Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Mol Pharm ; 20(3): 1480-1489, 2023 03 06.
Article in English | MEDLINE | ID: mdl-36702622

ABSTRACT

Monoclonal antibodies (mAbs) are an important and growing class of biotherapeutic drugs. Method development for the characterization of critical quality attributes, including higher-order structure (HOS), of mAbs remains an area of active inquiry. Recently, solution-state nuclear magnetic resonance (NMR) spectroscopy has received increased attention and is a means for reliable, high-resolution HOS characterization of aqueous-based preparations of mAbs. While mAbs are predominantly formulated in solution, up to 20% are prepared as solid amorphous powders and techniques for the robust characterization of HOS in the solid state remain limited. We propose here the use of solid-state NMR (ssNMR) fingerprinting to inform directly on the HOS of solid preparations of mAbs. Using lyophilized samples of the NISTmAb reference material prepared with different formulation conditions, we demonstrate that 1H-13C cross-polarization (hC-CP) buildup spectral series at natural isotopic abundance mAb samples are sensitive to differences in formulation. We also demonstrate that principal component analysis (PCA) can be used to differentiate the samples from one another in a user-independent manner while also highlighting areas where expert analysis can provide structural details about important molecular interactions in solid-phase protein formulations. Results from this study contribute to establishing the foundation for the use of ssNMR for HOS characterization of solid-phase biotherapeutics.


Subject(s)
Antibodies, Monoclonal , Magnetic Resonance Imaging , Antibodies, Monoclonal/chemistry , Magnetic Resonance Spectroscopy/methods
2.
Front Mol Biosci ; 9: 876780, 2022.
Article in English | MEDLINE | ID: mdl-35601836

ABSTRACT

Biopharmaceuticals such as monoclonal antibodies are required to be rigorously characterized using a wide range of analytical methods. Various material properties must be characterized and well controlled to assure that clinically relevant features and critical quality attributes are maintained. A thorough understanding of analytical method performance metrics, particularly emerging methods designed to address measurement gaps, is required to assure methods are appropriate for their intended use in assuring drug safety, stability, and functional activity. To this end, a series of interlaboratory studies have been conducted using NISTmAb, a biopharmaceutical-representative and publicly available monoclonal antibody test material, to report on state-of-the-art method performance, harmonize best practices, and inform on potential gaps in the analytical measurement infrastructure. Reported here is a summary of the study designs, results, and future perspectives revealed from these interlaboratory studies which focused on primary structure, post-translational modifications, and higher order structure measurements currently employed during biopharmaceutical development.

3.
J Pharm Sci ; 110(10): 3385-3394, 2021 10.
Article in English | MEDLINE | ID: mdl-34166704

ABSTRACT

The one-dimensional (1D) diffusion edited proton NMR method, Protein Fingerprint by Lineshape Enhancement (PROFILE) has been demonstrated to be suitable for higher order structure (HOS) characterization of protein therapeutics including monoclonal antibodies. Recent reports in the literature have demonstrated its advantages for HOS characterization over traditional methods such as circular dichroism and Fourier-transform infrared spectroscopy. Previously, we have demonstrated that the PROFILE method is complementary with high resolution 2D methyl correlated NMR methods and how both may be deployed as a multi-modal platform to further the utility of NMR for HOS characterization. A major limitation of the PROFILE method remains its need for high signal to noise data due to its reliance on convolution difference processing and linear correlation metrics to assess spectral similarity. Here we present an alternative method for analyzing 1D diffusion edited spectra, which overcomes this limitation by using nonlinear iterative partial least squares (NIPALS) principal component analysis, and which we dub PROtein Fingerprint Observed Using NIPALS Decomposition (PROFOUND). We demonstrate that results from the PROFOUND method are robust with respect to instrument, operator and in the presence of high experimental noise and how it may be employed to provide quantitative assessment of spectral similarity.


Subject(s)
Antibodies, Monoclonal , Circular Dichroism , Magnetic Resonance Spectroscopy , Principal Component Analysis , Proton Magnetic Resonance Spectroscopy
4.
J Biomol NMR ; 74(10-11): 643-656, 2020 Nov.
Article in English | MEDLINE | ID: mdl-32700053

ABSTRACT

Protein therapeutics have numerous critical quality attributes (CQA) that must be evaluated to ensure safety and efficacy, including the requirement to adopt and retain the correct three-dimensional fold without forming unintended aggregates. Therefore, the ability to monitor protein higher order structure (HOS) can be valuable throughout the lifecycle of a protein therapeutic, from development to manufacture. 2D NMR has been introduced as a robust and precise tool to assess the HOS of a protein biotherapeutic. A common use case is to decide whether two groups of spectra are substantially different, as an indicator of difference in HOS. We demonstrate a quantitative use of principal component analysis (PCA) scores to perform this decision-making, and demonstrate the effect of acquisition and processing details on class separation using samples of NISTmAb monoclonal antibody Reference Material subjected to two different oxidative stress protocols. The work introduces an approach to computing similarity from PCA scores based upon the technique of histogram intersection, a method originally developed for retrieval of images from large databases. Results show that class separation can be robust with respect to random noise, reconstruction method, and analysis region selection. By contrast, details such as baseline distortion can have a pronounced effect, and so must be controlled carefully. Since the classification approach can be performed without the need to identify peaks, results suggest that it is possible to use even more efficient measurement strategies that do not produce spectra that can be analyzed visually, but nevertheless allow useful decision-making that is objective and automated.


Subject(s)
Antibodies, Monoclonal/chemistry , Automation/methods , Nuclear Magnetic Resonance, Biomolecular/methods , Principal Component Analysis/methods , Biological Products , Fourier Analysis , Magnetic Resonance Spectroscopy/methods
5.
Curr Protoc Protein Sci ; 100(1): e105, 2020 06.
Article in English | MEDLINE | ID: mdl-32407007

ABSTRACT

Characterization of the higher-order structure (HOS) of protein therapeutics, and in particular of monoclonal antibodies, by 2D 1 H-13 C methyl correlated NMR has been demonstrated as precise and robust. Such characterization can be greatly enhanced when collections of spectra are analyzed using multivariate approaches such as principal component analysis (PCA), allowing for the detection and identification of small structural differences in drug substance that may otherwise fall below the limit of detection of conventional spectral analysis. A major limitation to this approach is the presence of aliphatic signals from formulation or excipient components, which result in spectral interference with the protein signal of interest; however, the recently described Selective Excipient Reduction and Removal (SIERRA) filter greatly reduces this issue. Here we will outline how basic 2D 1 H-13 C methyl-correlated NMR may be combined with the SIERRA approach to collect 'clean' NMR spectra of formulated monoclonal antibody therapeutics (i.e., drug substance spectra free of interfering component signals), and how series of such spectra may be used for HOS characterization by direct PCA of the series spectral matrix. © 2020 U.S. Government. Basic Protocol 1: NMR data acquisition Basic Protocol 2: Full spectral matrix data processing and analysis Support Protocol: Data visualization and cluster analysis.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Nuclear Magnetic Resonance, Biomolecular , Antibodies, Monoclonal, Murine-Derived/analysis , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Humans , Principal Component Analysis
6.
Anal Chem ; 92(9): 6366-6373, 2020 05 05.
Article in English | MEDLINE | ID: mdl-32267681

ABSTRACT

The use of NMR spectroscopy has emerged as a premier tool to characterize the higher order structure of protein therapeutics and in particular IgG1 monoclonal antibodies (mAbs). Due to their large size, traditional 1H-15N correlation experiments have proven exceedingly difficult to implement on mAbs, and a number of alternative techniques have been proposed, including the one-dimensional (1D) 1H protein fingerprint by line shape enhancement (PROFILE) method and the two-dimensional (2D) 1H-13C methyl correlation-based approach. Both 1D and 2D approaches have relative strengths and weaknesses, related to the inherent sensitivity and resolution of the respective methods. To further increase the utility of NMR to the biopharmaceutical community, harmonized criteria for decision making in employing 1D and 2D approaches for mAb characterization are warranted. To this end, we have conducted an interlaboratory comparative study of the 1D PROFILE and 2D methyl methods on several mAbs samples to determine the degree to which each method is suited to detect spectral difference between the samples and the degree to which results from each correlate with one another. Results from the study demonstrate both methods provide statistical data highly comparable to one another and that each method is capable of complementing the limitations commonly associated with the other, thus providing a better overall picture of higher order structure.


Subject(s)
Immunoglobulin G/analysis , Nuclear Magnetic Resonance, Biomolecular , Carbon Isotopes , Protons
7.
J Chem Inf Model ; 60(4): 2339-2355, 2020 04 27.
Article in English | MEDLINE | ID: mdl-32249579

ABSTRACT

Quality attributes (QAs) are measureable parameters of a biologic that impact product safety and efficacy and are essential characteristics that are linked to positive patient health outcomes. One QA, higher order structure (HOS), is directly coupled to the function of protein biologics, and deviations in this QA may cause adverse effects. To address the critical need for HOS assessment, methods for analyzing structural fingerprints from 2D nuclear magnetic resonance spectroscopy (2D-NMR) spectra have been established for drug substances as large as monoclonal antibody therapeutics. Here, chemometric analyses have been applied to 2D 1H,13C-methyl NMR correlation spectra of the IgG1κ NIST monoclonal antibody (NISTmAb), recorded at natural isotopic abundance, to benchmark the performance and robustness of the methods. In particular, a variety of possible spectral input schemes (e.g., chemical shift, peak intensity, and total spectral matrix) into chemometric algorithms are examined using two case studies: (1) a large global 2D-NMR interlaboratory study and (2) a blended series of enzymatically glycan-remodeled NISTmAb isoforms. These case studies demonstrate that the performance of chemometric algorithms using either peak positions or total spectral matrix as the input will depend on the study design and likely be product-specific. In general, peak positions are found to be a more robust spectral parameter for input into chemometric algorithms, whereas the total spectral matrix approach lends itself to easier automation and requires less user intervention. Analysis with different input data also shows differences in sensitivity to certain changes in HOS, highlighting that product knowledge will further guide appropriate method selection based on the fit-for-purpose application in the context of biopharmaceutical development, production, and quality control.


Subject(s)
Biological Products , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Algorithms , Antibodies, Monoclonal , Humans
8.
Article in English | MEDLINE | ID: mdl-34135539

ABSTRACT

Protein therapeutics are vitally important clinically and commercially, with monoclonal antibody (mAb) therapeutic sales alone accounting for $115 billion in revenue for 2018.[1] In order for these therapeutics to be safe and efficacious, their protein components must maintain their high order structure (HOS), which includes retaining their three-dimensional fold and not forming aggregates. As demonstrated in the recent NISTmAb Interlaboratory nuclear magnetic resonance (NMR) Study[2], NMR spectroscopy is a robust and precise approach to address this HOS measurement need. Using the NISTmAb study data, we benchmark a procedure for automated outlier detection used to identify spectra that are not of sufficient quality for further automated analysis. When applied to a diverse collection of all 252 1H,13C gHSQC spectra from the study, a recursive version of the automated procedure performed comparably to visual analysis, and identified three outlier cases that were missed by the human analyst. In total, this method represents a distinct advance in chemometric detection of outliers due to variation in both measurement and sample.

9.
J Magn Reson ; 308: 106588, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31561023

ABSTRACT

A pulsed element is proposed allowing the selective inversion of a single 1H nucleus, without regard to the presence of other degenerate 1H nuclei, provided that it is coupled to a heteronuclear spin with adequate chemical shift resolution in a 2D heteronuclear correlation spectrum. The approach is based on selective cross polarization, in which matched weak RF fields of specific amplitude are applied on resonance to the targeted 1H-X spin pair. It is shown theoretically that when the RF field amplitudes are set to specific values (ie. at a magic field), transfer of coherence can be fruitfully achieved transverse to the applied RF fields in addition to the normal longitudinal transfers. This enables the construction of a pulsed element which has the characteristics of a BIRDr,X element (Uhrin et al., 1993), yet with 2D frequency selectivity. Demonstration of the pulsed element is made in the context of selective spin inversion, by which all 1H spins are inverted except the targeted 1H spin.

10.
MAbs ; 11(1): 94-105, 2019 01.
Article in English | MEDLINE | ID: mdl-30570405

ABSTRACT

The increased interest in using monoclonal antibodies (mAbs) as a platform for biopharmaceuticals has led to the need for new analytical techniques that can precisely assess physicochemical properties of these large and very complex drugs for the purpose of correctly identifying quality attributes (QA). One QA, higher order structure (HOS), is unique to biopharmaceuticals and essential for establishing consistency in biopharmaceutical manufacturing, detecting process-related variations from manufacturing changes and establishing comparability between biologic products. To address this measurement challenge, two-dimensional nuclear magnetic resonance spectroscopy (2D-NMR) methods were introduced that allow for the precise atomic-level comparison of the HOS between two proteins, including mAbs. Here, an inter-laboratory comparison involving 26 industrial, government and academic laboratories worldwide was performed as a benchmark using the NISTmAb, from the National Institute of Standards and Technology (NIST), to facilitate the translation of the 2D-NMR method into routine use for biopharmaceutical product development. Two-dimensional 1H,15N and 1H,13C NMR spectra were acquired with harmonized experimental protocols on the unlabeled Fab domain and a uniformly enriched-15N, 20%-13C-enriched system suitability sample derived from the NISTmAb. Chemometric analyses from over 400 spectral maps acquired on 39 different NMR spectrometers ranging from 500 MHz to 900 MHz demonstrate spectral fingerprints that are fit-for-purpose for the assessment of HOS. The 2D-NMR method is shown to provide the measurement reliability needed to move the technique from an emerging technology to a harmonized, routine measurement that can be generally applied with great confidence to high precision assessments of the HOS of mAb-based biotherapeutics.


Subject(s)
Antibodies, Monoclonal/chemistry , Biopharmaceutics/standards , Laboratories/standards , Magnetic Resonance Spectroscopy/methods , Humans , Reproducibility of Results
11.
J Biomol NMR ; 72(3-4): 149-161, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30483914

ABSTRACT

While the use of 1H-13C methyl correlated NMR spectroscopy at natural isotopic abundance has been demonstrated as feasible on protein therapeutics as large as monoclonal antibodies, spectral interference from aliphatic excipients remains a significant obstacle to its widespread application. These signals can cause large baseline artifacts, obscure protein resonances, and cause dynamic range suppression of weak peaks in non-uniform sampling applications, thus hampering both traditional peak-based spectral analyses as well as emerging chemometric methods of analysis. Here we detail modifications to the 2D 1H-13C gradient-selected HSQC experiment that make use of selective pulsing techniques for targeted removal of interfering excipient signals in spectra of the NISTmAb prepared in several different formulations. This approach is demonstrated to selectively reduce interfering excipient signals while still yielding 2D spectra with only modest losses in protein signal. Furthermore, it is shown that spectral modeling based on the SMILE algorithm can be used to simulate and subtract any residual excipient signals and their attendant artifacts from the resulting 2D NMR spectra.


Subject(s)
Biological Products/chemistry , Excipients/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Algorithms , Carbon Isotopes , Methylation , Proteins/chemistry , Proteins/therapeutic use , Protons
12.
MAbs ; 10(6): 922-933, 2018.
Article in English | MEDLINE | ID: mdl-29958062

ABSTRACT

The successful development and regulatory approval of originator and biosimilar therapeutic proteins requires a systems approach to upstream and downstream processing as well as product characterization and quality control. Innovation in process design and control, product characterization strategies, and data integration represent an ecosystem whose concerted advancement may reduce time-to-market and further improve comparability and biosimilarity programs. The biopharmaceutical community has made great strides to this end, yet there currently exists no pre-competitive monoclonal antibody (mAb) expression platform for open innovation. Here, we describe the development and initial expression of an intended copy of the NISTmAb using three non-originator murine cell lines. It was found that, without optimization and in culture flasks, all three cell lines produce approximately 100 mg mAb per liter of culture. Sodium dodecyl sulfate polyacrylamide gel electrophoresis, size-exclusion chromatography, nuclear magnetic resonance spectroscopy, intact mass spectrometry, and surface plasmon resonance were used to demonstrate that the products of all three cell lines embody quality attributes with a sufficient degree of sameness to the NISTmAb Reference Material 8671 to warrant further bioreactor studies, process improvements and optimization. The implications of the work with regard to pre-competitive innovation to support process design and feedback control, comparability and biosimilarity assessments, and process analytical technologies are discussed.


Subject(s)
Antibodies, Monoclonal/chemistry , Biosimilar Pharmaceuticals/chemistry , Immunoglobulin G/chemistry , Recombinant Proteins/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , Biosimilar Pharmaceuticals/standards , Cell Line , Chromatography, Liquid , Electrophoresis, Polyacrylamide Gel , Humans , Magnetic Resonance Spectroscopy , Quality Control , Recombinant Proteins/standards , Reference Standards , Tandem Mass Spectrometry
13.
Anal Chem ; 89(21): 11839-11845, 2017 11 07.
Article in English | MEDLINE | ID: mdl-28937210

ABSTRACT

Two-dimensional (2D) 1H-13C methyl NMR provides a powerful tool to probe the higher order structure (HOS) of monoclonal antibodies (mAbs), since spectra can readily be acquired on intact mAbs at natural isotopic abundance, and small changes in chemical environment and structure give rise to observable changes in corresponding spectra, which can be interpreted at atomic resolution. This makes it possible to apply 2D NMR spectral fingerprinting approaches directly to drug products in order to systematically characterize structure and excipient effects. Systematic collections of NMR spectra are often analyzed in terms of the changes in specifically identified peak positions, as well as changes in peak height and line widths. A complementary approach is to apply principal component analysis (PCA) directly to the matrix of spectral data, correlating spectra according to similarities and differences in their overall shapes, rather than according to parameters of individually identified peaks. This is particularly well-suited for spectra of mAbs, where some of the individual peaks might not be well resolved. Here we demonstrate the performance of the PCA method for discriminating structural variation among systematic sets of 2D NMR fingerprint spectra using the NISTmAb and illustrate how spectral variability identified by PCA may be correlated to structure.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Carbon-13 Magnetic Resonance Spectroscopy , Glycosylation , Multivariate Analysis
15.
Methods Enzymol ; 566: 3-34, 2016.
Article in English | MEDLINE | ID: mdl-26791974

ABSTRACT

Methods for characterizing the higher-order structure of protein therapeutics are in great demand for establishing consistency in drug manufacturing, for detecting drug product variations resulting from modifications in the manufacturing process, and for comparing a biosimilar to an innovator reference product. In principle, solution NMR can provide a robust approach for characterization of the conformation(s) of protein therapeutics in formulation at atomic resolution. However, molecular weight limitations and the perceived need for stable isotope labeling have to date limited its practical applications in the biopharmaceutical industry. Advances in NMR magnet and console technologies, cryogenically cooled probes, and new rapid acquisition methodologies, particularly selective optimized flip-angle short transient pulse schemes and nonuniform sampling, have greatly ameliorated these limitations. Here, we describe experimental methods for the collection and analysis of 2D (1)H(N)-(15)N-amide- and (1)H-(13)C-methyl-correlated spectra applied to protein drug products at natural isotopic abundance, including representatives from the rapidly growing class of monoclonal antibody (mAb) therapeutics. Practical aspects of experimental setup and data acquisition for both standard and rapid acquisition NMR techniques are described. Furthermore, strategies for the statistical comparison of 2D (1)H(N)-(15)N-amide- and (1)H-(13)C-methyl-correlated spectra are detailed.


Subject(s)
Chemistry, Pharmaceutical/methods , Nuclear Magnetic Resonance, Biomolecular/methods , Proteins/chemistry , Proton Magnetic Resonance Spectroscopy/methods , Antibodies, Monoclonal , Carbon Radioisotopes/chemistry , Humans , Magnetic Resonance Spectroscopy , Nitrogen Radioisotopes/chemistry , Protein Conformation
16.
Pharm Res ; 33(2): 462-75, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26453189

ABSTRACT

PURPOSE: High-resolution nuclear magnetic resonance spectroscopy (NMR) provides a robust approach for producing unique spectral signatures of protein higher order structure at atomic resolution. Such signatures can be used as a tool to establish consistency of protein folding for the assessment of monoclonal antibody (mAb) drug quality and comparability. METHODS: Using the NIST monoclonal antibody (NISTmAb) and a commercial-sourced polyclonal antibody, both IgG1κ isotype, we apply 2D NMR methods at natural abundance for the acquisition and unbiased statistical analysis of (1)H(N) -(15)N correlated spectra of intact antibody (Ab) and protease-cleaved Fab and Fc fragments. RESULTS: The study demonstrates the feasibility of applying 2D NMR techniques to Abs and the precision with which these methods can be used to map structure and establish comparability between samples at atomic resolution. CONCLUSIONS: The statistical analyses suggests that, within the limit of detection, no significant structural differences are observed between the Fab and Fc domains of each respective intact Ab and its corresponding fragments. Discrimination between dissimilar species, such as between the Fab domains of both Abs or between the glycosylated and deglycosylated Fc domains, was further demonstrated. As such, these methods should find general utility for the assessment of mAb higher order structure.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular/methods , Antibodies, Monoclonal/chemistry , Glycosylation , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin G/chemistry , Protein Conformation
17.
Anal Chem ; 87(7): 3556-61, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25728213

ABSTRACT

Monoclonal antibodies (mAbs) represent an important and rapidly growing class of biotherapeutics. Correct folding of a mAb is critical for drug efficacy, while misfolding can impact safety by eliciting unwanted immune or other off-target responses. Robust methods are therefore needed for the precise measurement of mAb structure for drug quality assessment and comparability. To date, the perception in the field has been that NMR could not be applied practically to mAbs due to the size (∼150 kDa) and complexity of these molecules, as well as the insensitivity of the method. The feasibility of applying NMR methods to stable isotope-labeled, protease-cleaved, mAb domains (Fab and Fc) has been demonstrated from both E. coli and Chinese hamster ovaries (CHO) cell expression platforms; however, isotopic labeling is not typically available when analyzing drug products. Here, we address the issue of feasibility of NMR-based mapping of mAb structure by demonstrating for the first time the application of a 2D (13)C NMR methyl fingerprint method for structural mapping of an intact mAb at natural isotopic abundance. Further, we show that 2D (13)C NMR spectra of protease-cleaved Fc and Fab fragments can provide accurate reporters on the domain structures that can be mapped directly to the intact mAb. Through combined use of rapid acquisition and nonuniform sampling techniques, we show that these Fab and Fc fingerprint spectra can be rapidly acquired in as short as approximately 30 min.


Subject(s)
Antibodies, Monoclonal/chemistry , Carbon-13 Magnetic Resonance Spectroscopy , Immunoglobulin Fab Fragments/chemistry , Immunoglobulin Fc Fragments/chemistry , Nuclear Magnetic Resonance, Biomolecular , Animals , CHO Cells , Cricetinae , Cricetulus , Models, Molecular , Protein Conformation
18.
J Magn Reson ; 235: 26-31, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23917309

ABSTRACT

Long-range residual dipolar couplings (lrRDCs) have the potential to serve as powerful structural restraints in protein NMR spectroscopy as they can provide both distance and orientation information about nuclei separate in sequence but close in space. Current nonselective methods for their measurement are limited to moderate alignment strengths due to the sheer abundance of active couplings at stronger alignment. This limits the overall magnitude and therefore distance across which couplings can be measured. We have developed a double resonance technique for the inversion of individual coupled spin pairs, called Selective Inversion by Single Transition Cross Polarization (SIST-CP). This technique enables the selective recoupling of lrRDCs, thus allowing the complex multiplets occurring in strongly aligned systems to be disentangled. This technique is demonstrated in the context of an application to the measurement of (13)C'-(1)H(N) lrRDCs in strongly aligned proteins.


Subject(s)
Nuclear Magnetic Resonance, Biomolecular/methods , Proteins/chemistry , Algorithms , Carbon/chemistry , Carbon Isotopes , Indicators and Reagents , Nuclear Magnetic Resonance, Biomolecular/instrumentation , Protein Conformation , Protons , Ubiquitins/chemistry
19.
J Biomol NMR ; 46(2): 175-89, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20012159

ABSTRACT

A pair of 3D HNCO-based experiments have been developed with the aim of optimizing the precision of measurement of (1)J(NH) couplings. Both pulse sequences record (1)J(NH) coupling evolution during the entire constant time interval that (15)N magnetization is dephasing or rephasing with respect to the directly bonded (13)C' nucleus, with (15)N(13)C' multiple quantum coherence maintained during the (13)C' evolution period. The first experiment, designed for smaller proteins, produces an apparent doubling of the (1)J(NH) coupling without any accompanying increases in line width. The second experiment is a J-scaled TROSY-HNCO experiment in which the (1)J(NH) coupling is measured by frequency difference between resonances offset symmetrically about the position of the downfield component of the (15)N doublet (i.e. the TROSY resonance). This experiment delivers significant gains in precision of (1)J(NH) coupling measurement compared to existing J-scaled TROSY-HNCO experiments. With the proper choice of acquisition parameters and sufficient sensitivity to acquire a 3D TROSY-HNCO experiment, it is shown that (1)J(NH) couplings can be measured with a precision which approaches or exceeds the precision of measurement with which the frequency of the TROSY resonance itself can be determined.


Subject(s)
Amides/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Heme Oxygenase (Decyclizing)/chemistry , Neisseria meningitidis/enzymology , Nerve Tissue Proteins/chemistry , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...