Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Am Chem Soc ; 142(14): 6750-6760, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32203657

RESUMO

Post-translational modifications create a diverse mixture of proteoforms, leading to substantial challenges in linking proteomic information to disease. Top-down sequencing of intact proteins and multiprotein complexes offers significant advantages in proteoform analysis, but achieving complete fragmentation for such precursor ions remains challenging. Intact proteins that undergo slow-heating generally fragment via charge directed (i.e., mobile proton) or charge remote fragmentation pathways. Our efforts seek to alter this paradigm by labeling proteins with trimethyl pyrylium (TMP), which forms a stable, positively charged label at lysine residues. Fixing positive charges to the protein sequence reduces the availability of mobile protons, driving fragmentation to charge remote channels. Furthermore, we demonstrate that capping acidic side chains with carbodiimide chemistry obstructs this pathway, restoring charge-directed fragmentation and resulting in more even coverage of the protein sequence. With large amounts of fixed charge and few mobile protons, we demonstrate that it is also possible to direct fragmentation almost exclusively to lysine residues containing the charged label. Finally, our data indicate that when electrosprayed under native conditions, protein ions possess an immense capacity to accommodate excess positive charge. Molecular dynamics simulations of such ions bearing intrinsically charged labels reveal evidence of numerous charge solvation processes, including the preferential formation of helices that solvate charged labels through interactions with their macro-dipoles. Thus, these studies reveal the extent to which intact gas-phase protein ions are capable of solvating charge, and provide the most complete indication to date of the extensive physical forces opposing the goal of comprehensive top-down protein sequencing.


Assuntos
Íons/química , Simulação de Dinâmica Molecular/normas , Proteínas/química , Proteômica/métodos , Sequência de Aminoácidos
2.
Anal Chem ; 91(4): 3147-3155, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30668913

RESUMO

Ion mobility-mass spectrometry (IM-MS) has become an important addition to the structural biology toolbox, but separating closely related protein conformations remain challenging. Collision-induced unfolding (CIU) has emerged as a valuable technique for distinguishing iso-cross-sectional protein and protein complex ions through their distinct unfolding pathways in the gas phase. The speed and sensitivity of CIU analyses, coupled with their information-rich data sets, have resulted in the rapid growth of CIU for applications, ranging from the structural assessment of protein complexes to the characterization of biotherapeutics. This growth has occurred despite a lag in the capabilities of informatics tools available to process the complex data sets generated by CIU experiments, resulting in laborious manual analysis remaining commonplace. Here, we present CIUSuite 2, a software suite designed to enable robust, automated analysis of CIU data across the complete range of current CIU applications and to support the implementation of CIU as a true high-throughput technique. CIUSuite 2 uses statistical fitting and modeling methods to reliably quantify features of interest within CIU data sets, particularly in data with poor signal quality that cannot be interpreted with existing analysis tools. By reducing the signal-to-noise requirements for handling CIU data, we are able to demonstrate reductions in acquisition time of up to 2 orders of magnitude over current workflows. CIUSuite 2 also provides the first automated system for classifying CIU fingerprints, enabling the next generation of ligand screening and structural analysis experiments to be accomplished in a high-throughput fashion.


Assuntos
Proteínas/análise , Software , Gases/análise , Humanos , Espectrometria de Massas , Modelos Moleculares , Conformação Proteica , Desdobramento de Proteína
3.
Anal Chem ; 91(16): 10407-10412, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31310505

RESUMO

Collision-induced unfolding (CIU) has emerged as a valuable method for distinguishing iso-cross-sectional protein ions through their distinct gas-phase unfolding trajectories. CIU shows promise as a high-throughput, structure-sensitive screening technique with potential applications in drug discovery and biotherapeutic characterization. We recently developed a CIU classification workflow to support screening applications that utilized CIU data acquired from a single protein charge state to distinguish immunoglobulin (IgG) subtypes and membrane protein lipid binding. However, distinguishing highly similar protein structures, such as those associated with biotherapeutics, can be challenging. Here, we present an expansion of this classification method that includes CIU data from multiple charge states, or indeed any perturbation to protein structure that differentially affects CIU, into a combined classifier. Using this improved method, we are able to improve the accuracy of existing, single-state classifiers for IgG subtypes and develop an activation-state-sensitive classifier for selected Src kinase inhibitors when data from a single charge state was insufficient to do so. Finally, we employ the combination of multiple charge states and stress conditions to distinguish a highly similar innovator/biosimilar biotherapeutic pair, demonstrating the potential of CIU as a rapid screening tool for drug discovery and biotherapeutic analysis.


Assuntos
Anilidas/química , Dasatinibe/química , Imidazóis/química , Isotipos de Imunoglobulinas/isolamento & purificação , Inibidores de Proteínas Quinases/química , Piridazinas/química , Quinolinas/química , Estaurosporina/química , Quinases da Família src/isolamento & purificação , Algoritmos , Anilidas/farmacologia , Dasatinibe/farmacologia , Escherichia coli/genética , Escherichia coli/metabolismo , Ensaios de Triagem em Larga Escala , Humanos , Imidazóis/farmacologia , Isotipos de Imunoglobulinas/química , Isotipos de Imunoglobulinas/classificação , Isotipos de Imunoglobulinas/imunologia , Modelos Moleculares , Mieloma Múltiplo/química , Inibidores de Proteínas Quinases/farmacologia , Estrutura Secundária de Proteína , Piridazinas/farmacologia , Quinolinas/farmacologia , Proteínas Recombinantes/análise , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Eletricidade Estática , Estaurosporina/farmacologia , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/genética , Quinases da Família src/metabolismo
4.
Anal Chem ; 91(24): 15469-15476, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31743004

RESUMO

Membrane proteins represent most current therapeutic targets, yet remain understudied due to their insolubility in aqueous solvents and generally low yields during purification and expression. Ion mobility-mass spectrometry and collision induced unfolding experiments have recently garnered attention as methods capable of directly detecting and quantifying ligand binding within a wide range of membrane protein systems. Despite prior success, ionized surfactant often creates chemical noise patterns resulting in significant challenges surrounding the study of small membrane protein-ligand complexes. Here, we present a new data analysis workflow that overcomes such chemical noise and then utilize this approach to quantify and classify ligand binding associated with the 36 kDa dimer of translocator protein (TSPO). Following our denoising protocol, we detect separate gas-phase unfolding signatures for lipid and protoporphyrin TSPO binders, molecular classes that likely interact with separate regions of the protein surface. Further, a detailed classification analysis reveals that lipid alkyl chain saturation levels can be detected within our gas-phase protein unfolding data. We combine these data and classification schemes with mass spectra acquired directly from liquid-liquid extracts to propose an identity for a previously unknown endogenous TSPO ligand.


Assuntos
Proteínas de Membrana/química , Desdobramento de Proteína , Rhodobacter sphaeroides/química , Sítios de Ligação , Proteínas de Transporte , Ligantes , Mutação Puntual , Porfirinas/química , Porfirinas/metabolismo , Conformação Proteica
5.
Anal Chem ; 89(11): 5669-5672, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28471653

RESUMO

High mass accuracy, data-dependent acquisition is the current standard method in mass spectrometry-based peptide annotation and quantification. In high complexity samples, limited instrument scan speeds often result in under-sampling. In contrast, all-ion data-independent acquisition methods bypass precursor selection, alternating high and low collision energies to analyze product and precursor ions across wide mass ranges. Despite capturing data for all events, peptide annotation is limited by inadequate alignment algorithms or overlapping ions. Ion mobility separation can add an orthogonal analytical dimension, reducing ion interference to improve reproducibility, peak capacity, and peptide identifications to rival modern hybrid quadrupole orbitrap systems. Despite the advantages of ion mobility separation in complex proteomics analyses, there has been no quantitative measure of ion mobility resolution in a complex proteomic sample. Here, we present TWIMExtract, a data extraction tool to export defined slices of liquid chromatography/ion mobility/mass spectrometry (LC-IM-MS) data, providing a route to quantify ion mobility resolution from a commercial traveling-wave ion mobility time-of-flight mass spectrometer. Using standard traveling-wave ion mobility parameters (600 m/s, 40 V), 90% of the annotated peptides occupied just 23% of the ion mobility drift space, yet inclusion of ion mobility nearly doubled the overall peak capacity. Relative to fixed velocity traveling-wave ion mobility settings, ramping the traveling-wave velocity increased drift space occupancy, amplifying resolution by 16%, peak capacity by nearly 50%, and peptide/protein identifications by 40%. Overall, variable-velocity traveling-wave ion mobility-mass spectrometry significantly enhances proteomics analysis in all-ion fragmentation acquisition.


Assuntos
Espectrometria de Mobilidade Iônica/métodos , Proteômica/métodos , Cromatografia Líquida , Células HeLa , Humanos , Peptídeos/análise , Proteínas/análise , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem
6.
J Am Soc Mass Spectrom ; 35(4): 784-792, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38489759

RESUMO

We previously discovered that electron attachment to gaseous peptide anions can occur within a relatively narrow electron energy range. The resulting charge-increased radical ions undergo dissociation analogous to conventional cation electron capture/transfer dissociation (ECD/ETD), thus enabling a novel tandem mass spectrometry (MS/MS) technique that we termed negative ion electron capture dissociation (niECD). We proposed that gaseous zwitterionic structures are required for niECD with electron capture either occurring at or being directed by a positively charged site. Here, we further evaluate this zwitterion mechanism by performing niECD of peptides derivatized to alter their ability to form zwitterionic gaseous structures. Introduction of a fixed positive charge tag, a highly basic guanidino group, or a highly acidic sulfonate group to promote zwitterionic structures in singly charged anions, rescued the niECD ability of a peptide refractory to niECD in its unmodified form. We also performed a systematic study of five sets of synthetic peptides with decreasing zwitterion propensity and found that niECD efficiency decreased accordingly, further supporting the zwitterion mechanism. However, traveling-wave ion mobility-mass spectrometry experiments, performed to gain further insight into the gas-phase structures of peptides showing high niECD efficiency, exhibited an inverse correlation between the orientationally averaged collision cross sections and niECD efficiency. These results indicate that compact salt-bridged structures are also a requirement for effective niECD.


Assuntos
Gases , Espectrometria de Massas em Tandem , Gases/química , Espectrometria de Massas em Tandem/métodos , Elétrons , Ânions/química , Peptídeos/química , Cátions , Cloreto de Sódio
7.
J Am Soc Mass Spectrom ; 31(4): 880-887, 2020 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-32134265

RESUMO

The widespread use of traveling wave ion mobility (TWIM) technology in fields such as omics and structural biology motivates efforts to deepen our understanding of ion transport within such devices. Here, we describe a new advancement in TWIM theory, where pseudo-trapping within TW ion guides is characterized in detail. During pseudo-trapping, ions with different mobilities can travel with the same mean velocity, leaving others within the same TWIM experiment to separate as normal. Furthermore, pseudo-trapping limits typical band broadening experienced by ions during TWIM, manifesting as peaks with apparently improved IM resolving power, but all ions that undergo pseudo trapping are unable to separate by IM. SIMION simulations show that ions become locked into a repeated pattern of motion with respect to the TW reference frame during pseudo-trapping. We developed a simplified model capable of reproducing TW pseudo-trapping and reproducing trends observed in experimental data. Our model and simulations suggest that pseudo-trapping occurs only during experiments performed under static TWIM conditions, to an extent that depends on the detailed shape of the traveling wave. We show that pseudo-trapping alters the ion transit times and can adversely affect calibrated CCS measurements. Finally, we provide recommendations for avoiding unintentional pseudo-trapping in TWIM in order to obtain optimal separations and CCS determinations.


Assuntos
Espectrometria de Mobilidade Iônica/métodos , Modelos Químicos , Proteínas/química , Citocromos c/química , Íons/química
8.
J Am Soc Mass Spectrom ; 30(6): 956-966, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30815838

RESUMO

The inherent structural heterogeneity of biomolecules is an important biophysical property that is essential to their function, but is challenging to characterize experimentally. We present a workflow that rapidly and quantitatively assesses the conformational heterogeneity of peptides and proteins in the gas phase using traveling wave ion mobility (TWIM) arrival time distributions (ATDs). We have established a set of semi-empirical equations that model the TWIM ATD peak width and resolution across a wide range of wave amplitudes (V) and wave velocities (v). In addition, a conformational broadening parameter, δ, can be extracted from this analysis that reports on the contribution of conformational heterogeneity to the broadening of TWIM ATD peak width during ion mobility separation. We use this δ value to evaluate the conformational heterogeneity of a set of helical peptides, and our analysis correlates well with previous peak width observations reported for these ions. Furthermore, we use molecular dynamics simulations to independently investigate the general flexibility of these peptides in the gas phase, and generate similar trends found in experimental TWIM data. Finally, we extended our analysis to Avidin, a 64-kDa homotetramer, and quantify the structural heterogeneity of this intact complex using TWIM ATD data as a function of cross-linking. We observe an initial reduction in δ values as a function of cross-linker concentration, demonstrating the sensitivity of our δ value analysis to changes in flexibility of the assembly.

9.
Curr Opin Chem Biol ; 42: 93-100, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29207278

RESUMO

Rapidly characterizing the three-dimensional structures of proteins and the multimeric machines they form remains one of the great challenges facing modern biological and medical sciences. Ion mobility-mass spectrometry based techniques are playing an expanding role in characterizing these functional complexes, especially in drug discovery and development workflows. Despite this expansion, ion mobility-mass spectrometry faces many challenges, especially in the context of detecting small differences in protein tertiary structure that bear functional consequences. Collision induced unfolding is an ion mobility-mass spectrometry method that enables the rapid differentiation of subtly-different protein isoforms based on their unfolding patterns and stabilities. In this review, we summarize the modern implementation of such gas-phase unfolding experiments and provide an overview of recent developments in both methods and applications.


Assuntos
Gases/química , Espectrometria de Mobilidade Iônica/métodos , Espectrometria de Massas/métodos , Isoformas de Proteínas/química , Proteínas/química , Produtos Biológicos/análise , Produtos Biológicos/uso terapêutico , Descoberta de Drogas , Ensaios de Triagem em Larga Escala , Conformação Proteica , Estrutura Secundária de Proteína , Desdobramento de Proteína
10.
Structure ; 26(4): 599-606.e3, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29576318

RESUMO

The synthesis of active Klebsiella aerogenes urease via an 18-subunit enzyme apoprotein-accessory protein pre-activation complex has been well studied biochemically, but thus far this complex has remained refractory to direct structural characterization. Using ion mobility-mass spectrometry, we characterized several protein complexes between the core urease apoprotein and its accessory proteins, including the 610-kDa (UreABC)3(UreDFG)3 complex. Using our recently developed computational modeling workflow, we generated ensembles of putative (UreABC)3(UreDFG)3 species consistent with experimental restraints and characterized the structural ambiguity present in these models. By integrating structural information from previous studies, we increased the resolution of the ion mobility-mass spectrometry-derived models substantially, and we observe a discrete population of structures consistent with all of the available data for this complex.


Assuntos
Proteínas de Bactérias/química , Enterobacter aerogenes/química , Subunidades Proteicas/química , Urease/química , Motivos de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação , Clonagem Molecular , Enterobacter aerogenes/enzimologia , Ativação Enzimática , Escherichia coli/genética , Escherichia coli/metabolismo , Expressão Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Espectrometria de Mobilidade Iônica , Cinética , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homologia Estrutural de Proteína , Urease/genética , Urease/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA