Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 137
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Anal Chem ; 95(18): 7178-7185, 2023 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-37102678

RESUMO

Membrane proteins are vital in the human proteome for their cellular functions and make up a majority of drug targets in the U.S. However, characterizing their higher-order structures and interactions remains challenging. Most often membrane proteins are studied in artificial membranes, but such artificial systems do not fully account for the diversity of components present in cell membranes. In this study, we demonstrate that diethylpyrocarbonate (DEPC) covalent labeling mass spectrometry can provide binding site information for membrane proteins in living cells using membrane-bound tumor necrosis factor α (mTNFα) as a model system. Using three therapeutic monoclonal antibodies that bind TNFα, our results show that residues that are buried in the epitope upon antibody binding generally decrease in DEPC labeling extent. Additionally, serine, threonine, and tyrosine residues on the periphery of the epitope increase in labeling upon antibody binding because of a more hydrophobic microenvironment that is created. We also observe changes in labeling away from the epitope, indicating changes to the packing of the mTNFα homotrimer, compaction of the mTNFα trimer against the cell membrane, and/or previously uncharacterized allosteric changes upon antibody binding. Overall, DEPC-based covalent labeling mass spectrometry offers an effective means of characterizing structure and interactions of membrane proteins in living cells.


Assuntos
Proteínas de Membrana , Tirosina , Humanos , Dietil Pirocarbonato/química , Espectrometria de Massas/métodos , Membrana Celular , Ligação Proteica
2.
Mass Spectrom Rev ; 41(1): 51-69, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33145813

RESUMO

Membrane proteins are incredibly important biomolecules because they mediate interactions between a cell's external and internal environment. Obtaining information about membrane protein structure and interactions is thus important for understanding these essential biomolecules. Compared with the analyses of water-soluble proteins, the structural analysis of membrane proteins is more challenging owing to their unique chemical properties and the presence of lipid components that are necessary to solubilize them. The combination of covalent labeling (CL) and mass spectrometry (MS) has recently been applied with great success to study membrane protein structure and interactions. These studies have demonstrated the many advantages that CL-MS methods have over other traditional biophysical techniques. In this review, we discuss both amino acid-specific and non-specific labeling approaches and the special considerations needed to address the unique challenges associated with interrogating membrane proteins. This review highlights the aspects of this approach that require special care to be applied correctly and provides a comprehensive review of the membrane protein systems that have been studied by CL-MS. © 2020 John Wiley & Sons Ltd. Mass Spec Rev.


Assuntos
Aminoácidos , Proteínas de Membrana , Espectrometria de Massas/métodos
3.
Bioconjug Chem ; 34(6): 1130-1138, 2023 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-37220065

RESUMO

Targeted modification of endogenous proteins without genetic manipulation of protein expression machinery has a range of applications from chemical biology to drug discovery. Despite being demonstrated to be effective in various applications, target-specific protein labeling using ligand-directed strategies is limited by stringent amino acid selectivity. Here, we present highly reactive ligand-directed triggerable Michael acceptors (LD-TMAcs) that feature rapid protein labeling. Unlike previous approaches, the unique reactivity of LD-TMAcs enables multiple modifications on a single target protein, effectively mapping the ligand binding site. This capability is attributed to the tunable reactivity of TMAcs that enable the labeling of several amino acid functionalities via a binding-induced increase in local concentration while remaining fully dormant in the absence of protein binding. We demonstrate the target selectivity of these molecules in cell lysates using carbonic anhydrase as the model protein. Furthermore, we demonstrate the utility of this method by selectively labeling membrane-bound carbonic anhydrase XII in live cells. We envision that the unique features of LD-TMAcs will find use in target identification, investigation of binding/allosteric sites, and studying membrane proteins.


Assuntos
Aminoácidos , Proteínas de Membrana , Ligantes , Sítios de Ligação , Ligação Proteica
4.
Analyst ; 148(18): 4479-4488, 2023 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-37575048

RESUMO

Nanomaterials have been employed in many biomedical applications, and their distributions in biological systems can provide an understanding of their behavior in vivo. Laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) can be used to determine the distributions of metal-based NMs in biological systems. However, LA-ICP-MS has not commonly been used to quantitatively measure the cell-specific or sub-organ distributions of nanomaterials in tissues. Here, we describe a new platform that uses spiked gelatin standards with control tissues on top to obtain an almost perfect tissue mimic for quantitative imaging purposes. In our approach, gelatin is spiked with both nanomaterial standards and an internal standard to improve quantitation and image quality. The value of the developed approach is illustrated by determining the sub-organ distributions of different metal-based and metal-tagged polymeric nanomaterials in mice organs. The LA-ICP-MS images reveal that the chemical and physical properties of the nanomaterials cause them to distribute in quantitatively different extents in spleens, kidneys, and tumors, providing new insight into the fate of nanomaterials in vivo. Furthermore, we demonstrate that this approach enables quantitative co-localization of nanomaterials and their cargo. We envision this method being a valuable tool in the development of nanomaterial drug delivery systems.


Assuntos
Gelatina , Terapia a Laser , Camundongos , Animais , Espectrometria de Massas/métodos , Terapia a Laser/métodos , Metais/análise , Análise Espectral
5.
J Am Chem Soc ; 144(48): 22128-22139, 2022 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-36414315

RESUMO

Proteins can adopt different conformational states that are important for their biological function and, in some cases, can be responsible for their dysfunction. The essential roles that proteins play in biological systems make distinguishing the structural differences between these conformational states both fundamentally and practically important. Here, we demonstrate that collision-induced unfolding (CIU), in combination with ion mobility-mass spectrometry (IM-MS) measurements, distinguish subtly different conformational states for protein complexes. Using the open and closed states of the ß-lactoglobulin (ßLG) dimer as a model, we show that these two conformational isomers unfold during collisional activation to generate distinct states that are readily separated by IM-MS. Extensive molecular modeling of the CIU process reproduces the distinct unfolding intermediates and identifies the molecular details that explain why the two conformational states unfold in distinct ways. Strikingly, the open conformational state forms new electrostatic interactions upon collisional heating, while the closed state does not. These newly formed electrostatic interactions involve residues on the loop differentially positioned in the two ßLG conformational isomers, highlighting that gas-phase unfolding pathways reflect aspects of solution structure. This combination of experiment and theory provides a path forward for distinguishing subtly different conformational isomers for protein complexes via gas-phase unfolding experiments. Our results also have implications for understanding how protein complexes dissociate in the gas phase, indicating that current models need to be refined to explain protein complex dissociation.

6.
Anal Chem ; 94(2): 1003-1010, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-34962759

RESUMO

In this work, we use diethylpyrocarbonate (DEPC)-based covalent labeling together with LC-MS/MS analysis to distinguish the two sidechain tautomers of histidine residues in peptides and proteins. From labeling experiments on model peptides, we demonstrate that DEPC reacts equally with both tautomeric forms to produce chemically different products with distinct dissociation patterns and LC retention times, allowing the ratios of the two tautomers to be determined in peptides and proteins. Upon measuring the tautomer ratios of several histidine residues in myoglobin, we find good agreement with previous 2D NMR data on this protein. Because our DEPC labeling/MS approach is simpler, faster, and more precise than 2D NMR, our method will be a valuable way to determine how protein structure enforces histidine sidechain tautomerization. Because the tautomeric state of histidine residues is often important for protein structure and function, the ability of DEPC labeling/MS to distinguish histidine tautomers should equip researchers with a tool to understand the histidine residue structure and function more deeply in proteins.


Assuntos
Histidina , Espectrometria de Massas em Tandem , Cromatografia Líquida , Dietil Pirocarbonato/química , Isomerismo
7.
Anal Chem ; 94(22): 7901-7908, 2022 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-35612963

RESUMO

Polymeric nanocarriers (PNCs) are versatile drug delivery vehicles capable of delivering a variety of therapeutics. Quantitatively monitoring their uptake in biological systems is essential for realizing their potential as next-generation delivery systems; however, existing quantification strategies are limited due to the challenges of detecting polymeric materials in complex biological samples. Here, we describe a metal-coded mass tagging approach that enables the multiplexed quantification of the PNC uptake in cells using mass spectrometry (MS). In this approach, PNCs are conjugated with ligands that bind strongly to lanthanide ions, allowing the PNCs to be sensitively quantitated by inductively coupled plasma-MS. The metal-coded tags have little effect on the properties or toxicity of the PNCs, making them biocompatible. We demonstrate that the conjugation of different metals to the PNCs enables the multiplexed analysis of cellular uptake of multiple distinct PNCs at the same time. This multiplexing capability should improve the design and optimization of PNCs by minimizing biological variability and reducing analysis time, effort, and cost.


Assuntos
Elementos da Série dos Lantanídeos , Polímeros , Elementos da Série dos Lantanídeos/química , Espectrometria de Massas/métodos , Polímeros/química , Análise Espectral
8.
Anal Chem ; 94(2): 1052-1059, 2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-34932327

RESUMO

Antigen-antibody epitope mapping is essential for understanding binding mechanisms and developing new protein therapeutics. In this study, we investigate diethylpyrocarbonate (DEPC) covalent labeling-mass spectrometry as a means of analyzing antigen-antibody interactions using the well-characterized model system of TNFα in complex with three different antibodies. Results show that residues buried in the epitope undergo substantial decreases in labeling, as expected. Interestingly, serine, threonine, and tyrosine residues at the edges of the epitope undergo unexpected increases in labeling. The increased labeling of these weakly nucleophilic residues is caused by the formation of hydrophobic pockets upon antibody binding that presumably increase local DEPC concentrations. Residues that are distant from the epitope generally do not undergo changes in labeling extent; however, some that do change experience variations in their local microenvironment due to side-chain reorganization or stabilization of the TNFα trimer that occurs upon binding. Overall, DEPC labeling of antigen-antibody complexes is found to depend on both changes in solvent exposure and changes to the residue microenvironment.


Assuntos
Treonina , Tirosina , Dietil Pirocarbonato/química , Mapeamento de Epitopos , Espectrometria de Massas/métodos
9.
Anal Chem ; 93(23): 8188-8195, 2021 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-34061512

RESUMO

Diethylpyrocarbonate (DEPC) labeling analyzed with mass spectrometry can provide important insights into higher order protein structures. It has been previously shown that neighboring hydrophobic residues promote a local increase in DEPC concentration such that serine, threonine, and tyrosine residues are more likely to be labeled despite low solvent exposure. In this work, we developed a Rosetta algorithm that used the knowledge of labeled and unlabeled serine, threonine, and tyrosine residues and assessed their local hydrophobic environment to improve protein structure prediction. Additionally, DEPC-labeled histidine and lysine residues with higher relative solvent accessible surface area values (i.e., more exposed) were scored favorably. Application of our score term led to reductions of the root-mean-square deviations (RMSDs) of the lowest scoring models. Additionally, models that scored well tended to have lower RMSDs. A detailed tutorial describing our protocol and required command lines is included. Our work demonstrated the considerable potential of DEPC covalent labeling data to be used for accurate higher order structure determination.


Assuntos
Proteínas , Tirosina , Dietil Pirocarbonato , Histidina , Espectrometria de Massas
10.
Analyst ; 146(24): 7720-7729, 2021 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-34821231

RESUMO

Laser ablation inductively-coupled plasma mass spectrometry (LA-ICP-MS) imaging and matrix assisted laser desorption ionization mass spectrometry imaging (MALDI-MSI) are complementary methods that measure distributions of elements and biomolecules in tissue sections. Quantitative correlations of the information provided by these two imaging modalities requires that the datasets be registered in the same coordinate system, allowing for pixel-by-pixel comparisons. We describe here a computational workflow written in Python that accomplishes this registration, even for adjacent tissue sections, with accuracies within ±50 µm. The value of this registration process is demonstrated by correlating images of tissue sections from mice injected with gold nanomaterial drug delivery systems. Quantitative correlations of the nanomaterial delivery vehicle, as detected by LA-ICP-MS imaging, with biochemical changes, as detected by MALDI-MSI, provide deeper insight into how nanomaterial delivery systems influence lipid biochemistry in tissues. Moreover, the registration process allows the more precise images associated with LA-ICP-MS imaging to be leveraged to achieve improved segmentation in MALDI-MS images, resulting in the identification of lipids that are most associated with different sub-organ regions in tissues.


Assuntos
Terapia a Laser , Nanoestruturas , Animais , Ouro , Camundongos , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Distribuição Tecidual
11.
Biochemistry ; 59(44): 4294-4301, 2020 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-33135889

RESUMO

Cells rely on protein degradation by AAA+ proteases. A well-known example is the hexameric ClpX unfoldase, which captures ATP hydrolysis to feed substrates into the oligomeric ClpP peptidase. Recent studies show that an asymmetric ClpX spiral cycles protein translocation upon ATP hydrolysis. However, how this cycle affects peptide products is less explored in part because ClpP cleavage is thought to be solely defined by sequence constraints. Here, we comprehensively characterize peptides from Caulobacter crescentus ClpXP degradation of three different substrates using high-resolution mass spectrometry and find that cleavage of translocated substrates is driven by factors other than sequence. We report that defined locations in a translocated protein are especially sensitive to cleavage spaced on average every 10-13 residues. These sites are not exclusively controlled by sequence and are independent of bulk changes in catalytic peptidase sites, ATP hydrolysis, or the efficiency of initial recognition. These results fit a model in which processive translocation through ClpX starts at a specific location in a polypeptide and pauses during reset of the ClpX hexamer after a cycle of translocation. Our work suggests that defined peptides, which could be used as signaling molecules, can be generated from a given substrate by a nonspecific peptidase.


Assuntos
Caulobacter crescentus/enzimologia , Endopeptidase Clp/metabolismo , Proteólise , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Endopeptidase Clp/química , Hidrólise , Modelos Moleculares , Conformação Proteica
12.
Biochemistry ; 59(10): 1093-1103, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32100530

RESUMO

Epigallocatechin-3-gallate (EGCG) is a catechin found in green tea that can inhibit the amyloid formation of a wide variety of proteins. EGCG's ability to prevent or redirect the amyloid formation of so many proteins may reflect a common mechanism of action, and thus, greater molecular-level insight into how it exerts its effect could have broad implications. Here, we investigate the molecular details of EGCG's inhibition of the protein ß-2-microglobulin (ß2m), which forms amyloids in patients undergoing long-term dialysis treatment. Using size-exclusion chromatography and a collection of mass spectrometry-based techniques, we find that EGCG prevents Cu(II)-induced ß2m amyloid formation by diverting the normal progression of preamyloid oligomers toward the formation of spherical, redissolvable aggregates. EGCG exerts its effect by binding with a micromolar affinity (Kd ≈ 5 µM) to the ß2m monomer on the edge of two ß-sheets near the N-terminus. This interaction destabilizes the preamyloid dimer and prevents the formation of a tetramer species previously shown to be essential for Cu(II)-induced ß2m amyloid formation. EGCG's binding at the edge of the ß-sheets in ß2m is consistent with a previous hypothesis that EGCG generally prevents amyloid formation by binding cross-ß-sheet aggregation intermediates.


Assuntos
Amiloide/química , Catequina/análogos & derivados , Microglobulina beta-2/química , Amiloide/metabolismo , Proteínas Amiloidogênicas/química , Amiloidose/metabolismo , Catequina/metabolismo , Catequina/farmacologia , Catequina/fisiologia , Cromatografia em Gel/métodos , Cobre/metabolismo , Humanos , Espectrometria de Massas/métodos , Modelos Moleculares , Conformação Proteica em Folha beta/fisiologia , Multimerização Proteica/efeitos dos fármacos , Microglobulina beta-2/antagonistas & inibidores , Microglobulina beta-2/metabolismo
13.
Anal Chem ; 92(7): 4731-4735, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32159946

RESUMO

Obtaining kinetic and thermodynamic information for protein amyloid formation can yield new insight into the mechanistic details of this biomedically important process. The kinetics of the structural change that initiates the amyloid pathway, however, has been challenging to access for any amyloid protein system. Here, using the protein ß-2-microglobulin (ß2m) as a model, we measure the kinetics and energy barrier associated with an initial amyloidogenic structural change. Using covalent labeling and mass spectrometry, we measure the decrease in solvent accessibility of one of ß2m's Trp residues, which is buried during the initial structural change, as a way to probe the kinetics of this structural change at different temperatures and under different amyloid forming conditions. Our results provide the first-ever measure of the activation barrier for a structural change that initiates the amyloid formation pathway. The results also yield new mechanistic insight into ß2m's amyloidogenic structural change, especially the role of Pro32 isomerization in this reaction.


Assuntos
Termodinâmica , Microglobulina beta-2/química , Humanos , Cinética , Modelos Moleculares , Conformação Proteica
14.
Anal Chem ; 92(9): 6637-6644, 2020 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-32250591

RESUMO

A new covalent labeling (CL) reagent based on an α,ß-unsaturated carbonyl scaffold has been developed for studying protein structure and protein-protein interactions when coupled with mass spectrometry. We show that this new reagent scaffold can react with up to 13 different types of residues on protein surfaces, thereby providing excellent structural resolution. To illustrate the value of this reagent scaffold, it is used to identify the residues involved in the protein-protein interface that is formed upon Zn(II) binding to the protein ß-2-microglobulin. The modular design of the α,ß-unsaturated carbonyl scaffold allows facile variation of the functional groups, enabling labeling kinetics and selectivity to be tuned. Moreover, by introducing isotopically enriched functional groups into the reagent structure, labeling sites can be more easily identified by MS and MS/MS. Overall, this reagent scaffold should be a valuable CL reagent for protein higher order structure characterization by MS.


Assuntos
Aminoácidos/química , Anidrases Carbônicas/química , Lactoglobulinas/química , Mioglobina/química , Microglobulina beta-2/química , Aminoácidos/síntese química , Animais , Bovinos , Cavalos , Humanos , Espectrometria de Massas , Modelos Moleculares , Ligação Proteica , Conformação Proteica
15.
Anal Chem ; 92(2): 2011-2018, 2020 01 21.
Artigo em Inglês | MEDLINE | ID: mdl-31825199

RESUMO

Nanomaterial-based drug delivery vehicles are able to deliver therapeutics in a controlled, targeted manner. Currently, however, there are limited analytical methods that can detect both nanomaterial distributions and their biochemical effects concurrently. In this study, we demonstrate that matrix assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) and laser ablation inductively coupled plasma mass spectrometry imaging (LA-ICP-MSI) can be used together to obtain nanomaterial distributions and biochemical consequences. These studies employ nanoparticle-stabilized capsules (NPSCs) loaded with siRNA as a testbed. MALDI-MSI experiments on spleen tissues from intravenously injected mice indicate that NPSCs loaded with anti-TNF-α siRNA cause changes to the lipid composition in white pulp regions of the spleen, as anticipated, based on pathways known to be affected by TNF-α, whereas NPSCs loaded with scrambled siRNA do not cause the predicted changes. Interestingly, LA-ICP-MSI experiments reveal that the NPSCs primarily localize in the red pulp, suggesting that the observed changes in lipid composition are due to diffusive rather than localized effects on TNF-α production. Such information is only accessible by combining data from the two modalities, which we accomplish by using the heme signals from MALDI-MSI and iron signals from LA-ICP-MSI to overlay the images. Several unexpected changes in lipid composition also occur in regions where the NPSCs are found, suggesting that the NPSCs themselves can influence tissue biochemistry as well.


Assuntos
Cápsulas/análise , Nanopartículas/análise , Baço/química , Animais , Cápsulas/administração & dosagem , Cápsulas/metabolismo , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/análise , Portadores de Fármacos/metabolismo , Injeções Intravenosas , Espectrometria de Massas , Camundongos , Nanopartículas/administração & dosagem , Nanopartículas/metabolismo , Baço/metabolismo , Distribuição Tecidual
16.
Analyst ; 145(10): 3705-3712, 2020 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-32270789

RESUMO

Laser ablation inductively coupled plasma mass spectrometry (LA-ICP-MS) imaging has been extensively used to determine the distributions of metals in biological tissues for a wide variety of applications. To be useful for identifying metal biodistributions, the acquired raw data needs to be reconstructed into a two-dimensional image. Several approaches have been developed for LA-ICP-MS image reconstruction, but less focus has been placed on software for more in-depth statistical processing of the imaging data. Yet, improved image processing can allow the biological ramifications of metal distributions in tissues to be better understood. In this work, we describe software written in Python that automatically reconstructs, analyzes, and segments images from LA-ICP-MS imaging data. Image segmentation is achieved using LA-ICP-MS signals from the biological metals Fe and Zn together with k-means clustering to automatically identify sub-organ regions in different tissues. Spatial awareness also can be incorporated into the images through a neighboring pixel evaluation that allows regions of interest to be identified that are at the limit of the LA-ICP-MS imaging resolution. The value of the described algorithms is demonstrated for LA-ICP-MS images of nanomaterial biodistributions. The developed image reconstruction and processing approach reveals that nanomaterials distribute in different sub-organ regions based on their chemical and physical properties, opening new possibilities for understanding the impact of such nanomaterials in vivo.


Assuntos
Análise de Dados , Processamento de Imagem Assistida por Computador/métodos , Lasers , Espectrometria de Massas , Linguagens de Programação , Animais , Bismuto/química , Feminino , Ouro/química , Nanopartículas Metálicas/química , Camundongos , Camundongos Endogâmicos BALB C , Nanotubos/química , Baço/metabolismo , Sulfetos/química
17.
Anal Bioanal Chem ; 412(5): 1027-1035, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31925489

RESUMO

Human breast milk is an understudied biological fluid that may be useful for early detection of breast cancer. Methods for enriching and detecting biomarkers in human breast milk, however, are not as well-developed as compared with other biological fluids. In this work, we demonstrate a new enrichment method based on polymeric nanoassemblies that is capable of enhancing the mass spectrometry-based detection of peptides and proteins in human breast milk. In this method, positively charged nanoassemblies are used to selectively deplete abundant proteins in milk based on electrostatic interactions, which simplifies the mixture and enhances detection of positively charged peptides and proteins. Negatively charged nanoassemblies are used in a subsequent enrichment step to further enhance the detection and quantification of trace-level peptides and proteins. Together the depletion and enrichment steps allow model biomarkers to be detected at low nM levels, which are close to instrumental limits of detection. This new method not only demonstrates the ability to detect proteins in human breast milk but also provides an alternative approach for targeted protein detection in complex biological matrices. Graphical abstract.


Assuntos
Leite Humano/química , Nanoestruturas/química , Peptídeos/análise , Polímeros/química , Proteínas/análise , Adulto , Sequência de Aminoácidos , Biomarcadores/análise , Cromatografia Líquida/métodos , Eletroforese em Gel de Poliacrilamida , Feminino , Humanos , Limite de Detecção , Proteômica/métodos , Espectrometria de Massas por Ionização por Electrospray/métodos
18.
Anal Chem ; 91(23): 15248-15254, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31664819

RESUMO

Hydrogen-deuterium exchange (HDX) mass spectrometry (MS) and covalent labeling (CL) MS are typically considered to be complementary methods for protein structural analysis, because one probes the protein backbone, while the other probes side chains. For protein-ligand interactions, we demonstrate in this work that the two labeling techniques can provide synergistic structural information about protein-ligand binding when reagents like diethylpyrocarbonate (DEPC) are used for CL because of the differences in the reaction rates of DEPC and HDX. Using three model protein-ligand systems, we show that the slower time scale for DEPC labeling makes it only sensitive to changes in solvent accessibility and insensitive to changes in protein structural fluctuations, whereas HDX is sensitive to changes in both solvent accessibility and structural fluctuations. When used together, the two methods more clearly reveal binding sites and ligand-induced changes to structural fluctuations that are distant from the binding site, which is more comprehensive information than either technique alone can provide. We predict that these two methods will find widespread usage together for more deeply understanding protein-ligand interactions.


Assuntos
Dietil Pirocarbonato/química , Espectrometria de Massa com Troca Hidrogênio-Deutério , Proteínas/química , Ligantes , Modelos Moleculares
19.
Anal Chem ; 91(13): 8516-8523, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31150223

RESUMO

Covalent labeling with mass spectrometry is increasingly being used for the structural analysis of proteins. Diethylpyrocarbonate (DEPC) is a simple to use, commercially available covalent labeling reagent that can readily react with a range of nucleophilic residues in proteins. We find that in intact proteins weakly nucleophilic side chains (Ser, Thr, and Tyr) can be modified by DEPC in addition to other residues such as His, Lys, and Cys, providing very good structural resolution. We hypothesize that the microenvironment around these side chains, as formed by a protein's higher order structure, tunes their reactivity such that they can be labeled. To test this hypothesis, we compare DEPC labeling reactivity of Ser, Thr, and Tyr residues in intact proteins with peptide fragments from the same proteins. Results indicate that these residues almost never react with DEPC in free peptides, supporting the hypothesis that a protein's local microenvironment tunes the reactivity of these residues. From a close examination of the structural features near the reactive residues, we find that nearby hydrophobic residues are essential, suggesting that the enhanced reactivity of certain Ser, Thr, and Tyr residues occurs due to higher local concentrations of DEPC.


Assuntos
Dietil Pirocarbonato/química , Hormônio do Crescimento Humano/análise , Fragmentos de Peptídeos/análise , Ubiquitina/análise , Microglobulina beta-2/análise , Hormônio do Crescimento Humano/química , Humanos , Espectrometria de Massas , Fragmentos de Peptídeos/química , Conformação Proteica , Serina/química , Treonina/química , Tirosina/química , Ubiquitina/química , Microglobulina beta-2/química
20.
Analyst ; 144(21): 6321-6326, 2019 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-31552921

RESUMO

Mass spectrometry (MS)-based analysis of glycoproteins and glycopeptides requires selective separation strategies to eliminate interferences from more abundant non-glycosylated biomolecules. In this work, we describe a two-phase liquid-liquid extraction method using supramolecular polymeric nanoassemblies that can selectively and efficiently enrich glycopeptides for enhanced MS detection. The polymeric nanoassemblies are made selective for glycopeptides via the incorporation of hydrazide functional groups that covalently bind to glycans. The enrichment efficiency is further enhanced via the incorporation of acidic functional groups that lead to a proximity-assisted catalysis of the hydrazide-glycan conjugation reaction. Our results further demonstrate the value of designer supramolecular nanomaterials for the selective enrichment of modified peptides from complicated mixtures.


Assuntos
Glicopeptídeos/análise , Extração Líquido-Líquido/métodos , Nanoestruturas/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Animais , Anticorpos Monoclonais/análise , Anticorpos Monoclonais/química , Armoracia/enzimologia , Bovinos , Glicopeptídeos/química , Peroxidase do Rábano Silvestre/análise , Peroxidase do Rábano Silvestre/química , Hidrazinas/química , Imunoglobulina G/análise , Imunoglobulina G/química , Oxirredução , Fragmentos de Peptídeos/análise , Poliestirenos/química , Proteólise , Soroalbumina Bovina/análise , Soroalbumina Bovina/química , Tripsina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA