Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Magn Reson ; 362: 107667, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38626504

RESUMEN

Solution NMR spectroscopy has tremendous potential for providing atomic resolution insights into the interactions between proteins and nucleic acids partitioned into condensed phases of phase-separated systems. However, the highly viscous nature of the condensed phase challenges applications, and in particular, the extraction of quantitative, site-specific information. Here, we present a delayed decoupling-based HMQC pulse sequence for methyl-TROSY studies of 'client' proteins and nucleic acids partitioned into 'scaffold' proteinaceous phase-separated solvents. High sensitivity and excellent quality spectra are recorded of a nascent form of superoxide dismutase and of a small RNA fragment partitioned into CAPRIN1 condensates.


Asunto(s)
Resonancia Magnética Nuclear Biomolecular , ARN , ARN/química , Resonancia Magnética Nuclear Biomolecular/métodos , Pliegue de Proteína , Proteínas/química , Superóxido Dismutasa/química , Condensados Biomoleculares/química , Algoritmos
2.
Chem Sci ; 15(10): 3502-3515, 2024 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-38455030

RESUMEN

Human serum albumin (HSA), the most abundant protein in plasma and cerebrospinal fluid, not only serves as a crucial carrier of various exogenous and endogenous ligands but also modulates the aggregation of amyloidogenic proteins, including alpha synuclein (αSyn), which is associated with Parkinson's disease and other α-synucleinopathies. HSA decreases αSyn toxicity through the direct binding to monomeric and oligomeric αSyn species. However, it is possible that HSA also sequesters metal ions that otherwise promote aggregation. Cu(ii) ions, for example, enhance αSyn fibrillization in vitro, while also leading to neurotoxicity by generating reactive oxygen species (ROS). However, it is currently unclear if and how HSA affects Cu(ii)-binding to αSyn. Using an integrated set of NMR experiments, we show that HSA is able to chelate Cu(ii) ions from αSyn more efficiently than standard chelators such as EDTA, revealing an unexpected cooperativity between the HSA metal-binding sites. Notably, fatty acid binding to HSA perturbs this cooperativity, thus interfering with the sequestration of Cu(ii) ions from αSyn. We also observed that glycation of HSA diminished Cu(ii)-binding affinity, while largely preserving the degree of cooperativity between the HSA metal-binding sites. Additionally, our results show that Cu(ii)-binding to HSA stabilizes the interactions of HSA with αSyn primarily at two different regions, i.e. the N-terminus, Tyr 39 and the majority of the C-terminus. Our study not only unveils the effect of fatty acid binding and age-related posttranslational modifications, such as glycation, on the neuroprotective mechanisms of HSA, but also highlights the potential of αSyn as a viable NMR-based sensor to investigate HSA-metal interactions.

3.
JACS Au ; 3(12): 3485-3493, 2023 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-38155658

RESUMEN

Alpha synuclein (αS) aggregates are the main component of Lewy bodies (LBs) associated with Parkinson's disease (PD). A longstanding question about αS and PD pertains to the autosomal dominant E46K αS mutant, which leads to the early onset of PD and LB dementias. The E46K mutation not only promotes αS aggregation but also stabilizes αS monomers in "closed" conformers, which are compact and aggregation-incompetent. Hence, the mechanism of action of the E46K mutation is currently unclear. Here, we show that αS monomers harboring the E46K mutation exhibit more extensive interactions with fibrils compared to those of WT. Such monomer-fibril interactions are sufficient to allosterically drive transitions of αS monomers from closed to open conformations, enabling αS aggregation. We also show that E46K promotes head-to-tail monomer-monomer interactions in early self-association events. This multipronged mechanism provides a new framework to explain how the E46K mutation and possibly other αS variants trigger early-onset PD.

4.
Chem Sci ; 14(36): 9933-9942, 2023 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-37736631

RESUMEN

The ability of Adenosine Triphosphate (ATP) to modulate protein solubility establishes a critical link between ATP homeostasis and proteinopathies, such as Parkinson's (PD). The most significant risk factor for PD is aging, and ATP levels decline dramatically with age. However, the mechanism by which ATP interacts with alpha-synuclein (αS), whose aggregation is characteristic of PD, is currently not fully understood, as is ATP's effect on αS aggregation. Here, we use nuclear magnetic resonance spectroscopy as well as fluorescence, dynamic light scattering and microscopy to show that ATP affects multiple species in the αS self-association cascade. The triphosphate moiety of ATP disrupts long-range electrostatic intramolecular contacts in αS monomers to enhance initial aggregation, while also inhibiting the formation of late-stage ß-sheet fibrils by disrupting monomer-fibril interactions. These effects are modulated by magnesium ions and early onset PD-related αS mutations, suggesting that loss of the ATP hydrotropic function on αS fibrillization may play a role in PD etiology.

5.
Cell Res ; 33(8): 583-584, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37016021
6.
Essays Biochem ; 66(7): 863-873, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36416859

RESUMEN

The spatial and temporal organization of interactions between proteins underlie the regulation of most cellular processes. The requirement for such interactions to be specific predisposes a view that protein-protein interactions are relatively static and are formed through the stable complementarity of the interacting partners. A growing body of reports indicate, however, that many interactions lead to fuzzy complexes with an ensemble of conformations in dynamic exchange accounting for the observed binding. Here, we discuss how NMR has facilitated the characterization of these discrete, dynamic complexes and how such characterization has aided the understanding of dynamic, condensed phases of phase-separating proteins with exchanging multivalent interactions.


Asunto(s)
Fenómenos Bioquímicos , Proteínas Intrínsecamente Desordenadas , Proteínas Intrínsecamente Desordenadas/química
7.
PLoS One ; 17(3): e0263671, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35275926

RESUMEN

Novel therapeutic strategies are needed to control the SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) pandemic. Here, we present a protocol to anchor the SARS-CoV-2 spike (S-)protein in the cytoplasmic membranes of erythrocyte liposomes. A surfactant was used to stabilize the S-protein's structure in the aqueous environment before insertion and to facilitate reconstitution of the S-proteins in the erythrocyte membranes. The insertion process was studied using coarse grained Molecular Dynamics (MD) simulations. Liposome formation and S-protein anchoring was studied by dynamic light scattering (DLS), ELV-protein co-sedimentation assays, fluorescent microcopy and cryo-TEM. The Erythro-VLPs (erythrocyte based virus like particles) have a well defined size of ∼200 nm and an average protein density on the outer membrane of up to ∼300 proteins/µm2. The correct insertion and functional conformation of the S-proteins was verified by dose-dependent binding to ACE-2 (angiotensin converting enzyme 2) in biolayer interferometry (BLI) assays. Seroconversion was observed in a pilot mouse trial after 14 days when administered intravenously, based on enzyme-linked immunosorbent assays (ELISA). This red blood cell based platform can open novel possibilities for therapeutics for the coronavirus disease (COVID-19) including variants, and other viruses in the future.


Asunto(s)
Vacunas contra la COVID-19 , COVID-19 , Membrana Eritrocítica , Simulación de Dinámica Molecular , SARS-CoV-2/inmunología , Glicoproteína de la Espiga del Coronavirus , Vacunas de Partículas Similares a Virus , Animales , COVID-19/inmunología , COVID-19/prevención & control , Vacunas contra la COVID-19/química , Vacunas contra la COVID-19/inmunología , Vacunas contra la COVID-19/farmacología , Membrana Eritrocítica/química , Membrana Eritrocítica/inmunología , Femenino , Liposomas , Ratones , Proyectos Piloto , Dominios Proteicos , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/farmacología , Vacunas de Partículas Similares a Virus/química , Vacunas de Partículas Similares a Virus/inmunología , Vacunas de Partículas Similares a Virus/farmacología
8.
Biophys Chem ; 282: 106743, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35093643

RESUMEN

Human Serum Albumin (HSA), the most abundant protein in plasma, serves a diverse repertoire of biological functions including regulation of oncotic pressure and redox potential, transport of serum solutes, but also chaperoning of misfolded proteins. Here we review how HSA interacts with a wide spectrum of client proteins including intrinsically disordered proteins (IDPs) such as Aß, the islet amyloid peptide (IAPP), alpha synuclein and stressed globular proteins such as insulin. The comparative analysis of the HSA chaperone - client interactions reveals that the amyloid-inhibitory function of HSA arises from at least four emerging mechanisms. Two mechanisms (the monomer stabilizer model and the monomer competitor model) involve the direct binding of HSA to either IDP monomers or oligomers, while other mechanisms (metal chelation and membrane protection) rely on the indirect modulation by HSA of other factors that drive IDP aggregation. While HSA is not the only extracellular chaperone, given its abundance, HSA is likely to account for a significant fraction of the chaperoning effects in plasma, thus opening new therapeutic opportunities in the context of the peripheral sink hypothesis.


Asunto(s)
Proteínas Intrínsecamente Desordenadas , Humanos , Amiloide , Péptidos beta-Amiloides/metabolismo , Proteínas Amiloidogénicas , Albúmina Sérica Humana
9.
J Biol Chem ; 298(2): 101502, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34929173

RESUMEN

The aberrant self-assembly of intrinsically disordered proteins (IDPs) into soluble oligomers and their interactions with biological membranes underlie the pathogenesis of numerous neurodegenerative diseases, including Alzheimer's disease. Catechins have emerged as useful tools to reduce the toxicity of IDP oligomers by modulating their interactions with membranes. However, the structural determinants of catechin binding to IDP oligomers and membranes remain largely elusive. Here, we assemble a catechin library by combining several naturally occurring chemical modifications and, using a coupled NMR-statistical approach, we map at atomic resolution the interactions of such library with the Alzheimer's-associated amyloid-beta (Aß) oligomers and model membranes. Our results reveal multiple catechin affinity drivers and show that the combination of affinity-reducing covalent changes may lead to unexpected net gains in affinity. Interestingly, we find that the positive cooperativity is more prevalent for Aß oligomers than membrane binding, and that the determinants underlying catechin recognition by membranes are markedly different from those dissected for Aß oligomers. Notably, we find that the unanticipated positive cooperativity arises from the critical regulatory role of the gallate catechin moiety, which recruits previously disengaged substituents into the binding interface and leads to an overall greater compaction of the receptor-bound conformation. Overall, the previously elusive structural attributes mapped here provide an unprecedented foundation to establish structure-activity relationships of catechins.


Asunto(s)
Enfermedad de Alzheimer , Péptidos beta-Amiloides , Catequina , Humanos , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Catequina/química , Catequina/metabolismo , Lípidos , Relación Estructura-Actividad , Proteínas Intrínsecamente Desordenadas/química , Proteínas Intrínsecamente Desordenadas/metabolismo
10.
J Am Chem Soc ; 143(12): 4668-4679, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33733753

RESUMEN

Soluble oligomers formed by amyloidogenic intrinsically disordered proteins are some of the most cytotoxic species linked to neurodegeneration. Due to the transient and heterogeneous nature of such oligomeric intermediates, the underlying self-association events often remain elusive. NMR relaxation measurements sensitive to zero-frequency spectral densities (J(0)), such as the 15N - R2 rates, are ideally suited to map sites of self-association at atomic resolution without the need of exogenous labels. Such experiments exploit the dynamic exchange between NMR visible monomers and slowly tumbling oligomers. However,15N - R2 rates are also sensitive to intrinsic monomer dynamics, and it is often difficult to discern these contributions from those arising from exchange with oligomers. Another challenge pertains to defining a hierarchy of self-association. Here, using the archetypical amyloidogenic protein alpha synuclein (αS), we show that the temperature-dependence of 15N - R2 effectively identifies self-association sites with reduced bias from internal dynamics. The key signature of the residues involved in self-association is a nonlinear temperature-dependence of 15N - R2 with a positive ΔR2/ΔT slope. These two hallmarks are systematically probed through a thermal R2 correlation matrix, from which the network of residues involved in self-association as well as the hierarchy of αS self-association sites is extracted through agglomerative clustering. We find that aggregation is initiated by residues within the NAC region that is solvent inaccessible in αS fibrils and eventually extends to the N-terminal segment harboring familial PD mutations. These hierarchical self-association maps help dissect the essential drivers of oligomerization and reveal how amyloid inhibitors affect oligomer formation.


Asunto(s)
Proteínas Intrínsecamente Desordenadas/química , Temperatura , alfa-Sinucleína/análisis , Humanos , Proteínas Intrínsecamente Desordenadas/síntesis química , Modelos Moleculares
11.
Biophys Chem ; 269: 106508, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33310607

RESUMEN

Given the breadth and depth of the scientific contributions of Sir Christopher Dobson, with over 870 publications to date, it is inconceivable to convey in a single review the impact of his work and its legacy. This review therefore primarily focuses on his contributions to the development of strategies for preventing aberrant protein misfolding. The first section of this review highlights his seminal work on the elucidation of the microscopic nucleation processes underlying protein aggregation. Next, we discuss the specific inhibition of these steps by candidate drugs and biologics, with a particular emphasis on the role of molecular chaperones. In the final section, we review how protein aggregation principles can be exploited for the rational design of novel and more potent aggregation inhibitors. These milestones serve as excellent examples of the profound impact of Dobson's seminal work on fundamental science and its translation into drug discovery.


Asunto(s)
Chaperonas Moleculares/metabolismo , Agregado de Proteínas , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/química , Péptidos beta-Amiloides/metabolismo , Humanos
12.
Molecules ; 25(16)2020 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-32781559

RESUMEN

Protein misfolding as well as the subsequent self-association and deposition of amyloid aggregates is implicated in the progression of several neurodegenerative disorders including Alzheimer's and Parkinson's diseases. Modulators of amyloidogenic aggregation serve as essential tools to dissect the underlying molecular mechanisms and may offer insight on potential therapeutic solutions. These modulators include green tea catechins, which are potent inhibitors of amyloid aggregation. Although catechins often exhibit poor pharmacokinetic properties and bioavailability, they are still essential tools for identifying the drivers of amyloid aggregation and for developing other aggregation modulators through structural mimicry. As an illustration of such strategies, here we review how catechins have been used to map the toxic surfaces of oligomeric amyloid-like species and develop catechin-based phenolic compounds with enhanced anti-amyloid activity.


Asunto(s)
Catequina/farmacología , Enfermedades Neurodegenerativas , Fármacos Neuroprotectores/farmacología , Catequina/uso terapéutico , Humanos , Terapia Molecular Dirigida , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Fármacos Neuroprotectores/uso terapéutico
13.
ACS Chem Neurosci ; 11(17): 2566-2576, 2020 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-32687307

RESUMEN

Alzheimer's disease (AD) is linked to the abnormal accumulation of amyloid ß peptide (Aß) aggregates in the brain. Silybin B, a natural compound extracted from milk thistle (Silybum marianum), has been shown to significantly inhibit Aß aggregation in vitro and to exert neuroprotective properties in vivo. However, further explorations of silybin B's clinical potential are currently limited by three main factors: (a) poor solubility, (b) instability in blood serum, and (c) only partial knowledge of silybin's mechanism of action. Here, we address these three limitations. We demonstrate that conjugation of a trehalose moiety to silybin significantly increases both water solubility and stability in blood serum without significantly compromising its antiaggregation properties. Furthermore, using a combination of biophysical techniques with different spatial resolution, that is, TEM, ThT fluorescence, CD, and NMR spectroscopy, we profile the interactions of the trehalose conjugate with both Aß monomers and oligomers and evidence that silybin may shield the "toxic" surfaces formed by the N-terminal and central hydrophobic regions of Aß. Finally, comparative analysis with silybin A, a less active diastereoisomer of silybin B, revealed how even subtle differences in chemical structure may entail different effects on amyloid inhibition. The resulting insight on the mechanism of action of silybins as aggregation inhibitors is anticipated to facilitate the future investigation of silybin's therapeutic potential.


Asunto(s)
Péptidos beta-Amiloides , Profármacos , Antioxidantes , Fragmentos de Péptidos , Silibina , Trehalosa
14.
J Am Chem Soc ; 142(21): 9686-9699, 2020 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-32383602

RESUMEN

Alpha synuclein (αS) oligomers are a key component of Lewy bodies implicated in Parkinson's disease (PD). Although primarily intracellular, extracellular αS exocytosed from neurons also contributes to PD pathogenesis through a prion-like transmission mechanism. Here, we show at progressive degrees of resolution that the most abundantly expressed extracellular protein, human serum albumin (HSA), inhibits αS oligomer (αSn) toxicity through a three-pronged mechanism. First, endogenous HSA targets αSn with sub-µM affinity via solvent-exposed hydrophobic sites, breaking the catalytic cycle that promotes αS self-association. Second, HSA remodels αS oligomers and high-MW fibrils into chimeric intermediates with reduced toxicity. Third, HSA unexpectedly suppresses membrane interactions with the N-terminal and central αS regions. Overall, our findings suggest that the extracellular proteostasis network may regulate αS cell-to-cell transmission not only by reducing the populations of membrane-binding competent αS oligomers but possibly also by shielding the membrane interface from residual toxic species.


Asunto(s)
Chaperonas Moleculares/metabolismo , Albúmina Sérica Humana/metabolismo , alfa-Sinucleína/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Chaperonas Moleculares/química , Albúmina Sérica Humana/química , alfa-Sinucleína/química
15.
Chem Sci ; 10(24): 6072-6082, 2019 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-31360412

RESUMEN

Soluble amyloid beta assemblies (Aß n ) are neurotoxic and play a central role in the early phases of the pathogenesis cascade leading to Alzheimer's disease. However, the current knowledge about the molecular determinants of Aß n toxicity is at best scant. Here, we comparatively analyze Aß n prepared in the absence or presence of a catechin library that modulates cellular toxicity. By combining solution NMR with dynamic light scattering, fluorescence spectroscopy, electron microscopy, wide-angle X-ray diffraction and cell viability assays, we identify a cluster of unique molecular signatures that distinguish toxic vs. nontoxic Aß assemblies. These include the exposure of a hydrophobic surface spanning residues 17-28 and the concurrent shielding of the highly charged N-terminus. We show that the combination of these two dichotomous structural transitions promotes the colocalization and insertion of ß-sheet rich Aß n into the membrane, compromising membrane integrity. These previously elusive toxic surfaces mapped here provide an unprecedented foundation to establish structure-toxicity relationships of Aß assemblies.

16.
J Med Chem ; 62(10): 5063-5079, 2019 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-31074269

RESUMEN

A common source of false positives in drug discovery is ligand self-association into large colloidal assemblies that nonspecifically inhibit target proteins. However, the mechanisms of aggregation-based inhibition (ABI) and ABI-attenuation by additives, such as Triton X-100 (TX) and human serum albumin (HSA), are not fully understood. Here, we investigate the molecular basis of ABI and ABI-attenuation through the lens of NMR and coupled thermodynamic cycles. We unexpectedly discover a new class of aggregating ligands that exhibit negligible interactions with proteins but act as competitive sinks for the free inhibitor, resulting in bell-shaped dose-response curves. TX attenuates ABI by converting inhibitory, protein-binding aggregates into nonbinding coaggregates, whereas HSA minimizes nonspecific ligand interactions by functioning as a reservoir for free inhibitor and preventing self-association. Hence, both TX and HSA are useful tools to minimize false positives arising from nonspecific binding but at the cost of potentially introducing false negatives due to suppression of specific interactions.


Asunto(s)
Descubrimiento de Drogas/métodos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Sitios de Unión , Tampones (Química) , Relación Dosis-Respuesta a Droga , Reacciones Falso Positivas , Humanos , Luz , Espectroscopía de Resonancia Magnética , Octoxinol/farmacología , Dispersión de Radiación , Albúmina Sérica Humana/química , Albúmina Sérica Humana/efectos de los fármacos , Espectrometría de Masa por Ionización de Electrospray , Resonancia por Plasmón de Superficie , Termodinámica
17.
RSC Adv ; 9(33): 18978-18988, 2019 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-35516872

RESUMEN

Degradable low-fouling hydrogels are ideal vehicles for drug and cell delivery. For each application, hydrogel degradation rate must be re-optimized for maximum therapeutic benefit. We developed a method to rapidly and predictably tune degradation rates of low-fouling poly(oligo(ethylene glycol)methyl ether methacrylate) (P(EG) x MA) hydrogels by modifying two interdependent variables: (1) base-catalysed crosslink degradation kinetics, dependent on crosslinker electronics (electron withdrawing groups (EWGs)); and, (2) polymer hydration, dependent on the molecular weight (M W) of poly(ethylene glycol) (PEG) pendant groups. By controlling PEG M W and EWG strength, P(EG) x MA hydrogels were tuned to degrade over 6 to 52 d. A 6-member P(EG) x MA copolymer library yielded slow and fast degrading low-fouling hydrogels suitable for short- and long-term delivery applications. The degradation mechanism was also applied to RGD-functionalized poly(carboxybetaine methacrylamide) (PCBMAA) hydrogels to achieve slow (∼50 d) and fast (∼13 d) degrading low-fouling, bioactive hydrogels.

18.
Chem Commun (Camb) ; 54(37): 4644-4652, 2018 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-29658548

RESUMEN

The self-association of the amyloid beta (Aß) peptide into toxic oligomers is implicated in the early events leading to Alzheimer's disease (AD). Blocking the formation of Aß oligomers and their interactions with the extracellular and cellular environment through small molecules and biopharmaceuticals is therefore a promising preventive strategy for AD. However, given the heterogeneity and transient nature of the Aß oligomeric species, detailed structural and kinetic characterizations of such oligomers and oligomer:inhibitor complexes have proven to be challenging. Here, we discuss recent advancements in solution NMR that have been instrumental in overcoming these limitations and we provide two representative examples of Aß inhibitors from our work to demonstrate the applications of such experiments, i.e. EGCG and human serum albumin.


Asunto(s)
Péptidos beta-Amiloides/antagonistas & inhibidores , Péptidos beta-Amiloides/metabolismo , Catequina/análogos & derivados , Fragmentos de Péptidos/antagonistas & inhibidores , Fragmentos de Péptidos/metabolismo , Multimerización de Proteína/efectos de los fármacos , Albúmina Sérica/metabolismo , Secuencia de Aminoácidos , Péptidos beta-Amiloides/química , Catequina/química , Catequina/metabolismo , Humanos , Espectroscopía de Resonancia Magnética/métodos , Fragmentos de Péptidos/química , Unión Proteica , Albúmina Sérica/química
19.
Methods Mol Biol ; 1688: 391-405, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29151219

RESUMEN

Mapping allosteric sites is emerging as one of the central challenges in physiology, pathology, and pharmacology. Nuclear Magnetic Resonance (NMR) spectroscopy is ideally suited to map allosteric sites, given its ability to sense at atomic resolution the dynamics underlying allostery. Here, we focus specifically on the NMR CHEmical Shift Covariance Analysis (CHESCA), in which allosteric systems are interrogated through a targeted library of perturbations (e.g., mutations and/or analogs of the allosteric effector ligand). The atomic resolution readout for the response to such perturbation library is provided by NMR chemical shifts. These are then subject to statistical correlation and covariance analyses resulting in clusters of allosterically coupled residues that exhibit concerted responses to the common set of perturbations. This chapter provides a description of how each step in the CHESCA is implemented, starting from the selection of the perturbation library and ending with an overview of different clustering options.


Asunto(s)
Sitio Alostérico , Espectroscopía de Resonancia Magnética/métodos , Modelos Moleculares , Ligandos , Unión Proteica
20.
Biochim Biophys Acta Proteins Proteom ; 1865(11 Pt B): 1529-1543, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28911813

RESUMEN

It is now established that understanding the molecular basis of biological function requires atomic resolution maps of both structure and dynamics. Here, we review several illustrative examples of functional dynamics selected from our work on cyclic nucleotide signaling and amyloid inhibition. Although fundamentally diverse, a central aspect common to both fields is that function can only be rationalized by considering dynamic equilibria between distinct states of the accessible free energy landscape. The dynamic exchange between ground and excited states of signaling proteins is essential to explain auto-inhibition and allosteric activation. The dynamic exchange between non-toxic monomeric species and toxic oligomers of amyloidogenic proteins provides a foundation to understand amyloid inhibition. NMR ideally probes both types of dynamic exchange at atomic resolution. Specifically, we will show how NMR was utilized to reveal the dynamical basis of cyclic nucleotide affinity, selectivity, agonism and antagonism in multiple eukaryotic cAMP and cGMP receptors. We will also illustrate how NMR revealed the mechanism of action of plasma proteins that act as extracellular chaperones and inhibit the self-association of the prototypical amyloidogenic Aß peptide. The examples outlined in this review illustrate the widespread implications of functional dynamics and the power of NMR as an indispensable tool in molecular pharmacology and pathology.


Asunto(s)
Amiloide/química , Proteínas Portadoras/química , AMP Cíclico/química , GMP Cíclico/química , Péptidos y Proteínas de Señalización Intracelular/química , Resonancia Magnética Nuclear Biomolecular , Receptores de AMP Cíclico/química , Amiloide/metabolismo , Animales , Proteínas Portadoras/metabolismo , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptores de AMP Cíclico/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...