Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Vis Exp ; (172)2021 06 04.
Article in English | MEDLINE | ID: mdl-34152327

ABSTRACT

Hydroxyl Radical Protein Footprinting (HRPF) is an emerging and promising higher order structural analysis technique that provides information on changes in protein structure, protein-protein interactions, or protein-ligand interactions. HRPF utilizes hydroxyl radicals (▪OH) to irreversibly label a protein's solvent accessible surface. The inherent complexity, cost, and hazardous nature of performing HRPF have substantially limited broad-based adoption in biopharma. These factors include: 1) the use of complicated, dangerous, and expensive lasers that demand substantial safety precautions; and 2) the irreproducibility of HRPF caused by background scavenging of ▪OH that limit comparative studies. This publication provides a protocol for operation of a laser-free HRPF system. This laser-free HRPF system utilizes a high energy, high-pressure plasma light source flash oxidation technology with in-line radical dosimetry. The plasma light source is safer, easier to use, and more efficient in generating hydroxyl radicals than laser-based HRPF systems, and the in-line radical dosimeter increases the reproducibility of studies. Combined, the laser-free HRPF system addresses and surmounts the mentioned shortcomings and limitations of laser-based techniques.


Subject(s)
Hydroxyl Radical , Protein Footprinting , Lasers , Oxidation-Reduction , Proteins , Reproducibility of Results
2.
J Am Soc Mass Spectrom ; 32(7): 1601-1609, 2021 Jul 07.
Article in English | MEDLINE | ID: mdl-33872496

ABSTRACT

Hydroxyl radical protein footprinting (HRPF) is a powerful and flexible technique for probing changes in protein topography. With the development of the fast photochemical oxidation of proteins (FPOP), it became possible for researchers to perform HRPF in their laboratory on a very short time scale. While FPOP has grown significantly in popularity since its inception, adoption remains limited due to technical and safety issues involved in the operation of a hazardous Class IV UV laser and irreproducibility often caused by improper laser operation and/or differential radical scavenging by various sample components. Here, we present a new integrated FOX (Flash OXidation) Protein Footprinting System. This platform delivers sample via flow injection to a facile and safe-to-use high-pressure flash lamp with a flash duration of 10 µs fwhm. Integrated optics collect the radiant light and focus it into the lumen of a capillary flow cell. An inline radical dosimeter measures the hydroxyl radical dose delivered and allows for real-time compensation for differential radical scavenging. A programmable fraction collector collects and quenches only the sample that received the desired effective hydroxyl radical dose, diverting the carrier liquid and improperly oxidized sample to waste. We demonstrate the utility of the FOX Protein Footprinting System by determining the epitope of TNFα recognized by adalimumab. We successfully identify the surface of the protein that serves as the epitope for adalimumab, identifying four of the five regions previously noted by X-ray crystallography while seeing no changes in peptides not involved in the epitope interface. The FOX Protein Footprinting System allows for FPOP-like experiments with real-time dosimetry in a safe, compact, and integrated benchtop platform.


Subject(s)
Protein Footprinting/instrumentation , Protein Footprinting/methods , Chromatography, Liquid , Epitopes/chemistry , Equipment Design , HEK293 Cells , Humans , Oxidation-Reduction , Protein Conformation , Tandem Mass Spectrometry , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/chemistry , Tumor Necrosis Factor-alpha/genetics
3.
J Am Soc Mass Spectrom ; 31(2): 169-172, 2020 Feb 05.
Article in English | MEDLINE | ID: mdl-32031409

ABSTRACT

Fast photochemical oxidation of proteins (FPOP) is a powerful covalent labeling tool that uses hydroxyl radicals generated by laser flash photolysis of hydrogen peroxide to footprint protein surfaces. Because radical production varies with many experimental parameters, hydroxyl radical dosimeters have been introduced to track the effective radical dosage experienced by the protein analyte. FPOP experiments performed using adenine optical radical dosimetry containing protein in Tris buffer demonstrated unusual dosimetry behavior. We have investigated the behavior of Tris under oxidative conditions in detail. We find that Tris can act as a novel gain-of-signal optical hydroxyl radical dosimeter in FPOP experiments. This new dosimeter is also amenable to inline real-time monitoring, thereby allowing real-time adjustments to compensate for differences in samples for their quenching ability.


Subject(s)
Hydroxyl Radical/analysis , Hydroxyl Radical/chemistry , Mass Spectrometry/methods , Tromethamine/chemistry , Oxidation-Reduction , Photolysis , Proteins/analysis , Proteins/chemistry , Proteomics/methods
4.
AAPS J ; 21(5): 87, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31297623

ABSTRACT

Unlike small molecule drugs, therapeutic proteins must maintain the proper higher-order structure (HOS) in order to maintain safety and efficacy. Due to the sensitivity of many protein systems, even small changes due to differences in protein expression or formulation can alter HOS. Previous work has demonstrated how hydroxyl radical protein footprinting (HRPF) can sensitively detect changes in protein HOS by measuring the average topography of the protein monomers, as well as identify specific regions of the therapeutic protein impacted by the conformational changes. However, HRPF is very sensitive to the radical scavenging capacity of the buffer; addition of organic buffers and/or excipients can dramatically alter the HRPF footprint without affecting protein HOS. By compensating for the radical scavenging effects of different adalimumab biosimilar formulations using real-time adenine dosimetry, we identify that sodium citrate buffer causes a modest decrease in average solvent accessibility compared to sodium phosphate buffer at the same pH. We find that the addition of polysorbate 80 does not alter the conformation of the biosimilar in either buffer, but it does provide substantial protection from protein conformational perturbation during short periods of exposure to high temperature. Compensated HRPF measurements are validated and contextualized by dynamic light scattering (DLS), which suggests that changes in adalimumab biosimilar aggregation are major drivers in measured changes in protein topography. Overall, compensated HRPF accurately measured conformational changes in adalimumab biosimilar that occurred during formulation changes and identified the effect of formulation changes on protection of HOS from temperature extremes.


Subject(s)
Adalimumab/administration & dosage , Biosimilar Pharmaceuticals/administration & dosage , Excipients/chemistry , Protein Footprinting/methods , Adalimumab/chemistry , Biosimilar Pharmaceuticals/chemistry , Buffers , Dynamic Light Scattering , Hydrogen-Ion Concentration , Hydroxyl Radical/chemistry , Phosphates/chemistry , Polysorbates/chemistry , Protein Conformation , Sodium Citrate/chemistry , Temperature
5.
Anal Chem ; 90(21): 12625-12630, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30290117

ABSTRACT

Hydroxyl radical protein footprinting (HRPF) is a powerful method for measuring protein topography, allowing researchers to monitor events that alter the solvent accessible surface of a protein (e.g., ligand binding, aggregation, conformational changes, etc.) by measuring changes in the apparent rate of reaction of portions of the protein to hydroxyl radicals diffusing in solution. Fast Photochemical Oxidation of Proteins (FPOP) offers an ultrafast benchtop method for radical generation for HRPF, photolyzing hydrogen peroxide using a UV laser to generate high concentrations of hydroxyl radicals that are consumed on roughly a microsecond time scale. The broad reactivity of hydroxyl radicals means that almost anything added to the solution (e.g., ligands, buffers, excipients, etc.) will scavenge hydroxyl radicals, altering their half-life and changing the effective radical concentration experienced by the protein. Similarly, minute changes in peroxide concentration, laser fluence, and buffer composition can alter the effective radical concentration, making reproduction of data challenging. Here, we present a simple method for radical dosimetry that can be carried out as part of the FPOP workflow, allowing for measurement of effective radical concentration in real time. Additionally, by modulating the amount of radical generated, we demonstrate that effective hydroxyl radical yields in FPOP HRPF experiments carried out in buffers with widely differing levels of hydroxyl radical scavenging capacity can be compensated on the fly, yielding statistically indistinguishable results for the same conformer. This method represents a major step in transforming FPOP into a robust and reproducible technology capable of probing protein structure in a wide variety of contexts.


Subject(s)
Adenine/chemistry , Fibrinopeptide B/chemistry , Hydroxyl Radical/chemistry , Myoglobin/chemistry , Protein Footprinting/methods , Adenine/analysis , Hydroxyl Radical/radiation effects , Oxidation-Reduction , Spectrophotometry, Ultraviolet , Ultraviolet Rays
6.
J Am Chem Soc ; 132(31): 10620-2, 2010 Aug 11.
Article in English | MEDLINE | ID: mdl-20681677

ABSTRACT

The dissociation constant K(d) of the photosystem I (PSI):ferredoxin complex has been measured by backscattering interferometry (BSI) with cyanobacterial PSI (350 kDa) and ferredoxin (10.5 kDa). The BSI signal, consisting of shifts for interference fringes resulting from a change in refractive index due to complex formation, was monitored as ferredoxin concentration was titrated. K(d) values of 0.14-0.38 microM were obtained with wild-type PSI whereas no complex was detectable with a PSI mutant containing a single mutation (R39Q) in the PsaE extrinsic subunit. These results are in quantitative agreement with previous functional determinations consisting in the detection of fast electron transfer within the complex. They provide evidence that the main contribution for the high affinity binding of ferredoxin to PSI is due to a single region of PsaE comprising arginine 39. They do not support the existence of a secondary binding site that could have escaped functional detection.


Subject(s)
Ferredoxins/chemistry , Photosystem I Protein Complex/chemistry , Cyanobacteria/chemistry , Cyanobacteria/genetics , Models, Molecular , Mutation
7.
Expert Rev Proteomics ; 2(6): 847-62, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16307515

ABSTRACT

Protein expression profiling is increasingly being used to discover, validate and characterize biomarkers that can potentially be used for diagnostic purposes and to aid in pharmaceutical development. Correct analysis of data obtained from these experiments requires an understanding of the underlying analytic procedures used to obtain the data, statistical principles underlying high-dimensional data and clinical statistical tools used to determine the utility of the interpreted data. This review summarizes each of these steps, with the goal of providing the nonstatistician proteomics researcher with a working understanding of the various approaches that may be used by statisticians. Emphasis is placed on the process of mining high-dimensional data to identify a specific set of biomarkers that may be used in a diagnostic or other assay setting.


Subject(s)
Computational Biology/methods , Disease , Proteins/metabolism , Proteomics/methods , Electrophoresis, Gel, Two-Dimensional , Humans , Mass Spectrometry , Proteins/chemistry
8.
Int J Cancer ; 115(5): 783-9, 2005 Jul 10.
Article in English | MEDLINE | ID: mdl-15704152

ABSTRACT

Protein expression profiling has been increasingly used to discover and characterize biomarkers that can be used for diagnostic, prognostic or therapeutic purposes. Most proteomic studies published to date have identified relatively abundant host response proteins as candidate biomarkers, which are often dismissed because of an apparent lack of specificity. We demonstrate that 2 host response proteins previously identified as candidate markers for early stage ovarian cancer, transthyretin and inter-alpha trypsin inhibitor heavy chain 4 (ITIH4), are posttranslationally modified. These modifications include proteolytic truncation, cysteinylation and glutathionylation. Assays using Surface Enhanced Laser Desorption/Ionization Time of Flight Mass Spectrometry (SELDI-TOF-MS) may provide a means to confer specificity to these proteins because of their ability to detect and quantitate multiple posttranslationally modified forms of these proteins in a single assay. Quantitative measurements of these modifications using chromatographic and antibody-based ProteinChip array assays reveal that these posttranslational modifications occur to different extents in different cancers and that multivariate analysis permits the derivation of algorithms to improve the classification of these cancers. We have termed this process host response protein amplification cascade (HRPAC), since the process of synthesis, posttranslational modification and metabolism of host response proteins amplifies the signal of potentially low-abundant biologically active disease markers such as enzymes.


Subject(s)
Algorithms , Biomarkers, Tumor/analysis , Breast Neoplasms/classification , Breast Neoplasms/diagnosis , Colonic Neoplasms/classification , Colonic Neoplasms/diagnosis , Inflammation , Ovarian Neoplasms/classification , Ovarian Neoplasms/diagnosis , Prostatic Neoplasms/classification , Prostatic Neoplasms/diagnosis , Protein Array Analysis , Proteomics , Alpha-Globulins/analysis , Alpha-Globulins/biosynthesis , Breast Neoplasms/immunology , Colonic Neoplasms/immunology , Female , Humans , Male , Ovarian Neoplasms/immunology , Prealbumin/analysis , Prealbumin/biosynthesis , Prostatic Neoplasms/immunology , Protein Processing, Post-Translational , Sensitivity and Specificity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
9.
Mass Spectrom Rev ; 23(1): 34-44, 2004.
Article in English | MEDLINE | ID: mdl-14625891

ABSTRACT

The overall history and recent advancements in Surface-Enhanced Laser Desorption/Ionization (SELDI) affinity technology is reviewed. A detailed account of SELDI technology, utilizing Immobilized-Metal Affinity surfaces, pseudo-specific chromatographic surfaces, and biospecific interactive surfaces, is presented with particular emphasis placed upon examination of fundamental characteristics as well as specific applications for each. Finally, a detailed review of the specific use of such affinity surfaces in fundamental aspects of clinical, process, and research proteomics activity is presented.


Subject(s)
Molecular Probes/chemistry , Protein Array Analysis/methods , Protein Array Analysis/trends , Proteins/analysis , Proteins/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/trends , Molecular Probe Techniques/trends , Molecular Probes/chemical synthesis
11.
Electrophoresis ; 23(18): 3182-92, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12298090

ABSTRACT

A new global protein digestion and selective peptide extraction strategy for the purpose of monitoring differential protein expression, coined as tagless extraction-retentate chromatography, is introduced. Target protein populations are firstly digested under reduced and alkylated conditions, and resultant peptides selectively extracted via covalent attachment to methionine residues by bromoacetyl reactive groups tethered to the surface of glass beads packed in small reaction vessels. After conjugation, reactive beads are stringently washed to remove nonspecifically bound peptides and then later treated with beta-mercaptoethanol to release captured methionine peptides in their nascent state, without complicating affinity tags. Recovered methionine containing peptides are profiled using the surface-enhanced laser desorption/ionization (SELDI) retentate chromatography mass spectrometry (RCMS) method. Selected peptides are further studied employing ProteinChip tandem mass spectrometry (MS/MS) analysis to identify their parent proteins. This approach has been applied to an Escherichia coli lysate model system and has demonstrated facility in reducing global digest complexity, sensitivity to low protein expression levels, and significant quantitative capability. It is envisioned that tagless extraction-RCMS will evolve to be a valuable approach for both basic research and clinical proteomics endeavors.


Subject(s)
Chromatography/methods , Mass Spectrometry/methods , Mass Spectrometry/statistics & numerical data , Protein Array Analysis/methods , Proteins/isolation & purification , Amino Acid Sequence , Animals , Cattle , Chromatography/statistics & numerical data , Escherichia coli Proteins/genetics , Escherichia coli Proteins/isolation & purification , Molecular Sequence Data , Peptide Fragments/genetics , Peptide Fragments/isolation & purification , Protein Array Analysis/statistics & numerical data , Proteomics/methods , Proteomics/statistics & numerical data , Sensitivity and Specificity , Serum Albumin, Bovine/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/statistics & numerical data
12.
Curr Opin Chem Biol ; 6(1): 86-91, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11827829

ABSTRACT

Because of its inherent flexibility, the ProteinChip Array platform has demonstrated utility into basic research as well as clinical research. In the domain of basic research, it has been used to examine protein modifications, characterize protein-protein interactions and study signal transduction and enzymatic pathways. In clinical research, it has been used to elucidate and identify biomarkers of disease, and as a platform for predictive medicine.


Subject(s)
Miniaturization/methods , Proteins/analysis , Animals , Biotechnology/instrumentation , Biotechnology/methods , Humans , Microchemistry , Proteome/analysis , Specimen Handling/methods , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL
...