Fibronectina Tipo III/genética , Humanos , Conformação Proteica , Proteínas Quinases/genética
12.
Nat Catal ; 2(8): 726-734, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32159076

RESUMO

Protein conformational changes are frequently essential for enzyme catalysis, and in several cases, shown to be the limiting factor for overall catalytic speed. However, a structural understanding of corresponding transition states, needed to rationalize the kinetics, remains obscure due to their fleeting nature. Here, we determine the transition-state ensemble of the rate-limiting conformational transition in the enzyme adenylate kinase, by a synergistic approach between experimental high-pressure NMR relaxation during catalysis and molecular dynamics simulations. By comparing homologous kinases evolved under ambient or high pressure in the deep-sea, we detail transition state ensembles that differ in solvation as directly measured by the pressure dependence of catalysis. Capturing transition-state ensembles begins to complete the catalytic energy landscape that is generally characterized by structures of all intermediates and frequencies of transitions among them.

13.
Nat Commun ; 9(1): 4507, 2018 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-30375376

RESUMO

Protein tyrosine phosphatase SHP2 functions as a key regulator of cell cycle control, and activating mutations cause several cancers. Here, we dissect the energy landscape of wild-type SHP2 and the oncogenic mutation E76K. NMR spectroscopy and X-ray crystallography reveal that wild-type SHP2 exchanges between closed, inactive and open, active conformations. E76K mutation shifts this equilibrium toward the open state. The previously unknown open conformation is characterized, including the active-site WPD loop in the inward and outward conformations. Binding of the allosteric inhibitor SHP099 to E76K mutant, despite much weaker, results in an identical structure as the wild-type complex. A conformational selection to the closed state reduces drug affinity which, combined with E76K's much higher activity, demands significantly greater SHP099 concentrations to restore wild-type  activity levels. The differences in structural ensembles and drug-binding kinetics of cancer-associated SHP2 forms may stimulate innovative ideas for developing more potent inhibitors for activated SHP2 mutants.


Assuntos
Regulação Alostérica/genética , Mutação , Piperidinas/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Pirimidinas/metabolismo , Cristalografia por Raios X , Humanos , Espectroscopia de Ressonância Magnética , Piperidinas/farmacologia , Conformação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 11/antagonistas & inibidores , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/ultraestrutura , Pirimidinas/farmacologia
14.
Elife ; 72018 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-29901437

RESUMO

Protein kinases are major drug targets, but the development of highly-selective inhibitors has been challenging due to the similarity of their active sites. The observation of distinct structural states of the fully-conserved Asp-Phe-Gly (DFG) loop has put the concept of conformational selection for the DFG-state at the center of kinase drug discovery. Recently, it was shown that Gleevec selectivity for the Tyr-kinase Abl was instead rooted in conformational changes after drug binding. Here, we investigate whether protein dynamics after binding is a more general paradigm for drug selectivity by characterizing the binding of several approved drugs to the Ser/Thr-kinase Aurora A. Using a combination of biophysical techniques, we propose a universal drug-binding mechanism, that rationalizes selectivity, affinity and long on-target residence time for kinase inhibitors. These new concepts, where protein dynamics in the drug-bound state plays the crucial role, can be applied to inhibitor design of targets outside the kinome.


Assuntos
Aurora Quinase A/antagonistas & inibidores , Mesilato de Imatinib/farmacologia , Simulação de Dinâmica Molecular , Inibidores de Proteínas Quinases/farmacologia , Aurora Quinase A/química , Aurora Quinase A/metabolismo , Cristalografia por Raios X , Descoberta de Drogas/métodos , Humanos , Mesilato de Imatinib/química , Mesilato de Imatinib/metabolismo , Cinética , Ligação Proteica , Conformação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/metabolismo
15.
Nat Commun ; 9(1): 1314, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29615624

RESUMO

Rational design and directed evolution have proved to be successful approaches to increase catalytic efficiencies of both natural and artificial enzymes. Protein dynamics is recognized as important, but due to the inherent flexibility of biological macromolecules it is often difficult to distinguish which conformational changes are directly related to function. Here, we use directed evolution on an impaired mutant of the proline isomerase CypA and identify two second-shell mutations that partially restore its catalytic activity. We show both kinetically, using NMR spectroscopy, and structurally, by room-temperature X-ray crystallography, how local perturbations propagate through a large allosteric network to facilitate conformational dynamics. The increased catalysis selected for in the evolutionary screen is correlated with an accelerated interconversion between the two catalytically essential conformational sub-states, which are both captured in the high-resolution X-ray ensembles. Our data provide a glimpse of an evolutionary trajectory and show how subtle changes can fine-tune enzyme function.


Assuntos
Ciclofilina A/química , Evolução Molecular Direcionada , Catálise , Domínio Catalítico , Cristalografia por Raios X , Escherichia coli/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Modelos Moleculares , Método de Monte Carlo , Mutação , Prolina/química , Especificidade por Substrato , Temperatura
16.
Science ; 355(6322): 289-294, 2017 01 20.
Artigo em Inglês | MEDLINE | ID: mdl-28008087

RESUMO

With early life likely to have existed in a hot environment, enzymes had to cope with an inherent drop in catalytic speed caused by lowered temperature. Here we characterize the molecular mechanisms underlying thermoadaptation of enzyme catalysis in adenylate kinase using ancestral sequence reconstruction spanning 3 billion years of evolution. We show that evolution solved the enzyme's key kinetic obstacle-how to maintain catalytic speed on a cooler Earth-by exploiting transition-state heat capacity. Tracing the evolution of enzyme activity and stability from the hot-start toward modern hyperthermophilic, mesophilic, and psychrophilic organisms illustrates active pressure versus passive drift in evolution on a molecular level, refutes the debated activity/stability trade-off, and suggests that the catalytic speed of adenylate kinase is an evolutionary driver for organismal fitness.


Assuntos
Adenilil Ciclases/química , Biocatálise , Termotolerância , Adenilil Ciclases/classificação , Adenilil Ciclases/genética , Evolução Molecular , Temperatura Alta , Cinética , Mutação , Filogenia
17.
Mol Cell Oncol ; 3(4): e1046580, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27652309

RESUMO

Design of specific kinase inhibitors is an appealing approach for developing new anticancer treatments. However, only a few success stories have been reported to date. Here we demonstrate how the combination of old-fashioned and new biophysical tools together with recent advances in genomics and molecular evolution can aid in overcoming existing limitations.

18.
J Mol Biol ; 428(9 Pt A): 1742-59, 2016 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-26996940

RESUMO

The molecular mechanism by which the microtubule-associated protein (MAP) tau regulates the formation of microtubules (MTs) is poorly understood. The activity of tau is controlled via phosphorylation at specific Ser/Thr sites. Of those phosphorylation sites, 17 precede a proline, making them potential recognition sites for the peptidyl-prolyl isomerase Pin1. Pin1 binding and catalysis of phosphorylated tau at the AT180 epitope, which was implicated in Alzheimer's disease, has been reported to be crucial for restoring tau's ability to promote MT polymerization in vitro and in vivo [1]. Surprisingly, we discover that Pin1 does not promote phosphorylated tau-induced MT formation in vitro, refuting the commonly accepted model in which Pin1 binding and catalysis on the A180 epitope restores the function of the Alzheimer's associated phosphorylated tau in tubulin assembly [1, 2]. Using turbidity assays, time-resolved small angle X-ray scattering (SAXS), and time-resolved negative stain electron microscopy (EM), we investigate the mechanism of tau-mediated MT assembly and the role of the Thr231 and Ser235 phosphorylation on this process. We discover novel GTP-tubulin ring-shaped species, which are detectable in the earliest stage of tau-induced polymerization and may play a crucial role in the early nucleation phase of MT assembly. Finally, by NMR and SAXS experiments, we show that the tau molecules must be located on the surface of MTs and tubulin rings during the polymerization reaction. The interaction between tau and tubulin is multipartite, with a high affinity interaction of the four tubulin-binding repeats, and a weaker interaction with the proline-rich sequence and the termini of tau.


Assuntos
Microtúbulos/metabolismo , Peptidilprolil Isomerase de Interação com NIMA/metabolismo , Multimerização Proteica , Proteínas tau/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Microscopia Eletrônica , Microtúbulos/química , Microtúbulos/ultraestrutura , Espalhamento a Baixo Ângulo
19.
J Mol Biol ; 428(9 Pt A): 1760-75, 2016 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-26996941

RESUMO

Human peptidyl-prolyl isomerase (PPIase) Pin1 plays key roles in developmental processes, cell proliferation, and neuronal function. Extensive phosphorylation of the microtubule binding protein tau has been implicated in neurodegeneration and Alzheimer's disease. For the past 15years, these two players have been the focus of an enormous research effort to unravel the biological relevance of their interplay in health and disease, resulting in a series of proposed molecular mechanism of how Pin1 catalysis of tau results in biological phenotypes. Our results presented here refute these mechanisms of Pin1 action. Using NMR, isothermal calorimetry (ITC), and small angle x-ray scattering (SAXS), we dissect binding and catalysis on multiple phosphorylated tau with particular emphasis toward the Alzheimer's associated AT180 tau epitope containing phosphorylated THR231 and SER235. We find that phosphorylated (p-) SER235-PRO, but not pTHR231-PRO, is exclusively catalyzed by full-length Pin1 and isolated PPIase domain. Importantly, site-specific measurements of Pin1-catalysis of CDK2/CycA-phosphorylated full-length tau reveal a number of sites that are catalyzed simultaneously with different efficiencies. Furthermore, we show that the turnover efficiency at pSER235 by Pin1 is independent of both the WW domain and phosphorylation on THR231. Our mechanistic results on site-specific binding and catalysis together with the lack of an increase of dephosphorylation rates by PP2A counter a series of previously published models for the role of Pin1 catalysis of tau in Alzheimer's disease. Together, our data reemphasize the complicated scenario between binding and catalysis of multiple phosphorylated tau by Pin1 and the need for directly linking biological phenotypes and residue-specific turnover in Pin1 substrates.


Assuntos
Peptidilprolil Isomerase de Interação com NIMA/química , Peptidilprolil Isomerase de Interação com NIMA/metabolismo , Proteínas tau/química , Proteínas tau/metabolismo , Calorimetria , Humanos , Espectroscopia de Ressonância Magnética , Ligação Proteica , Processamento de Proteína Pós-Traducional , Espalhamento a Baixo Ângulo
20.
Cell Rep ; 14(1): 32-42, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26725117

RESUMO

Molecular recognition plays a central role in biology, and protein dynamics has been acknowledged to be important in this process. However, it is highly debated whether conformational changes happen before ligand binding to produce a binding-competent state (conformational selection) or are caused in response to ligand binding (induced fit). Proposals for both mechanisms in protein/protein recognition have been primarily based on structural arguments. However, the distinction between them is a question of the probabilities of going via these two opposing pathways. Here, we present a direct demonstration of exclusive conformational selection in protein/protein recognition by measuring the flux for rhodopsin kinase binding to its regulator recoverin, an important molecular recognition in the vision system. Using nuclear magnetic resonance (NMR) spectroscopy, stopped-flow kinetics, and isothermal titration calorimetry, we show that recoverin populates a minor conformation in solution that exposes a hydrophobic binding pocket responsible for binding rhodopsin kinase. Protein dynamics in free recoverin limits the overall rate of binding.


Assuntos
Receptor Quinase 1 Acoplada a Proteína G/química , Recoverina/química , Escherichia coli , Receptor Quinase 1 Acoplada a Proteína G/genética , Receptor Quinase 1 Acoplada a Proteína G/metabolismo , Humanos , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Estrutura Quaternária de Proteína , Recoverina/genética , Recoverina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...