Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Vis Exp ; (193)2023 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-37602868

RESUMEN

ARTICLES DISCUSSED: Espino, J. A., Jones, L. M. In vivo hydroxyl radical protein footprinting for the study of protein interactions in Caenorhabditis elegans. Journal of Visualized Experiments. (158), e60910 (2020). Chea, E. E., Rinas, A., Espino, J. A., Jones, L. M. Characterizing cellular proteins with in-cell fast photochemical oxidation of proteins. Journal of Visualized Experiments. (157), e60911 (2020). Moorthy, B. S., Iyer, L. K., Topp, E. M. Mass spectrometric approaches to study protein structure and interactions in lyophilized powders. Journal of Visualized Experiments. (98), e52503 (2015). Habibi, Y., Thibodeaux, C. J. A hydrogen-deuterium exchange mass spectrometry (HDX-MS) platform for investigating peptide biosynthetic enzymes. Journal of Visualized Experiments. (159), e61053 (2020). Kirsch, Z. J., Arden, B. G., Vachet, R. W., Limpikirati, P. Covalent labeling with diethylpyrocarbonate for studying protein higher-order structure by mass spectrometry. Journal of Visualized Experiments. (172), e61983 (2021). Johnson, D., Punshon-Smith, B., Espino, J. A., Gershenson, A., Jones, L. M. Platform incubator with movable XY stage: A new platform for implementing in-cell fast photochemical oxidation of proteins. Journal of Visualized Experiments. (171), e62153 (2021). Haupt, C. et al. Combining chemical cross-linking and mass spectrometry of intact protein complexes to study the architecture of multi-subunit protein assemblies. Journal of Visualized Experiments. (129), e56747 (2017). Lento, C. et al. Time-resolved electrospray ionization hydrogen-deuterium exchange mass spectrometry for studying protein structure and dynamics. Journal of Visualized Experiments. (122), e55464 (2017). Weinberger, S. R., Chea, E. E., Sharp, J. S., Misra, S. K. Laser-free hydroxyl radical protein footprinting to perform higher order structural analysis of proteins. Journal of Visualized Experiments. (172), e61861 (2021). Misra, S. K., Sharp, J. S. Enabling real-time compensation in fast photochemical oxidations of proteins for the determination of protein topography changes. Journal of Visualized Experiments. (163), e61580 (2020). Chaihu, L. et al. Capillary electrophoresis-based hydrogen/deuterium exchange for conformational characterization of proteins with top-down mass spectrometry. Journal of Visualized Experiments. (172), e62672 (2021).


Asunto(s)
Espectrometría de Masas de Intercambio de Hidrógeno-Deuterio , Radical Hidroxilo , Animales , Deuterio , Espectrometría de Masas , Caenorhabditis elegans , Hidrógeno
2.
J Pharm Biomed Anal ; 215: 114754, 2022 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-35427962

RESUMEN

Monoclonal antibody (mAb) has grown to be the major asset in protein therapeutics market since its initial introduction in 1980s. To identify a suitable mAb as a drug candidate for development requires deep understanding of disease biology and attribute sciences, including epitope-paratope binding recognition. Mass spectrometry (MS) has become a critical technology platform in epitope mapping. MS-based approaches utilize chemical labeling to assess the changes of solvent accessibilities during binding interactions, and the labeling can be either reversible or irreversible. Reversible labeling is represented by hydrogen/deuterium exchange (HDX), which probes the changes via exchange between backbone amide hydrogen and deuterium in the solvent. Irreversible labeling targets amino acid residue side chains and involves chemical based labeling such as glycine ethyl ester labeling, radical based labeling such as fast photochemical oxidation of proteins (FPOP), and chemical cross-linking. All these methods have been developed extensively for characterization of binding interface within an immunocomplex. This review covers the fundamentals of these different MS-based methods and highlights recent case studies to illustrate unique capabilities of MS-based approaches in epitope mapping of protein therapeutics.


Asunto(s)
Anticuerpos Monoclonales , Medición de Intercambio de Deuterio , Anticuerpos Monoclonales/química , Deuterio , Medición de Intercambio de Deuterio/métodos , Mapeo Epitopo/métodos , Espectrometría de Masas/métodos , Solventes
3.
Nat Commun ; 12(1): 7270, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34907205

RESUMEN

Mass spectrometry-based footprinting can probe higher order structure of soluble proteins in their native states and serve as a complement to high-resolution approaches. Traditional footprinting approaches, however, are hampered for integral membrane proteins because their transmembrane regions are not accessible to solvent, and they contain hydrophobic residues that are generally unreactive with most chemical reagents. To address this limitation, we bond photocatalytic titanium dioxide (TiO2) nanoparticles to a lipid bilayer. Upon laser irradiation, the nanoparticles produce local concentrations of radicals that penetrate the lipid layer, which is made permeable by a simultaneous laser-initiated Paternò-Büchi reaction. This approach achieves footprinting for integral membrane proteins in liposomes, helps locate both ligand-binding residues in a transporter and ligand-induced conformational changes, and reveals structural aspects of proteins at the flexible unbound state. Overall, this approach proves effective in intramembrane footprinting and forges a connection between material science and biology.


Asunto(s)
Proteínas de la Membrana/química , Nanopartículas/química , Huella de Proteína/métodos , Sitios de Unión , Ligandos , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Liposomas , Espectrometría de Masas , Proteínas de la Membrana/metabolismo , Procesos Fotoquímicos , Conformación Proteica , Especies Reactivas de Oxígeno/química , Titanio/química
4.
Nat Protoc ; 15(12): 3942-3970, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33169002

RESUMEN

The higher-order structure (HOS) of proteins plays a critical role in their function; therefore, it is important to our understanding of their function that we have as much information as possible about their three-dimensional structure and how it changes with time. Mass spectrometry (MS) has become an important tool for determining protein HOS owing to its high throughput, mid-to-high spatial resolution, low sample amount requirement and broad compatibility with various protein systems. Modern MS-based protein HOS analysis relies, in part, on footprinting, where a reagent reacts 'to mark' the solvent-accessible surface of the protein, and MS-enabled proteomic analysis locates the modifications to afford a footprint. Fast photochemical oxidation of proteins (FPOP), first introduced in 2005, has become a powerful approach for protein footprinting. Laser-induced hydrogen peroxide photolysis generates hydroxyl radicals that react with solvent-accessible side chains (14 out of 20 amino acid side chains) to fulfill the footprinting. The reaction takes place at sub-milliseconds, faster than most of labeling-induced protein conformational changes, thus enabling a 'snapshot' of protein HOS in solution. As a result, FPOP has been employed in solving several important problems, including mapping epitopes, following protein aggregation, locating small molecule binding, measuring ligand-binding affinity, monitoring protein folding and unfolding and determining hidden conformational changes invisible to other methods. Broader adoption will be promoted by dissemination of the technical details for assembling the FPOP platform and for dealing with the complexities of analyzing FPOP data. In this protocol, we describe the FPOP platform, the conditions for successful footprinting and its examination by mass measurements of the intact protein, the post-labeling sample handling and digestion, the liquid chromatography-tandem MS analysis of the digested sample and the data analysis with Protein Metrics Suite. This protocol is intended not only as a guide for investigators trying to establish an FPOP platform in their own lab but also for those willing to incorporate FPOP as an additional tool in addressing their questions of interest.


Asunto(s)
Espectrometría de Masas/métodos , Procesos Fotoquímicos , Proteínas/química , Proteínas/metabolismo , Cinética , Modelos Moleculares , Oxidación-Reducción , Conformación Proteica
5.
EMBO J ; 39(18): e105246, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32974937

RESUMEN

Tetraspanins, including CD53 and CD81, regulate a multitude of cellular processes through organizing an interaction network on cell membranes. Here, we report the crystal structure of CD53 in an open conformation poised for partner interaction. The large extracellular domain (EC2) of CD53 protrudes away from the membrane surface and exposes a variable region, which is identified by hydrogen-deuterium exchange as the common interface for CD53 and CD81 to bind partners. The EC2 orientation in CD53 is supported by an extracellular loop (EC1). At the closed conformation of CD81, however, EC2 disengages from EC1 and rotates toward the membrane, thereby preventing partner interaction. Structural simulation shows that EC1-EC2 interaction also supports the open conformation of CD81. Disrupting this interaction in CD81 impairs the accurate glycosylation of its CD19 partner, the target for leukemia immunotherapies. Moreover, EC1 mutations in CD53 prevent the chemotaxis of pre-B cells toward a chemokine that supports B-cell trafficking and homing within the bone marrow, a major CD53 function identified here. Overall, an open conformation is required for tetraspanin-partner interactions to support myriad cellular processes.


Asunto(s)
Movimiento Celular , Células Precursoras de Linfocitos B/metabolismo , Tetraspanina 25 , Tetraspanina 28 , Animales , Antígenos CD19/química , Antígenos CD19/genética , Antígenos CD19/metabolismo , Humanos , Ratones , Ratones Noqueados , Dominios Proteicos , Tetraspanina 25/química , Tetraspanina 25/genética , Tetraspanina 25/metabolismo , Tetraspanina 28/química , Tetraspanina 28/genética , Tetraspanina 28/metabolismo
6.
Blood Adv ; 4(10): 2202-2212, 2020 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-32433747

RESUMEN

Dose control of warfarin is a major complication in anticoagulation therapy and overdose is reversed by the vitamin K antidote. Improving the dosage management and antidotal efficacy requires mechanistic understanding. Here we find that effects of the major predictor of warfarin dosage, SNP -1639 G>A, follow a general correlation that warfarin 50% inhibitory concentration decreases with cellular level of vitamin K epoxide reductase (VKORC1), suggesting stoichiometric inhibition. Characterization of the inhibition kinetics required the use of microsomal VKORC1 with a native reductant, glutathione, that enables effective warfarin inhibition in vitro. The kinetics data can be fitted with the Morrison equation, giving a nanomolar inhibition constant and demonstrating that warfarin is a tight-binding inhibitor. However, warfarin is released from purified VKORC1-warfarin complex with increasing amount of vitamin K, indicating competitive inhibition. The competition occurs also in cells, resulting in rescued VKORC1 activity that augments the antidotal effects of vitamin K. Taken together, warfarin is a competitive inhibitor that binds VKORC1 tightly and inhibits at a stoichiometric (1:1) concentration, whereas exceeding the VKORC1 level results in warfarin overdose. Thus, warfarin dosage control should use VKORC1 level as a major indicator, and improved antidotes may be designed based on their competition with warfarin.


Asunto(s)
Antídotos , Warfarina , Vitamina K Epóxido Reductasas/genética , Warfarina/farmacología
7.
Chem Rev ; 120(10): 4355-4454, 2020 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-32319757

RESUMEN

Proteins adopt different higher-order structures (HOS) to enable their unique biological functions. Understanding the complexities of protein higher-order structures and dynamics requires integrated approaches, where mass spectrometry (MS) is now positioned to play a key role. One of those approaches is protein footprinting. Although the initial demonstration of footprinting was for the HOS determination of protein/nucleic acid binding, the concept was later adapted to MS-based protein HOS analysis, through which different covalent labeling approaches "mark" the solvent accessible surface area (SASA) of proteins to reflect protein HOS. Hydrogen-deuterium exchange (HDX), where deuterium in D2O replaces hydrogen of the backbone amides, is the most common example of footprinting. Its advantage is that the footprint reflects SASA and hydrogen bonding, whereas one drawback is the labeling is reversible. Another example of footprinting is slow irreversible labeling of functional groups on amino acid side chains by targeted reagents with high specificity, probing structural changes at selected sites. A third footprinting approach is by reactions with fast, irreversible labeling species that are highly reactive and footprint broadly several amino acid residue side chains on the time scale of submilliseconds. All of these covalent labeling approaches combine to constitute a problem-solving toolbox that enables mass spectrometry as a valuable tool for HOS elucidation. As there has been a growing need for MS-based protein footprinting in both academia and industry owing to its high throughput capability, prompt availability, and high spatial resolution, we present a summary of the history, descriptions, principles, mechanisms, and applications of these covalent labeling approaches. Moreover, their applications are highlighted according to the biological questions they can answer. This review is intended as a tutorial for MS-based protein HOS elucidation and as a reference for investigators seeking a MS-based tool to address structural questions in protein science.


Asunto(s)
Proteínas/química , Medición de Intercambio de Deuterio , Espectrometría de Masas , Conformación Proteica
8.
ACS Chem Biol ; 15(6): 1473-1486, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32293871

RESUMEN

Class II lanthipeptides belong to a diverse group of natural products known as ribosomally synthesized and post-translationally modified peptides (RiPPs). Most RiPP precursor peptides contain an N-terminal recognition sequence known as the leader peptide, which is typically recognized by biosynthetic enzymes that catalyze modifications on the C-terminal core peptide. For class II lanthipeptides, these are carried out by a bifunctional lanthipeptide synthetase (LanM) that catalyzes dehydration and cyclization reactions on peptidic substrates to generate thioether-containing, macrocyclic molecules. Some lanthipeptide synthetases are extraordinarily substrate tolerant, making them promising candidates for biotechnological applications such as combinatorial biosynthesis and cyclic peptide library construction. In this study, we characterized the mode of leader peptide recognition by HalM2, the lanthipeptide synthetase responsible for the production of the antimicrobial peptide haloduracin ß. Using NMR spectroscopic techniques, in vitro binding assays, and enzyme activity assays, we identified substrate residues that are important for binding to HalM2 and for post-translational modification of the peptide substrates. Additionally, we provide evidence of the binding site on the enzyme using binding assays with truncated enzyme variants, hydrogen-deuterium exchange mass spectrometry, and photoaffinity labeling. Understanding the mechanism by which lanthipeptide synthetases recognize their substrate will facilitate their use in biotechnology, as well as further our general understanding of how RiPP enzymes recognize their substrates.


Asunto(s)
Péptido Sintasas/metabolismo , Secuencia de Aminoácidos , Ciclización , Resonancia Magnética Nuclear Biomolecular , Péptido Sintasas/química , Procesamiento Proteico-Postraduccional , Especificidad por Sustrato
9.
Anal Chem ; 91(19): 12560-12567, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31487155

RESUMEN

Signaling proteins exemplified by calmodulin usually bind cooperatively to multiple ligands. Intermediate states and allosteric behavior are difficult to characterize. Here we extend a recently reported mass spectrometry (MS)-based method named LITPOMS (ligand titration, fast photochemical oxidation of proteins and mass spectrometry) that characterizes complex binding systems typically found as signaling proteins. As reported previously, calmodulin's response to binding four Ca2+ can be determined by LITPOMS to reveal binding sites, binding order, and most importantly composite binding behavior. Modeling this behavior provides site-specific binding affinities. In this article, we dissect the composite, peptide-level conformational changes at several regions either by digestion with a different protease or by tandem MS of LITPOMS behavior at the amino-acid residue level. Such dissection greatly elevates spatial resolution and increases the confidence of binding-order assignment. These complementary views of complex protein conformational change recapitulate the cumulative understanding via a single approach, providing new insights on poorly understood yet important allostery and underpin an approach applicable for exploring other signaling systems.


Asunto(s)
Calcio/metabolismo , Calcio/farmacología , Calmodulina/química , Calmodulina/metabolismo , Ligandos , Modelos Moleculares , Unión Proteica , Conformación Proteica/efectos de los fármacos , Proteolisis
10.
Anal Chem ; 91(14): 9238-9245, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31241913

RESUMEN

Fast photochemical oxidation of protein (FPOP) has become an important mass spectrometry-based protein footprinting approach. Although the hydroxyl radical (•OH) generated by photolysis of hydrogen peroxide (H2O2) is most commonly used, the pathways for its reaction with amino-acid side chains remain unclear. Here, we report a systematic study of •OH oxidative modification of 13 amino acid residues by using 18O isotopic labeling. The results differentiate three classes of residues on the basis of their oxygen uptake preference toward different oxygen sources. Histidine, arginine, tyrosine, and phenylalanine residues preferentially take oxygen from H2O2. Methionine residues competitively take oxygen from H2O2 and dissolved oxygen (O2), whereas the remaining residues take oxygen exclusively from O2. Results reported in this work deepen the understanding of •OH labeling pathway on a FPOP platform, opening new possibilities for tailoring FPOP conditions in addressing many biological questions in a profound way.


Asunto(s)
Marcaje Isotópico/métodos , Isótopos de Oxígeno/química , Fragmentos de Péptidos/química , Albúmina Sérica Bovina/química , Aminoácidos/química , Animales , Bovinos , Peróxido de Hidrógeno/química , Peróxido de Hidrógeno/efectos de la radiación , Radical Hidroxilo/química , Oxidación-Reducción , Oxígeno/química , Oxígeno/efectos de la radiación , Fotólisis , Huella de Proteína/métodos , Rayos Ultravioleta
11.
Anal Chem ; 91(9): 5508-5512, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30963760

RESUMEN

We found that a newly developed method named LITPOMS (ligand titration, fast photochemical oxidation of proteins and mass spectrometry) can characterize section-by-section of a protein the conformational changes induced by metal-ion binding. Peptide-level LITPOMS applied to Ca2+ binding to calmodulin reveals binding order and site-specific affinity, providing new insights on the behavior of proteins upon binding Ca2+. We established that EF hand-4 (EF-4) binds calcium first, followed by EF-3, EF-2, and EF-1 and determined the four affinity constants by modeling the extent-of-modification curves. We also found positive cooperativity between EF-4, EF-3 and EF-2, EF-1 and allostery involving the four EF-hands. LITPOMS recapitulates via one approach the calcium-calmodulin binding that required decades of sophisticated development to afford versatility, comprehensiveness, and outstanding spatial resolution.


Asunto(s)
Calcio/metabolismo , Calmodulina/metabolismo , Calmodulina/química , Modelos Moleculares , Unión Proteica , Conformación Proteica
12.
J Am Soc Mass Spectrom ; 30(2): 213-217, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30484077

RESUMEN

We report a novel method named LITPOMS (ligand titration, fast photochemical oxidation of proteins and mass spectrometry) to characterize protein-ligand binding stoichiometry, binding sites, and site-specific binding constants. The system used to test the method is melittin-calmodulin, in which the peptide melittin binds to calcium-bound calmodulin. Global-level measurements reveal the binding stoichiometry of 1:1 whereas peptide-level data coupled with fitting reveal the binding sites and the site-specific binding affinity. Moreover, we extended the analysis to the residue level and identified six critical binding residues. The results show that melittin binds to the N-terminal, central linker, and C-terminal regions of holo-calmodulin with an affinity of 4.6 nM, in agreement with results of previous studies. LITPOMS, for the first time, brings high residue-level resolution to affinity measurements, providing simultaneously qualitative and quantitative understanding of protein-ligand binding. The approach can be expanded to other binding systems without tagging the protein to give high spatial resolution. Graphical Abstract.


Asunto(s)
Espectrometría de Masas/métodos , Fotoquímica/métodos , Proteínas/análisis , Proteínas/metabolismo , Sitios de Unión , Calcio/metabolismo , Calmodulina/metabolismo , Ligandos , Meliteno/metabolismo , Oxidación-Reducción , Proteínas/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...